1
|
Du F, Ju J, Zheng F, Gao S, Yuan P. The Identification of Novel Prognostic and Predictive Biomarkers in Breast Cancer via the Elucidation of Tumor Ecotypes Using Ecotyper. CANCER INNOVATION 2025; 4:e70013. [PMID: 40432877 PMCID: PMC12107130 DOI: 10.1002/cai2.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/08/2025] [Accepted: 03/18/2025] [Indexed: 05/29/2025]
Abstract
Background Breast cancer is a highly heterogeneous disease, characterized by tumor and nontumor cells at various cell states. Ecotyper is an innovative machine learning framework that quantifies the tumor microenvironment and delineates the tumor ecosystem, demonstrating clinical significance. However, further validation is needed in breast cancer. Methods Ecotyper was applied to identify multiple cellular states and tumor ecotypes using large-scale breast cancer bulk sequencing data, followed by a detailed analysis of their associations with clinical classification, molecular subtypes, survival prognosis, and immunotherapy response. Identified subtypes were further characterized using single-cell and spatial data sets to reveal molecular profiles. Results In a comprehensive analysis of 6578 breast cancer samples from four data sets, Ecotyper identified 69 cellular states and 10 tumor ecotypes. Of these, 37 cellular states significantly correlated with overall survival. Notably, specific states within epithelial cells, macrophages/monocytes, and fibroblasts were linked to a worse prognosis. CE2 abundance was identified as the most significant marker indicating unfavorable prognosis and was further validated in an additional data set of 116 HER2-negative patients. These biomarkers also indicated the efficacy of neoadjuvant immunotherapy in breast cancer. CE2-high cancers were characterized by an abundance of basal-like epithelial cells, scant lymphocytic infiltration, and activation of hypoxia signaling. Single-cell analysis showed that CE2-high areas were rich in SPP1-positive tumor-associated macrophages(TAM), basal-like epithelial cells, and hypoxic cancer-associated fibroblasts(CAF). Spatially, these regions were often peripheral in triple-negative breast cancer, adjacent to fibrotic/necrotic zones. Multiplex immunofluorescence confirmed the enrichment of SPP1+CD68+TAM and HIF1A+SMA+CAF in hypoxic triple-negative breast cancer (TNBC) regions. Conclusions Ecotyper identified novel biomarkers for breast cancer prognosis and treatment prediction. The CE2-high region may represent a hypoxic immune-suppressive niche.
Collapse
Affiliation(s)
- Feng Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), The VIPII Gastrointestinal Cancer Division of Medical DepartmentPeking University Cancer Hospital and InstituteBeijingChina
| | - Jie Ju
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Day CarePeking University Cancer Hospital and InstituteBeijingChina
| | - Fangchao Zheng
- Department of Medical Oncology, Cancer Research Center, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanShandong ProvinceChina
| | - Songlin Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), The VIPII Gastrointestinal Cancer Division of Medical DepartmentPeking University Cancer Hospital and InstituteBeijingChina
| | - Peng Yuan
- Department of VIP Medical Services, National Cancer Centre/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
2
|
Ma L, Mao JH, Barcellos-Hoff MH. Systemic inflammation in response to radiation drives the genesis of an immunosuppressed tumor microenvironment. Neoplasia 2025; 64:101164. [PMID: 40184664 PMCID: PMC11999686 DOI: 10.1016/j.neo.2025.101164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
The composition of the tumor immune microenvironment has become a major determinant of response to therapy, particularly immunotherapy. Clinically, a tumor microenvironment lacking lymphocytes, so-called "cold" tumors, are considered poor candidates for immune checkpoint inhibition. In this review, we describe the diversity of the tumor immune microenvironment in breast cancer and how radiation exposure alters carcinogenesis. We review the development and use of a radiation-genetic mammary chimera model to clarify the mechanism by which radiation acts. Using the chimera model, we demonstrate that systemic inflammation elicited by a low dose of radiation is key to the construction of an immunosuppressive tumor microenvironment, resulting in aggressive, rapidly growing tumors lacking lymphocytes. Our experimental studies inform the non-mutagenic mechanisms by which radiation affects cancer and provide insight into the genesis of cold tumors.
Collapse
Affiliation(s)
- Lin Ma
- Department of Stomatology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, China
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology, School of Medicine, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94143 USA.
| |
Collapse
|
3
|
Jia J, Wang J, Zhang Y, Bai G, Han L, Niu Y. Deep Learning and Radiomic Signatures Associated with Tumor Immune Heterogeneity Predict Microvascular Invasion in Colon Cancer. Acad Radiol 2025:S1076-6332(25)00432-5. [PMID: 40413149 DOI: 10.1016/j.acra.2025.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/28/2025] [Accepted: 05/04/2025] [Indexed: 05/27/2025]
Abstract
RATIONALE AND OBJECTIVES This study aims to develop and validate a deep learning radiomics signature (DLRS) that integrates radiomics and deep learning features for the non-invasive prediction of microvascular invasion (MVI) in patients with colon cancer (CC). Furthermore, the study explores the potential association between DLRS and tumor immune heterogeneity. MATERIALS AND METHODS This study is a multi-center retrospective study that included a total of 1007 patients with colon cancer (CC) from three medical centers and The Cancer Genome Atlas (TCGA-COAD) database. Patients from Medical Centers 1 and 2 were divided into a training cohort (n = 592) and an internal validation cohort (n = 255) in a 7:3 ratio. Medical Center 3 (n = 135) and the TCGA-COAD database (n = 25) were used as external validation cohorts. Radiomics and deep learning features were extracted from contrast-enhanced venous-phase CT images. Feature selection was performed using machine learning algorithms, and three predictive models were developed: a radiomics model, a deep learning (DL) model, and a combined deep learning radiomics (DLR) model. The predictive performance of each model was evaluated using multiple metrics, including the area under the curve (AUC), sensitivity, and specificity. Additionally, differential gene expression analysis was conducted on RNA-seq data from the TCGA-COAD dataset to explore the association between the DLRS and tumor immune heterogeneity within the tumor microenvironment. RESULTS Compared to the standalone radiomics and deep learning models, DLR fusion model demonstrated superior predictive performance. The AUC for the internal validation cohort was 0.883 (95% CI: 0.828-0.937), while the AUC for the external validation cohort reached 0.855 (95% CI: 0.775-0.935). Furthermore, stratifying patients from the TCGA-COAD dataset into high-risk and low-risk groups based on the DLRS revealed significant differences in immune cell infiltration and immune checkpoint expression between the two groups (P < 0.05). CONCLUSION The contrast-enhanced CT-based DLR fusion model developed in this study effectively predicts the MVI status in patients with CC. This model serves as a non-invasive preoperative assessment tool and reveals a potential association between the DLRS and immune heterogeneity within the tumor microenvironment, providing insights to optimize individualized treatment strategies.
Collapse
Affiliation(s)
- Jianye Jia
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong'an Road, Xicheng District, Beijing 100050, China (J.J., J.W., Y.N.)
| | - Jiahao Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong'an Road, Xicheng District, Beijing 100050, China (J.J., J.W., Y.N.)
| | - Yongxian Zhang
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, No.1 DongJiaoMinXiang Street, DongCheng District, Beijing 100730, China (Y.Z.)
| | - Genji Bai
- Department of Medical Imaging Center, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian 223300, Jiangsu, PR China (G.B.)
| | - Lei Han
- Department of Medical Imaging, Huaian Hospital Affiliated to Xuzhou Medical University, Huaian 223001, Jiangsu, China (L.H.)
| | - Yantao Niu
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong'an Road, Xicheng District, Beijing 100050, China (J.J., J.W., Y.N.).
| |
Collapse
|
4
|
Pozo-Agundo E, Álvarez-González M, Rivera-García I, García-de-la-Fuente V, de Martino A, Tejedor JR, de Vicente JC, Rodrigo JP, García-Pedrero JM, Álvarez-Fernández M. Expression of MASTL (Greatwall) associates with good prognosis and response to radiotherapy in pharyngeal squamous cell carcinoma. Transl Oncol 2025; 58:102417. [PMID: 40398127 DOI: 10.1016/j.tranon.2025.102417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 04/16/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are highly heterogeneous in both disease progression and treatment outcome, with hardly any molecular biomarker in clinical practice. Thus, the aim of this study was to investigate the potential prognostic and predictive value of MASTL/Greatwall, a mitotic kinase also involved in PI3K-mTOR signaling and the DNA damage response, in HNSCC. MASTL expression was evaluated by immunohistochemistry in a cohort of 346 surgically treated HPV-negative pharyngeal and laryngeal squamous cell carcinoma patients, as well as in pre-treatment biopsies from a separate cohort of 64 patients treated with induction chemotherapy (ICT). In addition, MASTL mRNA expression was analyzed in 135 patients from The Cancer Genome Atlas (TCGA) database. High MASTL expression was significantly associated with improved disease-specific survival (DSS) (P = 0.029), specifically in well-differentiated pharyngeal squamous cell carcinoma (PSCC) tumors (P = 0.002). Notably, this association was restricted to patients who received adjuvant radiotherapy (RT) (P = 0.009). Consistently, a similar correlation was found at the mRNA level in PSCC tumors from the TCGA dataset. Moreover, the combined expression of MASTL and p21 was significantly associated with better DSS, specifically among patients receiving RT (P = 0.014). Multivariate Cox regression analysis further confirmed that high MASTL expression was independently associated with favorable prognosis in patients who received post-operative RT (HR= 0.65; 95 % CI: 0.45-0.94; P = 0.021). Collectively, these findings unprecedentedly revealed the association between high MASTL expression and favorable outcome in advanced HPV-negative PSCC, in marked contrast to previous reports in other tumor types. Importantly, MASTL expression emerges as an independent predictor of good prognosis in RT-treated PSCC patients.
Collapse
Affiliation(s)
- Esperanza Pozo-Agundo
- Health Research Institute of Asturias (ISPA), Oviedo, Spain; University Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain; Spanish Biomedical Research Network in Cancer (CIBERONC), ISCIII, Madrid, Spain
| | - Miguel Álvarez-González
- Health Research Institute of Asturias (ISPA), Oviedo, Spain; University Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain; Spanish Biomedical Research Network in Cancer (CIBERONC), ISCIII, Madrid, Spain
| | - Israel Rivera-García
- Health Research Institute of Asturias (ISPA), Oviedo, Spain; University Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - Vanessa García-de-la-Fuente
- Health Research Institute of Asturias (ISPA), Oviedo, Spain; University Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - Alba de Martino
- Histopathology Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Juan Ramón Tejedor
- Health Research Institute of Asturias (ISPA), Oviedo, Spain; University Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain; Nanomaterials and Nanotechnology Research Centre (CINN-CSIC), Principality of Asturias, Oviedo, Spain
| | - Juan Carlos de Vicente
- Health Research Institute of Asturias (ISPA), Oviedo, Spain; University Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain; Department of Oral and Maxillofacial Surgery, Central University Hospital of Asturias (HUCA), Oviedo, Spain
| | - Juan P Rodrigo
- Health Research Institute of Asturias (ISPA), Oviedo, Spain; University Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain; Spanish Biomedical Research Network in Cancer (CIBERONC), ISCIII, Madrid, Spain; Department of Otolaryngology, Central University Hospital of Asturias (HUCA), Oviedo, Spain.
| | - Juana M García-Pedrero
- Health Research Institute of Asturias (ISPA), Oviedo, Spain; University Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain; Spanish Biomedical Research Network in Cancer (CIBERONC), ISCIII, Madrid, Spain
| | - Mónica Álvarez-Fernández
- Health Research Institute of Asturias (ISPA), Oviedo, Spain; University Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain.
| |
Collapse
|
5
|
Xi W, Sun X, Wang M, Wang X, Li K, Jiang R, Jia X, Wang W. Identification of progression related LncRNAs in colorectal cancer aggressiveness. Sci Rep 2025; 15:17258. [PMID: 40383716 PMCID: PMC12086236 DOI: 10.1038/s41598-025-02096-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025] Open
Abstract
Colorectal cancer (CRC) progression involves complex molecular alterations, including the dysregulation of long non-coding RNAs (lncRNAs). In this study, we identified key progression-related lncRNAs in CRC by integrating transcriptomic data from TCGA and single-cell RNA sequencing (scRNA-seq). Differential expression analysis revealed numerous lncRNAs associated with CRC progression. To systematically prioritize these lncRNAs, we developed a scoring system incorporating multiple progression-related signatures, differential expression, and survival analysis. This approach identified 198 key lncRNAs, including both known (e.g., LINC01615) and novel candidates (e.g., AC007998.3). Experimental validation confirmed that LINC01615 was significantly upregulated in CRC tissues, whereas AC007998.3 was downregulated. Further analyses indicated that these lncRNAs influence CRC progression through cis-, trans-, and post-transcriptional regulation. Patients were classified into distinct molecular subgroups based on lncRNA expression, exhibiting significant differences in prognosis and immune microenvironment composition. The enrichment of progression-related lncRNAs among differentially expressed lncRNAs was statistically significant, reinforcing their functional relevance. Validation across independent datasets demonstrated the robustness of our findings. Our research provides novel insights into the molecular mechanisms underlying CRC progression and highlights the potential of progression-related lncRNAs as prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Wei Xi
- Department of Oncology, Liaocheng People's Hospital, Liaocheng, 252000, Shandong, China
| | - Xinxin Sun
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, 252000, Shandong, China
| | - Mingwei Wang
- Traditional Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, 250035, Shandong, China
| | - Xizi Wang
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, 252000, Shandong, China
| | - Kun Li
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, 252000, Shandong, China
| | - Runze Jiang
- Traditional Chinese Medicine Innovation Research Institute, Shandong University of Traditional Chinese Medicine, Jinan, 250035, Shandong, China
| | - Xiaodong Jia
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, 252000, Shandong, China
| | - Wenxiao Wang
- Department of Gastrointestinal Surgery, Liaocheng People's Hospital, Liaocheng, 252000, Shandong, China.
| |
Collapse
|
6
|
Zhang C, Li S, Guo J, Pan T, Zhang Y, Gao Y, Pan J, Liu M, Yang Q, Yu J, Xu J, Li Y, Li X. Multi-dimensional characterization of cellular states reveals clinically relevant immunological subtypes and therapeutic vulnerabilities in ovarian cancer. J Transl Med 2025; 23:519. [PMID: 40340848 PMCID: PMC12063340 DOI: 10.1186/s12967-025-06521-3] [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: 12/23/2024] [Accepted: 04/22/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Diverse cell types and cellular states in the tumor microenvironment (TME) are drivers of biological and therapeutic heterogeneity in ovarian cancer (OV). Characterization of the diverse malignant and immunology cellular states that make up the TME and their associations with clinical outcomes are critical for cancer therapy. However, we are still lack of knowledge about the cellular states and their clinical relevance in OV. METHODS We manually collected the comprehensive transcriptomes of OV samples and characterized the cellular states and ecotypes based on a machine-learning framework. The robustness of the cellular states was validated in independent cohorts and single-cell transcriptomes. The functions and regulators of cellular states were investigated. Meanwhile, we thoroughly examined the associations between cellular states and various clinical factors, including clinical prognosis and drug responses. RESULTS We depicted and characterized an immunophenotypic landscape of 3,099 OV samples and 80,044 cells based on a machine learning framework. We identified and validated 32 distinct transcriptionally defined cellular states from 12 cell types and three cellular communities or ecotypes, extending the current immunological subtypes in OV. Functional enrichment and upstream transcriptional regulator analyses revealed cancer hallmark-related pathways and potential immunological biomarkers. We further investigated the spatial patterns of identified cellular states by integrating the spatially resolved transcriptomes. Moreover, prognostic landscape and drug sensitivity analysis exhibited clinically relevant immunological subtypes and therapeutic vulnerabilities. CONCLUSION Our comprehensive analysis of TME helps leveraging various immunological subtypes to highlight new directions and targets for the treatment of cancer.
