1
|
Ma H, Srivastava S, Ho SWT, Xu C, Lian BSX, Ong X, Tay ST, Sheng T, Lum HYJ, Abdul Ghani SAB, Chu Y, Huang KK, Goh YT, Lee M, Hagihara T, Ng CSY, Tan ALK, Zhang Y, Ding Z, Zhu F, Ng MSW, Joseph CRC, Chen H, Li Z, Zhao JJ, Rha SY, Teh M, Yeong J, Yong WP, So JBY, Sundar R, Tan P. Spatially Resolved Tumor Ecosystems and Cell States in Gastric Adenocarcinoma Progression and Evolution. Cancer Discov 2025; 15:767-792. [PMID: 39774838 PMCID: PMC11962405 DOI: 10.1158/2159-8290.cd-24-0605] [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: 05/03/2024] [Revised: 10/17/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025]
Abstract
SIGNIFICANCE Integration of spatial transcriptomic (GeoMx Digital Spatial Profiler) and single-cell RNA sequencing data from multiple gastric cancers identifies spatially resolved expression-based intratumoral heterogeneity, associated with distinct immune microenvironments. We uncovered two separate evolutionary trajectories associated with specific molecular subtypes, clinical prognoses, stromal neighborhoods, and genetic drivers. Tumor-stroma interfaces emerged as a unique state of tumor ecology.
Collapse
Affiliation(s)
- Haoran Ma
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Supriya Srivastava
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shamaine Wei Ting Ho
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Chang Xu
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | | | - Xuewen Ong
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Su Ting Tay
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Taotao Sheng
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | | | | | - Yunqiang Chu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Kie Kyon Huang
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Yeek Teck Goh
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Minghui Lee
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Takeshi Hagihara
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Clara Shi Ya Ng
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Angie Lay Keng Tan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Yanrong Zhang
- Department of Information Systems and Analytics, School of Computing, National University of Singapore, Singapore, Singapore
| | - Zichen Ding
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Zhu
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Michelle Shu Wen Ng
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Craig Ryan Cecil Joseph
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Hui Chen
- MGI Tech Singapore Pte. Ltd., Singapore, Singapore
| | - Zhen Li
- MGI Tech Singapore Pte. Ltd., Singapore, Singapore
| | - Joseph J. Zhao
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Sun Young Rha
- Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ming Teh
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Joe Yeong
- Department of Pathology, National University Hospital, Singapore, Singapore
- Bioinformatics Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Wei Peng Yong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
| | - Jimmy Bok-Yan So
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
- Department of Surgery, University Surgical Cluster, National University Health System, Singapore, Singapore
- Division of Surgical Oncology, National University Cancer Institute, Singapore, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Raghav Sundar
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore
| | - Patrick Tan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cellular and Molecular Research, National Cancer Centre, Singapore, Singapore
- Singhealth/Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore, Singapore
| |
Collapse
|
2
|
Zhong Y, Zhang W, Zheng C, Wu H, Luo J, Yuan Z, Zhang H, Wang C, Feng H, Wang M, Zhang Q, Ju H, Wang G. Multi-omic analyses reveal PTPN6's impact on tumor immunity across various cancers. Sci Rep 2025; 15:11025. [PMID: 40164665 PMCID: PMC11958644 DOI: 10.1038/s41598-025-96302-1] [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: 01/02/2024] [Accepted: 03/27/2025] [Indexed: 04/02/2025] Open
Abstract
Protein Tyrosine Phosphatase Non-Receptor Type 6 (PTPN6) plays a crucial regulatory role in cellular processes and has been implicated in oncogenesis. This pan-cancer analysis aimed to elucidate PTPN6's involvement across various cancer types, with a particular emphasis on its association with tumor immunity. We analyzed PTPN6 expression data from open access databases using various statistical techniques, including survival analysis, genetic heterogeneity analysis, immune profiling, single-cell analysis, drug sensitivity analysis, and protein interaction analysis. We also conducted in vitro experiments utilizing colorectal cancer cell lines to validate PTPN6's functional role. PTPN6 exhibited distinct expression patterns across cancers, and its prognostic significance was apparent in several cancer types, particularly in glioblastoma, sarcoma, and melanoma. We observed correlations between PTPN6 and immune genes/cell infiltration in these cancers, suggesting a potential role in modulating the tumor immune microenvironment. Single-cell analysis revealed that PTPN6 is predominantly localized in macrophages, B cells, and dendritic cells within the tumor microenvironment, implying its involvement in regulating immune cell function. Enrichment analysis highlighted PTPN6's role in immune-related pathways. Drug sensitivity analysis identified specific drugs, including PAC-1, SNX-2112, BELINOSTAT, VORINOSTAT, TPCA-1, and PHA-893,888, whose efficacy may be influenced by PTPN6 expression. Knocking down PTPN6 expression inhibited the proliferation and migration of colorectal cancer cells in vitro, confirming its oncogenic role in this cancer type. This pan-cancer analysis establishes PTPN6's multifaceted influence on tumor immunity and its potential as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Yuchen Zhong
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China
| | - Weiyuan Zhang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Chaojing Zheng
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Hongyu Wu
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Jun Luo
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Ziming Yuan
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Hao Zhang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Chunlin Wang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Haiyang Feng
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Meng Wang
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Qian Zhang
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China.
| | - Haixing Ju
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China.
| | - Guiyu Wang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China.
| |
Collapse
|
3
|
Yu Q, Li YY, Chen Y. scMalignantFinder distinguishes malignant cells in single-cell and spatial transcriptomics by leveraging cancer signatures. Commun Biol 2025; 8:504. [PMID: 40148533 PMCID: PMC11950360 DOI: 10.1038/s42003-025-07942-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) is a powerful tool for characterizing tumor heterogeneity, yet accurately identifying malignant cells remains challenging. Here, we propose scMalignantFinder, a machine learning tool specifically designed to distinguish malignant cells from their normal counterparts using a data- and knowledge-driven strategy. To develop the tool, multiple cancer datasets were collected, and the initially annotated malignant cells were calibrated using nine carefully curated pan-cancer gene signatures, resulting in over 400,000 single-cell transcriptomes for training. The union of differentially expressed genes across datasets was taken as the features for model construction to comprehensively capture tumor transcriptional diversity. scMalignantFinder outperformed existing automated methods across two gold-standard and eleven patient-derived scRNA-seq datasets. The capability to predict malignancy probability empowers scMalignantFinder to capture dynamic characteristics during tumor progression. Furthermore, scMalignantFinder holds the potential to annotate malignant regions in tumor spatial transcriptomics. Overall, we provide an efficient tool for detecting heterogeneous malignant cell populations.
Collapse
Affiliation(s)
- Qiaoni Yu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
- Shanghai Genbase Biotechnology Co., Ltd, Shanghai, China
| | - Yuan-Yuan Li
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China.
| | - Yunqin Chen
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China.
- Shanghai Genbase Biotechnology Co., Ltd, Shanghai, China.
| |
Collapse
|
4
|
Azadegan C, Santoro J, Whetstine JR. CONNECTING THE DOTS: EPIGENETIC REGULATION OF EXTRACHROMOSOMAL AND INHERITED DNA AMPLIFICATIONS. J Biol Chem 2025:108454. [PMID: 40154613 DOI: 10.1016/j.jbc.2025.108454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/20/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025] Open
Abstract
DNA amplification has intrigued scientists for decades. Since its discovery, significant progress has been made in understanding the mechanisms promoting DNA amplification and their associated function(s). While DNA copy gains were once thought to be regulated purely by stochastic processes, recent findings have revealed the important role of epigenetic modifications in driving these amplifications and their integration into the genome. Furthermore, advances in genomic technology have enabled detailed characterization of these genomic events in terms of size, structure, formation, and regulation. This review highlights how our understanding of DNA amplifications has evolved over time, tracing its trajectory from initial discovery to the contemporary landscape. We describe how recent discoveries have started to uncover how these genomic events occur by controlled biological processes rather than stochastic mechanisms, presenting opportunities for therapeutic modulation.
Collapse
Affiliation(s)
- Chloe Azadegan
- Drexel University, College of Medicine, Philadelphia, PA, 19111; Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia PA, 19111
| | - John Santoro
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia PA, 19111
| | - Johnathan R Whetstine
- Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia PA, 19111.
| |
Collapse
|
5
|
Chen F, Wen X, Li S, Wu J, Luo Y, Gao Y, Yu X, Chen L. Targeting hypoxia-mediated chemo-immuno resistance by a hybrid NBDHEX-Pt(IV) prodrug via declining nuclear STING1-promoted AhR-CIN in human lung squamous cell carcinoma. Transl Oncol 2025; 55:102350. [PMID: 40138855 DOI: 10.1016/j.tranon.2025.102350] [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: 08/12/2024] [Revised: 02/19/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
As found in human lung squamous cell carcinoma (LUSC), STING1 involved in ER-Golgi intermediate compartment (ERGIC) could coordinate immune responses to ectopic DNA triggered by DNA-targeted chemotherapy. ERGIC STING1 is considered to compete with nuclear STING1 to decline aryl hydrocarbon receptor (AhR)-chromosomal instability (CIN)-triggered chronic STING activation which could cause therapeutic resistance. Moreover, GSTP1 was proved to inhibit ERGIC-STING1 via promoting S-glutathione modification of STING1. Hence, a potent GSTP1-targeted Pt(IV) hybrid NBDHEX-DN604, was designed via conjugating a GSTP1 inhibitor NBDHEX to the axial position of Pt(IV) prodrug. As mentioned, hypoxia is mainly observed in malignant tumors and develops acquired drug resistance. In vitro bio-properties of hypoxic SK-MES-1/cDDP cells demonstrated that NBDHEX-DN604 could reverse chemo-immuno resistance via intercepting GSTP1 to activate ERGIC STING1, leading to the decrease of nuclear STING1. The mechanistic data indicated that NBDHEX-DN604 could elevate ERGIC STING1 to mitigate nuclear STING1-mediated AhR-TLS-CIN-chronic activation. Meanwhile, NBDHEX-DN604 was found to decline STING1-AhR-CIN to circumvent chemo-immuno resistance, resulting in predominant in vivo antitumor effect in HY-KLN-205/cDDP-inoculated BALB/c mice. The data provide a novel rationale for the mixed chemo-immunotherapy of NBDHEX-DN604 as a potent Pt(IV) therapeutic method for patients with resistant LUSC.
Collapse
Affiliation(s)
- Feihong Chen
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China.
| | - Xin Wen
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Shan Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Jiani Wu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yaxuan Luo
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yuan Gao
- Senior Department of Obstetrics & Gynecology, the Seventh Medical Center of PLA General Hospital, Beijing 100700, China.
| | - Xiaoxuan Yu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, PR China.
| | - Li Chen
- Suzhou Institute for Drug Control, Suzhou 215104, PR China.
| |
Collapse
|
6
|
Haykal MM, Rodrigues-Ferreira S, Botty RE, Sourd L, Marangoni E, Varin M, Denis A, Nahmias C. Targeting WEE1 kinase as a therapeutic strategy in ATIP3-deficient breast cancers. Cancer Lett 2025:217665. [PMID: 40127815 DOI: 10.1016/j.canlet.2025.217665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
ATIP3-deficient breast cancers represent a subset of aggressive tumors with limited therapeutic options and poor prognosis. Here, we screened a panel of cell cycle kinase inhibitors to identify novel targets for these tumors. We show that loss of ATIP3 sensitizes breast cancer cells to WEE1 inhibition, resulting in aberrant mitoses characterized by detachment of centromere proteins from DNA and chromosome pulverization. This phenotype arises from excessive replication stress and DNA damage in S-phase, combined with premature mitotic entry driven by untimely CDK1 activation. Mechanistically, we identify DNA2 helicase/nuclease as a key mediator of chromosome pulverization. Importantly, the heightened sensitivity of ATIP3-deficient cells to WEE1 inhibition provides a strong rationale for clinical exploration of WEE1-targeted therapies. Furthermore, combining WEE1 and PKMYT1 inhibitors enhances therapeutic efficacy, offering a promising strategy for personalized treatment in ATIP3-deficient breast cancers.
Collapse
Affiliation(s)
- Maria M Haykal
- Institut Gustave Roussy, Inserm U981, Biomarqueurs prédictifs et nouvelles stratégies thérapeutiques en oncologie, 94800 Villejuif, France; Université Paris-Saclay, 91400 Orsay, France
| | - Sylvie Rodrigues-Ferreira
- Institut Gustave Roussy, Inserm U981, Biomarqueurs prédictifs et nouvelles stratégies thérapeutiques en oncologie, 94800 Villejuif, France; Université Paris-Saclay, 91400 Orsay, France; Inovarion, 75005 Paris, France
| | - Rania El Botty
- Translational Research Department, Institut Curie, PSL University, 75005 Paris, France
| | - Laura Sourd
- Translational Research Department, Institut Curie, PSL University, 75005 Paris, France
| | - Elisabetta Marangoni
- Translational Research Department, Institut Curie, PSL University, 75005 Paris, France
| | | | | | - Clara Nahmias
- Institut Gustave Roussy, Inserm U981, Biomarqueurs prédictifs et nouvelles stratégies thérapeutiques en oncologie, 94800 Villejuif, France; Université Paris-Saclay, 91400 Orsay, France.
| |
Collapse
|
7
|
Souza VGP, Benard KH, Stewart GL, Enfield KSS, Lam WL. Identification of Genomic Instability-Associated LncRNAs as Potential Therapeutic Targets in Lung Adenocarcinoma. Cancers (Basel) 2025; 17:996. [PMID: 40149330 PMCID: PMC11940503 DOI: 10.3390/cancers17060996] [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: 02/12/2025] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Non-small cell lung cancer (NSCLC) is the most common type of cancer, with lung adenocarcinoma (LUAD) as the predominant subtype. Despite advancements in targeted therapies, many NSCLC patients still experience poor outcomes due to treatment resistance and disease progression. Genomic instability (GI), a hallmark of cancer, defined as the increased tendency of DNA mutations and alterations, is closely linked to cancer initiation, progression, and resistance to therapy. Emerging evidence suggests that long non-coding RNAs (lncRNAs)-molecules longer than 200 nucleotides that do not encode proteins but regulate gene expression-play critical roles in cancer biology and are associated with GI. However, the relationship between GI and lncRNA expression in LUAD remains poorly understood. METHODS In this study, we analyzed the transcript profiles of lncRNAs and mRNAs from LUAD samples in The Cancer Genome Atlas (TCGA) database and classified them based on their Homologous Recombination Deficiency (HRD) score. The HRD score is an unweighted sum of three independent DNA-based measures of genomic instability: loss of heterozygosity, telomeric allelic imbalance, and large-scale transitions. We then performed a differential gene expression analysis to identify lncRNAs and mRNAs that were either upregulated or downregulated in samples with high HRD scores compared to those with low HRD scores. Following this, we conducted a correlation analysis to assess the significance of the association between HRD scores and the expression of both lncRNAs and mRNAs. RESULTS We identified 30 differentially expressed lncRNAs and 200 mRNAs associated with genomic instability. Using an RNA interactome database from sequencing experiments, we found evidence of interactions between GI-associated lncRNAs (GI-lncRNAs) and GI-associated mRNAs (GI-mRNAs). Further investigation showed that some GI-lncRNAs play regulatory and functional roles in LUAD and other diseases. We also found that GI-lncRNAs have potential as prognostic biomarkers, particularly when integrated with HRD stratification. The expression of specific GI-lncRNAs was associated with primary therapy response and immune infiltration in LUAD. Additionally, we identified existing drugs that could modulate GI-lncRNAs, offering potential therapeutic strategies to address GI in LUAD. CONCLUSIONS Our findings suggest that GI-associated lncRNAs could serve as valuable biomarkers for LUAD prognosis and therapeutic response. Furthermore, modulating these lncRNAs presents potential treatment avenues to address genomic instability in LUAD.
