51
|
Zhang X, Yang Z, Xie X, Li J, Xiao Q, Xu G, Ma B, Xie X, Liu Y, Zhai L, Tang Y, Fu H, He S, Liu T, Huang D, Zeng C, Zhou Y, Hu R, Guo B, Wang C, Liang S, Luo Q, Lv J, Nan Y, Li J, Li Q, Wang S, Wu Y, Liu Y. The single-cell immune landscape of HIV-associated aggressive B-cell lymphoma. JOURNAL OF THE NATIONAL CANCER CENTER 2025; 5:221-235. [PMID: 40265092 PMCID: PMC12010387 DOI: 10.1016/j.jncc.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/27/2025] [Accepted: 02/08/2025] [Indexed: 04/24/2025] Open
Abstract
Background Human immunodeficiency virus (HIV)-associated lymphomas (HAL), mainly aggressive B-cell lymphomas, pose a significant challenge in cancer research due to their multifaceted pathogenesis and aggressive clinical course. Despite the clinical importance, the genomic and immune characteristics of these lymphomas remain poorly elucidated. Methods We employed single-cell RNA sequencing (scRNA-seq) on lymph node samples from aggressive B-cell lymphomas, mainly including 6 cases of diffuse large B-cell lymphoma (DLBCL) and 5 cases of Burkitt lymphoma (BL) from people living with HIV (PLWH), along with 3 DLBCL cases from individuals without HIV for comparison. Results Malignant B cells in HAL consistently exhibited high proliferative and oxidative phosphorylation (OXPHOS)-type metabolic signatures. Moreover, these cells demonstrated loss expression of major histocompatibility complex class I (MHC-I), strategically reducing tumor immunogenicity. HAL harbors special populations of naive and atypical memory B cells that exhibited high metabolic and immune-activated transcriptional profiles. Additionally, HAL exhibited senescence-like dysfunction in T cells, characterized by the reductions in regulatory activity of Treg and cytotoxic activity of CD8+ T cells, as well as decreases expression of IL7R genes and increases expression of FOS and FOSB genes. Our immunofluorescence results showed that the cytotoxic CD8+ T cells in HAL may have a dysfunction of lytic granule polarization. Furthermore, macrophages from HAL exhibited stronger immunosuppressive transcriptional characteristics, and a robust immunosuppressive SPP1-CD44 interaction was predicted between C1QA+ macrophages and T cells. Conclusions Our findings clearly indicate that HAL differs significantly from non-HAL, ranging from malignant B cells to the immune microenvironment. This study provides a comprehensive single-cell atlas of HIV-associated aggressive B-cell lymphomas, offering new insights into aggressiveness and immune evasion observed in HAL.
Collapse
Affiliation(s)
- Xiaomei Zhang
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Zailin Yang
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaoqing Xie
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Jun Li
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Qing Xiao
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Guofa Xu
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
- Department of Hematology and Medical Oncology, Chongqing University Fuling Hospital, Chongqing, China
| | - Ben Ma
- Department of Integrated, Chongqing University Cancer Hospital, Chongqing, China
| | - Xudong Xie
- Department of Integrated, Chongqing University Cancer Hospital, Chongqing, China
| | - Yi Liu
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Liuyue Zhai
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yifeng Tang
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Huihui Fu
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Sanxiu He
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Tingting Liu
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Dehong Huang
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Censi Zeng
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yixing Zhou
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Renzhi Hu
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Binling Guo
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Chaoyu Wang
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Shunsi Liang
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Qin Luo
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Jing Lv
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yingyu Nan
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Jieping Li
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Qiying Li
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Shengqiang Wang
- Department of Integrated, Chongqing University Cancer Hospital, Chongqing, China
| | - Yongzhong Wu
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yao Liu
- Department of Hematology and Oncology, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
52
|
Moon CY, Belabed M, Park MD, Mattiuz R, Puleston D, Merad M. Dendritic cell maturation in cancer. Nat Rev Cancer 2025; 25:225-248. [PMID: 39920276 PMCID: PMC11954679 DOI: 10.1038/s41568-024-00787-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 02/09/2025]
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that are present at low abundance in the circulation and tissues; they serve as crucial immune sentinels by continually sampling their environment, migrating to secondary lymphoid organs and shaping adaptive immune responses through antigen presentation. Owing to their ability to orchestrate tolerogenic or immunogenic responses to a specific antigen, DCs have a pivotal role in antitumour immunity and the response to immune checkpoint blockade and other immunotherapeutic approaches. The multifaceted functions of DCs are acquired through a complex, multistage process called maturation. Although the role of inflammatory triggers in driving DC maturation was established decades ago, less is known about DC maturation in non-inflammatory contexts, such as during homeostasis and in cancer. The advent of single-cell technologies has enabled an unbiased, high-dimensional characterization of various DC states, including mature DCs. This approach has clarified the molecular programmes associated with DC maturation and also revealed how cancers exploit these pathways to subvert immune surveillance. In this Review, we discuss the mechanisms by which cancer disrupts DC maturation and highlight emerging therapeutic opportunities to modulate DC states. These insights could inform the development of DC-centric immunotherapies, expanding the arsenal of strategies to enhance antitumour immunity.
Collapse
Affiliation(s)
- Chang Yoon Moon
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meriem Belabed
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Park
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raphaël Mattiuz
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Puleston
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
53
|
Gu Y, Zhang Z, Huang H, Zhu W, Liu H, Zhang R, Weng N, Sun X. The dual role of CXCL9/SPP1 polarized tumor-associated macrophages in modulating anti-tumor immunity in hepatocellular carcinoma. Front Immunol 2025; 16:1528103. [PMID: 40230843 PMCID: PMC11994707 DOI: 10.3389/fimmu.2025.1528103] [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: 11/14/2024] [Accepted: 03/13/2025] [Indexed: 04/16/2025] Open
Abstract
Introduction The main challenge for cancer therapy lies in immuno-suppressive tumor micro-environment. Reprogramming tumor-associated macrophages (TAMs) into an anti-tumor phenotype is a promising strategy. Methods A comprehensive analysis by combing multi-regional single-cell, bulk and spatial transcriptome profiling with radiomics characterization was conducted to dissect the heterogeneity of TAMs and resolve the landscape of the CXCL9:SPP1 (CS) macrophage polarity in HCC. Results TAMs were particularly increased in HCC. SPP1+ TAMs and CXCL9+ TAMs were identified as the dominant subtypes with different evolutionary trajectories. SPP1+ TAMs, located in the tumor core, co-localized with cancer-associated fibroblasts to promote tumor growth and further contributed to worse prognosis. In contrast, CXCL9+ TAMs, located in the peritumoral region, synergized with CD8+ T cells to create an immunostimulatory micro-environment. For the first time, we explored the applicability of CS polarity in HCC tumors and revealed several key transcription factors involved in shaping this polarity. Moreover, CS polarity could serve as a potential indicator of prognostic and micro-environmental status for HCC patients. Based on medical imaging data, we developed a radiomics tool, RCSP (Radiogenomics-based CXCL9/SPP1 Polarity), to assist in non-invasively predicting the CS polarity in HCC patients. Conclusion Our research sheds light on the regulatory roles of SPP1+ TAMs and CXCL9+ TAMs in the micro-environment and provides new therapeutic targets or insights for the reprogramming of targeted macrophages in HCC.
Collapse
Affiliation(s)
- Yu Gu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Zhihui Zhang
- College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hao Huang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Wenyong Zhu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Hongjia Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Rongxin Zhang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Nan Weng
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Xiao Sun
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| |
Collapse
|
54
|
Lee OV, Ji DX, Rosa BA, Jaye DL, Suliman S, Mitreva M, Gabay C, Vance RE, Kotov DI. Interleukin-1 receptor antagonist is a conserved early factor for exacerbating tuberculosis susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.27.564420. [PMID: 37961447 PMCID: PMC10634924 DOI: 10.1101/2023.10.27.564420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Mycobacterium tuberculosis (Mtb) causes 1.25 million deaths a year. However, tuberculosis (TB) pathogenesis remains poorly understood and is not fully recapitulated in standard mouse models. Here we find that gene signatures from three different Mtb-susceptible mouse models predict active TB disease in humans significantly better than a signature from resistant C57BL/6 (B6) mice. Conserved among susceptible mice, non-human primates, and humans, but largely absent from B6 mice, was Mtb-induced differentiation of macrophages into an Spp1 + differentiation state. Spp1 + macrophages expressed high levels of immunosuppressive molecules including IL-1 receptor antagonist (IL-1Ra). IL-1Ra was previously reported to cause Mtb susceptibility in one mouse model, but whether IL-1Ra is broadly important remains uncertain. Here we report that enhancement of IL-1 signaling via deletion of IL-Ra promoted bacterial control across three susceptible mouse models. We found IL-1 signaling amplified production of multiple cytokines by lymphoid and stromal cells, providing a multifactorial mechanism for how IL-1 promotes Mtb control. Our results indicate that myeloid cell expression of immunosuppressive molecules, in particular IL-1 receptor antagonist, is a conserved early mechanism limiting Mtb control in mice, non-human primates, and humans.