Collapse
Affiliation(s)
- Can Zhang
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, 571199, China
| | - Si Li
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150081, China
| | - Jiyu Guo
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150081, China
| | - Tao Pan
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, 571199, China
| | - Ya Zhang
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, 571199, China
| | - Yueying Gao
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150081, China
| | - Jiwei Pan
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, 571199, China
| | - Meng Liu
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, 571199, China
| | - Qingyi Yang
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150081, China
| | - Jinyang Yu
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, 571199, China
| | - Juan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.
| | - Yongsheng Li
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150081, China.
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150040, Heilongjiang, China.
- Department of Anesthesiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Xia Li
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, 571199, China.
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.
| |
Collapse
|
7
|
Laman Trip DS, van Oostrum M, Memon D, Frommelt F, Baptista D, Panneerselvam K, Bradley G, Licata L, Hermjakob H, Orchard S, Trynka G, McDonagh EM, Fossati A, Aebersold R, Gstaiger M, Wollscheid B, Beltrao P. A tissue-specific atlas of protein-protein associations enables prioritization of candidate disease genes. Nat Biotechnol 2025:10.1038/s41587-025-02659-z. [PMID: 40316700 DOI: 10.1038/s41587-025-02659-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 03/28/2025] [Indexed: 05/04/2025]
Abstract
Despite progress in mapping protein-protein interactions, their tissue specificity is understudied. Here, given that protein coabundance is predictive of functional association, we compiled and analyzed protein abundance data of 7,811 proteomic samples from 11 human tissues to produce an atlas of tissue-specific protein associations. We find that this method recapitulates known protein complexes and the larger structural organization of the cell. Interactions of stable protein complexes are well preserved across tissues, while cell-type-specific cellular structures, such as synaptic components, are found to represent a substantial driver of differences between tissues. Over 25% of associations are tissue specific, of which <7% are because of differences in gene expression. We validate protein associations for the brain through cofractionation experiments in synaptosomes, curation of brain-derived pulldown data and AlphaFold2 modeling. We also construct a network of brain interactions for schizophrenia-related genes, indicating that our approach can functionally prioritize candidate disease genes in loci linked to brain disorders.
Collapse
Affiliation(s)
- Diederik S Laman Trip
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Marc van Oostrum
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
- Biozentrum, University of Basel, Basel, Switzerland
| | - Danish Memon
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - Fabian Frommelt
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Delora Baptista
- Gulbenkian Institute for Molecular Medicine, Oeiras, Portugal
| | - Kalpana Panneerselvam
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - Glyn Bradley
- Computational Biology, Functional Genomics, GSK, Stevenage, UK
| | - Luana Licata
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Henning Hermjakob
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - Sandra Orchard
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - Gosia Trynka
- Open Targets, Wellcome Genome Campus, Cambridge, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Ellen M McDonagh
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Andrea Fossati
- Science for Life Laboratory, Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Solna, Sweden
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Matthias Gstaiger
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Bernd Wollscheid
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
| | - Pedro Beltrao
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK.
- Open Targets, Wellcome Genome Campus, Cambridge, UK.
- Gulbenkian Institute for Molecular Medicine, Oeiras, Portugal.
| |
Collapse
|
8
|
Hui T, Zhou J, Yao M, Xie Y, Zeng H. Advances in Spatial Omics Technologies. SMALL METHODS 2025; 9:e2401171. [PMID: 40099571 DOI: 10.1002/smtd.202401171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 03/03/2025] [Indexed: 03/20/2025]
Abstract
Rapidly developing spatial omics technologies provide us with new approaches to deeply understanding the diversity and functions of cell types within organisms. Unlike traditional approaches, spatial omics technologies enable researchers to dissect the complex relationships between tissue structure and function at the cellular or even subcellular level. The application of spatial omics technologies provides new perspectives on key biological processes such as nervous system development, organ development, and tumor microenvironment. This review focuses on the advancements and strategies of spatial omics technologies, summarizes their applications in biomedical research, and highlights the power of spatial omics technologies in advancing the understanding of life sciences related to development and disease.
Collapse
Affiliation(s)
- Tianxiao Hui
- State Key Laboratory of Gene Function and Modulation Research, College of Future Technology, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Jian Zhou
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Muchen Yao
- College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Yige Xie
- School of Nursing, Peking University, Beijing, 100871, China
| | - Hu Zeng
- State Key Laboratory of Gene Function and Modulation Research, College of Future Technology, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, 100871, China
| |
Collapse
|
9
|
Tay RYK, Sachdeva M, Ma H, Kim YW, Kook MC, Kim H, Cheong JH, Hewitt LC, Nekolla K, Schmidt G, Yoshikawa T, Oshima T, Arai T, Srivastava S, Teh M, Ong X, Tay ST, Sheng T, Zhao JJ, Tan P, Grabsch HI, Sundar R. Spatial organization of B lymphocytes and prognosis prediction in patients with gastric cancer. Gastric Cancer 2025; 28:384-396. [PMID: 39971854 PMCID: PMC11993452 DOI: 10.1007/s10120-025-01593-y] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Within the tumor microenvironment (TME), the association of B lymphocytes (B cells) with prognosis and therapy response in gastric cancer (GC) remains poorly characterized. We investigated the predictive and prognostic value of B cells, including their spatial organization within the TME, in one of the largest multi-cohort studies to date. METHODS Using CD20 immunohistochemistry, we evaluated B cell density in resection specimens from 977 patients with resectable GC across three cohorts, including the randomized phase III Korean CLASSIC trial. The relationship between CD20 density, clinicopathological characteristics, and overall survival (OS) was analyzed. Digital spatial profiling of 1063 regions of interest from 15 patients was performed to characterize B cell distribution within different regions of interest (ROIs) using the NanoString GeoMx platform. RESULTS CD20 density was significantly higher in diffuse-type GC compared to intestinal-type (p = 0.000012). Patients with CD20-low diffuse-type GC had the shortest OS in the CLASSIC trial (median OS: 49 vs 62 months, HR: 1.9, 95% CI: 1.2-3.0, p = 0.003) and in a Japanese cohort (median OS: 49 vs 67 months, HR: 2.2, 95% CI: 1.2-4.0, p = 0.011). This survival difference was not seen in patients treated with adjuvant chemotherapy (median OS: 62 vs 63 months, HR: 1.8, 95% CI: 0.88-3.5, p = 0.108). Spatial profiling revealed significant B cell enrichment within tumor ROIs compared to the stroma, particularly in diffuse-type GC. CONCLUSIONS Low CD20 positivity, especially in diffuse-type GC, is linked to poor prognosis and may identify patients who could benefit from chemotherapy. These findings underscore the role of B cells in GC.
Collapse
Affiliation(s)
- Ryan Yong Kiat Tay
- Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore, 119228, Singapore
| | - Manavi Sachdeva
- Department of Haematology-Oncology, National University Cancer Institute, National University Hospital, Singapore, Singapore
| | - Haoran Ma
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | - Young-Woo Kim
- Department of Cancer Policy and Population Health, National Cancer Center Graduate School of Cancer Science and Policy and Center for Gastric Cancer and Department of Surgery, National Cancer Center, Goyang, Republic of Korea
| | - Myeong-Cherl Kook
- Center for Gastric Cancer, Department of Pathology, National Cancer Center, Goyang, Republic of Korea
| | - Hyunki Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Ho Cheong
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Lindsay C Hewitt
- Department of Pathology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Precision Medicine, GROW School for Oncology and Reproduction, Maastricht University Center+, Maastricht, The Netherlands
| | | | - Günter Schmidt
- Computational Pathology, Oncology R&D, AstraZeneca, Munich, Germany
| | | | - Takashi Oshima
- Department of Surgery, Yokohama City University, Yokohama, Japan
- Department of Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Tomio Arai
- Department of Pathology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Supriya Srivastava
- Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Ming Teh
- Department of Pathology, National University Hospital, Singapore, Singapore
| | - Xuewen Ong
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | - Su Ting Tay
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | - Taotao Sheng
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Joseph J Zhao
- Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore, 119228, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Hospital, Singapore, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | - Patrick Tan
- Cancer and Stem Cell Biology Program, 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
- Cellular and Molecular Research, National Cancer Centre, Singapore, Singapore
- Singhealth/Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore, Singapore
| | - Heike I Grabsch
- Department of Pathology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands.
- Pathology and Data Analytics, Leeds Institute of Medical Research at St. James'S, University of Leeds, Leeds, UK.
| | - Raghav Sundar
- Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore, 119228, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Hospital, Singapore, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
| |
Collapse
|
10
|
Turdo A, Tulone G, Di Bella S, Porcelli G, D'Accardo C, Gaggianesi M, Modica C, Di Franco S, Angeloro F, Bozzari G, Pantina VD, Lo Iacono M, Minasola C, Giaimo R, Martorana A, Pavan N, Todaro M, Simonato A, Stassi G. Heightened IDO1 levels predict Bacillus Calmette-Guèrin failure in high-risk non-muscle-invasive bladder cancer patients. Cell Death Discov 2025; 11:203. [PMID: 40287406 PMCID: PMC12033280 DOI: 10.1038/s41420-025-02489-7] [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: 01/23/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Recent studies have indicated a potential link between immune-related gene expression and Bacillus Calmette-Guèrin (BCG) treatment response in non-muscle-invasive bladder cancer (NMIBC) patients, however, prognostic gene signatures have not significantly improved risk stratification beyond clinical characteristics. To identify predictive biomarkers in T1 high-risk (HR) bladder cancer (BC) patients responding to BCG treatment, a gene signature was derived from a discovery cohort of 73 BCG-naïve patients, both responders and non-responders, using the publicly available dataset GSE1542618. Among the identified genes, Indoleamine 2,3-dioxygenase (IDO1), an immunosuppressive enzyme, emerged as a crucial determinant of treatment outcomes. The association between IDO1 expression and worse prognosis was subsequently validated in a cohort of 75 BC patients using formalin-fixed paraffin-embedded (FFPE) BC specimens collected prior BCG treatment. This research revealed significant insights into the mechanisms underlying unsatisfactory responses to BCG treatment in HR patients, posing IDO1 as a promising prognostic biomarker and therapeutic target for NMIBC.
Collapse
Affiliation(s)
- Alice Turdo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Gabriele Tulone
- Department of Precision Medicine in Medical, Surgical, and Critical Areas, University of Palermo, Palermo, Italy
- Azienda Ospedaliera Universitaria Policlinico (AOUP) "Paolo Giaccone", Palermo, Italy
| | - Sebastiano Di Bella
- Department of Precision Medicine in Medical, Surgical, and Critical Areas, University of Palermo, Palermo, Italy
| | - Gaetana Porcelli
- Department of Precision Medicine in Medical, Surgical, and Critical Areas, University of Palermo, Palermo, Italy
| | - Caterina D'Accardo
- Department of Precision Medicine in Medical, Surgical, and Critical Areas, University of Palermo, Palermo, Italy
| | - Miriam Gaggianesi
- Department of Precision Medicine in Medical, Surgical, and Critical Areas, University of Palermo, Palermo, Italy
| | - Chiara Modica
- Department of Precision Medicine in Medical, Surgical, and Critical Areas, University of Palermo, Palermo, Italy
| | - Simone Di Franco
- Department of Precision Medicine in Medical, Surgical, and Critical Areas, University of Palermo, Palermo, Italy
| | - Francesca Angeloro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Giulia Bozzari
- Department of Precision Medicine in Medical, Surgical, and Critical Areas, University of Palermo, Palermo, Italy
| | - Vincenzo Davide Pantina
- Department of Precision Medicine in Medical, Surgical, and Critical Areas, University of Palermo, Palermo, Italy
| | - Melania Lo Iacono
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Cristina Minasola
- Department of Precision Medicine in Medical, Surgical, and Critical Areas, University of Palermo, Palermo, Italy
- Azienda Ospedaliera Universitaria Policlinico (AOUP) "Paolo Giaccone", Palermo, Italy
| | - Rosa Giaimo
- Department of Precision Medicine in Medical, Surgical, and Critical Areas, University of Palermo, Palermo, Italy
- Azienda Ospedaliera Universitaria Policlinico (AOUP) "Paolo Giaccone", Palermo, Italy
| | - Anna Martorana
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
- Azienda Ospedaliera Universitaria Policlinico (AOUP) "Paolo Giaccone", Palermo, Italy
| | - Nicola Pavan
- Department of Precision Medicine in Medical, Surgical, and Critical Areas, University of Palermo, Palermo, Italy
- Azienda Ospedaliera Universitaria Policlinico (AOUP) "Paolo Giaccone", Palermo, Italy
| | - Matilde Todaro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
- Azienda Ospedaliera Universitaria Policlinico (AOUP) "Paolo Giaccone", Palermo, Italy
| | - Alchiede Simonato
- Department of Precision Medicine in Medical, Surgical, and Critical Areas, University of Palermo, Palermo, Italy.
- Azienda Ospedaliera Universitaria Policlinico (AOUP) "Paolo Giaccone", Palermo, Italy.
| | - Giorgio Stassi
- Department of Precision Medicine in Medical, Surgical, and Critical Areas, University of Palermo, Palermo, Italy.
| |
Collapse
|
11
|
Fan X, He Z, Guo J, Bu D, Han D, Qu X, Li Q, Cheng S, Han A, Guo J. Leveraging TME features and multi-omics data with an advanced deep learning framework for improved Cancer survival prediction. Sci Rep 2025; 15:14282. [PMID: 40275021 PMCID: PMC12022115 DOI: 10.1038/s41598-025-98565-0] [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: 02/14/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
Glioma, a malignant intracranial tumor with high invasiveness and heterogeneity, significantly impacts patient survival. This study integrates multi-omics data to improve prognostic prediction and identify therapeutic targets. Using single-cell data from glioblastoma (GBM) and low-grade glioma (LGG) samples, we identified 55 distinct cell states via the EcoTyper framework, validated for stability and prognostic impact in an independent cohort. We constructed multi-omics datasets of 620 samples, integrating transcriptomic, copy number variation (CNV), somatic mutation (MUT), Microbe (MIC), EcoTyper result data. A scRNA-seq enhanced Self-Normalizing Network-based glioma prognosis model achieved a C-index of 0.822 (training) and 0.817 (test), with AUC values of 0.867, 0.876, and 0.844 at 1, 3, and 5 years in the training set, and 0.820, 0.947, and 0.936 in the test set. Gradient attribution analysis enhanced the interpretability of the model and identified key molecular markers. The classification into high- and low-risk groups was validated as an independent prognostic factor. HDAC inhibitors are proposed as potential treatments. This study demonstrates the potential of integrating scRNA-seq and multi-omics data for robust glioma prognosis and clinical decision-making support.
Collapse
Affiliation(s)
- Xuan Fan
- School of Management, Beijing University of Chinese Medicine, Ningbo, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Ningbo, China
- Beijing University of Chinese Medicine, Ningbo, China
| | - Zihao He
- Ningbo No. 2 Hospital, Ningbo, 315010, China
| | - Jing Guo
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, China
| | - Dechao Bu
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
| | - Dongchen Han
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Ningbo, China
- Beijing University of Chinese Medicine, Ningbo, China
| | - Xinchi Qu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Ningbo, China
- Beijing University of Chinese Medicine, Ningbo, China
| | - Qihang Li
- Henan University, Kaifeng, 475004, China
| | - Sen Cheng
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China.
| | - Aiqing Han
- School of Management, Beijing University of Chinese Medicine, Ningbo, China.
- Beijing University of Chinese Medicine, Ningbo, China.
| | - Jincheng Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Ningbo, China.
- Beijing University of Chinese Medicine, Ningbo, China.
| |
Collapse
|
12
|
Su A, Lee H, Tran M, Dela Cruz RC, Sathe A, Bai X, Wichmann I, Pflieger L, Moulton B, Barker T, Haslem D, Jones D, Nadauld L, Nguyen Q, Ji HP, Rhodes T. The single-cell spatial landscape of stage III colorectal cancers. NPJ Precis Oncol 2025; 9:101. [PMID: 40189697 PMCID: PMC11973205 DOI: 10.1038/s41698-025-00853-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
We conducted a spatial analysis of stage III colorectal adenocarcinomas using Hyperion Imaging Mass Cytometry, examining 52 tumors to assess the tumor microenvironment at the single-cell level. This approach identified 10 distinct cell phenotypes in the tumor microenvironment, including stromal and immune cells, with a subset showing a proliferative phenotype. By focusing on spatial neighborhood interactions and tissue niches, particularly regions with tumor-infiltrating lymphocytes, we investigated how cellular organization relates to clinicopathological and molecular features such as microsatellite instability (MSI) and recurrence. We determined that microsatellite stable (MSS) colorectal cancers had an increased risk of recurrence if they had the following features: 1) a low level of stromal tumor-infiltrating lymphocytes, and 2) low interactions between CD4 + T cells and stromal cells. Our results point to the utility of spatial single-cell interaction analysis in defining novel features of the tumor immune microenvironments and providing useful clinical cell-related spatial biomarkers.