Collapse
Affiliation(s)
- Vanessa G. P. Souza
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada (W.L.L.)
| | - Katya H. Benard
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada (W.L.L.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Greg L. Stewart
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada (W.L.L.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Katey S. S. Enfield
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada (W.L.L.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| | - Wan L. Lam
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada (W.L.L.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| |
Collapse
|
8
|
Pilard C, Roncarati P, Ancion M, Luyckx M, Renard M, Reynders C, Lerho T, Poulain F, Bruyere D, Lebeau A, Hendrick E, Crake R, Peiffer R, Nokin MJ, Peulen O, Delvenne P, Hubert P, Herfs M. RANKL blockade inhibits cancer growth through reversing the tolerogenic profile of tumor-infiltrating (plasmacytoid) dendritic cells. J Immunother Cancer 2025; 13:e010753. [PMID: 40081943 PMCID: PMC11907081 DOI: 10.1136/jitc-2024-010753] [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: 10/08/2024] [Accepted: 02/26/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Originally identified for its involvement in bone remodeling, accumulating data emerged in the past years indicating that receptor activator of nuclear factor κB ligand (RANKL) actually acts as a multifunctional soluble molecule that influences various physiological and pathological processes. Regarding its role in carcinogenesis, while direct effects on tumor cell behavior have been precisely characterized, the impact of the RANKL/RANK system (and its inhibition) on the intratumoral immune landscape remains unclear. METHODS After various in silico/in situ/in vitro analyses, the immunotherapeutic efficacy of RANKL blockade (alone and in combination with immune checkpoint inhibitors (anti-programmed cell death protein-1 (PD-1)) or doxorubicin/paclitaxel-based chemotherapy) was investigated using different syngeneic mouse models of triple-negative breast cancer (4T1, 67NR and E0771). Isolated from retrieved tumors, 14 immune cell (sub)populations, along with the activation status of antigen-presenting cells, were thoroughly analyzed in each condition. Finally, the impact of RANKL on the functionality of both dendritic cells (DC) and plasmacytoid dendritic cells (pDC) was determined. RESULTS A drastic tumor growth inhibition was reproductively observed following RANKL inhibition. Strikingly, this antitumor activity was not detected in immunocompromised mice, demonstrating its dependence on the adaptive immune responses and justifying the diverse enriched signatures linked to immune cell regulation/differentiation detected in RANKLhigh-expressing human neoplasms. Interestingly, neoadjuvant chemotherapy (but not PD-1 checkpoint inhibition) potentiated the anticancer effects of RANKL blockade by priming effector T cells and increasing their infiltration within the tumor microenvironment. Mechanistically, we highlighted that RANKL indirectly promotes regulatory T cell differentiation and suppressive function by inhibiting the mTOR signaling pathway on antigen-presenting cells. CONCLUSIONS Taken together, this study provides insight into the role of RANKL/RANK axis in immune tolerance, demonstrates the significant impact of RANKL-dependent impairment of T cell-DC/pDC crosstalk on tumor development and, ultimately, supports that this ligand could be an interesting actionable target for cancer immunotherapy.
Collapse
Affiliation(s)
- Charlotte Pilard
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Patrick Roncarati
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Marie Ancion
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Margaux Luyckx
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Michael Renard
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Celia Reynders
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Thomas Lerho
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Florian Poulain
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Diane Bruyere
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Alizee Lebeau
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Elodie Hendrick
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Rebekah Crake
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Raphael Peiffer
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Marie-Julie Nokin
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Philippe Delvenne
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
- Department of Pathology, University Hospital Center of Liege, Liege, Belgium
| | - Pascale Hubert
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
| | - Michael Herfs
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liege, Belgium
| |
Collapse
|
9
|
Chen X, Xu S, Pan J, Xu W, Yang H, Chen X, Chen R, Wang Y, Qiu S. Integrative single cell transcriptomic analysis reveals 3p deletion associated tumor microenvironment and chemoresistance in head and neck squamous cell carcinoma. Sci Rep 2025; 15:8224. [PMID: 40064955 PMCID: PMC11893908 DOI: 10.1038/s41598-025-92078-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) remains a prevalent and lethal malignancy, with a five-year survival rate of just 50% for cases of locally advanced disease. Chromosomal aberrations, particularly the deletion of the short arm of chromosome 3 (3p), have been strongly associated with poor prognosis and more aggressive tumor phenotypes. The tumor microenvironment (TME) plays a pivotal role in tumor progression and resistance to therapy. This study aims to elucidate the impact of 3p deletion on the TME, immune cell infiltration, and treatment response in HNSCC, to identify novel therapeutic targets to improve patient outcomes. We analyzed single-cell RNA sequencing (scRNA-seq) data from the Gene Expression Omnibus (GEO) and bulk transcriptome data from The Cancer Genome Atlas (TCGA). Pseudo-time trajectory and cell-cell communication analyses were performed with the Monocle and CellChat packages. The Wilcoxon test was used to evaluate the differential gene expression between wild-type (wt) and mutant (mut) groups. Prognostic models were developed using the Least Absolute Shrinkage and Selection Operator (LASSO) algorithm and Cox regression analyses to find the genes related to survival, with survival analysis conducted via Kaplan-Meier curves. Gene set enrichment analysis (GSEA) was employed to investigate pathway dysregulation, and immune cell infiltration was assessed using various immune scoring methodologies to explore the differences immune environment. The Tumor Immune Dysfunction and Exclusion (TIDE) database was utilized to predict the potential efficacy of immune checkpoint inhibitors. mRNA and protein expression levels of SPP1 were examined by RT-qPCR and Western blotting, while cell proliferation was assessed using the CCK8 assay. The mut group demonstrated significant alterations in cellular composition, characterized by increased endothelial cells and macrophages and decreased fibroblasts and CD8 + T cells, indicative of an immunosuppressive TME. Differential expression analysis revealed downregulation of immune pathways, including antigen processing and presentation, T cell receptor signaling, and B cell receptor signaling pathways in the mut group, along with enhanced metabolic activity in glycolysis and lipid metabolism. The prognostic model identified nine key genes associated with poor survival in HNSCC. The mut group exhibited poorer overall survival and a more immunosuppressive microenvironment compared to the wt group, which correlated with the outcomes observed in high-risk versus low-risk groups. High-risk patients also showed a diminished response to immunotherapy compared to low-risk patients. Additionally, SPP1 emerged as a critical gene associated with chemotherapy resistance and macrophage M2 polarization. This study demonstrates that 3p deletion significantly reshapes the TME, contributing to poor prognosis in HNSCC by fostering an immunosuppressive environment and enhancing chemoresistance. These findings highlight the potential for developing targeted therapies that address the genetic and immunological landscape of HNSCC.
Collapse
Affiliation(s)
- Xiaochuan Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Siqi Xu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Junping Pan
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Wenqian Xu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Hanxuan Yang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Xin Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Ronghui Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yuan Wang
- The school of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Sufang Qiu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
| |
Collapse
|
10
|
Mercadante M, Scheben A, Estrada J, Savas-Carstens J, Sullivan W, Housel N, Volpari T, Hebner J, Sapar M, Rusielewicz T, Monsma FJ, Semrau S, Wang Y, Martin LA. A patient-derived ovarian cancer organoid platform to study susceptibility to natural killer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641285. [PMID: 40093054 PMCID: PMC11908259 DOI: 10.1101/2025.03.06.641285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Intratumoral heterogeneity drives therapy resistance and relapses in advanced stage cancers, such as ovarian cancer. Here, we present a live cell imaging assay using patient-derived ovarian cancer organoids for real time capture and quantification of natural killer cell-mediated apoptotic events in >500 organoids simultaneously. Our assay revealed significant inter- and intratumor response heterogeneity and identified a rare resistant organoid population, opening avenues to test immunomodulatory strategies that overcome resistance.
Collapse
Affiliation(s)
| | - Armin Scheben
- The New York Stem Cell Foundation Research Institute, New York, NY
| | - Jacob Estrada
- The New York Stem Cell Foundation Research Institute, New York, NY
| | | | - William Sullivan
- The New York Stem Cell Foundation Research Institute, New York, NY
| | | | - Tatiana Volpari
- The New York Stem Cell Foundation Research Institute, New York, NY
| | - Jax Hebner
- The New York Stem Cell Foundation Research Institute, New York, NY
| | - Maria Sapar
- The New York Stem Cell Foundation Research Institute, New York, NY
| | - Tom Rusielewicz
- The New York Stem Cell Foundation Research Institute, New York, NY
| | | | - Stefan Semrau
- The New York Stem Cell Foundation Research Institute, New York, NY
| | - Yinan Wang
- The New York Stem Cell Foundation Research Institute, New York, NY
| | - Laura A Martin
- The New York Stem Cell Foundation Research Institute, New York, NY
| |
Collapse
|
11
|
Rymuza J, Woroniecka R, Grygalewicz B, Bujko M. Pivotal role of biallelic frequency analysis in identifying copy number alterations using genome-wide methods in tumors with a high level of aneuploidy. J Appl Genet 2025:10.1007/s13353-025-00951-x. [PMID: 40036001 DOI: 10.1007/s13353-025-00951-x] [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: 05/28/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 03/06/2025]
Abstract
Chromosome number abnormalities is one of the hallmarks of cancer. DNA copy number alterations (CNA) are studied using various genome-wide methods. In our study, we investigated CNA in human pituitary tumors using three platforms CytoSNP-850 K microarrays, low-pass whole-genome sequencing (average × 7 coverage, LPWGS), and Infinium Methylation EPIC array. Virtual karyotypes based on each dataset were generated using open-source software packages for each sample. Concordant CNA profiles were found for most of tumor. Surprisingly, substantial discrepancies between results from SNP arrays and LPWGS/EPIC arrays were identified in 20% of tumors, for which discrimination of true karyotype was required. B-allelic frequency data from SNP arrays was crucial to adjust normal ploidy level as ultimately verified with FISH. The discrepancy between virtual karyotypes was more pronounced the more CNAs were found. When CNAs covered around half of genome, the level of normal/diploid copy number was incorrectly set with methods, based solely on signal intensity/read-counts coverage. To conclude, CNA analyses with methods such as LPWGS and methylation arrays in highly aneuploid tumors are prone to a bias from improper normal ploidy level setting. These methods are commonly used therefore, we aimed to aware the scientific community about this underestimated methodological problem.
Collapse
Affiliation(s)
- Julia Rymuza
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Renata Woroniecka
- Cytogenetic Laboratory, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Beata Grygalewicz
- Cytogenetic Laboratory, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Mateusz Bujko
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.
| |
Collapse
|
12
|
Sokolowski D, Mai M, Verma A, Morgenshtern G, Subasri V, Naveed H, Yampolsky M, Wilson M, Goldenberg A, Erdman L. iModEst: disentangling -omic impacts on gene expression variation across genes and tissues. NAR Genom Bioinform 2025; 7:lqaf011. [PMID: 40041206 PMCID: PMC11879402 DOI: 10.1093/nargab/lqaf011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/16/2025] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
Many regulatory factors impact the expression of individual genes including, but not limited, to microRNA, long non-coding RNA (lncRNA), transcription factors (TFs), cis-methylation, copy number variation (CNV), and single-nucleotide polymorphisms (SNPs). While each mechanism can influence gene expression substantially, the relative importance of each mechanism at the level of individual genes and tissues is poorly understood. Here, we present the integrative Models of Estimated gene expression (iModEst), which details the relative contribution of different regulators to the gene expression of 16,000 genes and 21 tissues within The Cancer Genome Atlas (TCGA). Specifically, we derive predictive models of gene expression using tumour data and test their predictive accuracy in cancerous and tumour-adjacent tissues. Our models can explain up to 70% of the variance in gene expression across 43% of the genes within both tumour and tumour-adjacent tissues. We confirm that TF expression best predicts gene expression in both tumour and tumour-adjacent tissue whereas methylation predictive models in tumour tissues does not transfer well to tumour adjacent tissues. We find new patterns and recapitulate previously reported relationships between regulator and gene-expression, such as CNV-predicted FGFR2 expression and SNP-predicted TP63 expression. Together, iModEst offers an interactive, comprehensive atlas of individual regulator-gene-tissue expression relationships as well as relationships between regulators.