Collapse
Affiliation(s)
- Ophelia V. Lee
- Divison of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Daisy X. Ji
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Bruce A. Rosa
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - David L. Jaye
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Sara Suliman
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, 94115, USA
| | - Makedonka Mitreva
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Cem Gabay
- Division of Rheumatology, Department of Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Russell E. Vance
- Divison of Immunology and Molecular Medicine, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Dmitri I. Kotov
- Division of Infectious Diseases, Department of Medicine, Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
55
|
Kumagai S, Momoi Y, Nishikawa H. Immunogenomic cancer evolution: A framework to understand cancer immunosuppression. Sci Immunol 2025; 10:eabo5570. [PMID: 40153489 DOI: 10.1126/sciimmunol.abo5570] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 06/26/2024] [Accepted: 03/05/2025] [Indexed: 03/30/2025]
Abstract
The process of tumor development involves tumor cells eluding detection and suppression of immune responses, which can cause decreased tumor cell antigenicity, expression of immunosuppressive molecules, and immunosuppressive cell recruitment to the tumor microenvironment (TME). Immunologically and genomically integrated analysis (immunogenomic analysis) of patient specimens has revealed that oncogenic aberrant signaling is involved in both carcinogenesis and immune evasion. In noninflamed cancers such as epidermal growth factor receptor (EGFR)-mutated lung cancers, genetic abnormalities in cancer cells contribute to the formation of an immunosuppressive TME by recruiting immunosuppressive cells, which cannot be fully explained by the cancer immunoediting hypothesis. This review summarizes the latest findings regarding the links between cancer genetic abnormalities and immunosuppression causing clinical resistance to immunotherapy. We propose the concepts of immunogenomic cancer evolution, in which cancer cell genomic evolution shapes the immunosuppressive TME, and immunogenomic precision medicine, in which cancer immunotherapy can be combined with molecularly targeted reagents that modulate the immunosuppressive TME.
Collapse
Affiliation(s)
- Shogo Kumagai
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
- Division of Cancer Immunology, Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Chiba 277-8577, Japan
- Division of Cellular Signaling, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
| | - Yusaku Momoi
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
- Department of Tumor Pathology, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Division of Cancer Immune Multicellular System Regulation, Center for Cancer Immunotherapy and Immunology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
- Kindai University Faculty of Medicine, Osaka-sayama 589-8511, Japan
| |
Collapse
|
56
|
Golfinos-Owens AE, Lozar T, Khatri P, Hu R, Harari PM, Lambert PF, Fitzpatrick MB, Dinh HQ. Integrated single-cell and spatial analysis identifies context-dependent myeloid-T cell interactions in head and neck cancer immune checkpoint blockade response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.644582. [PMID: 40196610 PMCID: PMC11974738 DOI: 10.1101/2025.03.24.644582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Background Approximately 15-20% of head and neck cancer squamous cell carcinoma (HNSCC) patients respond favorably to immune checkpoint blockade (ICB). Previous single-cell RNA-Seq (scRNA-Seq) studies identified immune features, including macrophage subset ratios and T-cell subtypes, in HNSCC ICB response. However, the spatial features of HNSCC-infiltrated immune cells in response to ICB treatment need to be better characterized. Methods Here, we perform a systematic evaluation of cell interactions between immune cell types within the tumor microenvironment using spatial omics data using complementary techniques from both 10X Visium spot-based spatial transcriptomics and Nanostring CosMx single-cell spatial omics with RNA gene panel including 435 ligands and receptors. In this study, we used integrated bioinformatics analyses to identify cellular neighborhoods of co-localizing cell types in single-cell spatial transcriptomics and proteomics data. In addition, we used both publicly available scRNA-Seq and in-house spatial RNA-Seq data to identify spatially constrained Ligand-Receptor interactions in Responder patients. Results With 522,399 single cells profiled with both RNA and protein from 26 patients, in addition to spot-resolved spatial RNA-Seq from 8 patients treated with ICB together with bioinformatics analysis of publicly available single-cell and bulk RNA-Seq, we have identified a spatial and cell-type specific context-dependency of myeloid and T cell interaction difference between Responders and Non-Responders. We defined further cellular neighborhood and the sources of chemokine CXCL9/10-CXCR3 interactions in Responders, emerging targets in ICB, as well as CXCL16-CXCR6, CCL4/5-CCR5, and other underappreciated and potential markers and targets for ICB response in HNSCC. In addition, we have contributed a rich data resource of cell-cell Ligand Receptor interactions for the immunotherapy and HNSCC research community. Discussion Our work provides a comprehensive single-cell and spatial atlas of immune cell interactions that correlate with response to ICB in HNSCC. We showcase how integrating multiple technologies and bioinformatics approaches can provide new insights into potential immune-based biomarkers of ICB response. Our results suggested refining future studies using preclinical animal models in a more context-specific manner to elucidate potential underlying mechanisms that lead to improved ICB responses.
Collapse
Affiliation(s)
- Athena E Golfinos-Owens
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Taja Lozar
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
- University of Ljubljana, Ljubljana, Slovenia
- Department of Surgical Oncology, Institute of Oncology Ljubljana, Ljubljana Slovenia
| | - Parth Khatri
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Rong Hu
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Paul M Harari
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Paul F Lambert
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Megan B Fitzpatrick
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Huy Q Dinh
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| |
Collapse
|
57
|
Wang Q, Yu Y, Ruan L, Huang M, Chen W, Sun X, Liu J, Jiang Z. Integrated single-cell and bulk transcriptomic analysis identifies a novel macrophage subtype associated with poor prognosis in breast cancer. Cancer Cell Int 2025; 25:119. [PMID: 40148933 PMCID: PMC11948682 DOI: 10.1186/s12935-025-03750-w] [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: 11/20/2024] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) are pivotal components of the breast cancer (BC) tumor microenvironment (TME), significantly influencing tumor progression and response to therapy. However, the heterogeneity and specific roles of TAM subpopulations in BC remain inadequately understood. METHODS We performed an integrated analysis of single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing (RNA-seq) data from BC patients to comprehensively characterize TAM heterogeneity. Utilizing the MetaTiME computational framework and consensus clustering, we identified distinct TAM subtypes and assessed their associations with clinical outcomes and treatment responses. A machine learning-based predictive model was developed to evaluate the prognostic significance of TAM-related gene expression profiles. RESULTS Our analysis revealed three distinct TAM subgroups. Notably, we identified a novel macrophage subtype, M_Macrophage-SPP1-C1Q, characterized by high expression of SPP1 and C1QA, representing an intermediate differentiation state with unique proliferative and oncogenic properties. High infiltration of M_Macrophage-SPP1-C1Q was significantly associated with poor overall survival (OS) and chemotherapy resistance in BC patients. We developed a Random Forest (RF)-based predictive model, Macro.RF, which accurately stratified patients based on survival outcomes and chemotherapy responses, independent of established prognostic parameters. CONCLUSION This study uncovers a previously unrecognized TAM subtype that drives poor prognosis in BC. The identification of M_Macrophage-SPP1-C1Q enhances our understanding of TAM heterogeneity within the TME and offers a novel prognostic biomarker. The Macro.RF model provides a robust tool for predicting clinical outcomes and guiding personalized treatment strategies in BC patients.
Collapse
Affiliation(s)
- Qing Wang
- Fujian Medical University, Fuzhou, 350011, China
| | - Yushuai Yu
- Fujian Medical University, Fuzhou, 350011, China
| | - Liqiong Ruan
- Department of Clinical Laboratory, Ningde Municipal Hospital of Ningde Normal University, Ningde, 352100, China
| | | | - Wei Chen
- Department of Breast Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Xiaomei Sun
- Department of Pathology, Ningde Municipal Hospital of Ningde Normal University, Ningde, 352100, China
| | - Jun Liu
- Department of Thyroid and Breast Surgery, Ningde Municipal Hospital of Ningde Normal University, Ningde, 352100, China.
- Department of Breast-Thyroid Surgery, Shanghai General Hospital, Shanghai, 200000, China.
| | - Zirong Jiang
- Department of Thyroid and Breast Surgery, Ningde Municipal Hospital of Ningde Normal University, Ningde, 352100, China.
- Ningde Clinical Medical College of Fujian Medical University, Ningde, 352100, China.
| |
Collapse
|
58
|
Ramirez CFA, Akkari L. Myeloid cell path to malignancy: insights into liver cancer. Trends Cancer 2025:S2405-8033(25)00054-8. [PMID: 40140328 DOI: 10.1016/j.trecan.2025.02.006] [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: 12/03/2024] [Revised: 02/12/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025]
Abstract
Clinically approved treatments for advanced liver cancer often lack potency because of the heterogeneous characteristics of hepatocellular carcinoma (HCC). This complexity is largely driven by context-dependent inflammatory responses brought on by diverse etiologies, such as metabolic dysfunction-associated steatohepatitis (MASH), the genetic makeup of cancer cells, and the versatile adaptability of immune cells, such as myeloid cells. In this review, we discuss the evolutionary dynamics of the immune landscape, particularly that of liver-resident Kupffer cells (KCs), TREM2+, and SPP1+ macrophages with an active role during liver disease progression, which eventually fuels hepatocarcinogenesis. We highlight exploitable immunomodulatory avenues amenable to mitigate both the inherent pathological characteristics of liver cancers and the associated external factors that favor malignancy, paving a roadmap toward improving the management and therapeutic outcome for patients with HCC.