Collapse
Affiliation(s)
- Andrew Su
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - HoJoon Lee
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Minh Tran
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | | | - Anuja Sathe
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Xiangqi Bai
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ignacio Wichmann
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Division of Obstetrics and Gynecology, Department of Obstetrics, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | | | - Bryce Moulton
- Intermountain Healthcare, Saint George, UT, 84770, USA
| | - Tyler Barker
- Intermountain Healthcare, Saint George, UT, 84770, USA
| | | | - David Jones
- Intermountain Healthcare, Saint George, UT, 84770, USA
| | | | - Quan Nguyen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Hanlee P Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | | |
Collapse
|
13
|
Ou W, Zhang XX, Li B, Tuo Y, Lin RX, Liu PF, Guo JP, Un HC, Li MH, Lei JH, Gao XJ, Zheng FF, Chen LW, Long LL, Wang ZR. Integrated proteogenomic characterization of localized prostate cancer identifies biological insights and subtype-specific therapeutic strategies. Nat Commun 2025; 16:3189. [PMID: 40180929 PMCID: PMC11968977 DOI: 10.1038/s41467-025-58569-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 03/21/2025] [Indexed: 04/05/2025] Open
Abstract
Localized prostate cancer (PCa) is highly variable in their response to therapies. Although a fraction of this heterogeneity can be explained by clinical factors or genomic and transcriptomic profiling, the proteomic-based profiling of aggressive PCa remains poorly understood. Here, we profiled the genome, transcriptome, proteome and phosphoproteome of 145 cases of localized PCa in Chinese patients. Proteome-based stratification of localized PCa revealed three subtypes with distinct molecular features: immune subgroup, arachidonic acid metabolic subgroup and sialic acid metabolic subgroup with highest biochemical recurrence (BCR) rates. Further, we nominated NANS protein, a key enzyme in sialic acid synthesis as a potential prognostic biomarker for aggressive PCa and validated in two independent cohorts. Finally, taking advantage of cell-derived orthotopic transplanted mouse models, single-cell RNA sequencing (scRNA-seq) and immunofluorescence analysis, we revealed that targeting NANS can reverse the immunosuppressive microenvironment through restricting the sialoglycan-sialic acid-recognizing immunoglobulin superfamily lectin (Siglec) axis, thereby inhibiting tumor growth of PCa. In sum, we integrate multi-omic data to refine molecular subtyping of localized PCa, and identify NANS as a potential prognostic biomarker and therapeutic option for aggressive PCa.
Collapse
Affiliation(s)
- Wei Ou
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xin-Xin Zhang
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Bin Li
- Clinical Trials Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ying Tuo
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ren-Xuan Lin
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Peng-Fei Liu
- Shanghai Applied Protein Technology Co., Ltd, Shanghai, 201100, China
| | - Jian-Ping Guo
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
| | - Hio-Cheng Un
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ming-Hao Li
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jia-Hao Lei
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiao-Jing Gao
- Shanghai Applied Protein Technology Co., Ltd, Shanghai, 201100, China
| | - Fu-Fu Zheng
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ling-Wu Chen
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Ling-Li Long
- Clinical Trials Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Zong-Ren Wang
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
14
|
Byun H, Lee HS, Song YS, Park YJ. Transcriptome of Anaplastic Thyroid Cancer Reveals Two Molecular Subtypes with Distinct Tumor Microenvironment and Prognosis. Thyroid 2025; 35:367-378. [PMID: 39869083 DOI: 10.1089/thy.2024.0266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Background: Although patients with anaplastic thyroid cancer (ATC) generally have a poor prognosis and there are currently no effective treatment options, survival and response to therapy vary between patients. Genomic and transcriptomic profiles of ATC have been reported; however, a comprehensive study of the tumor microenvironment (TME) of ATC is still lacking. This study aimed to elucidate the TME characteristics associated with ATC and their prognostic implications. Methods: We analyzed bulk RNA transcriptomic data from 1,634 samples-including 476 normal thyroid tissues, 25 benign thyroid adenomas, 340 RAS-like and 719 BRAFV600E-like differentiated thyroid cancers (DTC-R and DTC-B, respectively), and 74 ATCs. We assessed the TME and molecular characteristics of these thyroid cancer subtypes using deconvolution analysis. Results: The TME of ATC was characterized by a high abundance of immune cells and fibroblasts and a low abundance of epithelial cells compared to other thyroid histologies. During its malignant evolution, ATC exhibited an ecotype more closely related to DTC-B than RAS-like DTC (DTC-R). Furthermore, we identified two distinct molecular subtypes within ATC with significant differences in their TMEs. We termed the subtype with increased immune cells and fibroblasts as ATC-immune-fibroblast (ATC-IF) and the subtype with elevated epithelial and endothelial cells as ATC-epithelial-endothelial (ATC-E). The ATC-IF group had worse disease-specific survival (log-rank p = 0.035), higher ERK scores, and lower thyroid differentiation scores than the ATC-E group. While both ATC subtypes had elevated immune cells and fibroblasts compared to DTC-R and DTC-B, this increase was more pronounced in ATC-IF, with a marked rise in myeloid lineage cells and promigratory fibroblasts. Immune checkpoint gene expression and epithelial-mesenchymal transition scores were significantly higher in the ATC-IF group than in the ATC-E group. Conclusion: ATC shows a TME distinct from that of DTC and can be further divided into two molecular subtypes-each with its own unique TME. The ATC-IF group, with a poorer prognosis and higher ERK score, is enriched in immune cells and fibroblasts, which may represent potential therapeutic targets.
Collapse
Affiliation(s)
- Hyunjong Byun
- CHA University School of Medicine, Pocheon, Republic of Korea
| | - Han Sai Lee
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Gwanak-gu, Republic of Korea
| | - Young Shin Song
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Dongjak-gu, Republic of Korea
| | - Young Joo Park
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Gwanak-gu, Republic of Korea
- Department of Internal Medicine and Genomic Medicine Institute, Medical Research Center, Seoul National University College of Medicine, Jongno-gu, Republic of Korea
| |
Collapse
|
15
|
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] [Download PDF] [Figures] [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
|
16
|
Cen X, Lan Y, Zou J, Chen R, Hu C, Tong Y, Zhang C, Chen J, Wang Y, Zhou R, He W, Lu T, Dubee F, Jovic D, Dong W, Gao Q, Ma M, Lu Y, Xue Y, Cheng X, Li Y, Yang H. Pan-cancer analysis shapes the understanding of cancer biology and medicine. Cancer Commun (Lond) 2025. [PMID: 40120098 DOI: 10.1002/cac2.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 03/25/2025] Open
Abstract
Advances in multi-omics datasets and analytical methods have revolutionized cancer research, offering a comprehensive, pan-cancer perspective. Pan-cancer studies identify shared mechanisms and unique traits across different cancer types, which are reshaping diagnostic and treatment strategies. However, continued innovation is required to refine these approaches and deepen our understanding of cancer biology and medicine. This review summarized key findings from pan-cancer research and explored their potential to drive future advancements in oncology.
Collapse
Affiliation(s)
- Xiaoping Cen
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
- Guangzhou National Laboratory, Guangzhou, Guangdong, P. R. China
| | - Yuanyuan Lan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Jiansheng Zou
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. China
| | - Ruilin Chen
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. China
| | - Can Hu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yahan Tong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Chen Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Jingyue Chen
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yuanmei Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Run Zhou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Weiwei He
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Tianyu Lu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Fred Dubee
- BGI Research, Shenzhen, Guangdong, P. R. China
| | | | - Wei Dong
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- Clin Lab, BGI Genomics, Beijing, P. R. China
| | - Qingqing Gao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Man Ma
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Youyong Lu
- Laboratory of Molecular Oncology, Peking University Cancer Hospital and Institute, Beijing, P. R. China
| | - Yu Xue
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Xiangdong Cheng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yixue Li
- Guangzhou National Laboratory, Guangzhou, Guangdong, P. R. China
- GZMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Huanming Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- BGI, Shenzhen, Guangdong, P. R. China
- James D. Watson Institute of Genome Sciences, Hangzhou, Zhejiang, P. R. China
| |
Collapse
|
17
|
Veas Rodriguez J, Piñol M, Sorolla MA, Parisi E, Sorolla A, Santacana M, Ruiz M, Parra G, Bernabeu M, Iglesias M, Aracil C, Escartin A, Vilardell F, Matias-Guiu X, Salud A, Montal R. Comprehensive immunophenotyping of gastric adenocarcinoma identifies an inflamed class of tumors amenable to immunotherapies. J Immunother Cancer 2025; 13:e010024. [PMID: 40102027 PMCID: PMC11927434 DOI: 10.1136/jitc-2024-010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 02/22/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Gastric adenocarcinoma (GAC) imposes a considerable global health burden. Molecular profiling of GAC from the tumor microenvironment perspective through a multi-omics approach is eagerly awaited in order to allow a more precise application of novel therapies in the near future. METHODS To better understand the tumor-immune interface of GAC, we identified an internal cohort of 82 patients that allowed an integrative molecular analysis including mutational profiling by whole-exome sequencing, RNA gene expression of 770 genes associated with immune response, and multiplex protein expression at spatial resolution of 34 immuno-oncology targets at different compartments (tumorous cells and immune cells). Molecular findings were validated in 595 GAC from the TCGA and ACRG external cohorts with available multiomics data. Prediction of response to immunotherapies of the discovered immunophenotypes was assessed in 1039 patients with cancer from external cohorts with available transcriptome data. RESULTS Unsupervised clustering by gene expression identified a subgroup of GAC that includes 52% of the tumors, the so-called Inflamed class, characterized by high tumor immunogenicity and cytotoxicity, particularly in the tumor center at protein level, with enrichment of PIK3CA and ARID1A mutations and increased presence of exhausted CD8+ T cells as well as co-inhibitory receptors such as PD1, CTLA4, LAG3, and TIGIT. The remaining 48% of tumors were called non-inflamed based on the observed exclusion of T cell infiltration, with an overexpression of VEGFA and higher presence of TP53 mutations, resulting in a worse clinical outcome. A 10-gene RNA signature was developed for the identification of tumors belonging to these classes, demonstrating in evaluated datasets comparable clinical utility in predicting response to current immunotherapies when tested against other published gene signatures. CONCLUSIONS Comprehensive immunophenotyping of GAC identifies an inflamed class of tumors that complements previously proposed tumor-based molecular clusters. Such findings may provide the rationale for exploring novel immunotherapeutic approaches for biomarker-enriched populations in order to improve GAC patient's survival.
Collapse
Affiliation(s)
- Joel Veas Rodriguez
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Miquel Piñol
- Department of Pathology, Oncological Pathology Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Maria Alba Sorolla
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Eva Parisi
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Anabel Sorolla
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Maria Santacana
- Scientific and Technical Service of Immunohistochemistry, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Maria Ruiz
- Scientific and Technical Service of Biobank, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Genís Parra
- CNAG-Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mario Bernabeu
- CNAG-Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mar Iglesias
- Department of Pathology, Hospital del Mar, University Pompeu Fabra, Hospital del Mar Research Institute, CIBERONC, Barcelona, Spain
| | - Carles Aracil
- Department of Gastroenterology, Clinical and Experimental Research in Digestive and Hematological Pathology Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Alfredo Escartin
- Department of Surgery, Experimental Surgery Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Felip Vilardell
- Department of Pathology, Oncological Pathology Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Xavier Matias-Guiu
- Department of Pathology, Oncological Pathology Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Antonieta Salud
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Robert Montal
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| |
Collapse
|
18
|
Meng Z, Li J, Wang H, Cao Z, Lu W, Niu X, Yang Y, Li Z, Wang Y, Lu S. NLRP4 unlocks an NK/macrophages-centered ecosystem to suppress non-small cell lung cancer. Biomark Res 2025; 13:44. [PMID: 40087771 PMCID: PMC11909883 DOI: 10.1186/s40364-025-00756-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Tumor immune evasion extends beyond T cells, affecting innate immune elements like natural killer cells (NK) and macrophages within the tumor-immune microenvironment (TIME). Nevertheless, translational strategies to trigger collaboration of NK cells and macrophages to initiate sufficient anti-tumor cytoxicity remain scarce and are urgently needed. METHODS In this study, TCGA datasets was used to confirm the prognosis value of the expression level of NLR family pyrin domain containing 4 (NLRP4) in NSCLC and the tumor tissues microarray was used to further check its clinical-relevance at protein-level. Subsequently, a tumor cell line with stable NLRP4 overexpression was established and subcutaneous tumor models in C57BL/6J mice were used to validate the anti-tumor characteristics of NLRP4. After analyzing the tumor microenvironment using flow cytometry and multiplex immunofluorescence, we further validated our findings through co-culture transwell assays and TCGA analysis. Utilizing bulk-RNA sequencing, proteomics, and mass spectrometry of mouse tumor tissues, we innovatively identified the downstream pathways of NLRP4 and verified them through co-immunoprecipitation (co-IP) and Western blot (WB) experiments. RESULTS NLRP4 could trigger a distinct anti-tumor ecosystem organized by TIGIT+TNFA+ NK and iNOS+ M1 in lung cancer, discovered in TCGA analysis and verified in murine model. NLRP4-eco exerted tumor-suppression capacity through chemokine reprogramming including CCL5 and CXCL2. Meanwhile, the cytoxicity of NK could be facilitated by iNOS+M1. Mechanistically, NLRP4 stimulated PI3K/Akt-NF-kB axis through suppression of the activity of PP2A. Besides, knockdown of CCL5 and blockade of CXCL2-CXCR2 axis abolished chemotaxis of TIGIT+TNFA+ NK and iNOS+ M1 respectively, as well as for LB-100, a PP2A inhibitor. CONCLUSION Altogether, we delineated NLRP4's unexplored facets and discovered an NLRP4-driven anti-tumor ecosystem composed of TIGIT+TNFA+ NK and iNOS+ M1. Finally, targeting PP2A by its inhibitor successfully mimicked the anti-tumor capacity of the overexpression of NLRP4.
Collapse
Affiliation(s)
- Zhouwenli Meng
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Jian Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Hui Wang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Zhengqi Cao
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Wenqing Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Xiaomin Niu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Yi Yang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Ziming Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China.
| | - Ying Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China.
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China.
| |
Collapse
|
19
|
Abdulrahman Z, Slieker RC, McGuire D, Welters MJP, van Poelgeest MIE, van der Burg SH. Single-cell spatial transcriptomics unravels cell states and ecosystems associated with clinical response to immunotherapy. J Immunother Cancer 2025; 13:e011308. [PMID: 40081939 PMCID: PMC11907085 DOI: 10.1136/jitc-2024-011308] [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: 12/12/2024] [Accepted: 02/25/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND The tumor microenvironment (TME) is a complex and dynamic ecosystem that is known to influence responses to immunotherapy. We leveraged single-cell spatial transcriptomics to systematically dissect the intricate complexity of the TME, in particular the cellular heterogeneity and spatial interactions. Their collective impact on immunotherapy efficacy was studied in the context of a homogeneous group of patients with vulvar high-grade squamous intraepithelial lesions (vHSIL) treated with an immunotherapeutic tumor-specific peptide vaccine. METHODS We performed single-cell spatial transcriptomics on 20 pretreatment vHSIL lesions, stratified by clinical response to immunotherapeutic vaccination into complete responders (CR), partial responders (PR) and non-responders (NR). Using a 1,000-gene panel, we mapped over 274,000 single cells in situ, identifying 18 cell clusters and 99 distinct non-epithelial cell states. Findings were validated against public single-cell transcriptomic data sets to assess their broader relevance across tumor types. RESULTS Profound heterogeneity within the TME was detected across the response groups. CR lesions exhibited a higher ratio of immune-supportive to immune-suppressive cells-a pattern mirrored in other solid tumors following neoadjuvant checkpoint blockade. Key immune populations enriched in CRs included CD4+CD161+ effector T cells and chemotactic CD4+ and CD8+ T cells. Conversely, PRs were characterized by increased proportions of T helper 2 cells and CCL18-expressing macrophages, which are associated with the recruitment of type 2 T cells and regulatory T cells. NRs displayed preferential infiltration with immunosuppressive fibroblasts. Distinct spatial immune ecosystems further defined response groups. Although a number of immune cells were detected in all patients, type 1 effector cells dominated interactions in CRs, type 2 cells were prominently interacting in PRs, while NRs lacked organized immune cell interactions. CONCLUSIONS This study underscores the dual importance of both cellular composition and spatial organization in steering clinical response to immunotherapy.