Collapse
Affiliation(s)
- Dustin J Sokolowski
- Department of Molecular Genetics, University of Toronto, ON M5S 3K3, Canada
- Department of Computer Science, University of Toronto, ON M5S 2E4, Canada
| | - Mingjie Mai
- Department of Computer Science, University of Toronto, ON M5S 2E4, Canada
- SickKids Research Institute, Program in Genetics and Genome Biology, ON M5G 0A4, Canada
- Vector Institute
| | - Arnav Verma
- Department of Computer Science, University of Toronto, ON M5S 2E4, Canada
| | - Gabriela Morgenshtern
- Department of Computer Science, University of Toronto, ON M5S 2E4, Canada
- SickKids Research Institute, Program in Genetics and Genome Biology, ON M5G 0A4, Canada
- Vector Institute
| | - Vallijah Subasri
- SickKids Research Institute, Program in Genetics and Genome Biology, ON M5G 0A4, Canada
- Department of Medical Biophysics, University of Toronto, ON M5G 2C4, Canada
| | - Hareem Naveed
- Department of Computer Science, University of Toronto, ON M5S 2E4, Canada
- SickKids Research Institute, Program in Genetics and Genome Biology, ON M5G 0A4, Canada
| | - Maria Yampolsky
- SickKids Research Institute, Program in Genetics and Genome Biology, ON M5G 0A4, Canada
| | - Michael D Wilson
- Department of Molecular Genetics, University of Toronto, ON M5S 3K3, Canada
- SickKids Research Institute, Program in Genetics and Genome Biology, ON M5G 0A4, Canada
| | - Anna Goldenberg
- Department of Computer Science, University of Toronto, ON M5S 2E4, Canada
- SickKids Research Institute, Program in Genetics and Genome Biology, ON M5G 0A4, Canada
- Vector Institute
- CIFAR: Child and Brain Development, Toronto, ON M5G 1M1, Canada
| | - Lauren Erdman
- Department of Computer Science, University of Toronto, ON M5S 2E4, Canada
- SickKids Research Institute, Program in Genetics and Genome Biology, ON M5G 0A4, Canada
- Vector Institute
- James M. Anderson Center for Health Systems Excellence, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- College of Medicine, University of Cincinnati, OH 45267, United States
| |
Collapse
|
13
|
Machado GC, Ferrer VP. MUC1 and MUC4 expression are inversely correlated and trigger immunological response and transport pathways in adult-type diffuse gliomas. Comput Biol Med 2025; 187:109730. [PMID: 39889447 DOI: 10.1016/j.compbiomed.2025.109730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 02/03/2025]
Abstract
Adult-type diffuse gliomas arise from glial or progenitor cells. These tumors are currently classified as astrocytoma isocitrate dehydrogenase (IDH)-mutant or IDH-mutant oligodendroglioma with co-deletion of chromosomal arms 1p and 19q, both of which could be either slow-growing tumors, or glioblastoma (GBM), which is a more aggressive tumor. Despite advances in diagnosis and treatment, the median survival time after GBM diagnosis remains low at approximately 15 months, with a 5-year overall survival (OS) rate of 6.8 %. Therefore, new biomarker and therapeutic target discoveries are required to improve prognosis. Mucin 1 (MUC1) and MUC4 are membrane-bound mucins and potential biomarkers of several tumors. However, the role of these mucins in adult gliomas has not been well explored. In this retrospective study, in silico analysis of data from patients with adult-type diffuse glioma revealed differential methylation and expression patterns of MUC1 and MUC4 between GBM and non-GBM groups. In the GBM group, decreased methylation and elevated expression of MUC1 were observed (r = -0.25, p < 0.0001), whereas increased methylation and decreased expression of MUC4 were observed (r = -0.13, p = 0.1344). Conversely, in the non-GBM group, MUC1 exhibited higher methylation and lower expression (r = -0.27, p < 0.0001), whereas MUC4 showed lower methylation and higher expression (r = -0.32, p < 0.0001). The expression of these genes influenced OS in adult patients with glioma (p = 0.0344), with high MUC1 and low MUC4 expression associated with worse OS. MUC1 and MUC4 expression correlated with that of MUC20 in both GBM (r = 0.54) and non-GBM (r = 0.53) groups (p < 0.0001). Functional enrichment analysis identified the biological roles of MUC1-co-expressed genes as involvement in innate immunity, antigen processing, and proinflammatory responses in both the non-GBM and GBM groups, and integrin-based signaling pathways in the GBM group. MUC4-co-expressed genes are involved in ion transport in GBM patients. Using molecular docking, we observed that MUC1 domains physically interact with immune response-related proteins, such as receptors for advanced glycation end products (RAGE), major histocompatibility complex II (MHC-II), and extracellular matrix receptor integrin alpha 2 (ITGA2). To our knowledge, this is the first retrospective study and in silico analysis demonstrating the relevance and correlation of MUC1 and MUC4 in adult gliomas. These findings elucidate the molecular mechanisms underlying adult-type diffuse glioma progression and highlight MUC1 and MUC4 as potential prognostic markers and therapeutic targets for glioma management.
Collapse
Affiliation(s)
- Gabriel Cardoso Machado
- Graduate Program in Pathological Anatomy, Faculty of Medicine, Rio de Janeiro Federal University, Rio de Janeiro, Brazil; Laboratory of Cell and Molecular Biology of Tumors, Department of Cell and Molecular Biology, Biology Institute, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Valéria Pereira Ferrer
- Graduate Program in Pathological Anatomy, Faculty of Medicine, Rio de Janeiro Federal University, Rio de Janeiro, Brazil; Laboratory of Cell and Molecular Biology of Tumors, Department of Cell and Molecular Biology, Biology Institute, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil.
| |
Collapse
|
14
|
Weiner S, Bansal MS. DICE: fast and accurate distance-based reconstruction of single-cell copy number phylogenies. Life Sci Alliance 2025; 8:e202402923. [PMID: 39667913 PMCID: PMC11638338 DOI: 10.26508/lsa.202402923] [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: 07/02/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024] Open
Abstract
Somatic copy number alterations (sCNAs) are valuable phylogenetic markers for inferring evolutionary relationships among tumor cell subpopulations. Advances in single-cell DNA sequencing technologies are making it possible to obtain such sCNAs datasets at ever-larger scales. However, existing methods for reconstructing phylogenies from sCNAs are often too slow for large datasets. We propose two new distance-based methods, DICE-bar and DICE-star, for reconstructing single-cell tumor phylogenies from sCNA data. Using carefully simulated datasets, we find that DICE-bar matches or exceeds the accuracies of all other methods on noise-free datasets and that DICE-star shows exceptional robustness to noise and outperforms all other methods on noisy datasets. Both methods are also orders of magnitude faster than many existing methods. Our experimental analysis also reveals how noise/error in copy number inference, as expected for real datasets, can drastically impact the accuracies of most methods. We apply DICE-star, the most accurate method on error-prone datasets, to several real single-cell breast and ovarian cancer datasets and find that it rapidly produces phylogenies of equivalent or greater reliability compared with existing methods.
Collapse
Affiliation(s)
- Samson Weiner
- School of Computing, University of Connecticut, Storrs, CT, USA
| | - Mukul S Bansal
- School of Computing, University of Connecticut, Storrs, CT, USA
- The Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
15
|
Bökenkamp JE, Keuper K, Redel S, Barthel K, Johnson L, Becker A, Wieland A, Räschle M, Storchová Z. Proteogenomic analysis reveals adaptive strategies for alleviating the consequences of aneuploidy in cancer. EMBO J 2025; 44:1829-1865. [PMID: 39930267 PMCID: PMC11914506 DOI: 10.1038/s44318-025-00372-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 03/19/2025] Open
Abstract
Aneuploidy is prevalent in cancer and associates with fitness advantage and poor patient prognosis. Yet, experimentally induced aneuploidy initially leads to adverse effects and impaired proliferation, suggesting that cancer cells must adapt to aneuploidy. We performed in vitro evolution of cells with extra chromosomes and obtained cell lines with improved proliferation and gene expression changes congruent with changes in aneuploid cancers. Integrated analysis of cancer multi-omics data and model cells revealed increased expression of DNA replicative and repair factors, reduced genomic instability, and reduced lysosomal degradation. We identified E2F4 and FOXM1 as transcription factors strongly associated with adaptation to aneuploidy in vitro and in cancers and validated this finding. The adaptation to aneuploidy also coincided with specific copy number aberrations that correlate with poor patient prognosis. Chromosomal engineering mimicking these aberrations improved aneuploid cell proliferation, while loss of previously present extra chromosomes impaired it. The identified common adaptation strategies suggest replication stress, genomic instability, and lysosomal stress as common liabilities of aneuploid cancers.
Collapse
Affiliation(s)
- Jan-Eric Bökenkamp
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Kristina Keuper
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
- Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Stefan Redel
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Karen Barthel
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Leah Johnson
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Amelie Becker
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Angela Wieland
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Markus Räschle
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Zuzana Storchová
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany.
| |
Collapse
|
16
|
Curry RN, McDonald MF, He P, Lozzi B, Ko Y, O’Reilly I, Rosenbaum A, Kwon W, Fahim L, Marcus J, Powell N, Wang S, Ma J, Multani A, Choi DJ, Sardar D, Mohila C, Lee J, Gallo M, Harmanci A, Harmanci AS, Deneen B, Rao G. Mutant IDH impairs chromatin binding by PDGFB to promote chromosome instability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.20.639365. [PMID: 40060572 PMCID: PMC11888161 DOI: 10.1101/2025.02.20.639365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Non-canonical roles for growth factors in the nucleus have been previously described, but their mechanism of action and biological roles remain enigmatic. Platelet-derived growth factor B (PDGFB) can drive formation of low-grade glioma and here we show that it localizes to the nucleus of human glioma cells where it binds chromatin to preserve genome stability and cell lineage. Failure of PDGFB to localize to the nucleus leads to chromosomal abnormalities, aberrant heterochromatin architecture and accelerated tumorigenesis. Furthermore, nuclear localization of PDGFB is reliant upon the expression levels and mutation status of isocitrate dehydrogenase (IDH). Unexpectedly, we identified macrophages as the predominant source of PDGFB in human, finding that immune-derived PDGFB can localize to the nucleus of glioma cells. Collectively, these studies show that immune derived PDGFB enters the nucleus of glioma cells to maintain genomic stability, while identifying a new mechanism by which IDH mutations promote gliomagenesis.
Collapse
Affiliation(s)
- Rachel N. Curry
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
- Pediatric Neuro-Oncology Research Program, Texas Children’s Hospital, Houston, TX
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Malcolm F. McDonald
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX
- Program in Development, Disease Models, and Therapeutics, Baylor College of Medicine, Houston, TX
| | - Peihao He
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
- Program in Cancer Cell Biology, Baylor College of Medicine, Houston, TX, USA
| | - Brittney Lozzi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
- Program in Genetics and Genomics, Baylor College of Medicine, Houston, TX, USA
| | - Yeunjung Ko
- Program in Immunology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Isabella O’Reilly
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Anna Rosenbaum
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Wookbong Kwon
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Leyla Fahim
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Joshua Marcus
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Noah Powell
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Su Wang
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX
| | - Jin Ma
- Cytogenetics and Cell Authentication Core, MD Anderson Cancer Center, Houston, TX
| | - Asha Multani
- Cytogenetics and Cell Authentication Core, MD Anderson Cancer Center, Houston, TX
| | - Dong-Joo Choi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Debo Sardar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Carrie Mohila
- Department of Neuropatholgy, Texas Children’s Hospital, Houston, TX
| | - Jason Lee
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Marco Gallo
- Pediatric Neuro-Oncology Research Program, Texas Children’s Hospital, Houston, TX
| | - Arif Harmanci
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX
| | - Akdes Serin Harmanci
- Program in Genetics and Genomics, Baylor College of Medicine, Houston, TX, USA
- Program in Immunology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX
| | - Benjamin Deneen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
- Program in Development, Disease Models, and Therapeutics, Baylor College of Medicine, Houston, TX
- Program in Cancer Cell Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX
| | - Ganesh Rao
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX
| |
Collapse
|
17
|
Abdulrahman FA, Benford KA, Lin GT, Maroun AJ, Sammons C, Shirzad DN, Tsai H, Van Brunt VL, Jones Z, Marquez JE, Ratkus EC, Shehadeh AK, Abasto Valle H, Fejzo D, Gilbert AE, McWee CA, Underwood LF, Indico E, Rork BB, Nanjundan M. zDHHC-Mediated S-Palmitoylation in Skin Health and Its Targeting as a Treatment Perspective. Int J Mol Sci 2025; 26:1673. [PMID: 40004137 PMCID: PMC11854935 DOI: 10.3390/ijms26041673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
S-acylation, which includes S-palmitoylation, is the only known reversible lipid-based post-translational protein modification. S-palmitoylation is mediated by palmitoyl acyltransferases (PATs), a family of 23 enzymes commonly referred to as zDHHCs, which catalyze the addition of palmitate to cysteine residues on specific target proteins. Aberrant S-palmitoylation events have been linked to the pathogenesis of multiple human diseases. While there have been advances in elucidating the molecular mechanisms underlying the pathogenesis of various skin conditions, there remain gaps in the knowledge, specifically with respect to the contribution of S-palmitoylation to the maintenance of skin barrier function. Towards this goal, we performed PubMed literature searches relevant to S-palmitoylation in skin to define current knowledge and areas that may benefit from further research studies. Furthermore, to identify alterations in gene products that are S-palmitoylated, we utilized bioinformatic tools such as SwissPalm and analyzed relevant data from publicly available databases such as cBioportal. Since the targeting of S-palmitoylated targets may offer an innovative treatment perspective, we surveyed small molecules inhibiting zDHHCs, including 2-bromopalmitate (2-BP) which is associated with off-target effects, and other targeting strategies. Collectively, our work aims to advance both basic and clinical research on skin barrier function with a focus on zDHHCs and relevant protein targets that may contribute to the pathogenesis of skin conditions such as atopic dermatitis, psoriasis, and skin cancers including melanoma.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Meera Nanjundan
- Department of Molecular Biosciences, University of South Florida, 4202 East Fowler Avenue, ISA2015, Tampa, FL 33620, USA; (F.A.A.); (K.A.B.); (G.T.L.); (A.J.M.); (C.S.); (D.N.S.); (H.T.); (V.L.V.B.); (Z.J.); (J.E.M.); (E.C.R.); (A.K.S.); (H.A.V.); (D.F.); (A.E.G.); (C.A.M.); (L.F.U.); (E.I.); (B.B.R.)
| |
Collapse
|
18
|
Ellrott K, Wong CK, Yau C, Castro MAA, Lee JA, Karlberg BJ, Grewal JK, Lagani V, Tercan B, Friedl V, Hinoue T, Uzunangelov V, Westlake L, Loinaz X, Felau I, Wang PI, Kemal A, Caesar-Johnson SJ, Shmulevich I, Lazar AJ, Tsamardinos I, Hoadley KA, Robertson AG, Knijnenburg TA, Benz CC, Stuart JM, Zenklusen JC, Cherniack AD, Laird PW. Classification of non-TCGA cancer samples to TCGA molecular subtypes using compact feature sets. Cancer Cell 2025; 43:195-212.e11. [PMID: 39753139 PMCID: PMC11949768 DOI: 10.1016/j.ccell.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 08/26/2024] [Accepted: 12/05/2024] [Indexed: 02/12/2025]
Abstract
Molecular subtypes, such as defined by The Cancer Genome Atlas (TCGA), delineate a cancer's underlying biology, bringing hope to inform a patient's prognosis and treatment plan. However, most approaches used in the discovery of subtypes are not suitable for assigning subtype labels to new cancer specimens from other studies or clinical trials. Here, we address this barrier by applying five different machine learning approaches to multi-omic data from 8,791 TCGA tumor samples comprising 106 subtypes from 26 different cancer cohorts to build models based upon small numbers of features that can classify new samples into previously defined TCGA molecular subtypes-a step toward molecular subtype application in the clinic. We validate select classifiers using external datasets. Predictive performance and classifier-selected features yield insight into the different machine-learning approaches and genomic data platforms. For each cancer and data type we provide containerized versions of the top-performing models as a public resource.