Collapse
Affiliation(s)
- Christel F A Ramirez
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Leila Akkari
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
59
|
Zhao T, Luo Y, Sun Y, Wei Z. Characterizing macrophage diversity in colorectal malignancies through single-cell genomics. Front Immunol 2025; 16:1526668. [PMID: 40191203 PMCID: PMC11968368 DOI: 10.3389/fimmu.2025.1526668] [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: 11/12/2024] [Accepted: 03/10/2025] [Indexed: 04/09/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors of the digestive tract, with increasing incidence and mortality rates, posing a significant burden on human health. Its progression relies on various mechanisms, among which the tumor microenvironment and tumor-associated macrophages (TAMs) have garnered increasing attention. Macrophage infiltration in various solid tumors is associated with poor prognosis and is linked to chemotherapy resistance in many cancers. These significant biological behaviors depend on the heterogeneity of macrophages. Tumor-promoting TAMs comprise subpopulations characterized by distinct markers and unique transcriptional profiles, rendering them potential targets for anticancer therapies through either depletion or reprogramming from a pro-tumoral to an anti-tumoral state. Single-cell RNA sequencing technology has significantly enhanced our research resolution, breaking the traditional simplistic definitions of macrophage subtypes and deepening our understanding of the diversity within TAMs. However, a unified elucidation of the nomenclature and molecular characteristics associated with this diversity remains lacking. In this review, we assess the application of conventional macrophage polarization subtypes in colorectal malignancies and explore several unique subtypes defined from a single-cell omics perspective in recent years, categorizing them based on their potential functions.
Collapse
Affiliation(s)
- Tingshuo Zhao
- First Clinical Medical College, Shanxi Medical University, Tai Yuan, China
| | - Yinyi Luo
- First Clinical Medical College, Shanxi Medical University, Tai Yuan, China
| | - Yuanjie Sun
- First Clinical Medical College, Shanxi Medical University, Tai Yuan, China
| | - Zhigang Wei
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Shanxi Medical University, Tai Yuan, China
| |
Collapse
|
60
|
Jiang R, Yang L, Liu X, Xu Y, Han L, Chen Y, Gao G, Wang M, Su T, Li H, Fang L, Sun N, Du H, Zheng J, Wang G. Genetically engineered macrophages reverse the immunosuppressive tumor microenvironment and improve immunotherapeutic efficacy in TNBC. Mol Ther 2025:S1525-0016(25)00198-4. [PMID: 40119517 DOI: 10.1016/j.ymthe.2025.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 01/21/2025] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
The main challenges in current immunotherapy for triple-negative breast cancer (TNBC) lie in the immunosuppressive tumor microenvironment (TME). Considering tumor-associated macrophages (TAMs) are the most abundant immune cells in the TME, resetting TAMs is a promising strategy for ameliorating the immunosuppressive TME. Here, we developed genetically engineered macrophages (GEMs) with gene-carrying adenoviruses, to maintain the M1-like phenotype and directly deliver the immune regulators interleukin-12 and CXCL9 into local tumors, thereby reversing the immunosuppressive TME. In tumor-bearing mice, GEMs demonstrated targeted enrichment in tumors and successfully reprogramed TAMs to M1-like macrophages. Moreover, GEMs significantly enhanced the accumulation, proliferation, and activation of CD8+ T cells, mature dendritic cells, and natural killer cells within tumors, while diminishing M2-like macrophages, immunosuppressive myeloid-derived suppressor cells, and regulatory T cells. This treatment efficiently suppressed tumor growth. In addition, combination therapy with GEMs and anti-programmed cell death protein 1 further improved interferon-γ+CD8+ T cell percentages and tumor inhibition efficacy in an orthotopic murine TNBC model. Therefore, this study provides a novel strategy for reversing the immunosuppressive TME and improving immunotherapeutic efficacy through live macrophage-mediated gene delivery.
Collapse
Affiliation(s)
- Ranran Jiang
- Department of Oncology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China; Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Liechi Yang
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Xin Liu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Department of Urology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Yujun Xu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Lulu Han
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Yuxin Chen
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Ge Gao
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Meng Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Tong Su
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Lin Fang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Nan Sun
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Hongwei Du
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Junnian Zheng
- Department of Oncology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
61
|
Papayannakos CJ, Israr M, DeVoti JA, Lam F, Arazi A, Frank DK, Kamdar DP, Pereira LM, Seetharamu N, Steinberg BM, Bonagura VR. Oropharyngeal carcinomas induce circulating monocytes to express a TAM-like pro-tumor expression profile that suppresses T-cell proliferation. Front Immunol 2025; 16:1539780. [PMID: 40176808 PMCID: PMC11961958 DOI: 10.3389/fimmu.2025.1539780] [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: 12/04/2024] [Accepted: 02/07/2025] [Indexed: 04/04/2025] Open
Abstract
Introduction Tumor-associated macrophages (TAMs) recruited from circulating monocytes drive tumor-growth and establish an immunosuppressive tumor microenvironment (TME). Initial events in transition from resting monocytes to TAMs are poorly understood. Here, we report that monocytes from oropharyngeal cancer (OPC) patients and control monocytes treated with OPC-conditioned media (CM) express a repertoire of pro-tumor mediators that is characteristic of TAMs. Methods Monocytes were stimulated with OPC cell line CM, analyzed by single-cell RNAseq. Results of select genes were confirmed by qPCR with monocytes and analyzed in OPC tumors vs. clinically normal tissue. OPC spheroids containing control monocytes and T-cells were established, TAM phenotype characterized by flow analysis and qPCR, and T-cell proliferation assessed by flow. Results OPC-conditioned media induced multiple pro-tumor genes including CXCL1, CXCL5, CXCL8, SPP1, IL1B, GPNMB, and FABP5. Patient monocytes had higher baseline levels or achieved higher levels after stimulation than control monocytes. A subset of patient monocytes had high baseline levels of CXCL9/-10/-11 expression that resisted downregulation in response to stimulation, a potential sign of a more favorable TME. CXCL9/-10/-11 expression in OPC tumor biopsies compared to clinically normal tissue correlated with patient outcome. Spheroid TAMs derived from control monocytes maintained the pro-tumor repertoire seen with monocytes stimulated by tumor line conditioned media. These TAMs suppress T-cell proliferation. Inhibition of COX-2 or IL1 signaling during differentiation into TAMs partially blocked the suppression of T-cell proliferation. Conclusion Targeting the early transition of monocytes into pro-tumor TAMs could be used to develop new therapies for OPC.
Collapse
Affiliation(s)
- Christopher J. Papayannakos
- Northwell, New Hyde Park, NY, United States
- Northwell, New Hyde Park, NY and Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Mohd Israr
- Northwell, New Hyde Park, NY, United States
- Northwell, New Hyde Park, NY and Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - James A. DeVoti
- Northwell, New Hyde Park, NY, United States
- Northwell, New Hyde Park, NY and Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Northwell, New Hyde Park, NY and Cohen Children’s Medical Center, Queens, NY, United States
| | - Fung Lam
- Northwell, New Hyde Park, NY, United States
- Northwell, New Hyde Park, NY and Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Arnon Arazi
- Northwell, New Hyde Park, NY, United States
- Northwell, New Hyde Park, NY and Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Douglas K. Frank
- Northwell, New Hyde Park, NY, United States
- Northwell, New Hyde Park, NY and Department of Otolaryngology, Jong Island Jewish Medical Center, New Hyde Park, NY, United States
| | - Dev P. Kamdar
- Northwell, New Hyde Park, NY, United States
- Northwell, New Hyde Park, NY and Department of Otolaryngology, Jong Island Jewish Medical Center, New Hyde Park, NY, United States
| | - Lucio M. Pereira
- Northwell, New Hyde Park, NY, United States
- Northwell, New Hyde Park, NY and Department of Otolaryngology, Jong Island Jewish Medical Center, New Hyde Park, NY, United States
| | - Nagashree Seetharamu
- Northwell, New Hyde Park, NY, United States
- Northwell, New Hyde Park, NY and Department of Otolaryngology, Jong Island Jewish Medical Center, New Hyde Park, NY, United States
- Northwell, New Hyde Park, NY and Department of Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Bettie M. Steinberg
- Northwell, New Hyde Park, NY, United States
- Northwell, New Hyde Park, NY and Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Northwell, New Hyde Park, NY and Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Vincent R. Bonagura
- Northwell, New Hyde Park, NY, United States
- Northwell, New Hyde Park, NY and Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Northwell, New Hyde Park, NY and Cohen Children’s Medical Center, Queens, NY, United States
| |
Collapse
|
62
|
Allonsius L, Kelecom L, Laoui D. The IFN-high phenotype: A biomarker-driven breakthrough in colorectal cancer treatment. Cell Rep Med 2025; 6:102025. [PMID: 40107241 PMCID: PMC11970388 DOI: 10.1016/j.xcrm.2025.102025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Patient stratification is crucial for improving immunotherapy effectiveness. Acha-Sagredo et al. identified an interferon (IFN)-high immunophenotype and CD74 overexpression as predictors for immunotherapy response in colorectal cancer (CRC). This signature, involving cytotoxic T cells and antigen-presenting macrophages, was found in both mismatch repair-deficient and -proficient CRCs. CD74 overexpression could serve as a biomarker, enabling personalized CRC treatment.