Collapse
Affiliation(s)
- Ziena Abdulrahman
- Department of Medical Oncology, Leiden University Medical Center, Leiden, ZH, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Roderick C Slieker
- Department of Medical Oncology, Leiden University Medical Center, Leiden, ZH, Netherlands
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Marij J P Welters
- Department of Medical Oncology, Leiden University Medical Center, Leiden, ZH, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | | | - Sjoerd H van der Burg
- Department of Medical Oncology, Leiden University Medical Center, Leiden, ZH, Netherlands
- Oncode Institute, Utrecht, Netherlands
| |
Collapse
|
20
|
Rajagopal PS, Reid S, Fan R, Venton L, Weidner A, Roberson ML, Vadaparampil S, Wang X, Yoder S, Rosa M, Sanders M, Gonzalez-Ericsson P, Hirbo J, Whisenant JG, Pietenpol J, Ye F, Pal T, Lehmann BD. Population-specific patterns in assessing molecular subtypes of young black females with triple-negative breast cancer. NPJ Breast Cancer 2025; 11:28. [PMID: 40069179 PMCID: PMC11897140 DOI: 10.1038/s41523-025-00731-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 02/04/2025] [Indexed: 03/15/2025] Open
Abstract
We determined triple-negative breast cancer (TNBC) subtypes, genetic ancestry, and immune features in a cohort of self-reported Black females with TNBC diagnosed at or below age 50. Among 104 tumors, 34.6% were basal-like 1 (BL1), 17.3% basal-like 2 (BL2), 9.6% luminal androgen receptor (LAR), 26.9% mesenchymal (M), and 11.5% unsubtyped (UNS). Subtypes resembled those seen in Europeans or East Asians, with less LAR (9.6% vs. 14.6-24.4%) and more UNS (11.5% vs. 0-7.5%). "High" proportion of West African ancestry was associated with more LAR (14.9% vs. 4.9%) and less M (25.5% vs. 34.2%). M demonstrated reduced immune activity and was marginally associated with worse overall survival in a multivariate model including stage, West African ancestry, BMI, and TILs, meriting future research. Our study is the largest to date of TNBC subtypes in young Black females. These results reinforce TNBC subtypes' application across populations and potential use as a prognostic biomarker.
Collapse
Affiliation(s)
| | - Sonya Reid
- Vanderbilt University Medical Center; Department of Medicine, Nashville, TN, USA
| | - Run Fan
- Vanderbilt University Medical Center; Department of Biostatistics and Bioinformatics, Nashville, TN, USA
| | - Lindsay Venton
- Vanderbilt University Medical Center; Department of Medicine, Nashville, TN, USA
| | - Anne Weidner
- Vanderbilt University Medical Center; Department of Medicine, Nashville, TN, USA
| | - Mya L Roberson
- University of North Carolina; Department of Health Policy and Management, Chapel Hill, NC, USA
| | | | | | | | | | - Melinda Sanders
- Vanderbilt University Medical Center; Department of Medicine, Nashville, TN, USA
| | | | - Jibril Hirbo
- Vanderbilt University Medical Center; Department of Medicine, Nashville, TN, USA
| | - Jennifer G Whisenant
- Vanderbilt University Medical Center; Department of Medicine, Nashville, TN, USA
| | - Jennifer Pietenpol
- Vanderbilt University Medical Center; Department of Biochemistry, Nashville, TN, USA
| | - Fei Ye
- Vanderbilt University Medical Center; Department of Biostatistics and Bioinformatics, Nashville, TN, USA
| | - Tuya Pal
- Vanderbilt University Medical Center; Department of Medicine, Nashville, TN, USA.
| | - Brian D Lehmann
- Vanderbilt University Medical Center; Department of Medicine, Nashville, TN, USA
| |
Collapse
|
21
|
Liang Z, Li S, Pan Z, Duan Y, Ouyang Q, Zhu L, Song E, Chen K. Profiling Multiple CD8+ T-cell Functional Dimensions Enhances Breast Cancer Immune Assessment. Cancer Immunol Res 2025; 13:337-352. [PMID: 39715293 DOI: 10.1158/2326-6066.cir-24-0235] [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: 03/07/2024] [Revised: 08/19/2024] [Accepted: 12/20/2024] [Indexed: 12/25/2024]
Abstract
CD8+ T-cell abundance is insufficient to assess antitumor immunity and shows poor performance in predicting breast cancer prognosis and immunotherapy response, presumably owing to the complexity of CD8+ T-cell functionalities. Although single-cell RNA sequencing can dissect the multifaceted functions of CD8+ T cells for better immune assessment, its clinical application is limited. In this study, we developed bulk RNA sequencing-based FuncDimen models from integrative analysis of single-cell RNA sequencing and matched bulk RNA sequencing data to evaluate CD8+ T-cell functionalities across five dimensions: tumor reactivity, cytotoxicity, IFNγ secretion, proliferation, and apoptosis. The FuncDimen models quantifying different functional dimensions of CD8+ T cells were validated in our breast cancer cohort and external databases using immunofluorescence and imaging mass cytometry. We calculated the FuncAggre score by weighted aggregation of all five FuncDimen models to encapsulate the overall antitumor immunity. In our breast cancer cohort and external databases, the FuncAggre score demonstrated superior predictive performance for breast cancer prognosis (time-dependent AUC: 0.56-0.70) and immunotherapy response (AUC: 0.71-0.83) over other immune biomarkers, regardless of the breast cancer molecular subtype. Together, the FuncDimen models offer a refined assessment of antitumor immunity mediated by CD8+ T cells in the clinic, enhancing prognostic prediction and aiding personalized immunotherapy in breast cancer.
Collapse
Affiliation(s)
- Zhuozhi Liang
- School of Basic Medical Science, Southern Medical University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Zenith Institute of Medical Sciences, Guangzhou, China
| | - Shunrong Li
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhilong Pan
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yuanqiang Duan
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qian Ouyang
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Liling Zhu
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Erwei Song
- School of Basic Medical Science, Southern Medical University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Zenith Institute of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Kai Chen
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Artificial Intelligence Lab, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong, China
| |
Collapse
|
22
|
Deng F, Xiao G, Tanzhu G, Chu X, Ning J, Lu R, Chen L, Zhang Z, Zhou R. Predicting Survival Rates in Brain Metastases Patients from Non-Small Cell Lung Cancer Using Radiomic Signatures Associated with Tumor Immune Heterogeneity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412590. [PMID: 39840456 PMCID: PMC11904944 DOI: 10.1002/advs.202412590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/17/2024] [Indexed: 01/23/2025]
Abstract
Non-small cell lung cancer (NSCLC) frequently metastasizes to the brain, significantly worsened prognoses. This study aimed to develop an interpretable model for predicting survival in NSCLC patients with brain metastases (BM) integrating radiomic features and RNA sequencing data. 292 samples are collected and analyzed utilizing T1/T2 MRIs. Bidirectional stepwise logistic regression is employed to identify significant variables, facilitating the construction of a prognostic model, which is benchmarked against four machine learning algorithms. BM tissue samples are processed for RNA extraction and sequencing. The optimal model achieved an AUC of 0.96 and a C-index of 0.89 in the train set and an AUC of 0.84 with a C-index of 0.78 in the test set, indicating strong predictive performance and generalizability. Patients from Xiangya Hospital are stratified into high-risk (n = 11) and low-risk (n = 30) groups. RNA sequencing revealed an enrichment of immune-related pathways, particularly the interferon (IFN) pathway in the low-risk group. Immune cell infiltration analysis identified a significant presence of CD8+-T cells, IFNγ-6/-18 in the low-risk group, suggesting an immunologically favorable tumor microenvironment. These findings highlight the potential of combining radiomic and RNA sequencing data for improved survival predictions and personalized treatment strategies in BM patients from NSCLC.
Collapse
Affiliation(s)
- Fuxing Deng
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Gang Xiao
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Guilong Tanzhu
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Xianjing Chu
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Jiaoyang Ning
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Ruoyu Lu
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Liu Chen
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Zijian Zhang
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Rongrong Zhou
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
- Xiangya Lung Cancer CenterXiangya HospitalCentral South UniversityChangsha410008China
| |
Collapse
|
23
|
Ouyang Q, Cui J, Wang Y, Liu K, Zhan Y, Zhuo W, Chen J, Zhou H, Luo C, Xia J, Wang L, Guo C, Zhang J, Liu Z, Yin J. eIF3a function in immunity and protection against severe sepsis by regulating B cell quantity and function through m 6A modification. Acta Pharm Sin B 2025; 15:1571-1588. [PMID: 40370535 PMCID: PMC12069248 DOI: 10.1016/j.apsb.2025.02.005] [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: 06/17/2024] [Revised: 09/06/2024] [Accepted: 11/29/2024] [Indexed: 05/16/2025] Open
Abstract
eIF3a is a N 6-methyladenosine (m6A) reader that regulates mRNA translation by recognizing m6A modifications of these mRNAs. It has been suggested that eIF3a may play an important role in regulating translation initiation via m6A during infection when canonical cap-dependent initiation is inhibited. However, the death of animal model studies impedes our understanding of the functional significance of eIF3a in immunity and regulation in vivo. In this study, we investigated the in vivo function of eIF3a using eIF3a knockout and knockdown mouse models and found that eIF3a deficiency resulted in splenic tissue structural disruption and multi-organ damage, which contributed to severe sepsis induced by Lipopolysaccharide (LPS). Ectopic eIF3a overexpression in the eIF3a knockdown mice rescued mice from LPS-induced severe sepsis. We further showed that eIF3a maintains a functional and healthy immune system by regulating B cell function and quantity through m6A modification of mRNAs. These findings unveil a novel mechanism underlying sepsis, implicating the pivotal role of B cells in this complex disease process regulated by eIF3a. Furthermore, eIF3a may be used to develop a potential strategy for treating sepsis.
Collapse
Affiliation(s)
- Qianying Ouyang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Lymphoma & Hematology, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Jiajia Cui
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Geratic Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yang Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ke Liu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yan Zhan
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wei Zhuo
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Pharmacy, Hunan Traditional Chinese Medical College, Zhuzhou 412000, China
| | - Juan Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chenhui Luo
- Scientific Research Office, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Jianming Xia
- Department of Cardiac Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences/Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming 650102, China
| | - Liansheng Wang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chengxian Guo
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha 410017, China
| | - Jianting Zhang
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jiye Yin
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
24
|
Xiao L, Shen Z, Pan Z, Qiu Y, Huang D, Liu Y, Liu C, Zhang X. High-dimensional deconstruction of HNSC reveals clinically distinct cellular states and ecosystems that are associated with prognosis and therapy response. J Transl Med 2025; 23:254. [PMID: 40025504 PMCID: PMC11872339 DOI: 10.1186/s12967-025-06299-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 02/23/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Characterizing the variety of cell types in the tumor microenvironment (TME) and their organization into cellular communities is vital for elucidating the biological diversity of cancer and informing therapeutic strategies. METHODS Here, we employed a machine learning-based algorithm framework, EcoTyper, to analyze single-cell transcriptomes from 139 patients with head and neck squamous cell carcinoma (HNSC)and gene expression profiles from 983 additional HNSC patients, aiming to delineate the fundamental cell states and ecosystems integral to HNSC. RESULTS A diverse landscape of 66 cell states and 9 ecosystems within the HNSC microenvironment was identified, revealing classical cell types while also expanding upon previous immune classifications. Survival analysis revealed that specific cell states and ecotypes (ecosystems) are associated with patient prognosis, underscoring their potential as indicators of clinical outcomes. Moreover, distinct cell states and ecotypes exhibited varying responses to immunotherapy and chemotherapy, with several showing promise as predictive biomarkers for treatment efficacy. CONCLUSION Our large-scale integrative transcriptome analysis provides high-resolution insights into the cellular states and ecosystems of HNSC, facilitating the discovery of novel biomarkers and supporting the development of precision therapies.
Collapse
Affiliation(s)
- Lei Xiao
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Zhe Shen
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Zhaoyu Pan
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Yuanzheng Qiu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Donghai Huang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Yong Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Chao Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China.
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China.
| | - Xin Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China.
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China.
| |
Collapse
|
25
|
Conning-Rowland M, Cheng CW, Brown O, Giannoudi M, Levelt E, Roberts LD, Griffin KJ, Cubbon RM. Application of CIBERSORTx and BayesPrism to deconvolution of bulk RNA-seq data from human myocardium and skeletal muscle. Heliyon 2025; 11:e42499. [PMID: 40034311 PMCID: PMC11872574 DOI: 10.1016/j.heliyon.2025.e42499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 03/05/2025] Open
Abstract
RNA-sequencing (RNA-seq) is an important tool to explore molecular mechanisms of disease. Technological advances mean this can be performed at the single-cell level, but the large sample sizes needed in clinical studies are currently prohibitively expensive and complex. Deconvolution of bulk RNA-seq offers an opportunity to bridge this gap by defining the cell lineage composition of samples. This approach is widely used in immunology studies, but currently there are no validated pipelines for researchers analysing human myocardium or skeletal muscle. Here, we describe the application and in silico validation of two pipelines to deconvolute human right atrium, left ventricle and skeletal muscle bulk RNA-seq data. Specifically, we have defined the major cell lineages of these tissues using single cell/nucleus RNA-seq data from the Heart Cell Atlas, which are then applied during deconvolution using the CIBERSORTx or BayesPrism deconvolution packages. Both pipelines gave robust estimates of the proportion of all major cell lineages in these tissues. We demonstrate their value in defining age- and sex-differences in tissue composition using bulk RNA-seq data from the GTEx consortium. Our validated pipelines can be rapidly applied by researchers working with existing or novel bulk RNA-seq of myocardium or skeletal muscle to gain novel insights.
Collapse
Affiliation(s)
- Marcella Conning-Rowland
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds, United Kingdom
| | - Chew W. Cheng
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds, United Kingdom
| | - Oliver Brown
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds, United Kingdom
| | - Marilena Giannoudi
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds, United Kingdom
| | - Eylem Levelt
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds, United Kingdom
| | - Lee D. Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds, United Kingdom
| | - Kathryn J. Griffin
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds, United Kingdom
| | - Richard M. Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds, United Kingdom
| |
Collapse
|
26
|
Zhang P, Gao C, Zhang Z, Yuan Z, Zhang Q, Zhang P, Du S, Zhou W, Li Y, Li S. Systematic inference of super-resolution cell spatial profiles from histology images. Nat Commun 2025; 16:1838. [PMID: 39984438 PMCID: PMC11845739 DOI: 10.1038/s41467-025-57072-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 02/07/2025] [Indexed: 02/23/2025] Open
Abstract
Inferring cell spatial profiles from histology images is critical for cancer diagnosis and treatment in clinical settings. In this study, we report a weakly-supervised deep-learning method, HistoCell, to directly infer super-resolution cell spatial profiles consisting of cell types, cell states and their spatial network from histology images at the single-nucleus-level. Benchmark analysis demonstrates that HistoCell robustly achieves state-of-the-art performance in terms of cell type/states prediction solely from histology images across multiple cancer tissues. HistoCell can significantly enhance the deconvolution accuracy for the spatial transcriptomics data and enable accurate annotation of subtle cancer tissue architectures. Moreover, HistoCell is applied to de novo discovery of clinically relevant spatial organization indicators, including prognosis and drug response biomarkers, across diverse cancer types. HistoCell also enable image-based screening of cell populations that drives phenotype of interest, and is applied to discover the cell population and corresponding spatial organization indicators associated with gastric malignant transformation risk. Overall, HistoCell emerges as a powerful and versatile tool for cancer studies in histology image-only cohorts.