Collapse
Affiliation(s)
- Kyle Ellrott
- Oregon Health and Science University, Portland, OR 97239, USA.
| | - Christopher K Wong
- Biomolecular Engineering Department, School of Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Christina Yau
- University of California, San Francisco, Department of Surgery, San Francisco, CA 94158, USA; Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Mauro A A Castro
- Bioinformatics and Systems Biology Laboratory, Federal University of Paraná, Curitiba, PR 81520-260, Brazil
| | - Jordan A Lee
- Oregon Health and Science University, Portland, OR 97239, USA
| | | | - Jasleen K Grewal
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Vincenzo Lagani
- JADBio Gnosis DA, GR-700 13 Heraklion, Crete, Greece; Institute of Chemical Biology, Ilia State University, Tbilisi 0162, Georgia
| | - Bahar Tercan
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, WA 98109, USA
| | - Verena Friedl
- Biomolecular Engineering Department, School of Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Toshinori Hinoue
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Vladislav Uzunangelov
- Biomolecular Engineering Department, School of Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Lindsay Westlake
- The Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Xavier Loinaz
- The Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Ina Felau
- Center for Cancer Genomics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Peggy I Wang
- Center for Cancer Genomics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Anab Kemal
- Center for Cancer Genomics, National Cancer Institute, Bethesda, MD 20892, USA
| | | | - Ilya Shmulevich
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, WA 98109, USA
| | - Alexander J Lazar
- Departments of Pathology & Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ioannis Tsamardinos
- JADBio Gnosis DA, GR-700 13 Heraklion, Crete, Greece; Department of Computer Science, University of Crete, GR-700 13 Heraklion, Crete, Greece; Institute of Applied and Computational Mathematics, Foundation for Research and Technology Hellas (FORTH), GR-700 13 Heraklion, Crete, Greece
| | - Katherine A Hoadley
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27519, USA
| | - A Gordon Robertson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Theo A Knijnenburg
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, WA 98109, USA
| | | | - Joshua M Stuart
- Biomolecular Engineering Department, School of Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jean C Zenklusen
- Center for Cancer Genomics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Andrew D Cherniack
- The Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA.
| | - Peter W Laird
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
19
|
Henick BS, Taylor AM, Nakagawa H, Wong KK, Diehl JA, Rustgi AK. Squamous cell cancers of the aero-upper digestive tract: A unified perspective on biology, genetics, and therapy. Cancer Cell 2025; 43:178-194. [PMID: 39933897 PMCID: PMC11875029 DOI: 10.1016/j.ccell.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/23/2024] [Accepted: 01/10/2025] [Indexed: 02/13/2025]
Abstract
Squamous cell cancers (SCCs) of the head and neck, esophagus, and lung, referred to as aero-upper digestive SCCs, are prevalent in the United States and worldwide. Their incidence and mortality are projected to increase at alarming rates, posing diagnostic, prognostic, and therapeutic challenges. These SCCs share certain epigenetic, genomic, and genetic alterations, immunologic properties, environmental exposures, as well as lifestyle and nutritional risk factors, which may underscore common complex gene-environmental interactions across them. This review focuses upon the frequent shared epigenetic, genomic, and genetic alterations, emerging preclinical model systems, and how this collective knowledge can be leveraged into perspectives on standard of care therapies and mechanisms of resistance, nominating new potential directions in translational therapeutics.
Collapse
Affiliation(s)
- Brian S Henick
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Division of Hematology-Oncology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Alison M Taylor
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Hiroshi Nakagawa
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Kwok-Kin Wong
- Division of Hematology-Oncology, Department of Medicine, NYU Perlmutter Cancer Center, New York, NY, USA
| | - J Alan Diehl
- Department of Biochemistry, Case Western Reserve Comprehensive Cancer Center, Cleveland, OH, USA
| | - Anil K Rustgi
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
20
|
Ohkuni K, Au WC, Kazi A, Balachandra V, Basrai M. Oncohistone H3 E97K mutation facilitates CENP-A mislocalization and chromosomal instability in budding yeast. Nucleic Acids Res 2025; 53:gkaf083. [PMID: 39970289 PMCID: PMC11822376 DOI: 10.1093/nar/gkaf083] [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: 10/16/2024] [Revised: 01/14/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Mislocalization of overexpressed CENP-A (Cse4 in budding yeast) contributes to chromosomal instability (CIN) in yeasts, flies, and human cells. Overexpression of CENP-A is observed in many cancers and this correlates with poor prognosis. Here, we show that altered stoichiometry of histone H3 and expression of oncohistone mutation H3 E97K contributes to mislocalization of Cse4 and CIN. Oncohistone mutations in the globular domain of histone H3 such as H3 E97K occur in several cancers; however, their functional effects remain unexplored. We demonstrated that strains with reduced gene dosage of histone H3 (hht1Δ and hht2Δ) or oncohistone H3 E97K mutation exhibit enhanced Cse4-H4 interaction, an in vivo change in the conformational state of Cse4, and this contributes to mislocalization of Cse4. Oncohistone H3 E97K mutant protein was unstable and exhibited defects in interaction with histone H4. Notably, mislocalization of Cse4 and CIN phenotypes were observed in hht1Δ and oncohistone H3 E97K mutants expressing endogenous Cse4. In summary, our studies highlight the importance of histone H3 stoichiometry in preventing mislocalization of Cse4 for chromosomal stability and suggest that oncohistone H3 mutations may contribute to CIN in human cancers.
Collapse
Affiliation(s)
- Kentaro Ohkuni
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei-Chun Au
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Amira Z Kazi
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vinutha Balachandra
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Munira A Basrai
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
21
|
Hertel A, Storchová Z. The Role of p53 Mutations in Early and Late Response to Mitotic Aberrations. Biomolecules 2025; 15:244. [PMID: 40001547 PMCID: PMC11852650 DOI: 10.3390/biom15020244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Mutations in the TP53 gene and chromosomal instability (CIN) are two of the most common alterations in cancer. CIN, marked by changes in chromosome numbers and structure, drives tumor development, but is poorly tolerated in healthy cells, where developmental and tissue homeostasis mechanisms typically eliminate cells with chromosomal abnormalities. Mechanisms that allow cancer cells to acquire and adapt to CIN remain largely unknown. Tumor suppressor protein p53, often referred to as the "guardian of the genome", plays a critical role in maintaining genomic stability. In cancer, CIN strongly correlates with TP53 mutations, and recent studies suggest that p53 prevents the propagation of cells with abnormal karyotypes arising from mitotic errors. Furthermore, p53 dysfunction is frequent in cells that underwent whole-genome doubling (WGD), a process that facilitates CIN onset, promotes aneuploidy tolerance, and is associated with poor patient prognosis across multiple cancer types. This review summarizes current insights into p53's role in protecting cells from chromosome copy number alterations and discusses the implications of its dysfunction for the adaption and propagation of cancer cells.
Collapse
Affiliation(s)
| | - Zuzana Storchová
- Group Molecular Genetics, Faculty of Biology, RPTU Kaiserslautern-Landau, Paul Ehrlich Str. 24, 67663 Kaiserslautern, Germany
| |
Collapse
|
22
|
Liu Y, Lai J, Wood LD, Karchin R. SVCFit: Inferring structural variant cellular fraction in tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.01.636056. [PMID: 39975255 PMCID: PMC11838439 DOI: 10.1101/2025.02.01.636056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Many tumors evolve through cellular mutation and selection, where subpopulations of cells (subclones) with shared ancestry compete for dominance. Introduction of next generation sequencing enables subclone identification using small somatic. However, there are several advantages to marking subclones with structural variants: they have greater functional impact, play a crucial role in late-stage tumors, and provide a more complete view of genomic instability driving tumor evolution. Here, we present SVCFit, a scalable method to estimate the cellular prevalence of somatic deletions, duplications and inversions. We demonstrate that cellular prevalence estimation can be improved by incorporating distinct read patterns for each structural variant type. Additionally, this improvement is achieved without prior knowledge of tumor purity, which is often inaccurate. Using a combination of simulated data and patient-derived metastatic samples with known mixture proportions, we show that our algorithm achieves significantly greater accuracy than state-of-the-art in estimating the structural variants cellular prevalence (p<0.05). The speed of SVCFit estimation from cost-effective bulk whole-genome sequencing (WGS) makes it well-suited for analyzing large cohorts of sequenced tumor samples, enhancing the accessibility of SV-based clonal reconstruction.
Collapse
|
23
|
Zhang Y, Leung AK, Kang JJ, Sun Y, Wu G, Li L, Sun J, Cheng L, Qiu T, Zhang J, Wierbowski SD, Gupta S, Booth JG, Yu H. A multiscale functional map of somatic mutations in cancer integrating protein structure and network topology. Nat Commun 2025; 16:975. [PMID: 39856048 PMCID: PMC11760531 DOI: 10.1038/s41467-024-54176-3] [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: 03/11/2024] [Accepted: 11/04/2024] [Indexed: 01/27/2025] Open
Abstract
A major goal of cancer biology is to understand the mechanisms driven by somatically acquired mutations. Two distinct methodologies-one analyzing mutation clustering within protein sequences and 3D structures, the other leveraging protein-protein interaction network topology-offer complementary strengths. We present NetFlow3D, a unified, end-to-end 3D structurally-informed protein interaction network propagation framework that maps the multiscale mechanistic effects of mutations. Built upon the Human Protein Structurome, which incorporates the 3D structures of every protein and the binding interfaces of all known protein interactions, NetFlow3D integrates atomic, residue, protein and network-level information: It clusters mutations on 3D protein structures to identify driver mutations and propagates their impacts anisotropically across the protein interaction network, guided by the involved interaction interfaces, to reveal systems-level impacts. Applied to 33 cancer types, NetFlow3D identifies 2 times more 3D clusters and incorporates 8 times more proteins in significantly interconnected network modules compared to traditional methods.
Collapse
Affiliation(s)
- Yingying Zhang
- Department of Computational Biology, Cornell University, Ithaca, 14853, NY, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, 14853, NY, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, 14853, NY, USA
| | - Alden K Leung
- Department of Computational Biology, Cornell University, Ithaca, 14853, NY, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, 14853, NY, USA
| | - Jin Joo Kang
- Department of Computational Biology, Cornell University, Ithaca, 14853, NY, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, 14853, NY, USA
| | - Yu Sun
- Department of Computational Biology, Cornell University, Ithaca, 14853, NY, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, 14853, NY, USA
| | - Guanxi Wu
- College of Agriculture and Life Sciences, Cornell University, Ithaca, 14853, NY, USA
| | - Le Li
- Department of Computational Biology, Cornell University, Ithaca, 14853, NY, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, 14853, NY, USA
| | - Jiayang Sun
- Department of Computational Biology, Cornell University, Ithaca, 14853, NY, USA
| | - Lily Cheng
- Department of Science and Technology Studies, Cornell University, Ithaca, 14853, NY, USA
| | - Tian Qiu
- School of Electrical and Computer Engineering, Cornell University, Ithaca, 14853, NY, USA
| | - Junke Zhang
- Department of Computational Biology, Cornell University, Ithaca, 14853, NY, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, 14853, NY, USA
| | - Shayne D Wierbowski
- Department of Computational Biology, Cornell University, Ithaca, 14853, NY, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, 14853, NY, USA
| | - Shagun Gupta
- Department of Computational Biology, Cornell University, Ithaca, 14853, NY, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, 14853, NY, USA
| | - James G Booth
- Department of Computational Biology, Cornell University, Ithaca, 14853, NY, USA
- Department of Statistics and Data Science, Cornell University, Ithaca, 14853, NY, USA
| | - Haiyuan Yu
- Department of Computational Biology, Cornell University, Ithaca, 14853, NY, USA.
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, 14853, NY, USA.
| |
Collapse
|
24
|
Gardner AL, Zheng L, Howland K, Saunders A, Ramirez A, Parker P, Iloegbunam C, Morgan D, Jost TA, Brock A. Mapping cell-cell fusion at single-cell resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.11.627873. [PMID: 39896473 PMCID: PMC11785005 DOI: 10.1101/2024.12.11.627873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Cell-cell fusion is a tightly controlled process in the human body known to be involved in fertilization, placental development, muscle growth, bone remodeling, and viral response. Fusion between cancer cells results first in a whole-genome doubled state, which may be followed by the generation of aneuploidies; these genomic alterations are known drivers of tumor evolution. The role of cell-cell fusion in cancer progression and treatment response has been understudied due to limited experimental systems for tracking and analyzing individual fusion events. To meet this need, we developed a molecular toolkit to map the origins and outcomes of individual cell fusion events within a tumor cell population. This platform, ClonMapper Duo ('CMDuo'), identifies cells that have undergone cell-cell fusion through a combination of reporter expression and engineered fluorescence-associated index sequences paired to randomly generated nucleotide barcodes. scRNA-seq of the indexed barcodes enables the mapping of each set of parental cells and fusion progeny throughout the cell population. In triple-negative breast cancer cells CMDuo uncovered subclonal transcriptomic hybridization and unveiled distinct cell-states which arise in direct consequence of homotypic cell-cell fusion. CMDuo is a platform that enables mapping of cell-cell fusion events in high-throughput single cell data and enables the study of cell fusion in disease progression and therapeutic response.
Collapse
Affiliation(s)
- Andrea L Gardner
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Lan Zheng
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Kennedy Howland
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Andrew Saunders
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Andrea Ramirez
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Patrik Parker
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Chisom Iloegbunam
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Daylin Morgan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Tyler A Jost
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Amy Brock
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
25
|
Bates BA, Bates KE, Boris SA, Wessman C, Stone D, Bryan J, Davis MF, Bailey MH. Intersection of rare pathogenic variants from TCGA in the All of Us Research Program v6. HGG ADVANCES 2025; 6:100405. [PMID: 39799398 PMCID: PMC11830373 DOI: 10.1016/j.xhgg.2025.100405] [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: 08/13/2024] [Revised: 01/09/2025] [Accepted: 01/08/2025] [Indexed: 01/15/2025] Open
Abstract
Using rare cancer predisposition alleles derived from The Cancer Genome Atlas (TCGA) and high cancer prevalence (14% of participants) in All of Us (version 6), we assessed the impact of these rare alleles on cancer occurrence in six broad groups of genetic similarity provided by All of Us: African/African American (AFR), Admixed American/Latino (AMR), East Asian (EAS), European (EUR), Middle Eastern (MID), or South Asian (SAS). We observed that germline susceptibility to cancer consistently replicates in EUR-like participants but less so in other participants. We found that All of Us participants from the EUR (p = 1.8 × 10-7), AFR (p = 0.018), and MID (p = 0.0083) genetic similarity groups who carry a rare pathogenic mutation are more likely to have cancer than those without a rare pathogenic mutation. With the advent of combining medical records and genetic mutations, we also performed a phenome-wide association study (PheWAS) to assess the effect of pathogenic variants on additional phenotypes. This analysis again showed several associations between predisposition variants and cancer in EUR-like participants, but fewer in those of the other genetic similarity groups. As All of Us grows to 1 million participants, our projections suggest sufficient power (>99%) to detect cancer-associated variants that are common, but limited power (∼28%) to detect rare mutations when using the entire cohort. This study provides preliminary insights into genetic predispositions to cancer across a diverse cohort and demonstrates the value of All of Us as a resource for cancer research.