Collapse
Affiliation(s)
- Lize Allonsius
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Luca Kelecom
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Damya Laoui
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
63
|
Qin T, Mattox AK, Campbell JS, Park JC, Shin KY, Li S, Sadow PM, Faquin WC, Micevic G, Daniels AJ, Haddad R, Garris CS, Pittet MJ, Mempel TR, ONeill A, Sartor MA, Pai SI. Epigenetic therapy sensitizes anti-PD-1 refractory head and neck cancers to immunotherapy rechallenge. J Clin Invest 2025; 135:e181671. [PMID: 40091844 PMCID: PMC11910227 DOI: 10.1172/jci181671] [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] [Indexed: 03/19/2025] Open
Abstract
BACKGROUNDImmune checkpoint blockade (ICB) is an effective treatment in a subset of patients diagnosed with head and neck squamous cell carcinoma (HNSCC); however, the majority of patients are refractory.METHODSIn a nonrandomized, open-label Phase 1b clinical trial, participants with recurrent and/or metastatic (R/M) HNSCC were treated with low-dose 5-azacytidine (5-aza) daily for either 5 or 10 days in combination with durvalumab and tremelimumab after progression on ICB. The primary objective was to assess the biologically effective dose of 5-aza as determined by molecular changes in paired baseline and on-treatment tumor biopsies; the secondary objective was safety.RESULTSThirty-eight percent (3 of 8) of participants with evaluable paired tissue samples had a greater-than 2-fold increase from baseline in IFN-γ signature and CD274 (programmed cell death protein 1 ligand, PD-L1) expression within the tumor microenvironment (TME), which was associated with increased CD8+ T cell infiltration and decreased infiltration of CD4+ T regulatory cells. The mean neutrophil-to-lymphocyte ratio (NLR) decreased by greater than 50%, from 14.2 (SD 22.6) to 6.9 (SD 5.2). Median overall survival (OS) was 16.3 months (95% CI 1.9, NA), 2-year OS rate was 24.7% (95% CI: 4.5%, 53.2%), and 58% (7 of 12) of treated participants demonstrated prolonged OS of greater than 12 months.CONCLUSIONOur findings suggest that low-dose 5-aza can reprogram systemic host immune responses and the local TME to increase IFN-γ and PD-L1 expression. The increased expression of these established biomarkers correlated with prolonged OS upon ICB rechallenge.TRIAL REGISTRATIONClinicalTrials.gov NCT03019003.FUNDINGNIH/NCI P01 CA240239.
Collapse
Affiliation(s)
- Tingting Qin
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Austin K. Mattox
- Department of Surgery, Division of Otolaryngology—Head and Neck Surgery, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Jong Chul Park
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kee-Young Shin
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Shiting Li
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Peter M. Sadow
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - William C. Faquin
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Goran Micevic
- Department of Dermatology, and
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Andrew J. Daniels
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Robert Haddad
- Department of Medical Oncology, Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Christopher S. Garris
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mikael J. Pittet
- University of Geneva, Geneva, Switzerland
- AGORA Cancer Center and Swiss Cancer Center Leman, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Thorsten R. Mempel
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Anne ONeill
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Maureen A. Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Sara I. Pai
- Department of Surgery, and
- Cancer Immunology Program, Dana-Farber Harvard Cancer Center, Boston, Massachusetts, USA
| |
Collapse
|
64
|
Espinoza FI, Tankov S, Chliate S, Pereira Couto J, Marinari E, Vermeil T, Lecoultre M, El Harane N, Dutoit V, Migliorini D, Walker PR. Targeting HIF-2α in glioblastoma reshapes the immune infiltrate and enhances response to immune checkpoint blockade. Cell Mol Life Sci 2025; 82:119. [PMID: 40095115 PMCID: PMC11914682 DOI: 10.1007/s00018-025-05642-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/31/2025] [Accepted: 02/25/2025] [Indexed: 03/19/2025]
Abstract
Glioblastoma (GBM) is an aggressive primary brain tumor with dismal clinical prognosis and resistance to current therapies. GBM progression is facilitated by the tumor microenvironment (TME), with an immune infiltrate dominated by tumor-associated microglia/macrophages (TAMs) and regulatory T cells (Tregs). The TME is also characterized by hypoxia and the expression of hypoxia-inducible factors (HIFs), with HIF-2α emerging as a potential regulator of tumor progression. However, its role in GBM immunosuppression remains unknown. Here, we investigate HIF-2α and the use of the HIF-2α inhibitor PT2385 to modulate the TME in the immunocompetent GL261 mouse GBM model. PT2385 administration in vivo decreased tumor volume and prolonged survival of tumor-bearing mice, without affecting GL261 viability in vitro. Notably, HIF-2α inhibition alleviated the immunosuppressive TME and synergized with immune checkpoint blockade (ICB) using αPD-1 and αTIM-3 antibodies to promote long-term survival. Comprehensive analysis of the immune infiltrate through single-cell RNA sequencing and flow cytometry revealed that combining PT2385 with ICB reduced numbers of pro-tumoral macrophages and Tregs while increasing numbers of microglia, with a corresponding transcriptional modulation towards an anti-tumoral profile of these TAMs. In vitro, deletion of HIF-2α in microglia impeded their polarization towards a pro-tumoral M2-like profile, and its inhibition impaired Treg migration. Our results show that targeting HIF-2α can switch an immunosuppressive TME towards one that favors a robust and sustained response to ICB based immunotherapy. These findings establish that clinically relevant HIF-2α inhibitors should be explored not only in malignancies with defects in the HIF-2α axis, but also in those exhibiting an immunosuppressive TME that limits immunotherapy responsiveness.
Collapse
Affiliation(s)
- Felipe I Espinoza
- Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
| | - Stoyan Tankov
- Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
| | - Sylvie Chliate
- Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
| | - Joana Pereira Couto
- Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
| | - Eliana Marinari
- Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Thibaud Vermeil
- Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
| | - Marc Lecoultre
- Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
| | - Nadia El Harane
- Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
| | - Valérie Dutoit
- Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Denis Migliorini
- Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Paul R Walker
- Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland.
| |
Collapse
|
65
|
Reggio A, Fuoco C, Deodati R, Palma A. SPP1 macrophages across diseases: A call for reclassification? FASEB J 2025; 39:e70448. [PMID: 40047497 PMCID: PMC11884386 DOI: 10.1096/fj.202403227r] [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: 12/13/2024] [Revised: 01/31/2025] [Accepted: 02/26/2025] [Indexed: 03/09/2025]
Abstract
SPP1+ macrophages, characterized by elevated expression of the osteopontin gene (secreted phosphoprotein 1, SPP1), have emerged as key players in various pathological contexts, including aging, chronic inflammatory diseases, and cancer. While frequently classified as a subclass of tumor-associated macrophages in oncological settings, their presence in noncancer conditions, such as aging-related disorders and muscular diseases, suggests a broader role beyond tumors. These macrophages share conserved traits, including fibrosis promotion, extracellular matrix remodeling, and immune modulation, often linked to poor clinical outcomes. This perspective explores the multifaceted roles of SPP1+ macrophages across diseases and advocates for their reclassification as a distinct macrophage subtype associated with chronic or prolonged inflammation. Recognizing their cross-disease relevance could reshape macrophage biology and inform targeted therapeutic strategies.
Collapse
Affiliation(s)
- Alessio Reggio
- Saint Camillus International University of Health SciencesRomeItaly
- Department of BiologyUniversity of Rome Tor VergataRomeItaly
| | - Claudia Fuoco
- Department of BiologyUniversity of Rome Tor VergataRomeItaly
| | - Rebecca Deodati
- Department of BiologyUniversity of Rome Tor VergataRomeItaly
| | - Alessandro Palma
- Department of Biology and Biotechnologies “Charles Darwin”Sapienza University of RomeRomeItaly
| |
Collapse
|
66
|
Hu L, Chen D, Yang J, Wang Y, Yu K, Wang C, Wang H. A single fluorescent probe for dual-color imaging and polarity precise analysis in lipid droplets and endoplasmic reticulum. Anal Chim Acta 2025; 1343:343703. [PMID: 39947794 DOI: 10.1016/j.aca.2025.343703] [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: 11/27/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Elaborating the subcellular polarity is pivotal for pathophysiological research and early disease diagnosis. Despite its significance, achieving simultaneous two-color fluorescence imaging and quantitative analysis of polarity across multiple organelles remains challenging. This limitation primarily stems from the lack of single fluorescent (SF) probes capable of such precise and multifaceted functionality. RESULTS We introduce a novel SF probe LE-TPA, designed for concurrent dual-color imaging and precise polarity determination in lipid droplets (LDs) and the endoplasmic reticulum (ER) under a single excitation. LE-TPA adopts a D-π-A-π-D configuration, which demonstrates highly selective and sensitive fluorescence response to polarity variations in Oleic acid-THF or THF-water mixtures, accompanied with an exceptional linearity (R2 > 0.99) between the max λem and polarity parameter Δf, paving the way for quantitative polarity analysis in live samples. Furthermore, LE-TPA enables simultaneous, real-time imaging of these organelles due to their different water contents, and provides compelling evidence for supporting the hypothesis that LDs derive from the ER. Importantly, LE-TPA effectively identifies polarity differences between healthy and cancerous cells at the subcellular level and allows precise polarity mapping of non-alcoholic fatty liver disease (NAFLD) tissues at different pathological stages. SIGNIFICANCE These findings highlight the versatility of probe LE-TPA as a powerful tool for subcellular polarity studies and related disease diagnosis.