Collapse
Affiliation(s)
- Peng Zhang
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing, China
| | - Chaofei Gao
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing, China
| | - Zhuoyu Zhang
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing, China
| | - Zhiyuan Yuan
- Institute of Science and Technology for Brain-Inspired Intelligence; MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence; MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Qian Zhang
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing, China
| | - Ping Zhang
- Department of Pathology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shiyu Du
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| | - Weixun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Li
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Shao Li
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing, China.
| |
Collapse
|
27
|
Lawrence ALE, Tan S. Building Spatiotemporal Understanding of Mycobacterium tuberculosis-Host Interactions. ACS Infect Dis 2025; 11:277-286. [PMID: 39847659 PMCID: PMC11828672 DOI: 10.1021/acsinfecdis.4c00840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Heterogeneity during Mycobacterium tuberculosis (Mtb) infection greatly impacts disease outcome and complicates treatment. This heterogeneity encompasses many facets, spanning local differences in the host immune response to Mtb and the environment experienced by the bacterium, to nonuniformity in Mtb replication state. All of these facets are interlinked and each can affect Mtb susceptibility to antibiotic treatment. In-depth spatiotemporal understanding of Mtb-host interactions is thus critical to both fundamental comprehension of Mtb infection biology and for the development of effective therapeutic regimens. Such spatiotemporal understanding dictates the need for analysis at the single bacterium/cell level in the context of intact tissue architecture, which has been a significant technical challenge. Excitingly, innovations in spatial single cell methodology have opened the door to such studies, beginning to illuminate aspects ranging from intergranuloma differences in cellular composition and phenotype, to sublocation differences in Mtb physiology and replication state. In this perspective, we discuss recent studies that demonstrate the potential of these methodological advancements to reveal critical spatiotemporal insight into Mtb-host interactions, and highlight future avenues of research made possible by these advances.
Collapse
Affiliation(s)
- Anna-Lisa E Lawrence
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts 02111, United States
| | - Shumin Tan
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts 02111, United States
| |
Collapse
|
28
|
Mukund K, Veraksa D, Frankhouser D, Yang L, Tomsic J, Pillai R, Atti S, Mesrizadeh Z, Schmolze D, Wu XC, LeBlanc MA, Miele L, Ochoa A, Seewaldt V, Subramaniam S. Spatially distinct cellular and molecular landscapes define prognosis in triple negative breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637503. [PMID: 39990419 PMCID: PMC11844391 DOI: 10.1101/2025.02.10.637503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Background- Triple-negative breast cancer is a prevalent breast cancer subtype with the lowest 5-year survival. Several factors contribute to its treatment response, but the inherent molecular and cellular tumor heterogeneity are increasingly acknowledged as crucial determinants. Methods- Spatial transcriptomic profiling was performed on FFPE tissues from a retrospective, treatment-naive group of women with differential prognoses (17 with >15 years survival- good prognosis (GPx) and 15 with <3 years survival-poor prognosis (PPx)) using GeoMX® Digital Spatial Profiler. Regions of interest were segmented on pan-cytokeratin and analyzed for tumor and stromal components, probed using GeoMx human whole transcriptome atlas (WTA) panel. Data quality control, normalization, and differential analysis was performed in R using GeomxTools and linear mixed models. Additional analyses including cell-type deconvolution, spatial entropy, functional enrichment, TF-target / ligand-receptor analysis and convolution neural networks were employed to identify significant gene signatures contributing to differential prognosis. Results- Here we report on the spatial and molecular heterogeneity underlying differential prognosis. We observe that the state of the epithelia and its microenvironment (TME) are transcriptionally distinct between the two groups. Invasive epithelia in GPx show a significant increase in immune transcripts with the TME exhibiting increased immune cell presence (via IF), while in PPx they are more metabolically and translationally active, with the TME being more mesenchymal/fibrotic. Specifically, pre-cancerous epithelia in PPx display a prescience of aggressiveness as evidenced by increased EMT-signaling. We identify distinct epithelial gene signatures for PPx and GPx, that can, with high accuracy, classify samples at the time of diagnosis and likely inform therapy. Conclusions- To the best of our knowledge, this is the first study to leverage spatial transcriptomics for an in-depth delineation of the cellular and molecular underpinnings of differential prognosis in TNBC. Our study highlights the potential of spatial transcriptomics to not only uncover the molecular drivers of differential prognosis in TNBC but also to pave the way for precision diagnostics and tailored therapeutic strategies, transforming the clinical landscape for this aggressive breast cancer subtype.
Collapse
Affiliation(s)
- Kavitha Mukund
- Department of Bioengineering, UC San Diego, Gilman Drive, La Jolla, CA 92093, USA
| | - Darya Veraksa
- Department of Bioengineering, UC San Diego, Gilman Drive, La Jolla, CA 92093, USA
| | - David Frankhouser
- City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Lixin Yang
- City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Jerneja Tomsic
- City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Raju Pillai
- City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Srijan Atti
- Del Norte High School, San Diego, CA 92127, USA
| | - Zahra Mesrizadeh
- Department of Bioengineering, UC San Diego, Gilman Drive, La Jolla, CA 92093, USA
| | - Daniel Schmolze
- City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Xiao-Cheng Wu
- LSU Health Sciences Center, School of Medicine, New Orleans, LA 70112, USA
| | - Mary-Anne LeBlanc
- LSU Health Sciences Center, School of Medicine, New Orleans, LA 70112, USA
| | - Lucio Miele
- LSU Health Sciences Center, School of Medicine, New Orleans, LA 70112, USA
| | - Augusto Ochoa
- LSU Health Sciences Center, School of Medicine, New Orleans, LA 70112, USA
| | - Victoria Seewaldt
- City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Shankar Subramaniam
- Department of Bioengineering, UC San Diego, Gilman Drive, La Jolla, CA 92093, USA
- Departments of Cellular & Molecular Medicine, Computer Science & Engineering, and Data Science, UC San Diego, Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
29
|
Gong W, Wang Z, Wei Y, Wang M, Li K, Chen X, Huang X, Zhou L, Gan Q, Xu X, Huang Z, Yao H, Wu N, Huang L, Yan B, Zhao B, Yang Z. Dynamic changes in peripheral blood immunophenotyping and its prognostic value in cervical cancer patients undergoing immune checkpoint blockade therapy. Discov Oncol 2025; 16:167. [PMID: 39937363 PMCID: PMC11822146 DOI: 10.1007/s12672-025-01943-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/05/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Immune checkpoint blockade (ICB) therapy, including antibodies targeting the programmed cell death protein 1 (PD-1) pathway, has significantly prolonged the overall survival (OS) in patients with advanced cervical cancer (CC). ICB treatment affects both target cells and various components released by immune cells, which can be observed in peripheral blood. However, there has been limited research on the dynamics of peripheral blood immunophenotyping and its association with OS in CC patients receiving ICB therapy. METHODS Patients with persistent, recurrent, or metastatic CC treated with ICB were enrolled between December 2019 and September 2022. The dynamic changes in peripheral blood immune cells, immunoglobulins, and complement components were analyzed at baseline (within 30 days prior to the first ICB cycle) and after the second cycle of ICB treatment (4-6 weeks after the first ICB treatment). Associations of the baseline levels of peripheral blood immune cells, immunoglobulins, complement components with OS were analyzed using multivariable Cox regression analysis. RESULTS In this retrospective cohort study, 119 patients who received at least two cycles of ICB were included. Data on peripheral blood immune cells, immunoglobulins, and complement components were available for 70 of these patients. The percentages of suppressor T (Ts) cells and natural killer (NK) cells in peripheral blood increased significantly post-ICB treatment, whereas the Th/Ts ratio and IgM levels decreased. The percentages of cytotoxic T (Tc) cells, Ts cells, the Th/Ts ratio, and levels of IgM, IgA, C3, and C4 were significantly associated with the OS of patients. Furthermore, multivariable Cox regression analysis found that a high level of IgA was associated with poor OS of the patients (HR = 2.918; 95% CI, 1.081-7.877, P = 0.035). CONCLUSION Our study demonstrated the potential proliferation of peripheral blood anti-tumor T cells in some CC patients undergoing ICB therapy. The observed associations between peripheral blood immunophenotyping and OS suggest that these biomarkers might have potential as prognostic tools.
Collapse
Affiliation(s)
- Wenjian Gong
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhi Wang
- Department of Gynecology and Obstetrics, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Yongqiang Wei
- Department of Obstetrics and Gynecology, Guangxi Medical University of Cancer Hospital, Nanning, 530021, Guangxi, China
| | - Maomao Wang
- Department of Obstetrics and Gynecology, Guangxi Medical University of Cancer Hospital, Nanning, 530021, Guangxi, China
| | - Kuina Li
- Department of Obstetrics and Gynecology, Guangxi Medical University of Cancer Hospital, Nanning, 530021, Guangxi, China
| | - Xiaoqi Chen
- Department of Obstetrics and Gynecology, Guangxi Medical University of Cancer Hospital, Nanning, 530021, Guangxi, China
| | - Xiaoling Huang
- Department of Obstetrics and Gynecology, Guangxi Medical University of Cancer Hospital, Nanning, 530021, Guangxi, China
| | - Lu Zhou
- Department of Obstetrics and Gynecology, Guangxi Medical University of Cancer Hospital, Nanning, 530021, Guangxi, China
| | - Qiuting Gan
- Department of Obstetrics and Gynecology, Guangxi Medical University of Cancer Hospital, Nanning, 530021, Guangxi, China
| | - Xiaoying Xu
- Department of Obstetrics and Gynecology, Guangxi Medical University of Cancer Hospital, Nanning, 530021, Guangxi, China
| | - Zhijiong Huang
- Department of Obstetrics and Gynecology, Guangxi Medical University of Cancer Hospital, Nanning, 530021, Guangxi, China
| | - Hongyu Yao
- Department of Obstetrics and Gynecology, Guangxi Medical University of Cancer Hospital, Nanning, 530021, Guangxi, China
| | - Nengxian Wu
- Department of Obstetrics and Gynecology, Guangxi Medical University of Cancer Hospital, Nanning, 530021, Guangxi, China
| | - Lu Huang
- Department of Obstetrics and Gynecology, Guangxi Medical University of Cancer Hospital, Nanning, 530021, Guangxi, China
| | - Bingbing Yan
- Department of Obstetrics and Gynecology, Guangxi Medical University of Cancer Hospital, Nanning, 530021, Guangxi, China
| | - Bingbing Zhao
- Department of Obstetrics and Gynecology, Guangxi Medical University of Cancer Hospital, Nanning, 530021, Guangxi, China.
- State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China.
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, Guangxi, China.
| | - Zhijun Yang
- Department of Obstetrics and Gynecology, Guangxi Medical University of Cancer Hospital, Nanning, 530021, Guangxi, China.
- State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, Guangxi, China.
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, Guangxi, China.
| |
Collapse
|
30
|
Lai X, Wu L, Lin P, You L, Ye J. Plasma miRNAs in polycystic ovary syndrome drive endometrial cancer progression: insights into molecular pathways and therapeutic targets. Discov Oncol 2025; 16:133. [PMID: 39920371 PMCID: PMC11806182 DOI: 10.1007/s12672-025-01861-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/03/2025] [Indexed: 02/09/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is a known risk factor for uterine endometrial cancer (UCEC), but its underlying mechanisms remain unclear. MicroRNAs (miRNAs) could provide insights into these mechanisms and reveal potential therapeutic targets. Differential miRNA expression was analyzed in plasma exosomes from 15 PCOS and 15 control samples. Survival analysis assessed the prognostic value of these miRNAs in UCEC. MiRNA-target gene interaction networks and gene co-expression analyses were used to explore molecular mechanisms. Validation was performed using experimental data from Ishikawa cells treated with six candidate drugs. Among the 15 differentially expressed miRNAs, 12 were up-regulated and 3 were down-regulated in PCOS. Twelve of these miRNAs were associated with UCEC overall survival, with miR-142, miR-424, and miR-331 acting as protective factors, while the remaining 9 miRNAs were identified as risk factors. MiRNA-target network highlighted key genes such as PHF8, LCOR, SFT2D3, E2F1, and ESR1, which were found to be prognostic for patient survival. Further gene expression and co-expression analyses based on miR-424 and miR-330 expression revealed significant alterations in gene expression and cellular processes related to UCEC. Two-sample Mendelian randomization analysis identified potential causal relationships between AURKA gene expression and PCOS or UCEC. Testosterone and estradiol might have adverse roles in UCEC, while drugs like troglitazone, valproic acid, retinoic acid, and progesterone demonstrated various effects on gene expression and cellular processes. Our findings suggest that aberrant miRNA expression, particularly miR-330 and miR-424, may play crucial roles in UCEC progression. The identified miRNAs and candidate drugs may serve as potential therapeutic targets for UCEC, but further research is required to validate and explore their clinical applications.
Collapse
Affiliation(s)
- Xuedan Lai
- Department of Gynaecology, Fuzhou First Hospital Affiliated to Fujian Medical University, Fuzhou, 350009, People's Republic of China
| | - Ling Wu
- Department of Gynaecology, Fuzhou First Hospital Affiliated to Fujian Medical University, Fuzhou, 350009, People's Republic of China
| | - Peihong Lin
- Department of Gynaecology, Fuzhou First Hospital Affiliated to Fujian Medical University, Fuzhou, 350009, People's Republic of China
| | - Lijiao You
- Department of Gynaecology, Fuzhou First Hospital Affiliated to Fujian Medical University, Fuzhou, 350009, People's Republic of China
| | - Jianwen Ye
- Department of Gynaecology, Fuzhou First Hospital Affiliated to Fujian Medical University, Fuzhou, 350009, People's Republic of China.
| |
Collapse
|
31
|
Chen YC, Hsu CL, Wang HM, Wu SG, Chang YL, Chen JS, Wu YC, Lin YT, Yang CY, Lin MW, Lee JM, Kuo SW, Chen KC, Hsu HH, Huang PM, Huang YL, Yu CJ, Pirooznia M, Huang BE, Yang R, Shih JY, Yang PC. Multiomics Analysis Reveals Molecular Changes during Early Progression of Precancerous Lesions to Lung Adenocarcinoma in Never-Smokers. Cancer Res 2025; 85:602-617. [PMID: 39570802 PMCID: PMC11786955 DOI: 10.1158/0008-5472.can-24-0821] [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: 03/14/2024] [Revised: 07/20/2024] [Accepted: 11/05/2024] [Indexed: 02/03/2025]
Abstract
Lung cancer is the most common cause of cancer mortality globally, and the prevalence of lung adenocarcinoma, the most common lung cancer subtype, has increased sharply in East Asia. Early diagnosis leads to better survival rates, but this requires an improved understanding of the molecular changes during early tumorigenesis, particularly in nonsmokers. In this study, we performed whole-exome sequencing and RNA sequencing of samples from 94 East Asian patients with precancerous lesions [25 with atypical adenomatous hyperplasia (AAH); 69 with adenocarcinoma in situ (AIS)] and 73 patients with early invasive lesions [minimally invasive adenocarcinoma (MIA)]. Cellular analysis revealed that the activities of endothelial and stromal cells could be used to categorize tumors into molecular subtypes within pathologically defined types of lesions. The subtypes were linked with the radiologically defined type of lesions and corresponded to immune cell infiltration throughout the early progression of lung adenocarcinoma. Spatial transcriptomic analysis revealed the distribution of epithelial cells, endothelial cells, fibroblasts, and plasma cells within MIA samples. Characterization of the molecular lesion subtypes identified positively selected mutational patterns and suggested that angiogenesis in the late-stage AIS type potentially contributes to tissue invasion of the MIA type. This study offers a resource that may help improve early diagnosis and patient prognosis, and the findings suggest possible approaches for early disease interception. Significance: Integrative analysis of multiomics data revealed coordination between immune and nonimmune cells during early progression of precancerous lesions to lung adenocarcinomas and shed light on the molecular characteristics of clinically defined subtypes.