Collapse
Affiliation(s)
- Blaine A Bates
- Department of Biology, Brigham Young University, Provo, UT 84061, USA; Department of Chemical Engineering, Brigham Young University, Provo, UT 84602, USA
| | - Kylee E Bates
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA
| | - Spencer A Boris
- Department of Biology, Brigham Young University, Provo, UT 84061, USA
| | - Colin Wessman
- Department of Biology, Brigham Young University, Provo, UT 84061, USA
| | - David Stone
- Department of Biology, Brigham Young University, Provo, UT 84061, USA
| | - Justin Bryan
- Department of Biology, Brigham Young University, Provo, UT 84061, USA
| | - Mary F Davis
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA; Department of Biomedical Informatics, Vanderbilt University, Nashville, TN 37203, USA
| | - Matthew H Bailey
- Department of Biology, Brigham Young University, Provo, UT 84061, USA; Simmons Center for Cancer Research, Brigham Young University, Provo, UT 84602, USA.
| |
Collapse
|
26
|
Voropaeva EN, Orlov YL, Loginova AB, Seregina OB, Maksimov VN, Pospelova TI. Deregulation mechanisms and therapeutic opportunities of p53-responsive microRNAs in diffuse large B-cell lymphoma. PeerJ 2025; 13:e18661. [PMID: 39802185 PMCID: PMC11720970 DOI: 10.7717/peerj.18661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/17/2024] [Indexed: 01/16/2025] Open
Abstract
Here, we have discussed the molecular mechanisms of p53-responsive microRNAs dysregulation in response to genotoxic stress in diffuse large B-cell lymphoma (DLBCL) patients. The role of micro ribonucleic acids (microRNAs) in p53-signaling cellular stress has been studied. MicroRNAs are the small non-coding RNAs, which regulate genes expression at post-transcriptional level. Many of them play a crucial role in carcinogenesis and may act as oncogenes or suppressor of tumor growth. The understanding of the effect of p53-responsive microRNA dysregulation on oncogenesis achieved in recent decades opens wide opportunities for the diagnosis, prediction and of microRNA-based cancer therapy. Development of new bioinformatics tools and databases for microRNA supports DLBCL research. We overview the studies on the role of miRNAs in regulating gene expression associated with tumorigenesis processes, with particular emphasis on their role as tumor growth-suppressing factors. The starting point is a brief description of the classical microRNA biogenesis pathway and the role of p53 in regulating the expression of these molecules. We analyze various molecular mechanisms leading to this dysregulation, including mutations in the TP53 gene, DNA methylation, changes in host-genes expression or microRNA gene copy number, mutations in microRNA and microRNA biogenesis genes.
Collapse
Affiliation(s)
- Elena N. Voropaeva
- Research Institute of Internal and Preventive Medicine - Branch of the Federal State Budget Scientific Institution “The Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences”, Novosibirsk, Russia
- Novosibirsk State Medical University of the Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Yuriy L. Orlov
- The Digital Health Center, I.M Sechenov First Moscow State Medical University, Moscow, Russia
- Agrarian and Technological Institute, Patrice Lumumba Peoples’ Friendship University of Russia, Moscow, Russia
| | - Anastasia B. Loginova
- Novosibirsk State Medical University of the Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Olga B. Seregina
- Novosibirsk State Medical University of the Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Vladimir N. Maksimov
- Research Institute of Internal and Preventive Medicine - Branch of the Federal State Budget Scientific Institution “The Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences”, Novosibirsk, Russia
- Novosibirsk State Medical University of the Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Tatiana I. Pospelova
- Novosibirsk State Medical University of the Ministry of Health of the Russian Federation, Novosibirsk, Russia
| |
Collapse
|
27
|
Zhang Q, Cao Z, Wang Y, Wu H, Zhang Z, Liu Z. Proteomic Analysis of Tissue Proteins Related to Lateral Lymph Node Metastasis in Papillary Thyroid Microcarcinoma. J Proteome Res 2025; 24:256-267. [PMID: 39600146 PMCID: PMC11705366 DOI: 10.1021/acs.jproteome.4c00737] [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: 08/27/2024] [Revised: 11/04/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024]
Abstract
Patients with lateral lymph node metastasis (LLNM) may experience higher locoregional recurrence rates and poorer prognoses compared to those without LLNM, highlighting the need for effective preoperative stratification to reliably assess risk LLNM. In this study, we collected PTMC samples from Peking Union Medical College Hospital and employed data-independent acquisition mass spectrometry proteomics technique to identify protein profiles in PTMC tissues with and without LLNM. Pseudo temporal analysis and single sample gene set enrichment analysis were conducted in combination with The Cancer Genome Atlas Thyroid Carcinoma for functional coordination analysis and the construction of a prediction model based on random forest. Non-negative matrix factorization (NMF) clustering was utilized to classify molecular subtypes of PTMC. Our findings revealed that the differential activation of pathways such as MAPK and PI3K was critical in enhancing the lateral lymph node metastatic potential of PTMC. We successfully screened biomarkers via machine learning and public databases, creating an effective prediction model for metastasis. Additionally, we explored the mechanism of metastasis-associated PTMC subtypes via NMF clustering. These insights into LLNM mechanisms in PTMC may contribute to future biomarker screening and the identification of therapeutic targets.
Collapse
Affiliation(s)
- Qiyao Zhang
- Department
of General Surgery, Peking Union Medical
College Hospital, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100730, P. R. China
| | - Zhen Cao
- Department
of General Surgery, Peking Union Medical
College Hospital, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100730, P. R. China
| | - Yuanyang Wang
- Department
of General Surgery, Peking Union Medical
College Hospital, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100730, P. R. China
| | - Hao Wu
- Department
of General Surgery, Peking Union Medical
College Hospital, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100730, P. R. China
| | - Zejian Zhang
- Institute
of Clinical Medicine, State Key Laboratory of Complex Severe and Rare
Diseases, National Infrastructure for Translational Medicine, Peking
Union Medical College Hospital, Chinese
Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P. R. China
| | - Ziwen Liu
- Department
of General Surgery, Peking Union Medical
College Hospital, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100730, P. R. China
| |
Collapse
|
28
|
Chen X, Agustinus AS, Li J, DiBona M, Bakhoum SF. Chromosomal instability as a driver of cancer progression. Nat Rev Genet 2025; 26:31-46. [PMID: 39075192 DOI: 10.1038/s41576-024-00761-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 07/31/2024]
Abstract
Chromosomal instability (CIN) refers to an increased propensity of cells to acquire structural and numerical chromosomal abnormalities during cell division, which contributes to tumour genetic heterogeneity. CIN has long been recognized as a hallmark of cancer, and evidence over the past decade has strongly linked CIN to tumour evolution, metastasis, immune evasion and treatment resistance. Until recently, the mechanisms by which CIN propels cancer progression have remained elusive. Beyond the generation of genomic copy number heterogeneity, recent work has unveiled additional tumour-promoting consequences of abnormal chromosome segregation. These mechanisms include complex chromosomal rearrangements, epigenetic reprogramming and the induction of cancer cell-intrinsic inflammation, emphasizing the multifaceted role of CIN in cancer.
Collapse
Affiliation(s)
- Xuelan Chen
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Albert S Agustinus
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Pharmacology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Jun Li
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Melody DiBona
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samuel F Bakhoum
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
29
|
Mertens F, Hofvander J, Mandahl N, Mitelman F. Aneuploidy in neoplasia: Single-cell data on 83,862 tumors. Int J Cancer 2025; 156:34-39. [PMID: 39222304 DOI: 10.1002/ijc.35163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/05/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Chromosomal aneuploidy, that is, numerical chromosome aberrations, is one of the molecular hallmarks of cancer. However, when neoplasms are studied with sequencing- and array-based approaches, chromosome numbers and ploidy states are typically inferred from bulk DNA data. Furthermore, published molecular estimates of neoplasia-associated aneuploidy often also include genomic imbalances resulting from various types of structural rearrangement, which likely result from other mechanisms than numerical chromosome aberrations. We thus analyzed chromosome numbers using single-cell cytogenetic data from 83,862 tumors, and show that both benign and malignant tumors are highly heterogeneous with regard to deviations from the normal, diploid state. Focusing on the chromosome numbers in 112 specific tumor types, defined by both exact morphologic diagnosis and organ location and from which data from ≥50 cases were available, we found two major clusters: one predominated by near-diploid neoplasms and one by neoplasms with extensive aneuploidy and one or more whole genome doublings. The former cluster included most benign solid tumors, myeloid neoplasms, and malignant gene fusion-associated solid tumors, whereas the latter was predominated by malignant solid tumors and lymphomas. For 16 malignant tumor types, the distribution of chromosome numbers could be compared to TCGA ploidy level data. Cytogenetic and molecular data correlated well, but the former indicates a higher level of clonal heterogeneity. The results presented here suggest shared pathogenetic mechanisms in certain tumor types and provide a reference for molecular analyses.
Collapse
Affiliation(s)
- Fredrik Mertens
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Department of Clinical Genetics, Pathology, and Molecular Diagnostics, Division of Laboratory Medicine, Lund, Sweden
| | - Jakob Hofvander
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Nils Mandahl
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Felix Mitelman
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
30
|
Egawa M, Uno N, Komazaki R, Ohkame Y, Yamazaki K, Yoshimatsu C, Ishizu Y, Okano Y, Miyamoto H, Osaki M, Suzuki T, Hosomichi K, Aizawa Y, Kazuki Y, Tomizuka K. Generation of Monosomy 21q Human iPS Cells by CRISPR/Cas9-Mediated Interstitial Megabase Deletion. Genes Cells 2025; 30:e13184. [PMID: 39581190 DOI: 10.1111/gtc.13184] [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: 09/16/2024] [Revised: 11/05/2024] [Accepted: 11/09/2024] [Indexed: 11/26/2024]
Abstract
Missing an entire chromosome or chromosome arm in normal diploid cells has a deleterious impact on cell viability, which may contribute to the development of specific birth defects. Nevertheless, the effects of chromosome loss in human cells have remained unexplored due to the lack of suitable model systems. Here, we developed an efficient, selection-free approach to generate partial monosomy in human induced pluripotent stem cells (iPSCs). The introduction of Cas9 proteins and a pair of gRNAs induces over megabase-sized interstitial chromosomal deletions. Using human chromosome 21 (HSA21) as a model, partial monosomy 21q (PM21q) iPSC lines with deletions ranging from 4.5 to 27.9 Mb were isolated. A 33.6 Mb deletion, encompassing all protein-coding genes on 21q, was also achieved, establishing the first 21q monosomy human iPSC line. Transcriptome and proteome analyses revealed that the abundances of mRNA and protein encoded by the majority of genes in the monosomic regions are half of the diploid expression level, indicating an absence of dosage compensation. The ability to generate customized partial monosomy cell lines on an isogenic, karyotypically normal background should facilitate the gain of novel insights into the impact of chromosome loss on cellular fitness.
Collapse
Affiliation(s)
- Masaya Egawa
- Laboratory of Bioengineering, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Narumi Uno
- Laboratory of Bioengineering, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Rina Komazaki
- Laboratory of Bioengineering, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yusuke Ohkame
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan
| | - Kyotaro Yamazaki
- Chromosome Engineering Research Group, the Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan
- Department of Homeostatic Regulation, National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences (NINS), Okazaki, Japan
| | - Chihiro Yoshimatsu
- Department of Chromosome Biomedical Engineering, Integrated Medical Sciences, Graduate School of Medical Sciences, Tottori University, Tottori, Japan
| | - Yuki Ishizu
- Laboratory of Bioengineering, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yusaku Okano
- Laboratory of Bioengineering, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Hitomaru Miyamoto
- Department of Chromosome Biomedical Engineering, Integrated Medical Sciences, Graduate School of Medical Sciences, Tottori University, Tottori, Japan
| | - Mitsuhiko Osaki
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan
- Division of Experimental Pathology, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Teruhiko Suzuki
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kazuyoshi Hosomichi
- Laboratory of Computational Genomics, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yasunori Aizawa
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
- Kanagawa Institute of Industrial Science and Technology, Ebina, Japan
| | - Yasuhiro Kazuki
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan
- Chromosome Engineering Research Group, the Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan
- Department of Chromosome Biomedical Engineering, Integrated Medical Sciences, Graduate School of Medical Sciences, Tottori University, Tottori, Japan
| | - Kazuma Tomizuka
- Laboratory of Bioengineering, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| |
Collapse
|
31
|
Wang J, Zhou W, Xu Y, Duan J, Zhou Q, Wang G, Li L, Xu C, Wang W, Cai S, Wang Z, Wang J. Antithetical impacts of deleterious LRP1B mutations in non-squamous and squamous NSCLCs on predicting benefits from immune checkpoint inhibitor alone or with chemotherapy over chemotherapy alone: retrospective analyses of the POPLAR/OAK and CHOICE-01 trials. SCIENCE CHINA. LIFE SCIENCES 2025; 68:249-262. [PMID: 39276256 DOI: 10.1007/s11427-023-2554-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 01/18/2024] [Indexed: 09/16/2024]
Abstract
In non-small cell lung cancers, the non-squamous and squamous subtypes (nsqNSCLC and sqNSCLC) exhibit disparities in pathophysiology, tumor immunology, and potential genomic correlates affecting responses to immune checkpoint inhibitor (ICI)-based treatments. In our in-house training cohort (n=85), the presence of the LRP1B deleterious mutation (LRP1B-del) was associated with longer and shorter progression-free survival (PFS) on ICIs alone in nsqNSCLCs and sqNSCLCs, respectively (Pinteraction=0.008). These results were validated using a larger public ICI cohort (n=208, Pinteraction<0.001). Multiplex immunofluorescence staining revealed an association between LRP1B-del and increased and decreased numbers of tumor-infiltrating CD8+ T cells in nsqNSCLCs (P=0.040) and sqNSCLCs (P=0.014), respectively. In the POPLAR/OAK cohort, nsqNSCLCs with LRP1B-del demonstrated improved PFS benefits from atezolizumab over docetaxel (hazard ratio (HR) =0.70, P=0.046), whereas this benefit was negligible in those without LRP1B-del (HR=1.05, P=0.64). Conversely, sqNSCLCs without LRP1B-del benefited more from atezolizumab (HR=0.60, P=0.002) than those with LRP1B-del (HR=1.30, P=0.31). Consistent results were observed in the in-house CHOICE-01 cohort, in which nsqNSCLCs with LRP1B-del and sqNSCLCs without LRP1B-del benefited more from toripalimab plus chemotherapy than from chemotherapy alone (Pinteraction=0.008). This multi-cohort study delineates the antithetical impacts of LRP1B-del in nsqNSCLCs and sqNSCLCs on predicting the benefits from ICI alone or with chemotherapy over chemotherapy alone. Our findings highlight the distinct clinical utility of LRP1B-del in guiding treatment choices for nsqNSCLCs and sqNSCLCs, emphasizing the necessity for a detailed analysis based on pathological subtypes when investigating biomarkers for cancer therapeutics.