Collapse
Affiliation(s)
- Lei Hu
- Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, 241002, PR China; School of Pharmacy, Wannan Medical College, Wuhu, 241002, PR China
| | - Dandan Chen
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, Huaxi MR Research Centre (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, 610000, PR China
| | - Jing Yang
- Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, 241002, PR China
| | - Yuqing Wang
- Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, 241002, PR China
| | - Kun Yu
- Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, 241002, PR China
| | - Chunfei Wang
- Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, 241002, PR China; School of Pharmacy, Wannan Medical College, Wuhu, 241002, PR China.
| | - Hui Wang
- Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical College, Wuhu, 241002, PR China; School of Pharmacy, Wannan Medical College, Wuhu, 241002, PR China.
| |
Collapse
|
67
|
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
|
68
|
Zhao R, Pan Z, Qiu J, Li B, Qi Y, Gao Z, Qiu W, Tang W, Guo X, Deng L, Li G, Xue H. Blocking ITGA5 potentiates the efficacy of anti-PD-1 therapy on glioblastoma by remodeling tumor-associated macrophages. Cancer Commun (Lond) 2025. [PMID: 40084746 DOI: 10.1002/cac2.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Glioblastoma (GBM) is largely refractory to antibodies against programmed cell death 1 (anti-PD-1) therapy. Fully understanding the cellular heterogeneity and immune adaptations in response to anti-PD-1 therapy is necessary to design more effective immunotherapies for GBM. This study aimed to dissect the molecular mechanisms of specific immunosuppressive subpopulations to drive anti-PD-1 resistance in GBM. METHODS We systematically analysed single-cell RNA sequencing and spatial transcriptomics data from GBM tissues receiving anti-PD-1 therapy to characterize the microenvironment alterations. The biological functions of a novel circular RNA (circRNA) were validated both in vitro and in vivo. Mechanically, co-immunoprecipitation, RNA immunoprecipitation and pull-down assays were conducted. RESULTS Mesenchymal GBM (MES-GBM) cells, which were associated with a poor prognosis, and secreted phosphoprotein 1 (SPP1)+ myeloid-derived macrophages (SPP1+ MDMs), a unique subpopulation of MDMs with complex functions, preferentially accumulated in non-responders to anti-PD-1 therapy, indicating that MES-GBM cells and SPP1+ MDMs were the main anti-PD-1-resistant cell subpopulations. Functionally, we determined that circular RNA succinate dehydrogenase complex assembly factor 2 (circSDHAF2), which was positively associated with the abundance of these two anti-PD-1-resistant cell subpopulations, facilitated the formation of a regional MES-GBM and SPP1+ MDM cell interaction loop, resulting in a spatially specific adaptive immunosuppressive microenvironment. Mechanically, we found that circSDHAF2 promoted MES-GBM cell formation by stabilizing the integrin alpha 5 (ITGA5) protein through N-glycosylation. Meanwhile, the N-glycosylation of the ITGA5 protein facilitated its translocation into exosomes and subsequent delivery to MDMs to induce the formation of SPP1+ MDMs, which in turn maintained the MES-GBM cell status and induced T-cell dysfunction via the SPP1-ITGA5 pathway, ultimately promoting GBM immune escape. Importantly, our findings demonstrated that antibody-mediated ITGA5 blockade enhanced anti-PD-1-mediated antitumor immunity. CONCLUSIONS This work elucidated the potential tissue adaptation mechanism of intratumoral dynamic interactions between MES-GBM cells, MDMs and T cells in anti-PD-1 non-responders and identified the therapeutic potential of targeting ITGA5 to reduce anti-PD-1 resistance in GBM.
Collapse
Affiliation(s)
- Rongrong Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Ziwen Pan
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Jiawei Qiu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Boyan Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Yanhua Qi
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Zijie Gao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Wei Qiu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Weijie Tang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Xiaofan Guo
- Department of Neurology, Loma Linda University Health, Loma Linda, California, USA
| | - Lin Deng
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Hao Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| |
Collapse
|
69
|
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
|
70
|
He D, Yang Z, Zhang T, Luo Y, Peng L, Yan J, Qiu T, Zhang J, Qin L, Liu Z, Sun M. Multi-omics and machine learning-driven CD8 + T cell heterogeneity score for head and neck squamous cell carcinoma. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102413. [PMID: 40027882 PMCID: PMC11869859 DOI: 10.1016/j.omtn.2024.102413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 12/03/2024] [Indexed: 03/05/2025]
Abstract
The heterogeneity of head and neck squamous cell carcinoma (HNSCC) poses a significant challenge to treatment, underscoring the urgent need for more precise and personalized therapeutic approaches. CD8+ T cells, integral components of the tumor immune microenvironment, have emerged as key targets for immunotherapy. Our research has established a correlation between a decrease in CD8+ T cell score and a poor clinical prognosis, highlighting the prognostic value of this biomarker. By analyzing the gene expression related to CD8+ T cells, we have differentiated HNSCC into cold and hot tumor subtypes, uncovering disparities in clinical prognosis and responses to immunotherapy. Utilizing eight machine learning methods, we identified the key gene OLR1. Single-cell analysis of HNSCC tissues and peripheral blood, along with spatial transcriptome analysis, revealed that OLR1 predominantly functions in macrophages, modulating the immune microenvironment of HNSCC. The expression level of OLR1 may serve as a predictive marker for immunotherapy responses. Moreover, drug sensitivity analysis and molecular docking studies have indicated that simvastatin and pazopanib are potential inhibitors of OLR1. These findings suggest that simvastatin and pazopanib could open up innovative potential therapeutic avenues for individuals with HNSCC.
Collapse
Affiliation(s)
- Di He
- Department of Oral and Maxillofacial Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Zhan Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Tian Zhang
- Department of Oral and Maxillofacial Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yaxian Luo
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Lianjie Peng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Jiatao Yan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Tao Qiu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Jingyu Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Luying Qin
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Zhichao Liu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Mouyuan Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| |
Collapse
|
71
|
Dong X, Wang X, Zheng X, Jiang H, Liu L, Ma N, Wang S. Targeted nanoparticle delivery system for tumor-associated macrophage reprogramming to enhance TNBC therapy. Cell Biol Toxicol 2025; 41:58. [PMID: 40056273 PMCID: PMC11890257 DOI: 10.1007/s10565-025-10001-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 02/12/2025] [Indexed: 03/10/2025]
Abstract
Triple-negative breast cancer (TNBC) poses as a daunting and intricate manifestation of breast cancer, highlighted by few treatment options and a poor outlook. The crucial element in fostering tumor growth and immune resistance is the polarization of tumor-associated macrophages (TAMs) into the M2 state within the tumor microenvironment (TME). To address this, we developed M2 targeting peptide-chitosan-curcumin nanoparticles (M2pep-Cs-Cur NPs), a targeted delivery system utilizing chitosan (Cs) as a carrier, curcumin (Cur) as a therapeutic agent, and targeting peptides for specificity. These NPs effectively inhibited TNBC cell proliferation (~ 70%) and invasion (~ 70%), while increasing the responsiveness of tumors to anti-PD-L1 treatment (~ 50% survival enhancement) in vitro and in vivo. Bioinformatics analysis suggested that Cur modulates TAM polarization by influencing key genes such as COX-2, offering insights into its underlying mechanisms. This study highlights the potential of M2pep-Cs-Cur NPs to reverse M2 polarization in TAMs, providing a promising targeted therapeutic strategy to overcome immunotherapy resistance and improve TNBC outcomes.
Collapse
Affiliation(s)
- Xiaoshen Dong
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, 155 North Nanjing St, Shenyang, 110001, China
| | - Xiaoou Wang
- Department of Geriatric Cardiovascular, The First Hospital of China Medical University, 155 North Nanjing St, Shenyang, 110001, China
| | - Xinyu Zheng
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, 155 North Nanjing St, Shenyang, 110001, China
- Lab 1, Cancer Institute, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Haiyang Jiang
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, 155 North Nanjing St, Shenyang, 110001, China
| | - Lu Liu
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, 155 North Nanjing St, Shenyang, 110001, China
| | - Ningye Ma
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning Province, China.
| | - Shuo Wang
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, 155 North Nanjing St, Shenyang, 110001, China.
| |
Collapse
|
72
|
Donat A, Xie W, Jiang S, Brylka LJ, Schinke T, Rolvien T, Frosch KH, Baranowsky A, Keller J. Cxcl9-deficiency attenuates the progression of post-traumatic osteoarthritis in mice. Inflamm Res 2025; 74:48. [PMID: 40047894 PMCID: PMC11885341 DOI: 10.1007/s00011-025-02013-8] [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: 05/15/2024] [Revised: 08/30/2024] [Accepted: 02/18/2025] [Indexed: 03/09/2025] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is one of the leading causes of disability in the aging population. While about 10% of the adult population is affected by OA, there is to date no curative treatment and joint replacement surgery remains the only option for treating end-stage OA. Previous studies found elevated levels of the chemokine C-X-C motif ligand 9 (CXCL9) in the synovial fluid of OA knees. However, the exact role of CXCL9 in OA progression is still unknown. METHODS Female wild-type and Cxcl9-deficient mice were challenged with a unilateral anterior cruciate ligament transection (ACLT). Joint destruction in early and late stages of experimental OA was assessed using micro-CT scanning, histological scoring, histomorphometry, and gene expression analysis. RESULTS Inactivation of Cxcl9 protected from cartilage destruction and osteophyte formation in post-traumatic OA in mice. Similarly, indices of joint inflammation including synovitis and expression of pro-inflammatory interleukin-1beta were reduced in OA knees of Cxcl9-deficient mice. However, bone erosion and pathophysiological changes in the subchondral bone compartment remained unaffected in Cxcl9-deficient mice with experimental OA. CONCLUSION Our results point towards a pro-inflammatory role of CXCL9 in OA and identify a potential new target for the pharmacological treatment of OA.