Collapse
Affiliation(s)
- Yun-Ching Chen
- Interventional Oncology, Johnson & Johnson Enterprise Innovation, Inc., Boston, Massachusetts
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Hui-Min Wang
- Interventional Oncology, Johnson & Johnson Enterprise Innovation, Inc., Boston, Massachusetts
| | - Shang-Gin Wu
- Department of Medicine, National Taiwan University Cancer Center and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yih-Leong Chang
- Department of Pathology, National Taiwan University Cancer Center and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jin-Shing Chen
- Department of Surgery Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Ching Wu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Ting Lin
- Department of Medicine, National Taiwan University Cancer Center and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ching-Yao Yang
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Mong-Wei Lin
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Jang-Ming Lee
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Shuenn-Wen Kuo
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Ke-Cheng Chen
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsao-Hsun Hsu
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Ming Huang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Lin Huang
- Department of Pathology, National Taiwan University Cancer Center and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chong-Jen Yu
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | - Mehdi Pirooznia
- Interventional Oncology, Johnson & Johnson Enterprise Innovation, Inc., Boston, Massachusetts
| | - Bevan E. Huang
- Interventional Oncology, Johnson & Johnson Enterprise Innovation, Inc., Boston, Massachusetts
| | - Rob Yang
- Interventional Oncology, Johnson & Johnson Enterprise Innovation, Inc., Boston, Massachusetts
| | - Jin-Yuan Shih
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Pan-Chyr Yang
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
32
|
To A, Yu Z, Sugimura R. Recent advancement in the spatial immuno-oncology. Semin Cell Dev Biol 2025; 166:22-28. [PMID: 39705969 DOI: 10.1016/j.semcdb.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
Recent advancements in spatial transcriptomics and spatial proteomics enabled the high-throughput profiling of single or multi-cell types and cell states with spatial information. They transformed our understanding of the higher-order architectures and paired cell-cell interactions within a tumor microenvironment (TME). Within less than a decade, this rapidly emerging field has discovered much crucial fundamental knowledge and significantly improved clinical diagnosis in the field of immuno-oncology. This review summarizes the conceptual frameworks to understand spatial omics data and highlights the updated knowledge of spatial immuno-oncology.
Collapse
Affiliation(s)
- Alex To
- School of Biomedical Sciences, University of Hong Kong, Hong Kong
| | - Zou Yu
- School of Biomedical Sciences, University of Hong Kong, Hong Kong
| | - Ryohichi Sugimura
- School of Biomedical Sciences, University of Hong Kong, Hong Kong; Centre for Translational Stem Cell Biology, Hong Kong.
| |
Collapse
|
33
|
Wang X, Li Z, Zhang C. Integrated Analysis of Serum and Tissue microRNA Transcriptome for Biomarker Discovery in Gastric Cancer. ENVIRONMENTAL TOXICOLOGY 2025; 40:281-290. [PMID: 39400980 DOI: 10.1002/tox.24430] [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: 03/19/2024] [Revised: 05/25/2024] [Accepted: 08/31/2024] [Indexed: 10/15/2024]
Abstract
Gastric cancer (GC) poses a significant global health challenge, demanding a detailed exploration of its molecular landscape. Studies suggest that exposure to environmental pollutants can lead to changes in microRNA (miRNA) expression patterns, which may contribute to the development and progression of GC. MiRNAs have emerged as crucial regulators implicated in GC pathogenesis. The largest GC serum miRNA dataset to date, comprising 1417 non-cancer controls and 1417 GC samples was used. We conducted a comprehensive analysis of miRNA expression profiles. Differential expression analysis, co-expression network construction, and machine learning models were employed to identify key serum miRNAs and their association with clinical parameters. Weighted Gene Co-expression Network Analysis (WGCNA) and immune infiltration analysis were used to validate the importance of the key miRNA. A total of 1766 differentially expressed miRNAs were identified, with miR-1290, miR-1246, and miR-451a among the top up-regulated, and miR-6875-5p, miR-6784-5p, miR-1228-5p, and miR-6765-5p among the top down-regulated. WGCNA revealed that modules M1 and M5 were significantly associated with GC subtypes and disease status. MiRNA-target gene network analysis identified prognostically significant genes TP53, EMCN, CBX8, and ALDH1A3. Machine learning models LASSO, SVM, randomforest, and XGBOOST demonstrated the diagnostic potential of miRNA profiles. Tissue and serum miR-187 emerged as an independent prognostic factor, influencing patient survival across clinical parameters. Gene expression and immune cell infiltration were different in tissues stratified by miR-187 expression. In summary, the integration of differential gene expression, co-expression analysis, and immune cell profiling provided insights into the molecular intricacies of GC progression.
Collapse
Affiliation(s)
- Xinfeng Wang
- Department of Pharmacy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Zhuoran Li
- Department of Optometry, Fenyang College of Shanxi Medical University, Fenyang, China
| | - Chengyan Zhang
- Department of Gastroenterology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
34
|
Mirza HB, Hunt A, Ennis DP, McDermott J, McNeish IA. Spatial transcriptomic analysis reveals significant differences in tumor microenvironment in HPV-dependent and HPV-independent vulvar squamous cell carcinoma. Gynecol Oncol 2025; 193:65-72. [PMID: 39787746 DOI: 10.1016/j.ygyno.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
OBJECTIVE Vulvar squamous cell carcinoma (VSCC) can be either HPV-dependent (HPVd) or HPV-independent (HPVi). HPVd VSCC typically occurs in younger women, has a more favorable prognosis, and develops from high-grade squamous intraepithelial lesions (HSIL). HPVi VSCC predominantly affects older women and arises within areas of chronic inflammation, particularly lichen sclerosis (LS). We utilized sequencing-based spatial transcriptomics to explore gene expression in a cohort of patients with HPVi and HPVd VSCC. METHODS We analysed gene expression in distinct areas (SCC, inflammation, LS, HSIL) from four early-stage VSCC cases (two HPVi, two HPVd) using the 10× Genomics Visium spatial transcriptomics platform. Cell-specific type expression was inferred using CIBERSORTx. RESULTS 28,183 Visium spots were detected; each contained an estimated 20-50 cells. Reads per spot ranged from 9903 to 68,527. More genes were upregulated in HPVd (N = 601) than HPVi (N = 72) with distinct differences in Keratin and Collagen genes between etiologies. Gene expression was strikingly similar between SCC and adjacent inflammatory areas, regardless of etiology. IL-17 signaling was upregulated in HPVd samples. Surprisingly, CIBERSORTx inferred significantly more CD45+ cells in HPVi tissues than HPVd, especially CD4+ resting memory and follicular helper T cells in SCC areas. Immune cells moved from resting states in the pre-invasive tissues to activated states in the SCC and peri-tumoral inflammatory areas. CONCLUSIONS This study represents the first application of spatial transcriptomics in VSCC, with significantly more immune cells identified in HPVi SCC than in HPVd SCC. These data will act as a baseline for future studies.
Collapse
Affiliation(s)
- Hasan B Mirza
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Ashton Hunt
- Department of Cellular Pathology, Imperial College Healthcare NHS Trust, London, UK
| | - Darren P Ennis
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Jacqueline McDermott
- Department of Cellular Pathology, Imperial College Healthcare NHS Trust, London, UK; Department of Pathology, Barts Healthcare NHS Trust, London, UK
| | - Iain A McNeish
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, UK.
| |
Collapse
|
35
|
Guo S, Liu X, Cheng X, Jiang Y, Ji S, Liang Q, Koval A, Li Y, Owen LA, Kim IK, Aparicio A, Lee S, Sood AK, Kopetz S, Shen JP, Weinstein JN, DeAngelis MM, Chen R, Wang W. A deconvolution framework that uses single-cell sequencing plus a small benchmark data set for accurate analysis of cell type ratios in complex tissue samples. Genome Res 2025; 35:147-161. [PMID: 39586714 PMCID: PMC11789644 DOI: 10.1101/gr.278822.123] [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: 12/05/2023] [Accepted: 11/19/2024] [Indexed: 11/27/2024]
Abstract
Bulk deconvolution with single-cell/nucleus RNA-seq data is critical for understanding heterogeneity in complex biological samples, yet the technological discrepancy across sequencing platforms limits deconvolution accuracy. To address this, we utilize an experimental design to match inter-platform biological signals, hence revealing the technological discrepancy, and then develop a deconvolution framework called DeMixSC using this well-matched, that is, benchmark, data. Built upon a novel weighted nonnegative least-squares framework, DeMixSC identifies and adjusts genes with high technological discrepancy and aligns the benchmark data with large patient cohorts of matched-tissue-type for large-scale deconvolution. Our results using two benchmark data sets of healthy retinas and ovarian cancer tissues suggest much-improved deconvolution accuracy. Leveraging tissue-specific benchmark data sets, we applied DeMixSC to a large cohort of 453 age-related macular degeneration patients and a cohort of 30 ovarian cancer patients with various responses to neoadjuvant chemotherapy. Only DeMixSC successfully unveiled biologically meaningful differences across patient groups, demonstrating its broad applicability in diverse real-world clinical scenarios. Our findings reveal the impact of technological discrepancy on deconvolution performance and underscore the importance of a well-matched data set to resolve this challenge. The developed DeMixSC framework is generally applicable for accurately deconvolving large cohorts of disease tissues, including cancers, when a well-matched benchmark data set is available.
Collapse
Affiliation(s)
- Shuai Guo
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Xiaoqian Liu
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Xuesen Cheng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Yujie Jiang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Statistics, Rice University, Houston, Texas 77005, USA
| | - Shuangxi Ji
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Qingnan Liang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Andrew Koval
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Statistics, Rice University, Houston, Texas 77005, USA
| | - Yumei Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Leah A Owen
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Engineering, SUNY University at Buffalo, Buffalo, New York 14209, USA
- Department of Population Health Sciences, University of Utah School of Medicine, Salt Lake City, Utah 84108, USA
- Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, Salt Lake City, Utah 84132, USA
| | - Ivana K Kim
- USA Retina Service, Harvard Medical School, Massachusetts Eye and Ear, Boston, Massachusetts 02114, USA
| | - Ana Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77230, USA
| | - Sanghoon Lee
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77230, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77230, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - John N Weinstein
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Margaret M DeAngelis
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Engineering, SUNY University at Buffalo, Buffalo, New York 14209, USA
- Department of Population Health Sciences, University of Utah School of Medicine, Salt Lake City, Utah 84108, USA
- Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, Salt Lake City, Utah 84132, USA
- VA Western New York Healthcare System, Buffalo, New York 14215, USA
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Wenyi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA;
| |
Collapse
|
36
|
Paul ED, Huraiová B, Valková N, Matyasovska N, Gábrišová D, Gubová S, Ignačáková H, Ondris T, Gala M, Barroso L, Bendíková S, Bíla J, Buranovská K, Drobná D, Krchňáková Z, Kryvokhyzha M, Lovíšek D, Mamoilyk V, Mancikova V, Vojtaššáková N, Ristová M, Comino-Méndez I, Andrašina I, Morozov P, Tuschl T, Pareja F, Kather JN, Čekan P. The spatially informed mFISHseq assay resolves biomarker discordance and predicts treatment response in breast cancer. Nat Commun 2025; 16:226. [PMID: 39747865 PMCID: PMC11696812 DOI: 10.1038/s41467-024-55583-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025] Open
Abstract
Current assays fail to address breast cancer's complex biology and accurately predict treatment response. On a retrospective cohort of 1082 female breast tissues, we develop and validate mFISHseq, which integrates multiplexed RNA fluorescent in situ hybridization with RNA-sequencing, guided by laser capture microdissection. This technique ensures tumor purity, unbiased whole transcriptome profiling, and explicitly quantifies intratumoral heterogeneity. Here we show mFISHseq has 93% accuracy compared to immunohistochemistry. Our consensus subtyping and risk groups mitigate single sample discordance, provide early and late prognostic information, and identify high risk patients with enriched immune signatures, which predict response to neoadjuvant immunotherapy in the multicenter, phase II, prospective I-SPY2 trial. We identify putative antibody-drug conjugate (ADC)-responsive patients, as evidenced by a 19-feature T-DM1 classifier, validated on I-SPY2. Deploying mFISHseq as a research-use only test on 48 patients demonstrates clinical feasibility, revealing insights into the efficacy of targeted therapies, like CDK4/6 inhibitors, immunotherapies, and ADCs.
Collapse
Affiliation(s)
- Evan D Paul
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia.
- MultiplexDX, Inc, Rockville, MD, USA.
| | - Barbora Huraiová
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Natália Valková
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Natalia Matyasovska
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
- Institute of Clinical Biochemistry and Diagnostics, University Hospital, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Daniela Gábrišová
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Soňa Gubová
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Helena Ignačáková
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Tomáš Ondris
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Michal Gala
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Liliane Barroso
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Silvia Bendíková
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Jarmila Bíla
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Katarína Buranovská
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Diana Drobná
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Zuzana Krchňáková
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Maryna Kryvokhyzha
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Daniel Lovíšek
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Viktoriia Mamoilyk
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Veronika Mancikova
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Nina Vojtaššáková
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
| | - Michaela Ristová
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia
- MultiplexDX, Inc, Rockville, MD, USA
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - Iñaki Comino-Méndez
- Hospital Universitario Virgen de la Victoria, The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), Málaga, Spain
| | - Igor Andrašina
- Department of Radiotherapy and Oncology, East Slovakia Institute of Oncology, Košice, Slovakia
| | - Pavel Morozov
- Laboratory for RNA Molecular Biology, The Rockefeller University, New York, NY, USA
| | - Thomas Tuschl
- Laboratory for RNA Molecular Biology, The Rockefeller University, New York, NY, USA
| | - Fresia Pareja
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Jakob N Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany.
- Department of Medicine I, University Hospital Dresden, Dresden, Germany.
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| | - Pavol Čekan
- MultiplexDX, s.r.o., Comenius University Science Park, Bratislava, Slovakia.
- MultiplexDX, Inc, Rockville, MD, USA.
| |
Collapse
|
37
|
Yan Y, Sun D, Hu J, Chen Y, Sun L, Yu H, Xiong Y, Huang Z, Xia H, Zhu X, Bian D, Sun F, Hou L, Wu C, Fan OR, Hu H, Zeng A, Zhang L, Sun YE, Wang C, Zhang P. Multi-omic profiling highlights factors associated with resistance to immuno-chemotherapy in non-small-cell lung cancer. Nat Genet 2025; 57:126-139. [PMID: 39658657 DOI: 10.1038/s41588-024-01998-y] [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: 04/27/2023] [Accepted: 10/18/2024] [Indexed: 12/12/2024]
Abstract
Although immune checkpoint blockade (ICB) therapies have shifted the treatment paradigm for non-small-cell lung cancer (NSCLC), many patients remain resistant. Here we characterize the tumor cell states and spatial cellular compositions of the NSCLC tumor microenvironment (TME) by analyzing single-cell transcriptomes of 232,080 cells and spatially resolved transcriptomes of tumors from 19 patients before and after ICB-chemotherapy. We find that tumor cells and secreted phosphoprotein 1-positive macrophages interact with collagen type XI alpha 1 chain-positive cancer-associated fibroblasts to stimulate the deposition and entanglement of collagen fibers at tumor boundaries, obstructing T cell infiltration and leading to poor prognosis. We also reveal distinct states of tertiary lymphoid structures (TLSs) in the TME. Activated TLSs are associated with improved prognosis, whereas a hypoxic microenvironment appears to suppress TLS development and is associated with poor prognosis. Our study provides novel insights into different cellular and molecular components corresponding to NSCLC ICB-chemotherapeutic responsiveness, which will benefit future individualized immuno-chemotherapy.