Collapse
Affiliation(s)
- Jinliang Wang
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Wenyong Zhou
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Yu Xu
- Burning Rock Biotech, Guangzhou, 510300, China
| | - Jianchun Duan
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | | | | | - Leo Li
- Burning Rock Biotech, Guangzhou, 510300, China
| | - Chunwei Xu
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310000, China
| | - Wenxian Wang
- Department of Clinical Trial, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, 310022, China
| | - Shangli Cai
- Burning Rock Biotech, Guangzhou, 510300, China.
| | - Zhijie Wang
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie Wang
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
32
|
Abbate JM, Mangraviti D, Brunetti B, Cafarella C, Rigano F, Iaria C, Marino F, Mondello L. Machine learning approach in canine mammary tumour classification using rapid evaporative ionization mass spectrometry. Anal Bioanal Chem 2025; 417:373-388. [PMID: 39562368 DOI: 10.1007/s00216-024-05656-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024]
Abstract
Rapid evaporative ionization mass spectrometry (REIMS) coupled with a monopolar handpiece used for surgical resection and combined with chemometrics has been previously explored by our research group (Mangraviti et al. in Int J Mol Sci 23(18):10562, 2022) to identify several mammary gland pathologies. Here, the increased sample size allowed the construction of three statistical models to distinguish between benign and malignant canine mammary tumours (CMTs), facilitating a more in-depth investigation of changes in cellular metabolic phenotype during neoplastic transformation and biological behaviour. The results demonstrate that REIMS is effective in identifying neoplastic tissues with an accuracy of 97%, with differences in MS spectra characterized by the relative abundance of phospholipids compared to triglycerides more commonly identified in normal mammary glands. The increased rate of phospholipid synthesis represents an informative feature for tumour recognition, with phosphatidylcholine and phosphatidylethanolamine, the two major phospholipid species identified here together with sphingolipids, playing a crucial role in carcinogenesis. REIMS technology allowed the classification of different histotypes of benign CMTs with an accuracy score of 95%, distinguishing them from normal glands based on the increase in sphingolipids, glycolipids, phospholipids, and arachidonic acid, demonstrating the close association between cancer and inflammation. Finally, dysregulation of fatty acid metabolism with increased signalling for saturated, mono- and polyunsaturated fatty acids characterized the metabolic phenotype of neoplastic cells and their malignant transformation, supporting the increased formation of new organelles for cell division. Further investigations on a more significant number of tumour histotypes will allow for the creation of a more extensive database and lay the basis for how understanding metabolic alterations in the tumour microenvironment can improve surgical precision.
Collapse
Affiliation(s)
- Jessica Maria Abbate
- Department of Veterinary Sciences, University of Messina, Polo Universitario Annunziata, 98168, Messina, Italy
| | - Domenica Mangraviti
- Messina Institute of Technology c/o Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, Former Veterinary School, University of Messina, Viale G. Palatucci snc, 98168, Messina, Italy.
| | - Barbara Brunetti
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, via Tolara di Sopra 50, 40064, Ozzano Emilia, BO, Italy
| | - Cinzia Cafarella
- Messina Institute of Technology c/o Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, Former Veterinary School, University of Messina, Viale G. Palatucci snc, 98168, Messina, Italy
| | - Francesca Rigano
- Messina Institute of Technology c/o Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, Former Veterinary School, University of Messina, Viale G. Palatucci snc, 98168, Messina, Italy
| | - Carmelo Iaria
- Institute for Comparative, Experimental, Forensic and Aquatic Pathology (ICEFAP) "Slavko Bambir", Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno D'Alcontres 31, 98166, Messina, Italy
| | - Fabio Marino
- Institute for Comparative, Experimental, Forensic and Aquatic Pathology (ICEFAP) "Slavko Bambir", Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno D'Alcontres 31, 98166, Messina, Italy
| | - Luigi Mondello
- Messina Institute of Technology c/o Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, Former Veterinary School, University of Messina, Viale G. Palatucci snc, 98168, Messina, Italy
- Chromaleont s.r.l., c/o Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, Former Veterinary School, University of Messina, Viale G. Palatucci snc, 98168, Messina, Italy
| |
Collapse
|
33
|
Liu Y, Carbonetto P, Willwerscheid J, Oakes SA, Macleod KF, Stephens M. Dissecting tumor transcriptional heterogeneity from single-cell RNA-seq data by generalized binary covariance decomposition. Nat Genet 2025; 57:263-273. [PMID: 39747597 DOI: 10.1038/s41588-024-01997-z] [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: 08/25/2023] [Accepted: 10/18/2024] [Indexed: 01/04/2025]
Abstract
Profiling tumors with single-cell RNA sequencing has the potential to identify recurrent patterns of transcription variation related to cancer progression, and to produce therapeutically relevant insights. However, strong intertumor heterogeneity can obscure more subtle patterns that are shared across tumors. Here we introduce a statistical method, generalized binary covariance decomposition (GBCD), to address this problem. We show that GBCD can decompose transcriptional heterogeneity into interpretable components-including patient-specific, dataset-specific and shared components relevant to disease subtypes-and that, in the presence of strong intertumor heterogeneity, it can produce more interpretable results than existing methods. Applied to data on pancreatic ductal adenocarcinoma, GBCD produced a refined characterization of existing tumor subtypes, and identified a gene expression program prognostic of poor survival independent of tumor stage and subtype. The gene expression program is enriched for genes involved in stress responses, and suggests a role for the integrated stress response in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Yusha Liu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Peter Carbonetto
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Jason Willwerscheid
- Department of Mathematics and Computer Science, Providence College, Providence, RI, USA
| | - Scott A Oakes
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Kay F Macleod
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Matthew Stephens
- Departments of Statistics and Human Genetics, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
34
|
Zhu S, Chen C, Wang M, Liu Y, Li B, Qi X, Song M, Liu X, Feng J, Liu J. Pan-cancer association of a mitochondrial function score with genomic alterations and clinical outcome. Sci Rep 2024; 14:31430. [PMID: 39733076 PMCID: PMC11682264 DOI: 10.1038/s41598-024-83022-1] [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: 09/18/2024] [Accepted: 12/11/2024] [Indexed: 12/30/2024] Open
Abstract
Mitochondria are pivotal in cellular energy metabolism and have garnered significant attention for their roles in cancer progression and therapy resistance. Despite this, the functional diversity of mitochondria across various cancer types remains inadequately characterized. This study seeks to fill this knowledge gap by introducing and validating MitoScore-a novel metric designed to quantitatively assess mitochondrial function across a wide array of cancers. Our investigation evaluates the capacity of MitoScore not only to distinguish between tumor and adjacent normal tissues but also to serve as a predictive marker for clinical outcomes. We analyzed gene expression data from 24 cancer types and corresponding normal tissues using the TCGA database. MitoScore was calculated by summing the normalized expression levels of six mitochondrial genes known to be consistently altered across multiple cancers. Differential gene expression was assessed using DESeq2, with a focus on identifying significant changes in mitochondrial function. MitoScore's associations with tumor proliferation, hypoxia, aneuploidy, and clinical outcomes were evaluated using Spearman's correlation, linear regression, and Kaplan-Meier survival analyses. MitoScore was significantly higher in tumor tissues compared to normal tissues across most cancer types (p < 0.001). It positively correlated with tumor proliferation rates (r = 0.46), hypoxia scores (r = 0.61), and aneuploidy (r = 0.44), indicating its potential as a marker of aggressive tumor behavior. High MitoScore was also associated with poorer prognosis in several cancer types, suggesting its utility as a predictive biomarker for clinical outcomes. This study introduces MitoScore, a metric for mitochondrial activity often elevated in tumors and linked to poor prognosis. It correlates positively with hypoxia and negatively with stromal and immune infiltration, highlighting mitochondria's role in the tumor microenvironment. MitoScore's association with genomic instability, such as aneuploidy, suggests mitochondrial dysfunction contributes to cancer progression. Despite challenges in mitochondrial-targeted therapies, MitoScore may identify tumors responsive to such treatments, warranting further research for clinical application.
Collapse
Affiliation(s)
- Shikun Zhu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Chen Chen
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Min Wang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Yue Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Baolin Li
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Xing Qi
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
- Ziyang People's Hospital, Ziyang, Sichaun, China
| | - Miao Song
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Xuexue Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China
| | - Jia Feng
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China.
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, China.
| |
Collapse
|
35
|
Mitsiades IR, Onozato M, Iafrate AJ, Hicks D, Gülhan DC, Sgroi DC, Rheinbay E. ERBB2/HOXB13 co-amplification with interstitial loss of BRCA1 defines a unique subset of breast cancers. Breast Cancer Res 2024; 26:185. [PMID: 39695741 DOI: 10.1186/s13058-024-01943-1] [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: 08/08/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The HOXB13/IL17RB gene expression biomarker has been shown to predict response to adjuvant and extended endocrine therapy in patients with early-stage ER+ HER2- breast tumors. HOXB13 gene expression is the primary determinant driving the prognostic and endocrine treatment-predictive performance of the biomarker. Currently, there is limited data on HOXB13 expression in HER2+ and ER- breast cancers. Herein, we studied the expression of HOXB13 in large cohorts of HER2+ and ER- breast cancers. METHODS We investigated gene expression, genomic copy number, mutational signatures, and clinical outcome data in the TGGA and METABRIC breast cancer cohorts. Genomic-based gene amplification data was validated with tri-colored fluorescence in situ hybridization. RESULTS In the TCGA breast cancer cohort, HOXB13 gene expression was significantly higher in HER2+ versus HER2- breast cancers, and its expression was also significantly higher in the ER- versus ER+ breast cancers. HOXB13 is frequently co-gained or co-amplified with ERBB2. Joint copy gains of HOXB13 and ERBB2 occurred with low-level co-gains or high-level co-amplifications (co-amp), the latter of which is associated with an interstitial loss that includes the tumor suppressor BRCA1. ERBB2/HOXB13 co-amp tumors with interstitial BRCA1 loss exhibit a mutational signature associated with APOBEC deaminase activity and copy number signatures associated with chromothripsis and genomic instability. Among ERBB2-amplified tumors of different tissue origins, ERBB2/HOXB13 co-amp with a BRCA1 loss appeared to be enriched in breast cancer compared to other tumor types. Lastly, patients with ERBB2/HOXB13 co-amplified and BRCA1 lost tumors displayed a significantly shorter progression-free survival (PFS) than those with ERBB2-only amplifications. The difference in PFS was restricted to the ER- subset patients and this difference in PFS was not solely driven by HOXB13 gene expression. CONCLUSIONS HOXB13 is frequently co-gained with ERBB2 at both low-copy number level or as complex high-level amplification with relative BRCA1 loss. ERBB2/HOXB13 amplified, BRCA1-lost tumors are strongly enriched in breast cancer, and patients with such breast tumors experience a shortened PFS.
Collapse
Affiliation(s)
- Irene Rin Mitsiades
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, 02129, USA
| | - Maristela Onozato
- Vertex Pharmaceuticals, Preclinical Safety Assessment, Pathology, 316 Northern Ave, Boston, MA, 02210, USA
| | - A John Iafrate
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, 02129, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Daniel Hicks
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, 02129, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Doğa C Gülhan
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, 02129, USA
- The Broad Institute or MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Dennis C Sgroi
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, 02129, USA.
- Department of Pathology, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| | - Esther Rheinbay
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, 02129, USA.
- The Broad Institute or MIT and Harvard, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
36
|
Eltohami Y, Eltayeb O, Suleiman A. Oral squamous cell carcinoma in the Sudan: clinical behavior, DNA ploidy and S-phase fraction characteristics. BMC Oral Health 2024; 24:1473. [PMID: 39633352 PMCID: PMC11619201 DOI: 10.1186/s12903-024-05270-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] [Received: 03/03/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND The DNA ploidy and the S phase fraction (SPF) are associated with advanced tumors progression. The present study aimed to investigate the clinicopathological behavior of oral squamous cell carcinoma(OSCC) and the prognostication of the DNA ploidy and the SPF on its clinical outcomes. METHODS This a prospective longitudinal study of 93 OSCC patients (186 specimens). Using high resolution flowcytometry, DNA analysis and SPF were performed for the primary tumor, field of cancerization and a control normal field. The patients were followed clinically for the development of recurrence or second primary tumor(SPT). RESULTS All the 93 patients are associated with wide field of cancerization. Out of the total number of cases, 28 (30%) patients developed recurrence. Of them 17 (18.3%) had regional recurrence, and eight (8.6%) had local recurrence. Second primary tumors were identified in 14 (15.1%) of the cases. Nodal metastasis, overall recurrence and SPTs were significantly associated with DNA aneuploidy and high SPF. High SPF and DNA hyperploidy showed a 28% increased risk of local recurrence (P.value = 0.032). CONCLUSION In a series of 93 oral cancers who had wide field of cancerization, 30% of the patients developed recurrence, and 15% developed second primary tumors. The DNA ploidy and the SPF were independent prognosticators for the prediction of OSCC outcomes.
Collapse
Affiliation(s)
- Yousif Eltohami
- Oral and maxillofacial surgery, Faculty of Dentistry, University of Khartoum, Khartoum, Sudan.
| | - Osama Eltayeb
- Flowcytometry Center and Sudan University of Science & Technology, Khartoum, Sudan
| | - Ahmed Suleiman
- Oral and maxillofacial surgery, Faculty of Dentistry, University of Khartoum, Khartoum, Sudan
| |
Collapse
|
37
|
Sturgill I, Raab J, Hoadley K. Expanded detection and impact of BAP1 alterations in cancer. NAR Cancer 2024; 6:zcae045. [PMID: 39554490 PMCID: PMC11567159 DOI: 10.1093/narcan/zcae045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/15/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024] Open
Abstract
Aberrant expression of the BAP1 (BRCA associated protein 1) tumor suppressor gene is a prominent risk factor for several tumor types and is important in tumor evolution and progression. Here we performed integrated multi-omics analyses using data from The Cancer Genome Atlas for 33 cancer types and over 10 000 individuals to identify alterations leading to BAP1 disruption. We combined existing variant calls and new calls derived from a de novo local realignment pipeline across multiple independent variant callers, increasing somatic variant detection by 41% from 182 to 257, including 11 indels ≥40 bp. The expanded detection of mutations highlights the power of new tools to uncover longer indels and impactful mutations. We developed an expression-based BAP1 activity score and identified a transcriptional profile associated with BAP1 disruption in cancer. BAP1 has been proposed to play a critical role in controlling tumor plasticity and normal cell fate. Leveraging human and mouse liver datasets, BAP1 loss in normal cells resulted in lower BAP1 activity scores and lower scores were associated with a less-differentiated phenotype in embryonic cells. Together, our expanded BAP1 mutant samples revealed a transcriptional signature in cancer cells, supporting BAP1's influences on cellular plasticity and cell identity maintenance.