Collapse
Affiliation(s)
- Antonia Donat
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251, Hamburg, Germany
| | - Weixin Xie
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251, Hamburg, Germany
| | - Shan Jiang
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251, Hamburg, Germany
| | - Laura Janina Brylka
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Tim Rolvien
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251, Hamburg, Germany
| | - Karl-Heinz Frosch
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251, Hamburg, Germany
- Department of Trauma Surgery, Orthopedics and Sports Traumatology, BG Hospital Hamburg, 21033, Hamburg, Germany
| | - Anke Baranowsky
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251, Hamburg, Germany
| | - Johannes Keller
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251, Hamburg, Germany.
| |
Collapse
|
73
|
Kroehling L, Chen A, Spinella A, Reed E, Kukuruzinka M, Varelas X, Monti S. A highly resolved integrated single-cell atlas of HPV-negative head and neck cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.02.640812. [PMID: 40093171 PMCID: PMC11908118 DOI: 10.1101/2025.03.02.640812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Head and Neck Squamous Cell Carcinomas (HNSCC) are the seventh most prevalent form of cancer and are associated with human papilloma virus infection (HPV-positive) or with tobacco and alcohol use (HPV-negative). HPV-negative HNSCCs have a high recurrence rate, and individual patients' responses to treatment vary greatly due to the high level of cellular heterogeneity of the tumor and its microenvironment. Here, we describe a HNSCC single cell atlas, which we created by integrating six publicly available datasets encompassing over 230,000 cells across 54 patients. We contextualized the relationships between existing signatures and cell populations, identified new subpopulations, and show the power of this large-scale resource to robustly identify associations between transcriptional signatures and clinical phenotypes that would not be possible to discover using fewer patients. We reveal a previously undefined myeloid population, sex-associated changes in cell type proportions, and novel interactions between CXCL8-positive fibroblasts and vascular endothelial cells. Beyond our findings, the atlas will serve as a public resource for the high-resolution characterization of tumor heterogeneity of HPV-negative HNSCC.
Collapse
Affiliation(s)
- Lina Kroehling
- Bioinformatics Program, Faculty of Computing and Data Science, Boston University, Boston, Massachusetts, USA
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Andrew Chen
- Bioinformatics Program, Faculty of Computing and Data Science, Boston University, Boston, Massachusetts, USA
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Anthony Spinella
- Department of Biochemistry and Cell Biology, Boston University Medical Center, Boston, MA, USA
| | - Eric Reed
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Maria Kukuruzinka
- Department of Molecular and Cell Biology, Department of Translational Dental Medicine, Boston University Medical Center, Boston, Massachusetts USA
| | - Xaralabos Varelas
- Department of Biochemistry and Cell Biology, Boston University Medical Center, Boston, MA, USA
| | - Stefano Monti
- Bioinformatics Program, Faculty of Computing and Data Science, Boston University, Boston, Massachusetts, USA
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, Massachusetts, USA
| |
Collapse
|
74
|
Alladina J, Medoff BD, Cho JL. Innate Immunity and Asthma Exacerbations: Insights From Human Models. Immunol Rev 2025; 330:e70016. [PMID: 40087882 PMCID: PMC11922041 DOI: 10.1111/imr.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/14/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
Asthma is a common chronic respiratory disease characterized by the presence of airway inflammation, airway hyperresponsiveness, and mucus hypersecretion. Repeated asthma exacerbations can lead to progressive airway remodeling and irreversible airflow obstruction. Thus, understanding and preventing asthma exacerbations are of paramount importance. Although multiple endotypes exist, asthma is most often driven by type 2 airway inflammation. New therapies that target specific type 2 mediators have been shown to reduce the frequency of asthma exacerbations but are incompletely effective in a significant number of asthmatics. Furthermore, it remains unknown whether current treatments lead to sustained changes in the airway or if targeting additional pathways may be necessary to achieve asthma remission. Activation of innate immunity is the initial event in the inflammatory sequence that occurs during an asthma exacerbation. However, there continue to be critical gaps in our understanding of the innate immune response to asthma exacerbating factors. In this review, we summarize the current understanding of the role of innate immunity in asthma exacerbations and the methods used to study them. We also identify potential novel therapeutic targets for asthma and future areas for investigation.
Collapse
Affiliation(s)
- Jehan Alladina
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Benjamin D. Medoff
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Josalyn L. Cho
- Division of Pulmonary, Critical Care and Occupational Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
75
|
Lee CYC, McCaffrey J, McGovern D, Clatworthy MR. Profiling immune cell tissue niches in the spatial -omics era. J Allergy Clin Immunol 2025; 155:663-677. [PMID: 39522655 DOI: 10.1016/j.jaci.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Immune responses require complex, spatially coordinated interactions between immune cells and their tissue environment. For decades, we have imaged tissue sections to visualize a limited number of immune-related macromolecules in situ, functioning as surrogates for cell types or processes of interest. However, this inevitably provides a limited snapshot of the tissue's immune landscape. Recent developments in high-throughput spatial -omics technologies, particularly spatial transcriptomics, and its application to human samples has facilitated a more comprehensive understanding of tissue immunity by mapping fine-grained immune cell states to their precise tissue location while providing contextual information about their immediate cellular and tissue environment. These data provide opportunities to investigate mechanisms underlying the spatial distribution of immune cells and its functional implications, including the identification of immune niches, although the criteria used to define this term have been inconsistent. Here, we review recent technological and analytic advances in multiparameter spatial profiling, focusing on how these methods have generated new insights in translational immunology. We propose a 3-step framework for the definition and characterization of immune niches, which is powerfully facilitated by new spatial profiling methodologies. Finally, we summarize current approaches to analyze adaptive immune repertoires and lymphocyte clonal expansion in a spatially resolved manner.
Collapse
Affiliation(s)
- Colin Y C Lee
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom; Cellular Genetics, the Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - James McCaffrey
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom; Cellular Genetics, the Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom; Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Dominic McGovern
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom; Cellular Genetics, the Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom; Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Menna R Clatworthy
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom; Cellular Genetics, the Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom; Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom.
| |
Collapse
|
76
|
Chang TG, Spathis A, Schäffer AA, Gavrielatou N, Kuo F, Jia D, Mukherjee S, Sievers C, Economopoulou P, Anastasiou M, Moutafi M, Pal LR, Vos J, Lee AS, Lam S, Zhao K, Jiang P, Allen CT, Foukas P, Gomatou G, Altan-Bonnet G, Morris LGT, Psyrri A, Ruppin E. Tumor and blood B-cell abundance outperforms established immune checkpoint blockade response prediction signatures in head and neck cancer. Ann Oncol 2025; 36:309-320. [PMID: 39551185 PMCID: PMC11845298 DOI: 10.1016/j.annonc.2024.11.008] [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: 07/19/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Immunotherapy has improved the outcomes for some patients with head and neck squamous-cell carcinoma (HNSCC). However, the low and variable response rates observed highlight the need for robust response biomarkers to select patients for treatment. PATIENTS AND METHODS We assembled and analyzed a large HNSCC dataset, encompassing 11 clinical cohorts including 1232 patient samples, spanning a variety of disease subtypes and immune checkpoint blockade (ICB) treatment types, tissue sources, data modalities, and timing of measurements. We conducted a comprehensive evaluation of the predictive power of various cell types, traditional biomarkers, and emerging predictors in both blood and tumor tissues of HNSCC patients. RESULTS Tumor B-cell infiltration emerged as a strong and robust predictor of both patient survival and ICB response. It outperformed all other established biomarkers of response to ICB, including the tertiary lymphoid structure signature and numerous T-cell-based signatures. B-cell infiltration was associated with a 'hot' antitumor microenvironment that promotes tumor eradication. Furthermore, B-cell levels in peripheral blood mononuclear cells (PBMCs) correlated strongly with tumor B-cell levels and demonstrated high predictive value for ICB response, with high odds ratios (≥7.8) in two independent clinical cohorts. CONCLUSION B-cell abundance, whether assessed in PBMCs or tumor tissues, is one of the strongest predictors of ICB response in HNSCC. For translation to patient care, measuring B-cell abundance in PBMCs via cytometry offers a practical and accessible tool for clinical decision making.