Collapse
Affiliation(s)
- Yilv Yan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dongqing Sun
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Life Science and Technology, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Junjie Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yue Chen
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Liangdong Sun
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huansha Yu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yicheng Xiong
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhida Huang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haoran Xia
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinsheng Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dongliang Bian
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fenghuan Sun
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Likun Hou
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Orion R Fan
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haiyang Hu
- Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - An Zeng
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| | - Lele Zhang
- Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Yi Eve Sun
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Chenfei Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Life Science and Technology, Tongji University, Shanghai, China.
- Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.
- National Key Laboratory of Autonomous Intelligent Unmanned Systems, Tongji University, Shanghai, China.
- Frontier Science Center for Intelligent Autonomous Systems, Tongji University, Shanghai, China.
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
- Department of Thoracic Surgery, The First Affiliated Hospital of Shihezi University Medical College, Shihezi, China.
| |
Collapse
|
38
|
Gulati GS, D'Silva JP, Liu Y, Wang L, Newman AM. Profiling cell identity and tissue architecture with single-cell and spatial transcriptomics. Nat Rev Mol Cell Biol 2025; 26:11-31. [PMID: 39169166 DOI: 10.1038/s41580-024-00768-2] [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: 07/16/2024] [Indexed: 08/23/2024]
Abstract
Single-cell transcriptomics has broadened our understanding of cellular diversity and gene expression dynamics in healthy and diseased tissues. Recently, spatial transcriptomics has emerged as a tool to contextualize single cells in multicellular neighbourhoods and to identify spatially recurrent phenotypes, or ecotypes. These technologies have generated vast datasets with targeted-transcriptome and whole-transcriptome profiles of hundreds to millions of cells. Such data have provided new insights into developmental hierarchies, cellular plasticity and diverse tissue microenvironments, and spurred a burst of innovation in computational methods for single-cell analysis. In this Review, we discuss recent advancements, ongoing challenges and prospects in identifying and characterizing cell states and multicellular neighbourhoods. We discuss recent progress in sample processing, data integration, identification of subtle cell states, trajectory modelling, deconvolution and spatial analysis. Furthermore, we discuss the increasing application of deep learning, including foundation models, in analysing single-cell and spatial transcriptomics data. Finally, we discuss recent applications of these tools in the fields of stem cell biology, immunology, and tumour biology, and the future of single-cell and spatial transcriptomics in biological research and its translation to the clinic.
Collapse
Affiliation(s)
- Gunsagar S Gulati
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Yunhe Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Aaron M Newman
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
39
|
Zhu AK, Li GY, Chen FC, Shan JQ, Shan YQ, Lv CX, Zhu ZQ, He YR, Zhai LL. Integrated Analysis of Single-Cell and Bulk RNA-Sequencing Based on EcoTyper Machine Learning Framework Identifies Cell-State-Specific M2 Macrophage Markers Associated with Gastric Cancer Prognosis. Immunotargets Ther 2024; 13:721-734. [PMID: 39678138 PMCID: PMC11646439 DOI: 10.2147/itt.s490075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 11/30/2024] [Indexed: 12/17/2024] Open
Abstract
Background Tumor is a complex and dynamic ecosystem formed by the interaction of numerous diverse cells types and the microenvironments they inhabit. Determining how cellular states change and develop distinct cellular communities in response to the tumor microenvironment is critical to understanding cancer progression. Tumour-associated macrophages (TAMs) are an important component of the tumour microenvironment and play a crucial role in cancer progression. This study was designed to identify cell-state-specific M2 macrophage markers associated with gastric cancer (GC) prognosis through integrative analysis of single-cell RNA sequencing (scRNA-seq) and bulk RNA-seq data using a machine learning framework named EcoTyper. Results The results showed that TAMs were classified into M1 macrophages, M2 macrophages, monocytes, undefined macrophages and dendritic cells, with M2 macrophages predominating. EcoTyper assigned macrophages to different cell states and ecotypes. A total of 168 cell-state-specific M2 macrophage markers were obtained by integrative analysis of scRNA-seq and bulk RNA-seq data. These markers could categorize GC patients into two clusters (clusters A and B) with different survival and M2 macrophages infiltration abundance. Cell adhesion molecules, cytokine-cytokine receptor interaction, JAK/STAT pathway, MAPK pathway were significantly enriched in cluster A, which had worse survival and higher M2 macrophages infiltration. Conclusion In conclusion, this study profiles a single-cell atlas of intratumor heterogeneity and defines the cell states and ecotypes of TAMs in GC. Furthermore, we have identified prognostically relevant cell-state-specific M2 macrophage markers. These findings provide novel insights into the tumor ecosystem and cancer progression.
Collapse
Affiliation(s)
- A-Kao Zhu
- Department of Colorectal Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, People’s Republic of China
| | - Guang-Yao Li
- Department of General Surgery, The Second People’s Hospital of Wuhu, Wuhu, 241000, People’s Republic of China
| | - Fang-Ci Chen
- The Fourth School of Clinical Medicine, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310006, People’s Republic of China
| | - Jia-Qi Shan
- The Fourth School of Clinical Medicine, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310006, People’s Republic of China
| | - Yu-Qiang Shan
- The Fourth School of Clinical Medicine, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310006, People’s Republic of China
- Department of Gastrointestinal Surgery, Hangzhou First People’s Hospital Affiliated to Westlake University School of Medicine, Hangzhou, 310006, People’s Republic of China
| | - Chen-Xi Lv
- The Fourth School of Clinical Medicine, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310006, People’s Republic of China
| | - Zhi-Qiang Zhu
- Department of General Surgery, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, People’s Republic of China
| | - Yi-Ren He
- Department of General Surgery, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, People’s Republic of China
| | - Lu-Lu Zhai
- Department of General Surgery, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, People’s Republic of China
| |
Collapse
|
40
|
Yao S, Li K, Li T, Yu X, Kuan PF, Wang X. GPS-Net: Discovering prognostic pathway modules based on network regularized kernel learning. Am J Hum Genet 2024; 111:2826-2838. [PMID: 39510078 PMCID: PMC11639089 DOI: 10.1016/j.ajhg.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 11/15/2024] Open
Abstract
The search for prognostic biomarkers capable of predicting patient outcomes, by analyzing gene expression in tissue samples and other molecular profiles, remains largely focused on single-gene-based or global-gene-search approaches. Gene-centric approaches, while foundational, fail to capture the higher-order dependencies that reflect the activities of co-regulated processes, pathway alterations, and regulatory networks, all of which are crucial in determining the patient outcomes in complex diseases like cancer. Here, we introduce GPS-Net, a computational framework that fills the gap in efficiently identifying prognostic modules by incorporating the holistic pathway structures and the network of gene interactions. By innovatively incorporating advanced multiple kernel learning techniques and network-based regularization, the proposed method not only enhances the accuracy of biomarker and pathway identification but also significantly reduces computational complexity, as demonstrated by extensive simulation studies. Applying GPS-Net, we identified key pathways that are predictive of patient outcomes in a cancer immunotherapy study. Overall, our approach provides a novel framework that renders genome-wide pathway-level prognostic analysis both feasible and scalable, synergizing both mechanism-driven and data-driven methodologies for precision genomics.
Collapse
Affiliation(s)
- Sijie Yao
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institution, Tampa, FL 33612, USA
| | - Kaiqiao Li
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Tingyi Li
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institution, Tampa, FL 33612, USA
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institution, Tampa, FL 33612, USA
| | - Pei Fen Kuan
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institution, Tampa, FL 33612, USA.
| |
Collapse
|
41
|
Busselaar J, Sijbranda M, Borst J. The importance of type I interferon in orchestrating the cytotoxic T-cell response to cancer. Immunol Lett 2024; 270:106938. [PMID: 39490629 DOI: 10.1016/j.imlet.2024.106938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Both type I interferon (IFN-I) and CD4+ T-cell help are required to generate effective CD8+ T-cell responses to cancer. We here outline based on existing literature how IFN-I signaling and CD4+ T-cell help are connected. Both impact on the functional state of dendritic cells (DCs), particularly conventional (c)DC1. The cDC1s are critical for crosspresentation of cell-associated antigens and for delivery of CD4+ T-cell help for cytotoxic T-lymphocyte (CTL) effector and memory differentiation. In infection, production of IFN-I is prompted by pathogen-associated molecular patterns (PAMPs), while in cancer it relies on danger-associated molecular patterns (DAMPs). IFN-I production by tumor cells and pDCs in the tumor micro-environment (TME) is often limited. IFN-I signals increase the ability of migratory cDC1s and cDC2s to transport tumor antigens to tumor-draining lymph nodes (tdLNs). IFN-I also enables cDC1s to form and sustain the platform for help delivery by stimulating the production of chemokines that attract CD4+ and CD8+ T cells. IFN-I promotes delivery of help in concert with CD40 signals by additive and synergistic impact on cross-presentation and provision of critical costimulatory and cytokine signals for CTL effector and memory differentiation. The scenario of CD4+ T-cell help therefore depends on IFN-I signaling. This scenario can play out in tdLNs as well as in the TME, thereby contributing to the cancer immunity cycle. The collective observations may explain why both IFN-I and CD4+ T-cell help signatures in the TME correlate with good prognosis and response to PD-1 targeting immunotherapy in human cancer. They also may explain why a variety of tumor types in which IFN-I signaling is attenuated, remain devoid of functional CTLs.
Collapse
Affiliation(s)
- Julia Busselaar
- Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Merel Sijbranda
- Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Jannie Borst
- Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| |
Collapse
|
42
|
Ren H, Du MZ, Liao Y, Zu R, Rao L, Xiang R, Zhang X, Liu S, Zhang P, Leng P, Qi L, Luo H. Deciphering the Significance of Platelet-Derived Chloride Ion Channel Gene (BEST3) Through Platelet-Related Subtypes Mining for Non-Small Cell Lung Cancer. J Cell Mol Med 2024; 28:e70233. [PMID: 39708330 DOI: 10.1111/jcmm.70233] [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: 03/21/2024] [Revised: 10/04/2024] [Accepted: 11/08/2024] [Indexed: 12/23/2024] Open
Abstract
This study investigates platelet-related subtypes in non-small cell lung cancer (NSCLC) and seeks to identify genes associated with prognosis, focusing on the clinical significance of the chloride ion channel gene BEST3. We utilised sequencing and clinical data from GEO, TCGA and the Xena platform, building a risk model based on genetic features. TCGA and GSE37745 served as training cohorts, while GSE50081, GSE13213, GSE30129 and GSE42127 were validation cohorts. Immunotherapy datasets (GSE135222, TCGA-SKCM) were also analysed. Differentially expressed genes (DEGs) were identified using Limma, subtypes through ConsensusClusterPlus and key prognostic genes using COX regression, Random Forest and LASSO-COX. BEST3 expression was validated by flow cytometry (FCM) and functional assays in A549 cells with lentiviral overexpression evaluated its impact on apoptosis, proliferation and migration. Three platelet-related subtypes were identified, with ten key prognostic genes (including BEST3). Gene Ontology (GO) analysis showed six genes involved in platelet pathways. BEST3 was highly expressed in the platelet subtype 1. Flow cytometry confirmed elevated BEST3 levels in NSCLC (35.9% vs. 27.3% in healthy individuals). Overexpression of BEST3 in NSCLC cells suppressed apoptosis and promoted proliferation and migration. The discovery of three platelet subtypes and the role of BEST3 in promoting tumour growth and migration highlights its potential as a therapeutic target and prognostic marker in NSCLC.
Collapse
Affiliation(s)
- Hanxiao Ren
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People's Republic of China
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of the University of Electronic Science and Technology of China, Chengdu, China
| | - Meng-Ze Du
- School of Health and Medical Technology, Chengdu Neusoft University, Chengdu, Sichuan Province, People's Republic of China
| | - Yulin Liao
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of the University of Electronic Science and Technology of China, Chengdu, China
| | - Ruiling Zu
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of the University of Electronic Science and Technology of China, Chengdu, China
| | - Lubei Rao
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of the University of Electronic Science and Technology of China, Chengdu, China
| | - Run Xiang
- Department of Thoracic Surgery, Sichuan Cancer Hospital, Affiliate to the School of Medicine, The University of Electronic Science and Technology of China, Chengdu, China
| | - Xingmei Zhang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People's Republic of China
| | - Shan Liu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People's Republic of China
| | - Peiyin Zhang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People's Republic of China
| | - Ping Leng
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People's Republic of China
| | - Ling Qi
- Department of Core Medical Laboratory, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Huaichao Luo
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of the University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
43
|
Xia S, Chen L, Yu M, Li J, Chen J, Xu F, Ni M, Liu C, Wu X, Chen X, Li J. Genetic and therapeutic heterogeneity shape the baseline and longitudinal immune ecosystem of ovarian clear cell carcinoma. J Immunother Cancer 2024; 12:e010069. [PMID: 39608974 PMCID: PMC11603735 DOI: 10.1136/jitc-2024-010069] [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: 07/11/2024] [Accepted: 11/06/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Ovarian clear cell carcinoma (OCCC) is a rare and chemo-resistant subtype of ovarian cancer. While immunotherapy has demonstrated effectiveness in some OCCC cases, the mechanisms for heterogeneous immunoreactivity and potential combinatory strategies remain unclear. METHODS Tumor samples from 13 patients with OCCC underwent single-cell mRNA-seq and TCR-seq to generate 1 40 683 cells transcriptome, while additionally 31 formalin-fixed paraffin-embedded samples were used for immunohistochemistry. Spatial transcriptomics of two OCCC samples and bulk RNA-seq of 58 patients were incorporated for spatial and interpatient level explorations. Serum tumor markers and radiologic images of three patients with OCCC who received combinatory VEGF and PD-1 inhibition were retrospectively analyzed. RESULTS OCCC exhibited a dynamic immune architecture shaped by genetic and therapeutic pressure. ARID1A mutation linked to baseline immune activation, correlated with an enrichment of neoantigen-reactive CXCL13+ CTLA4+ CD8+ T cells (p<0.001) and enhanced FASLG-FAS interactions. Recurrent OCCC was fibrotic, angiogenic, and immunosuppressive, exhibiting metabolic reprogramming towards activated activity in fatty acid metabolism. High CD36 (log-rank p=0.012, HR: 4.515) and CD47 expression (log-rank p=0.037, HR: 3.246) indicated worse progression-free survival. Treatment with bevacizumab increased intratumoral T cell infiltration and activated T cell interferon-γ signaling. Retrospective analysis of clinical cases revealed that combination therapy with anti-VEGF (vascular endothelial growth factor) and anti-PD-1 agents exerted clinical benefits in patients with OCCC with persistent, recurrent, and metastatic disease. CONCLUSIONS ARID1A mutation correlated with OCCC baseline immune activation. Stromal reconstruction and tumor metabolic reprogramming functioned as key processes of OCCC dynamic progression. VEGF inhibition remodeled OCCC stroma, restored T cell function and potentiated immunotherapy. CD36 and CD47 might be potential therapeutic targets for recurrent OCCC.
Collapse
Affiliation(s)
- Siyu Xia
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Lihua Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Min Yu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Jiana Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Jiaxin Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Fei Xu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Mengdong Ni
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Chaohua Liu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaohua Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaojun Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Jiajia Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| |
Collapse
|
44
|
Elorbany S, Berlato C, Carnevalli LS, Maniati E, Barry ST, Wang J, Manchanda R, Kzhyshkowska J, Balkwill F. Immunotherapy that improves response to chemotherapy in high-grade serous ovarian cancer. Nat Commun 2024; 15:10144. [PMID: 39578450 PMCID: PMC11584700 DOI: 10.1038/s41467-024-54295-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 11/01/2024] [Indexed: 11/24/2024] Open
Abstract
Single-cell RNA sequencing (scRNAseq) of tumour-infiltrating immune cells in high-grade serous ovarian cancer (HGSOC) omental biopsies reveals potential targets that could enhance response to neo-adjuvant chemotherapy (NACT). Analysis of 64,097 cells identifies NACT-induced overexpression of stabilin-1 (clever-1) on macrophages and FOXP3 in Tregs that is confirmed at the protein level. STAB1 inhibition in vitro induces anti-tumour macrophages. FOXP3 anti-sense oligonucleotide (FOXP3-ASO), repolarises Tregs to an effector T cell phenotype. ScRNAseq on 69,781 cells from an HGSOC syngeneic mouse model recapitulates the patients' data. Combining chemotherapy with anti-stabilin1 antibody and/or Foxp3-ASO significantly increases survival of mice with established peritoneal disease in two HGSOC syngeneic models and progression-free survival in a third model. Long-term survivors (300 days + ) are resistant to tumour rechallenge. Anti-stabilin1 antibody enriches the tumours with CXCL9+ macrophages and Foxp3-ASO increases TBET cell infiltration. Our results suggest that targeting these molecules in immune cells may improve chemotherapy response in patients.