Collapse
Affiliation(s)
- Ian R Sturgill
- Bioinformatics and Computational Biology Curriculum, Department of Genetics, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599, USA
| | - Jesse R Raab
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599, USA
| | - Katherine A Hoadley
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599, USA
| |
Collapse
|
38
|
Wang J, Du J, Luo X, Guo L, Liu Y, Zhou J, Zou Y, Lu Z, Pan X, Chen X, Zhong A, Wan X, Wang L, Liu H, Dai S, Zhang S, Xiong X, Tan P, Wang M, Wu B, Zhang Q, Wang Y, Zhang M, Lu R, Lin H, Li Y, Li Y, Han Z, Chen L, Hu B, Liu Y, Na F, Chen C. A platform of functional studies of ESCC-associated gene mutations identifies the roles of TGFBR2 in ESCC progression and metastasis. Cell Rep 2024; 43:114952. [PMID: 39527477 DOI: 10.1016/j.celrep.2024.114952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/31/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Genomics studies have detected numerous genetic alterations in esophageal squamous cell carcinoma (ESCC). However, the functions of these mutations largely remain elusive, partially due to a lack of feasible animal models. Here, we report a convenient platform with CRISPR-Cas9-mediated introduction of genetic alterations and orthotopic transplantation to generate a series of primary ESCC models in mice. With this platform, we validate multiple frequently mutated genes, including EP300, FAT1/2/4, KMT2D, NOTCH2, and TGFBR2, as tumor-suppressor genes in ESCC. Among them, TGFBR2 loss dramatically promotes tumorigenesis and multi-organ metastasis. Paradoxically, TGFBR2 deficiency leads to Smad3 activation, and disruption of Smad3 partially restrains the progression of Tgfbr2-mutated tumors. Drug screening with tumor organoids identifies that pinaverium bromide represses Smad3 activity and restrains Tgfbr2-deficient ESCC. Our studies provide a highly efficient platform to investigate the in vivo functions of ESCC-associated mutations and develop potential treatments for this miserable malignancy.
Collapse
Affiliation(s)
- Jian Wang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiajia Du
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiangmeng Luo
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Linjie Guo
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yixin Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianfeng Zhou
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yang Zou
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenghao Lu
- Chengdu OrganoidMed Medical Laboratory, West China Health Valley, Chengdu, Sichuan 610041, China
| | - Xiangyu Pan
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xuelan Chen
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ailing Zhong
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xudong Wan
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lu Wang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongyu Liu
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Siqi Dai
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shiyu Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xingyu Xiong
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ping Tan
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Manli Wang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Baohong Wu
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qi Zhang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yingjie Wang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Mengsha Zhang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Runda Lu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Huahang Lin
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Li
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yaxin Li
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zongkai Han
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Longqi Chen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Bing Hu
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Yu Liu
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Feifei Na
- Department of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Chong Chen
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, No387-201 Hemin st., Chengdu, Sichuan 610212, China; Children's Medicine Key Laboratory of Sichuan Province, Sichuan 610041, China.
| |
Collapse
|
39
|
Li Q, Li Z, Chen B, Zhao J, Yu H, Hu J, Lai H, Zhang H, Li Y, Meng Z, Hu Z, Huang S. RNA splicing junction landscape reveals abundant tumor-specific transcripts in human cancer. Cell Rep 2024; 43:114893. [PMID: 39446586 DOI: 10.1016/j.celrep.2024.114893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/08/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024] Open
Abstract
RNA splicing is a critical process governing gene expression and transcriptomic diversity. Despite its importance, a detailed examination of transcript variation at the splicing junction level remains scarce. Here, we perform a thorough analysis of RNA splicing junctions in 34,775 samples across multiple sample types. We identified 29,051 tumor-specific transcripts (TSTs) in pan-cancer, with a majority of these TSTs being unannotated. Our findings show that TSTs are positively correlated with tumor stemness and linked to unfavorable outcomes in cancer patients. Additionally, TSTs display mutual exclusivity with somatic mutations and are overrepresented in transposable-element-derived transcripts possessing oncogenic functions. Importantly, TSTs can generate putative neoantigens for immunotherapy. Moreover, TSTs can be detected in blood extracellular vesicles from cancer patients. Our results shed light on the intricacies of RNA splicing and offer promising avenues for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Qin Li
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, and Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Ziteng Li
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Bing Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jingjing Zhao
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Hongwu Yu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jia Hu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Hongyan Lai
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Hena Zhang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yan Li
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhiqiang Meng
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Zhixiang Hu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Shenglin Huang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
40
|
van der Sluis K, van Sandick JW, Koemans WJ, van den Bosch T, Broeks A, Peters D, Seignette IM, Rausch CR, van Dijk E, Snaebjornsson P, van den Berg JG, van Grieken NCT, Ylstra B, Carvalho B, Miedema DM, Kodach LL. Karyotype evolution in response to chemoradiotherapy and upon recurrence of esophageal adenocarcinomas. Cell Rep 2024; 43:114981. [PMID: 39535918 DOI: 10.1016/j.celrep.2024.114981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 09/06/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The genome of esophageal adenocarcinoma (EAC) is highly unstable and might evolve over time. Here, we track karyotype evolution in EACs in response to treatment and upon recurrence through multi-region and longitudinal analysis. To this end, we introduce L-PAC (low-purity inference of absolute copy-number alterations [CNAs]), a bio-informatics technique that allows inference of absolute CNAs of low-purity samples by leveraging the information of high-purity samples from the same cancer. Quantitative analysis of matched absolute CNAs reveals that the amount of karyotype evolution induced by chemoradiotherapy (CRT) is predictive for early recurrence and depends on the initial level of karyotype intra-tumor heterogeneity. We observe that CNAs acquired in response to CRT are partially reversed back to the initial state upon recurrence. Hence, CRT alters the fitness landscape to which tumors can adjust by adapting their karyotype. Together, our results indicate that karyotype plasticity contributes to the therapy resistance of EACs.
Collapse
Affiliation(s)
- Karen van der Sluis
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Johanna W van Sandick
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Willem J Koemans
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Tom van den Bosch
- Amsterdam UMC Location University of Amsterdam, Cancer Center Amsterdam & Amsterdam Gastroenterology Endocrinology Metabolism, Center for Experimental and Molecular Medicine, Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Dennis Peters
- Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Iris M Seignette
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Christian R Rausch
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Erik van Dijk
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Petur Snaebjornsson
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - José G van den Berg
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Nicole C T van Grieken
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Bauke Ylstra
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Beatriz Carvalho
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Daniël M Miedema
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Liudmila L Kodach
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
41
|
Jemma A, Ardizzoia A, Redaelli S, Bentivegna A, Lavitrano M, Conconi D. Prognostic Relevance of Copy Number Losses in Ovarian Cancer. Genes (Basel) 2024; 15:1487. [PMID: 39596687 PMCID: PMC11593593 DOI: 10.3390/genes15111487] [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: 10/03/2024] [Revised: 10/29/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND/OBJECTIVES Aneuploidy is a prevalent cancer feature that occurs in many solid tumors. For example, high-grade serous ovarian cancer shows a high level of copy number alterations and genomic rearrangements. This makes genomic variants appealing as diagnostic or prognostic biomarkers, as well as for their easy detection. In this study, we focused on copy number (CN) losses shared by ovarian cancer stem cells (CSCs) to identify chromosomal regions that may be important for CSC features and, in turn, for patients' prognosis. METHODS Array-CGH and bioinformatic analyses on three CSCs subpopulations were performed. RESULTS Pathway and gene ontology analyses on genes involved in copy number loss in all CSCs revealed a significant decrease in mRNA surveillance pathway, as well as miRNA-mediated gene silencing. Then, starting from these CN losses, we validated their potential prognostic relevance by analyzing the TCGA cohort. Notably, losses of 4q34.3-q35.2, 8p21.2-p21.1, and 18q12.2-q23 were linked to increased genomic instability. Loss of 18q12.2-q23 was also related to a higher tumor stage and poor prognosis. Finally, specific genes mapping in these regions, such as PPP2R2A and TPGS2A, emerged as potential biomarkers. CONCLUSIONS Our findings highlight the importance of genomic alterations in ovarian cancer and their impact on tumor progression and patients' prognosis, offering advance in understanding of the application of numerical aberrations as prognostic ovarian cancer biomarkers.
Collapse
Affiliation(s)
- Andrea Jemma
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (A.J.); (A.A.); (S.R.); (A.B.); (M.L.)
| | - Alessandra Ardizzoia
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (A.J.); (A.A.); (S.R.); (A.B.); (M.L.)
- Fondazione Istituto di Oncologia Molecolare ETS (IFOM), The AIRC Institute for Molecular Oncology, 20139 Milan, Italy
| | - Serena Redaelli
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (A.J.); (A.A.); (S.R.); (A.B.); (M.L.)
| | - Angela Bentivegna
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (A.J.); (A.A.); (S.R.); (A.B.); (M.L.)
| | - Marialuisa Lavitrano
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (A.J.); (A.A.); (S.R.); (A.B.); (M.L.)
| | - Donatella Conconi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (A.J.); (A.A.); (S.R.); (A.B.); (M.L.)
| |
Collapse
|
42
|
Xie X, Xi X, Zhao D, Zhao Y, Yi T, Chen D, Liu R, Qi L, Pan Z, Wang H, Zhang H, Ding R, Du H. Advancing pathogen and tumor copy number variation detection through simultaneous metagenomic next-generation sequencing: A comprehensive review. Heliyon 2024; 10:e38826. [PMID: 39568836 PMCID: PMC11577201 DOI: 10.1016/j.heliyon.2024.e38826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 11/22/2024] Open
Abstract
In clinical practice, timely and accurate diagnosis can effectively reduce unnecessary treatment, avoid high medical costs, and prevent adverse prognoses. However, some patients with malignant tumors and those with infection often exhibit similar symptoms, which are difficult to distinguish, posing challenges in accurate clinical diagnosis. Metagenomic next-generation sequencing (mNGS) technology has been widely applied to confirm the source of infection. Recent studies have shown that for pathogen detection, mNGS technology can be used to perform chromosomal copy number variations (CNVs) analysis in two different analytical pipelines using the same wet test. mNGS technology has further demonstrated its utility in not only the determination of pathogenic microorganisms but also of CNVs, thereby facilitating early differential diagnosis for malignant tumors. In this review, we aim to analyze the diagnostic performance of mNGS technology in the simultaneous detection of pathogenic microorganisms and CNVs in current clinical practice and discuss the advantages and limitations of mNGS-CNV dual-omics detection technology. Our review highlights the need for more large-scale prospective research data on current mNGS-CNV dual-omics detection technology to provide more evidence-based results for researchers and clinicians and to promote the greater role of this technology in future clinical practice.
Collapse
Affiliation(s)
- Xiaofang Xie
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College, Soochow University, China
| | - Xiaotong Xi
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
- Cancer Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Dan Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, China
| | - Yingyue Zhao
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
- Cancer Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Tiantian Yi
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, China
| | - Dongsheng Chen
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
- Cancer Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Rui Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, China
| | - Lin Qi
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, China
| | - Zhen Pan
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, China
| | - Hongqiu Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, China
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, China
| | - Ran Ding
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
- Cancer Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Hong Du
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, China
| |
Collapse
|
43
|
Hintzen DC, Schubert M, Soto M, Medema RH, Raaijmakers JA. Reduction of chromosomal instability and inflammation is a common aspect of adaptation to aneuploidy. EMBO Rep 2024; 25:5169-5193. [PMID: 39294502 PMCID: PMC11549362 DOI: 10.1038/s44319-024-00252-0] [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: 03/06/2024] [Revised: 08/20/2024] [Accepted: 08/28/2024] [Indexed: 09/20/2024] Open
Abstract
Aneuploidy, while detrimental to untransformed cells, is notably prevalent in cancer. Aneuploidy is found as an early event during tumorigenesis which indicates that cancer cells have the ability to surmount the initial stress responses associated with aneuploidy, enabling rapid proliferation despite aberrant karyotypes. To generate more insight into key cellular processes and requirements underlying adaptation to aneuploidy, we generated a panel of aneuploid clones in p53-deficient RPE-1 cells and studied their behavior over time. As expected, de novo-generated aneuploid clones initially display reduced fitness, enhanced levels of chromosomal instability (CIN), and an upregulated inflammatory response. Intriguingly, after prolonged culturing, aneuploid clones exhibit increased proliferation rates while maintaining aberrant karyotypes, indicative of an adaptive response to the aneuploid state. Interestingly, all adapted clones display reduced CIN and reduced inflammatory signaling, suggesting that these are common aspects of adaptation to aneuploidy. Collectively, our data suggests that CIN and concomitant inflammation are key processes that require correction to allow for fast proliferation in vitro. Finally, we provide evidence that amplification of oncogenic KRAS can promote adaptation.
Collapse
Affiliation(s)
- Dorine C Hintzen
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Michael Schubert
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Mar Soto
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - René H Medema
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
- Oncode Institute, Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands.
| | - Jonne A Raaijmakers
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
44
|
Ghosh MK, Kumar S, Begam S, Ghosh S, Basu M. GBM immunotherapy: Exploring molecular and clinical frontiers. Life Sci 2024; 356:123018. [PMID: 39214286 DOI: 10.1016/j.lfs.2024.123018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/21/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
GBM is the most common, aggressive, and intracranial primary brain tumor; it originates from the glial progenitor cells, has poor overall survival (OS), and has limited treatment options. In this decade, GBM immunotherapy is in trend and preferred over several conventional therapies, due to their better patient survival outcome. This review explores the clinical trials of several immunotherapeutic approaches (immune checkpoint blockers (ICBs), CAR T-cell therapy, cancer vaccines, and adoptive cell therapy) with their efficacy and safety. Despite significant progress, several challenges (viz., immunosuppressive microenvironment, heterogeneity, and blood-brain barrier (BBB)) were experienced that hamper their immunotherapeutic potential. Furthermore, these challenges were clinically studied to be resolved by multiple combinatorial approaches, discussed in the later part of the review. Thus, this review suggests the clinical use and potential of immunotherapy in GBM and provides the holistic recent knowledge and future perspectives.
Collapse
Affiliation(s)
- Mrinal K Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata 700091, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India.
| | - Sunny Kumar
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata 700091, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Sabana Begam
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata 700091, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Sayani Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata 700091, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Malini Basu
- Department of Microbiology, Dhruba Chand Halder College, Dakshin Barasat, South 24 Parganas, PIN-743372, India
| |
Collapse
|
45
|
Kumari L, Sreedharanunni S, Dahiya D, Dey P, Bhatia A. High prevalence of chromosome 17 in breast cancer micronuclei: a means to get rid of tumor suppressors? Hum Cell 2024; 38:5. [PMID: 39438374 DOI: 10.1007/s13577-024-01143-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Micronuclei (MN), defined as small extra-nuclear chromatin bodies enclosed by a nuclear envelope, serve as noticeable markers of chromosomal instability (CIN). The MN have been used for breast cancer (BC) screening, diagnosis, and prognosis. However, more recently they have gained attention as seats for active chromosomal rearrangements. BC subtypes exhibit differential CIN levels and aggressiveness. This study aimed to investigate MN chromosomal contents across BC subtypes, exploring its potential role in aggressiveness and pathogenesis. Immunostaining of BC cells was performed with anti-centromeric antibody followed by confocal microscopy. Further, fluorescence in situ hybridization (FISH) was done to check the presence of specific chromosomes in the MN. The real time PCR was also done from the RNA isolated from MN to check the expression of TP53 gene. BC cell lines (CLs) showed the presence of both centromere-positive ( +) and -negative ( -) MN, with significant variation in frequency among hormone and human epidermal growth factor receptor positive and triple-negative (TN) BC cells. FISH targeting chromosomes 1, 3, 8, 11, and 17 detected centromeric signals for all the above chromosomes in MN with a relatively higher prevalence of chromosome 17 in all the CLs. Out of all the CLs, TNBC cells demonstrated the highest frequency of centromere + and chromosome 17 + MN. TP53 expression could also be demonstrated inside the MN by FISH and real time PCR. Patient sample imprints also confirmed the presence of chromosome 17 in MN with polysomy of the same in corresponding nuclei. The high prevalence of chromosome 17 in BC MN may connote the importance of its rearrangements in the pathogenesis of BC. Further, the higher prevalence of chromosome 17 and 1 signals in TNBC MN point towards the significance of pathogenetic events involving the genes located in these chromosomes in evolution of this more aggressive phenotype.