Collapse
Affiliation(s)
- T-G Chang
- Cancer Data Science Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, USA
| | - A Spathis
- Department of Pathology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - A A Schäffer
- Cancer Data Science Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, USA
| | - N Gavrielatou
- Internal Medicine/Section of Department of Medical Oncology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - F Kuo
- Department of Surgery and Cancer Immunogenomics Research Program, Memorial Sloan Kettering Cancer Center, New York, USA
| | - D Jia
- Immunodynamics Group, Laboratory of Integrative Cancer Immunology, CCR, NCI, Bethesda, USA
| | - S Mukherjee
- Cancer Data Science Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, USA
| | - C Sievers
- Surgical Oncology Program, CCR, NCI, NIH, Bethesda, USA
| | - P Economopoulou
- Internal Medicine/Section of Department of Medical Oncology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - M Anastasiou
- Internal Medicine/Section of Department of Medical Oncology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - M Moutafi
- Internal Medicine/Section of Department of Medical Oncology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - L R Pal
- Cancer Data Science Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, USA
| | - J Vos
- Department of Surgery and Cancer Immunogenomics Research Program, Memorial Sloan Kettering Cancer Center, New York, USA
| | - A S Lee
- Department of Surgery and Cancer Immunogenomics Research Program, Memorial Sloan Kettering Cancer Center, New York, USA
| | - S Lam
- Department of Surgery and Cancer Immunogenomics Research Program, Memorial Sloan Kettering Cancer Center, New York, USA
| | - K Zhao
- Department of Surgery and Cancer Immunogenomics Research Program, Memorial Sloan Kettering Cancer Center, New York, USA
| | - P Jiang
- Cancer Data Science Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, USA
| | - C T Allen
- Surgical Oncology Program, CCR, NCI, NIH, Bethesda, USA; Center for Immune-Oncology, CCR, NCI, NIH, Bethesda, USA
| | - P Foukas
- Department of Pathology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - G Gomatou
- Internal Medicine/Section of Department of Medical Oncology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - G Altan-Bonnet
- Immunodynamics Group, Laboratory of Integrative Cancer Immunology, CCR, NCI, Bethesda, USA
| | - L G T Morris
- Department of Surgery and Cancer Immunogenomics Research Program, Memorial Sloan Kettering Cancer Center, New York, USA.
| | - A Psyrri
- Internal Medicine/Section of Department of Medical Oncology, Attikon University Hospital, National Kapodistrian University of Athens, Athens, Greece.
| | - E Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, USA.
| |
Collapse
|
77
|
Li C, Liu C, Ma H, Zhang Z, Zhang J. Lymphocytes-Associated Extracellular Vesicles Activate Natural Killer Cells in HNSCC. Cancer Sci 2025; 116:633-642. [PMID: 39749376 PMCID: PMC11875761 DOI: 10.1111/cas.16440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Small extracellular vesicles (sEVs) facilitate intercellular communication and play a pivotal role in tumor progression. Accumulated evidence has indicated the diversity of sEVs but with limited results revealing the landscape of heterogeneity of sEVs. The heterogeneity of cargo RNA in sEVs presents the different cell origins and indicates different functions. Here, we analyzed the heterogeneity of sEVs at droplet levels from single-cell RNA sequencing results of head and neck squamous cell carcinoma (HNSCC) with the previously reported algorithm SEVtras. With the sEVs secretion activity calculated by SEVtras, we also found that the T cells held the major role of sEVs secretion. In addition, we found these sEVs secreted by T cells increased the cytotoxic ability of natural killer cells (NK cells), which illustrated an indirect manner for the anti-tumor function of T cells. These results revealed the heterogeneity of cargo RNA of sEVs in HNSCC and underlined a sEVs-dependent manner in which T cells act on NK cells and anti-tumor immunity.
Collapse
Affiliation(s)
- Chuwen Li
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
| | - Chun Liu
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
| | - Hailong Ma
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
- Research Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghaiPeople's Republic of China
| | - Jianjun Zhang
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
| |
Collapse
|
78
|
Mariniello A, Borgeaud M, Weiner M, Frisone D, Kim F, Addeo A. Primary and Acquired Resistance to Immunotherapy with Checkpoint Inhibitors in NSCLC: From Bedside to Bench and Back. BioDrugs 2025; 39:215-235. [PMID: 39954220 PMCID: PMC11906525 DOI: 10.1007/s40259-024-00700-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2024] [Indexed: 02/17/2025]
Abstract
Immunotherapy with checkpoint inhibitors has become the cornerstone of systemic treatment for non-oncogene addicted non-small-cell lung cancer. Despite its pivotal role, a significant proportion of patients-approximately 70-85%-either exhibit primary resistance to PD-1 blockade or develop acquired resistance following an initial benefit, even in combination with chemotherapy and/or anti-CTLA-4 agents. The phenomenon of primary and acquired resistance to immunotherapy represents a critical clinical challenge, largely based on our incomplete understanding of the mechanisms of action of immunotherapy, and the resulting lack of accurate predictive biomarkers. Here, we review the definitions and explore the proposed mechanisms of primary and acquired resistance, including those related to the tumor microenvironment, systemic factors, and intrinsic tumor characteristics. We also discuss translational data on adaptive changes within tumor cells and the immune infiltrate following exposure to checkpoint inhibitors. Lastly, we offer a comprehensive overview of current and emerging therapeutic strategies designed to prevent primary resistance and counteract acquired resistance.
Collapse
Affiliation(s)
- Annapaola Mariniello
- Oncology Department, University Hospital Geneva, rue Perret-Gentil 4, 1205, Geneva, Switzerland
| | - Maxime Borgeaud
- Oncology Department, University Hospital Geneva, rue Perret-Gentil 4, 1205, Geneva, Switzerland
| | - Marc Weiner
- Oncology Department, University Hospital Geneva, rue Perret-Gentil 4, 1205, Geneva, Switzerland
| | - Daniele Frisone
- Oncology Department, University Hospital Geneva, rue Perret-Gentil 4, 1205, Geneva, Switzerland
| | - Floryane Kim
- Oncology Department, University Hospital Geneva, rue Perret-Gentil 4, 1205, Geneva, Switzerland
| | - Alfredo Addeo
- Oncology Department, University Hospital Geneva, rue Perret-Gentil 4, 1205, Geneva, Switzerland.
| |
Collapse
|
79
|
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
|
80
|
Feng B, Zhao D, Zhang Z, Jia R, Schuler PJ, Hess J. Ligand-receptor interactions combined with histopathology for improved prognostic modeling in HPV-negative head and neck squamous cell carcinoma. NPJ Precis Oncol 2025; 9:57. [PMID: 40021759 PMCID: PMC11871237 DOI: 10.1038/s41698-025-00844-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/20/2025] [Indexed: 03/03/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSC) is a prevalent malignancy, with HPV-negative tumors exhibiting aggressive behavior and poor prognosis. Understanding the intricate interactions within the tumor microenvironment (TME) is crucial for improving prognostic models and identifying therapeutic targets. Using BulkSignalR, we identified ligand-receptor interactions in HPV-negative TCGA-HNSC cohort (n = 395). A prognostic model incorporating 14 ligand-receptor pairs was developed using random forest survival analysis and LASSO-penalized Cox regression based on overall survival and progression-free interval of HPV-negative tumors from TCGA-HNSC. Multi-omics analysis revealed distinct molecular features between risk groups, including differences in extracellular matrix remodeling, angiogenesis, immune infiltration, and APOBEC enzyme activity. Deep learning-based tissue morphology analysis on HE-stained whole slide images further improved risk stratification, with region selection via Silicon enhancing accuracy. The integration of routine histopathology with deep learning and multi-omics data offers a clinically accessible tool for precise risk stratification, facilitating personalized treatment strategies in HPV-negative HNSC.
Collapse
Affiliation(s)
- Bohai Feng
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China.
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Heidelberg, Heidelberg, Germany.
| | - Di Zhao
- Department of Otorhinolaryngology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zheng Zhang
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ru Jia
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Patrick J Schuler
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Jochen Hess
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Heidelberg, Heidelberg, Germany.
- Division Radiooncology/Radiobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
81
|
Zuo Y, Jin Y, Li G, Ming Y, Fan T, Pan Y, Yao X, Peng Y. Spatial transcriptomic analysis of tumor microenvironment in esophageal squamous cell carcinoma with HIV infection. Mol Cancer 2025; 24:54. [PMID: 39994631 PMCID: PMC11853777 DOI: 10.1186/s12943-025-02248-3] [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: 12/02/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Human Immunodeficiency Virus (HIV) is one of the most prevalent viruses, causing significant immune depletion in affected individuals. Current treatments can control HIV and prolong patients' lives, but new challenges have emerged. Increasing incidence of cancers occur in HIV patients. Esophageal squamous cell carcinoma (ESCC) is one of the most common cancers observed in HIV patients. However, the spatial cellular characteristics of HIV-related ESCC have not been explored, and the differences between HIV-ESCC and typical ESCC remain unclear. METHODS We performed spatial transcriptome sequencing on HIV-ESCC samples to depict the microenvironment and employed cell communication analysis and multiplex immunofluorescence to investigate the molecular mechanism in HIV-ESCC. RESULTS We found that HIV-ESCC exhibited a unique cellular composition, with fibroblasts and epithelial cells intermixed throughout the tumor tissue, lacking obvious spatial separation, while other cell types were sparse. Besides, HIV-ESCC exhibited an immune desert phenotype, characterized by a low degree of immune cell infiltration, with only a few SPP1+ macrophages showing immune resistance functions. Cell communication analysis and multiplex immunofluorescence staining revealed that tumor fibroblasts in HIV-ESCC interact with CD44+ epithelial cells via COL1A2, promoting the expression of PIK3R1 in epithelial cells. This interaction activates the PI3K-AKT signaling pathway, which contributes to the progression of HIV-ESCC. CONCLUSIONS Our findings depict the spatial microenvironment of HIV-ESCC and elucidate a molecular mechanism in the progression of HIV-ESCC. This will provide us insights into the molecular basis of HIV-ESCC and potential treatment strategies.