Collapse
MESH Headings
- Female
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/pathology
- Animals
- Humans
- Mice
- Forkhead Transcription Factors/metabolism
- Forkhead Transcription Factors/genetics
- Immunotherapy/methods
- Cell Line, Tumor
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/drug effects
- Cell Adhesion Molecules, Neuronal/metabolism
- Cell Adhesion Molecules, Neuronal/genetics
- Cell Adhesion Molecules, Neuronal/immunology
- Macrophages/immunology
- Macrophages/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/drug effects
- Cystadenocarcinoma, Serous/drug therapy
- Cystadenocarcinoma, Serous/immunology
- Cystadenocarcinoma, Serous/pathology
- Neoadjuvant Therapy/methods
- Chemokine CXCL9/metabolism
- Chemokine CXCL9/genetics
- Single-Cell Analysis
Collapse
Affiliation(s)
- Samar Elorbany
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK.
| | - Chiara Berlato
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
| | | | - Eleni Maniati
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
| | - Simon T Barry
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | - Jun Wang
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
| | - Ranjit Manchanda
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
- Department of Gynaecological Oncology, Barts Health NHS Trust, London, UK
| | - Julia Kzhyshkowska
- Institute of Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunosciences (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Frances Balkwill
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
| |
Collapse
|
45
|
Wang MG, Chen L, Zhang XF. Dual decoding of cell types and gene expression in spatial transcriptomics with PANDA. Nucleic Acids Res 2024; 52:12173-12190. [PMID: 39404057 PMCID: PMC11551751 DOI: 10.1093/nar/gkae876] [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: 06/26/2024] [Revised: 08/24/2024] [Accepted: 09/24/2024] [Indexed: 11/12/2024] Open
Abstract
Sequencing-based spatial transcriptomics technologies have revolutionized our understanding of complex biological systems by enabling transcriptome profiling while preserving spatial context. However, spot-level expression measurements often amalgamate signals from diverse cells, obscuring potential heterogeneity. Existing methods aim to deconvolute spatial transcriptomics data into cell type proportions for each spot using single-cell RNA sequencing references but overlook cell-type-specific gene expression, essential for uncovering intra-type heterogeneity. We present PANDA (ProbAbilistic-based decoNvolution with spot-aDaptive cell type signAtures), a novel method that concurrently deciphers spot-level gene expression into both cell type proportions and cell-type-specific gene expression. PANDA integrates archetypal analysis to capture within-cell-type heterogeneity and dynamically learns cell type signatures for each spot during deconvolution. Simulations demonstrate PANDA's superior performance. Applied to real spatial transcriptomics data from diverse tissues, including tumor, brain, and developing heart, PANDA reconstructs spatial structures and reveals subtle transcriptional variations within specific cell types, offering a comprehensive understanding of tissue dynamics.
Collapse
Affiliation(s)
- Meng-Guo Wang
- School of Mathematics and Statistics, and Hubei Key Lab–Math. Sci., Central China Normal University, Wuhan 430079, Hubei, China
| | - Luonan Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, Zhejiang, China
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai 519031, Guangdong, China
| | - Xiao-Fei Zhang
- School of Mathematics and Statistics, and Hubei Key Lab–Math. Sci., Central China Normal University, Wuhan 430079, Hubei, China
- Key Laboratory of Nonlinear Analysis & Applications (Ministry of Education), Central China Normal University, Wuhan 430079, Hubei, China
| |
Collapse
|
46
|
Su A, Lee H, Tran M, Cruz RD, Sathe A, Bai X, Wichmann I, Pflieger L, Moulton B, Barker T, Haslem D, Jones D, Nadauld L, Nguyen Q, Ji HP, Rhodes T. The single-cell spatial landscape of stage III colorectal cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.07.622577. [PMID: 39605367 PMCID: PMC11601238 DOI: 10.1101/2024.11.07.622577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
We conducted a spatial analysis using imaging mass cytometry applied to stage III colorectal adenocarcinomas. This study used multiplexed markers to distinguish individual cells and their spatial organization from 52 colorectal cancers. We determined the landscape features of cellular spatial features in the CRC tumor microenvironment. This spatial single-cell analysis identified 10 unique cell phenotypes in the tumor microenvironment that included stromal and immune cells with a subset which had a proliferative phenotype. These special features included spatial neighborhood interactions between single cells as well as different tissue niches, especially the tumor infiltrating lymphocyte regions. We applied a robust statistical analysis to identify significant correlations of cell features with phenotypes such as microsatellite instability or recurrence. We determined that microsatellite stable (MSS) colorectal cancers had an increased risk of recurrence if they had the following features: 1) a low level of stromal tumor-infiltrating lymphocytes, and 2) low interactions between CD4+ T cells and stromal cells. Our results point to the utility of spatial single-cell interaction analysis in defining novel features of the tumor immune microenvironments and providing useful clinical cell-related spatial biomarkers.
Collapse
Affiliation(s)
- Andrew Su
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, United States
- Institute for Molecular Bioscience, The University of Queensland, QLD 4072, Australia
| | - HoJoon Lee
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, United States
| | - Minh Tran
- Institute for Molecular Bioscience, The University of Queensland, QLD 4072, Australia
| | - Richard D. Cruz
- Intermountain Healthcare, Saint George, UT, 84770, United States
| | - Anuja Sathe
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, United States
| | - Xiangqi Bai
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, United States
| | - Ignacio Wichmann
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, United States
- Division of Obstetrics and Gynecology, Department of Obstetrics, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - Lance Pflieger
- Intermountain Healthcare, Saint George, UT, 84770, United States
| | - Bryce Moulton
- Intermountain Healthcare, Saint George, UT, 84770, United States
| | - Tyler Barker
- Intermountain Healthcare, Saint George, UT, 84770, United States
| | - Derrick Haslem
- Intermountain Healthcare, Saint George, UT, 84770, United States
| | - David Jones
- Intermountain Healthcare, Saint George, UT, 84770, United States
| | - Lincoln Nadauld
- Intermountain Healthcare, Saint George, UT, 84770, United States
| | - Quan Nguyen
- Institute for Molecular Bioscience, The University of Queensland, QLD 4072, Australia
| | - Hanlee P. Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, United States
| | - Terence Rhodes
- Intermountain Healthcare, Saint George, UT, 84770, United States
| |
Collapse
|
47
|
Wang Y, Hu M, Finn OJ, Wang XS. Tumor-Associated Antigen Burden Correlates with Immune Checkpoint Blockade Benefit in Tumors with Low Levels of T-cell Exhaustion. Cancer Immunol Res 2024; 12:1589-1602. [PMID: 39137006 PMCID: PMC11534523 DOI: 10.1158/2326-6066.cir-23-0932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 03/20/2024] [Accepted: 08/09/2024] [Indexed: 09/14/2024]
Abstract
Tumor-associated antigens (TAA) are important targets for cancer vaccines. However, TAA-based vaccines have not yet achieved their full potential in clinical trials. In contrast, immune checkpoint blockade (ICB) has emerged as an effective therapy, leading to durable responses in selected patients with cancer. To date, few generalizable associations between TAAs and ICB benefit have been reported, with most studies focusing on melanoma, which has the highest mutation rate in cancer. In this study, we developed a TAA burden (TAB) algorithm based on known and putative TAAs and investigated the association of TAB with ICB benefit. Analysis of the IMvigor210 patient cohort of urothelial carcinoma treated with anti-PDL1 revealed that high tumor mutation burden weakened the association of TAB with ICB benefit. Furthermore, TAB correlated with ICB efficacy in tumors characterized by negative PDL1 staining on immune cells; however, high levels of PDL1 staining on immune cells were linked to T-cell exhaustion. Validation across independent clinical datasets-including urothelial carcinoma cohorts treated with anti-PD1/PDL1 agents and neoadjuvant anti-PD1 trials for head and neck cancers-corroborated the finding that TAB correlates with ICB benefit in tumors with low T-cell exhaustion. Pan-cancer analyses revealed that in most cancer entities, tumors with higher T-cell exhaustion exhibited lower TAB levels, implying possible immunoediting of TAAs in tumors with established antitumor immunity. Our study challenges the prevailing notion of a lack of association between TAAs and ICB response. It also underscores the need for future investigations into the immunogenicity of TAAs and TAA-based vaccine strategies in tumors with low levels of T-cell exhaustion.
Collapse
Affiliation(s)
- Yue Wang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
| | - Mengying Hu
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
| | - Olivera J Finn
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, U.S.A
| | - Xiao-Song Wang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, U.S.A
| |
Collapse
|
48
|
Dong W, Sheng J, Cui JZM, Zhao H, Wong STC. Systems immunology insights into brain metastasis. Trends Immunol 2024; 45:903-916. [PMID: 39443266 PMCID: PMC12049182 DOI: 10.1016/j.it.2024.09.010] [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: 08/05/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024]
Abstract
Brain metastasis poses formidable clinical challenges due to its intricate interactions with the brain's unique immune environment, often resulting in poor prognoses. This review delves into systems immunology's role in uncovering the dynamic interplay between metastatic cancer cells and brain immunity. Leveraging spatial and single-cell technologies, along with advanced computational modeling, systems immunology offers unprecedented insights into mechanisms of immune evasion and tumor proliferation. Recent studies highlight potential immunotherapeutic targets, suggesting strategies to boost antitumor immunity and counteract cancer cell evasion in the brain. Despite substantial progress, challenges persist, particularly in accurately simulating human conditions. This review underscores the need for interdisciplinary collaboration to harness systems immunology's full potential, aiming to dramatically improve outcomes for patients with brain metastasis.
Collapse
Affiliation(s)
- Wenjuan Dong
- Department of Systems Medicine and Bioengineering and T. T. and W. F. Chao Center for BRAIN, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Jianting Sheng
- Department of Systems Medicine and Bioengineering and T. T. and W. F. Chao Center for BRAIN, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Johnny Z M Cui
- Department of Systems Medicine and Bioengineering and T. T. and W. F. Chao Center for BRAIN, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Hong Zhao
- Department of Systems Medicine and Bioengineering and T. T. and W. F. Chao Center for BRAIN, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA.
| | - Stephen T C Wong
- Department of Systems Medicine and Bioengineering and T. T. and W. F. Chao Center for BRAIN, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA.
| |
Collapse
|
49
|
Fiorini MR, Dilliott AA, Thomas RA, Farhan SMK. Transcriptomics of Human Brain Tissue in Parkinson's Disease: a Comparison of Bulk and Single-cell RNA Sequencing. Mol Neurobiol 2024; 61:8996-9015. [PMID: 38578357 PMCID: PMC11496323 DOI: 10.1007/s12035-024-04124-5] [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: 10/11/2023] [Accepted: 03/12/2024] [Indexed: 04/06/2024]
Abstract
Parkinson's disease (PD) is a chronic and progressive neurodegenerative disease leading to motor dysfunction and, in some cases, dementia. Transcriptome analysis is one promising approach for characterizing PD and other neurodegenerative disorders by informing how specific disease events influence gene expression and contribute to pathogenesis. With the emergence of single-cell and single-nucleus RNA sequencing (scnRNA-seq) technologies, the transcriptional landscape of neurodegenerative diseases can now be described at the cellular level. As the application of scnRNA-seq is becoming routine, it calls to question how results at a single-cell resolution compare to those obtained from RNA sequencing of whole tissues (bulk RNA-seq), whether the findings are compatible, and how the assays are complimentary for unraveling the elusive transcriptional changes that drive neurodegenerative disease. Herein, we review the studies that have leveraged RNA-seq technologies to investigate PD. Through the integration of bulk and scnRNA-seq findings from human, post-mortem brain tissue, we use the PD literature as a case study to evaluate the compatibility of the results generated from each assay and demonstrate the complementarity of the sequencing technologies. Finally, through the lens of the PD transcriptomic literature, we evaluate the current feasibility of bulk and scnRNA-seq technologies to illustrate the necessity of both technologies for achieving a comprehensive insight into the mechanism by which gene expression promotes neurodegenerative disease. We conclude that the continued application of both assays will provide the greatest insight into neurodegenerative disease pathology, providing both cell-specific and whole-tissue level information.
Collapse
Affiliation(s)
- Michael R Fiorini
- The Montreal Neurological Institute-Hospital, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Allison A Dilliott
- The Montreal Neurological Institute-Hospital, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Rhalena A Thomas
- The Montreal Neurological Institute-Hospital, Montreal, QC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
| | - Sali M K Farhan
- The Montreal Neurological Institute-Hospital, Montreal, QC, Canada.
- Department of Human Genetics, McGill University, Montreal, QC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
| |
Collapse
|
50
|
Palaniappan A, Muthamilselvan S, Sarathi A. COADREADx: A comprehensive algorithmic dissection of colorectal cancer unravels salient biomarkers and actionable insights into its discrete progression. PeerJ 2024; 12:e18347. [PMID: 39484215 PMCID: PMC11526798 DOI: 10.7717/peerj.18347] [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: 06/05/2024] [Accepted: 09/27/2024] [Indexed: 11/03/2024] Open
Abstract
Background Colorectal cancer is a common condition with an uncommon burden of disease, heterogeneity in manifestation, and no definitive treatment in the advanced stages. Renewed efforts to unravel the genetic drivers of colorectal cancer progression are paramount. Early-stage detection contributes to the success of cancer therapy and increases the likelihood of a favorable prognosis. Here, we have executed a comprehensive computational workflow aimed at uncovering the discrete stagewise genomic drivers of colorectal cancer progression. Methods Using the TCGA COADREAD expression data and clinical metadata, we constructed stage-specific linear models as well as contrast models to identify stage-salient differentially expressed genes. Stage-salient differentially expressed genes with a significant monotone trend of expression across the stages were identified as progression-significant biomarkers. The stage-salient genes were benchmarked using normals-augmented dataset, and cross-referenced with existing knowledge. The candidate biomarkers were used to construct the feature space for learning an optimal model for the digital screening of early-stage colorectal cancers. The candidate biomarkers were also examined for constructing a prognostic model based on survival analysis. Results Among the biomarkers identified are: CRLF1, CALB2, STAC2, UCHL1, KCNG1 (stage-I salient), KLHL34, LPHN3, GREM2, ADCY5, PLAC2, DMRT3 (stage-II salient), PIGR, HABP2, SLC26A9 (stage-III salient), GABRD, DKK1, DLX3, CST6, HOTAIR (stage-IV salient), and CDH3, KRT80, AADACL2, OTOP2, FAM135B, HSP90AB1 (top linear model genes). In particular the study yielded 31 genes that are progression-significant such as ESM1, DKK1, SPDYC, IGFBP1, BIRC7, NKD1, CXCL13, VGLL1, PLAC1, SPERT, UPK2, and interestingly three members of the LY6G6 family. Significant monotonic linear model genes included HIGD1A, ACADS, PEX26, and SPIB. A feature space of just seven biomarkers, namely ESM1, DHRS7C, OTOP3, AADACL2, LPHN3, GABRD, and LPAR1, was sufficient to optimize a RandomForest model that achieved > 98% balanced accuracy (and performant recall) of cancer vs. normal on external validation. Design of an optimal multivariate model based on survival analysis yielded a prognostic panel of three stage-IV salient genes, namely HOTAIR, GABRD, and DKK1. Based on the above sparse signatures, we have developed COADREADx, a web-server for potentially assisting colorectal cancer screening and patient risk stratification. COADREADx provides uncertainty measures for its predictions and needs clinical validation. It has been deployed for experimental non-commercial use at: https://apalanialab.shinyapps.io/coadreadx/.
Collapse
Affiliation(s)
- Ashok Palaniappan
- Systems Computational Biology Lab, Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Sangeetha Muthamilselvan
- Systems Computational Biology Lab, Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Arjun Sarathi
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|