Collapse
Affiliation(s)
- Laxmi Kumari
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sreejesh Sreedharanunni
- Department of Haematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Divya Dahiya
- Department of General Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pranab Dey
- Department of Cytology and Gynaecological Pathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
46
|
Houlahan KE, Bihie M, Contreras JG, Fulop DJ, Lopez G, Huang HH, Van Loo P, Curtis C, Boutros PC, Huang KL. Deletions Rate-Limit Breast and Ovarian Cancer Initiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618945. [PMID: 39484366 PMCID: PMC11526986 DOI: 10.1101/2024.10.17.618945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Optimizing prevention and early detection of cancer requires understanding the number, types and timing of driver mutations. To quantify this, we exploited the elevated cancer incidence and mutation rates in germline BRCA1 and BRCA2 (gBRCA1/2) carriers. Using novel statistical models, we identify genomic deletions as the likely rate-limiting mutational processes, with 1-3 deletions required to initiate breast and ovarian tumors. gBRCA1/2-driven hereditary and sporadic tumors undergo convergent evolution to develop a similar set of driver deletions, and deletions explain the elevated cancer risk of gBRCA1/2-carriers. Orthogonal mutation timing analysis identifies deletions of chromosome 17 and 13q as early, recurrent events. Single-cell analyses confirmed deletion rate differences in gBRCA1/2 vs. non-carrier tumors as well as cells engineered to harbor gBRCA1/2. The centrality of deletion-associated chromosomal instability to tumorigenesis shapes interpretation of the somatic evolution of non-malignant tissue and guides strategies for precision prevention and early detection.
Collapse
Affiliation(s)
- Kathleen E. Houlahan
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USATable
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, CA, USA
- Vector Institute, Toronto, Canada
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | - Daniel J. Fulop
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gonzalo Lopez
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hsin-Hsiung Huang
- Department of Statistics, University of Central Florida, Orlando, FL, USA
| | - Peter Van Loo
- The Francis Crick Institute, London, UK
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christina Curtis
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Paul C. Boutros
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USATable
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, CA, USA
- Vector Institute, Toronto, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
- Department of Urology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kuan-lin Huang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
47
|
Muijlwijk T, Nauta IH, van der Lee A, Grünewald KJT, Brink A, Ganzevles SH, Baatenburg de Jong RJ, Atanesyan L, Savola S, van de Wiel MA, Peferoen LAN, Bloemena E, van de Ven R, Leemans CR, Poell JB, Brakenhoff RH. Hallmarks of a genomically distinct subclass of head and neck cancer. Nat Commun 2024; 15:9060. [PMID: 39428388 PMCID: PMC11491468 DOI: 10.1038/s41467-024-53390-3] [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: 02/07/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024] Open
Abstract
Cancer is caused by an accumulation of somatic mutations and copy number alterations (CNAs). Besides mutations, these copy number changes are key characteristics of cancer development. Nonetheless, some tumors show hardly any CNAs, a remarkable phenomenon in oncogenesis. Head and neck squamous cell carcinomas (HNSCCs) arise by either exposure to carcinogens, or infection with the human papillomavirus (HPV). HPV-negative HNSCCs are generally characterized by many CNAs and frequent mutations in CDKN2A, TP53, FAT1, and NOTCH1. Here, we present the hallmarks of the distinct subgroup of HPV-negative HNSCC with no or few CNAs (CNA-quiet) by genetic profiling of 802 oral cavity squamous cell carcinomas (OCSCCs). In total, 73 OCSCC (9.1%) are classified as CNA-quiet and 729 as CNA-other. The CNA-quiet group is characterized by wild-type TP53, frequent CASP8 and HRAS mutations, and a less immunosuppressed tumor immune microenvironment with lower density of regulatory T cells. Patients with CNA-quiet OCSCC are older, more often women, less frequently current smokers, and have a better 5-year overall survival compared to CNA-other OCSCC. This study demonstrates that CNA-quiet OCSCC should be considered as a distinct, clinically relevant subclass. Given the clinical characteristics, the patient group with these tumors will rapidly increase in the aging population.
Collapse
Affiliation(s)
- Tara Muijlwijk
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Otolaryngology / Head and Neck Surgery, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Irene H Nauta
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Otolaryngology / Head and Neck Surgery, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Anabel van der Lee
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Otolaryngology / Head and Neck Surgery, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Kari J T Grünewald
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Otolaryngology / Head and Neck Surgery, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Arjen Brink
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Otolaryngology / Head and Neck Surgery, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Sonja H Ganzevles
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Otolaryngology / Head and Neck Surgery, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | | | | | - Suvi Savola
- MRC Holland, Oncogenetics, Amsterdam, The Netherlands
| | - Mark A van de Wiel
- Amsterdam UMC, Epidemiology & Data Science, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Laura A N Peferoen
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Pathology, Amsterdam, The Netherlands
- Academic Center for Dentistry, Maxillofacial Surgery/ Oral Pathology, Amsterdam, The Netherlands
| | - Elisabeth Bloemena
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Pathology, Amsterdam, The Netherlands
- Academic Center for Dentistry, Maxillofacial Surgery/ Oral Pathology, Amsterdam, The Netherlands
| | - Rieneke van de Ven
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Otolaryngology / Head and Neck Surgery, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - C René Leemans
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Otolaryngology / Head and Neck Surgery, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Jos B Poell
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Otolaryngology / Head and Neck Surgery, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands.
| | - Ruud H Brakenhoff
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Otolaryngology / Head and Neck Surgery, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands.
| |
Collapse
|
48
|
Nair NU, Schäffer AA, Gertz EM, Cheng K, Zerbib J, Sahu AD, Leor G, Shulman ED, Aldape KD, Ben-David U, Ruppin E. Chromosome 7 Gain Compensates for Chromosome 10 Loss in Glioma. Cancer Res 2024; 84:3464-3477. [PMID: 39078448 PMCID: PMC11479827 DOI: 10.1158/0008-5472.can-24-1366] [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: 04/24/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 07/31/2024]
Abstract
The co-occurrence of chromosome 10 loss and chromosome 7 gain in gliomas is the most frequent loss-gain co-aneuploidy pair in human cancers. This phenomenon has been investigated since the late 1980s without resolution. Expanding beyond previous gene-centric studies, we investigated the co-occurrence in a genome-wide manner, taking an evolutionary perspective. Mining of large-scale tumor aneuploidy data confirmed the previous finding of a small-scale longitudinal study that the most likely order is chromosome 10 loss, followed by chromosome 7 gain. Extensive analysis of genomic and transcriptomic data from both patients and cell lines revealed that this co-occurrence can be explained by functional rescue interactions that are highly enriched on chromosome 7, which could potentially compensate for any detrimental consequences arising from the loss of chromosome 10. Transcriptomic data from various normal, noncancerous human brain tissues were analyzed to assess which tissues may be most predisposed to tolerate compensation of chromosome 10 loss by chromosome 7 gain. The analysis indicated that the preexisting transcriptomic states in the cortex and frontal cortex, where gliomas arise, are more favorable than other brain regions for compensation by rescuer genes that are active on chromosome 7. Collectively, these findings suggest that the phenomenon of chromosome 10 loss and chromosome 7 gain in gliomas is orchestrated by a complex interaction of many genes residing within these two chromosomes and provide a plausible reason why this co-occurrence happens preferentially in cancers originating in certain regions of the brain. Significance: Increased expression of multiple rescuer genes on the gained chromosome 7 could compensate for the downregulation of several vulnerable genes on the lost chromosome 10, resolving the long-standing mystery of this frequent co-occurrence in gliomas.
Collapse
Affiliation(s)
- Nishanth Ulhas Nair
- Computational Precision Oncology Section, Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alejandro A. Schäffer
- Computational Precision Oncology Section, Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - E. Michael Gertz
- Computational Precision Oncology Section, Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kuoyuan Cheng
- Computational Precision Oncology Section, Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- MSD, Beijing, China
| | - Johanna Zerbib
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Avinash Das Sahu
- The University of New Mexico, Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Gil Leor
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Eldad D. Shulman
- Computational Precision Oncology Section, Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth D. Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Uri Ben-David
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Eytan Ruppin
- Computational Precision Oncology Section, Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Lead contact
| |
Collapse
|
49
|
Requesens M, Foijer F, Nijman HW, de Bruyn M. Genomic instability as a driver and suppressor of anti-tumor immunity. Front Immunol 2024; 15:1462496. [PMID: 39544936 PMCID: PMC11562473 DOI: 10.3389/fimmu.2024.1462496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/23/2024] [Indexed: 11/17/2024] Open
Abstract
Genomic instability is a driver and accelerator of tumorigenesis and influences disease outcomes across cancer types. Although genomic instability has been associated with immune evasion and worsened disease prognosis, emerging evidence shows that genomic instability instigates pro-inflammatory signaling and enhances the immunogenicity of tumor cells, making them more susceptible to immune recognition. While this paradoxical role of genomic instability in cancer is complex and likely context-dependent, understanding it is essential for improving the success rates of cancer immunotherapy. In this review, we provide an overview of the underlying mechanisms that link genomic instability to pro-inflammatory signaling and increased immune surveillance in the context of cancer, as well as discuss how genomically unstable tumors evade the immune system. A better understanding of the molecular crosstalk between genomic instability, inflammatory signaling, and immune surveillance could guide the exploitation of immunotherapeutic vulnerabilities in cancer.
Collapse
Affiliation(s)
- Marta Requesens
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Hans W. Nijman
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marco de Bruyn
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
50
|
Pessei V, Macagno M, Mariella E, Congiusta N, Battaglieri V, Battuello P, Viviani M, Gionfriddo G, Lamba S, Lorenzato A, Oddo D, Idrees F, Cavaliere A, Bartolini A, Guarrera S, Linnebacher M, Monteonofrio L, Cardone L, Milella M, Bertotti A, Soddu S, Grassi E, Crisafulli G, Bardelli A, Barault L, Di Nicolantonio F. DNA demethylation triggers cell free DNA release in colorectal cancer cells. Genome Med 2024; 16:118. [PMID: 39385243 PMCID: PMC11462661 DOI: 10.1186/s13073-024-01386-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/18/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Liquid biopsy based on cell-free DNA (cfDNA) analysis holds significant promise as a minimally invasive approach for the diagnosis, genotyping, and monitoring of solid malignancies. Human tumors release cfDNA in the bloodstream through a combination of events, including cell death, active and passive release. However, the precise mechanisms leading to cfDNA shedding remain to be characterized. Addressing this question in patients is confounded by several factors, such as tumor burden extent, anatomical and vasculature barriers, and release of nucleic acids from normal cells. In this work, we exploited cancer models to dissect basic mechanisms of DNA release. METHODS We measured cell loss ratio, doubling time, and cfDNA release in the supernatant of a colorectal cancer (CRC) cell line collection (N = 76) representative of the molecular subtypes previously identified in cancer patients. Association analyses between quantitative parameters of cfDNA release, cell proliferation, and molecular features were evaluated. Functional experiments were performed to test the impact of modulating DNA methylation on cfDNA release. RESULTS Higher levels of supernatant cfDNA were significantly associated with slower cell cycling and increased cell death. In addition, a higher cfDNA shedding was found in non-CpG Island Methylator Phenotype (CIMP) models. These results indicate a positive correlation between lower methylation and increased cfDNA levels. To explore this further, we exploited methylation microarrays to identify a subset of probes significantly associated with cfDNA shedding and derive a methylation signature capable of discriminating high from low cfDNA releasers. We applied this signature to an independent set of 176 CRC cell lines and patient derived organoids to select 14 models predicted to be low or high releasers. The methylation profile successfully predicted the amount of cfDNA released in the supernatant. At the functional level, genetic ablation of DNA methyl-transferases increased chromatin accessibility and DNA fragmentation, leading to increased cfDNA release in isogenic CRC cell lines. Furthermore, in vitro treatment of five low releaser CRC cells with a demethylating agent was able to induce a significant increase in cfDNA shedding. CONCLUSIONS Methylation status of cancer cell lines contributes to the variability of cfDNA shedding in vitro. Changes in methylation pattern are associated with cfDNA release levels and might be exploited to increase sensitivity of liquid biopsy assays.
Collapse
Affiliation(s)
- Valeria Pessei
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Marco Macagno
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Elisa Mariella
- Department of Oncology, University of Torino, Turin, Italy
- IFOM, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Noemi Congiusta
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | - Vittorio Battaglieri
- Department of Oncology, University of Torino, Turin, Italy
- IFOM, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Paolo Battuello
- Department of Oncology, University of Torino, Turin, Italy
- IFOM, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Marco Viviani
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | - Giulia Gionfriddo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | - Simona Lamba
- Department of Oncology, University of Torino, Turin, Italy
| | | | - Daniele Oddo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | - Fariha Idrees
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | - Alessandro Cavaliere
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | - Alice Bartolini
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Simonetta Guarrera
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- IIGM-Italian Institute for Genomic Medicine, c/o IRCCS, Candiolo, Turin, Italy
| | - Michael Linnebacher
- Clinic of General Surgery, Molecular Oncology and Immunotherapy, UMR, Rostock, Germany
| | - Laura Monteonofrio
- Department of Research and Advanced Technologies, Regina Elena National Cancer Institute IRCCS, Rome, Italy
| | - Luca Cardone
- Department of Research and Advanced Technologies, Regina Elena National Cancer Institute IRCCS, Rome, Italy
| | - Michele Milella
- Section of Innovation Biomedicine - Oncology Area, Department of Engineering for Innovation Medicine, University of Verona and Verona University and Hospital Trust, Verona, Italy
| | - Andrea Bertotti
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | - Silvia Soddu
- Department of Research and Advanced Technologies, Regina Elena National Cancer Institute IRCCS, Rome, Italy
| | - Elena Grassi
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Turin, Italy
| | | | - Alberto Bardelli
- Department of Oncology, University of Torino, Turin, Italy
- IFOM, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Ludovic Barault
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.
- Department of Oncology, University of Torino, Turin, Italy.
| | - Federica Di Nicolantonio
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.
- Department of Oncology, University of Torino, Turin, Italy.
| |
Collapse
|