Collapse
Affiliation(s)
- Yuanli Zuo
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yang Jin
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Li
- Department of Thoracic Surgery, The Public Health Clinical Center of Chengdu, Chengdu, 610061, China
| | - Yue Ming
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Fan
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yitong Pan
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaojun Yao
- Department of Thoracic Surgery, The Public Health Clinical Center of Chengdu, Chengdu, 610061, China.
| | - Yong Peng
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
82
|
Liu F, Zhang T, Yang Y, Wang K, Wei J, Shi JH, Zhang D, Sheng X, Zhang Y, Zhou J, Zhao F. Integrated analysis of single-cell and bulk transcriptomics reveals cellular subtypes and molecular features associated with osteosarcoma prognosis. BMC Cancer 2025; 25:280. [PMID: 39962461 PMCID: PMC11834279 DOI: 10.1186/s12885-025-13714-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Osteosarcoma (OS) is the most common primary bone malignancy with variable molecular biology and prognosis. However, our understanding of the association between cell types and OS progression remains poor. METHODS We generated a human OS cell atlas by integrating over 110,000 single cells from 17 samples. Multiple machine learning algorithms were applied to develop tumor purity prediction models based on transcriptomic profile of OS. The Scissor algorithm and gene enrichment analyses were conducted to delve into cell-intrinsic molecular characteristics linked to OS prognosis. Moreover, the study investigated the impact of ATF6α in OS aggressiveness through genetic and pharmacological loss of function analyses. Lastly, the CellChat algorithm was employed to investigate cell-cell communications. RESULTS Utilizing the high-quality human OS cell atlas, we identified tumor purity as a prognostic indicator and developed a robust tumor purity prediction model. We respectively delineated cancer cell- and immune cell-intrinsic molecular characteristics associated with OS prognosis at single-cell resolution. Interestingly, tumor cells with activated unfolded protein response (UPR) pathway were significantly associated with disease aggressiveness. Notably, ATF6α emerged as the top-activated transcription factor for this tumor subcluster. Subsequently, we confirmed that ATF6α was markedly associated with OS progression, while both genetic and pharmacological inhibition of ATF6α impaired the survival of HOS cells. Lastly, we depicted the landscape of signal crosstalk between the UPR-related subcluster and other cell types within the tumor microenvironment. CONCLUSION In summary, our work provides novel insights into the molecular biology of OS, and offers valuable resource for OS biomarker discovery and treatment strategy development.
Collapse
Affiliation(s)
- Feng Liu
- Department of Hand/Foot/Ankle Surgery, Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655099, China
| | - Tingting Zhang
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
| | - Yongqiang Yang
- Department of Hand/Foot/Ankle Surgery, Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655099, China
| | - Kailun Wang
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
| | - Jinlan Wei
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
| | - Ji-Hua Shi
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Dong Zhang
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250021, China
| | - Xia Sheng
- Department of Hand/Foot/Ankle Surgery, Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655099, China
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
| | - Yi Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Jing Zhou
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Faming Zhao
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China.
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
83
|
Yong R, Mu R, Han C, Chao T, Liu Y, Dong L, Wang C. Optimizing a 5-factor cocktail to prepare reparative macrophages for wound healing. J Leukoc Biol 2025; 117:qiae096. [PMID: 38630870 DOI: 10.1093/jleuko/qiae096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/14/2024] [Accepted: 04/06/2024] [Indexed: 04/19/2024] Open
Abstract
The treatment of nonhealing wounds, such as diabetic ulcers, remains a critical clinical challenge. Recent breakthroughs in cell therapy have shown great promise, with one primary focus on preparing cells with comprehensive reparative functions and foreseeable safety. In our previous study, we recapitulated the proregenerative and immunosuppressive functions of tumor-associated macrophages in non-tumor-derived macrophages, endowing the latter with characteristics for promoting diabetic wound healing-termed tumor-associated macrophage-educated macrophages. To eliminate the use of tumor-derived sources and devise a more controllable method to prepare tumor-associated macrophage-educated macrophage-like cells, in this study, we identify a cocktail comprising 5 recombinant proteins as an essential condition to induce nonpolarized macrophages into therapeutic cells with prohealing functions. The screened 5 factors are osteopontin, macrophage inflammatory protein 2, chemokine (C-C motif) ligand 8, vascular endothelial growth factor B, and macrophage colony-stimulating factor. We demonstrate the rationale for screening these factors and the phenotype of the 5 factor-induced tumor-associated macrophage-educated macrophage-like macrophages prepared from murine bone marrow-derived macrophages, which exhibit angiogenic and immunomodulatory effects in vitro. Then, we induce primary human monocytes from periphery blood into the 5 factor-induced tumor-associated macrophage-educated macrophage-like macrophages, which show prohealing effects in a human primary cell-based ex vivo model (T-Skin™). Our study demonstrates a simple, effective, and controllable approach to induce primary macrophages to possess repairing activities, which may provide insights for developing cell-based therapeutics for nonhealing wounds clinically.
Collapse
Affiliation(s)
- Rong Yong
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| | - Ruoyu Mu
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| | - Congwei Han
- School of Life Sciences & State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, No. 163 Xianlin Avenue, 210023, Nanjing, China
| | - Tzuwei Chao
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| | - Yu Liu
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| | - Lei Dong
- School of Life Sciences & State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, No. 163 Xianlin Avenue, 210023, Nanjing, China
- Chemistry and Biomedicine Innovative Center, Nanjing University, No. 163 Xianlin Avenue, 210023, Nanjing, Jiangsu, China
| | - Chunming Wang
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| |
Collapse
|
84
|
Yu W, Gui S, Peng L, Luo H, Xie J, Xiao J, Yilamu Y, Sun Y, Cai S, Cheng Z, Tao Z. STAT3-controlled CHI3L1/SPP1 positive feedback loop demonstrates the spatial heterogeneity and immune characteristics of glioblastoma. Dev Cell 2025:S1534-5807(25)00034-6. [PMID: 39933531 DOI: 10.1016/j.devcel.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/30/2024] [Accepted: 01/17/2025] [Indexed: 02/13/2025]
Abstract
Proneural-mesenchymal transition (PMT) is a phenotypic alteration and contributes to the malignant progression of glioblastoma (GBM). Macrophages, as a main infiltrating component of the tumor immune microenvironment (TIM), control the biological processes of PMT; however, the mechanisms driving this process remain largely unknown. Here, the overall landscape of tumor and nontumor cells was described by scMulti-omics technology. Then, we demonstrated that chitinase-3-like protein 1 (CHI3L1) played a critical role in maintaining mesenchymal (MES) status and reprogramming macrophage phenotype using C57BL/6 and NSG mice models derived from PN20 cells. Mechanistically, osteopontin (OPN)/ITGB1 maintained the activation of nuclear factor κB (NF-κB) and signal transducer and activator of transcription 3 (STAT3) pathways by establishing a positive feedback loop with the CHI3L1-STAT3 axis, resulting in PMT. CHI3L1 enhanced the phosphorylation, nuclear localization, and transcriptional activity of STAT3 via directly binding its coiled-coil domain (CCD). Importantly, we screened and validated that hygromycin B (HB), an inhibitor of the STAT3-CCD domain, disrupted the CHI3L1-STAT3 interaction, thereby reducing the tumor burden in vitro and in vivo.
Collapse
Affiliation(s)
- Wanli Yu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang 330006, Jiangxi, China; JXHC key Laboratory of Neurological Medicine, Nanchang University, Nanchang 330006, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Shikai Gui
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang 330006, Jiangxi, China; JXHC key Laboratory of Neurological Medicine, Nanchang University, Nanchang 330006, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Lunshan Peng
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang 330006, Jiangxi, China; JXHC key Laboratory of Neurological Medicine, Nanchang University, Nanchang 330006, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Haitao Luo
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Jiabao Xie
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang 330006, Jiangxi, China; JXHC key Laboratory of Neurological Medicine, Nanchang University, Nanchang 330006, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Juexian Xiao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Yimuran Yilamu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Yi Sun
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Shihao Cai
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang 330006, Jiangxi, China; JXHC key Laboratory of Neurological Medicine, Nanchang University, Nanchang 330006, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Zujue Cheng
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China.
| | - Zhennan Tao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China; Neurosurgical Institute, Nanjing University, Nanjing 210008, Jiangsu, China.
| |
Collapse
|
85
|
Pan Z, Chen J, Xu T, Cai A, Han B, Li Y, Fang Z, Yu D, Wang S, Zhou J, Gong Y, Che Y, Zou X, Cheng L, Tan Z, Ge M, Huang P. VSIG4 + tumor-associated macrophages mediate neutrophil infiltration and impair antigen-specific immunity in aggressive cancers through epigenetic regulation of SPP1. J Exp Clin Cancer Res 2025; 44:45. [PMID: 39920772 PMCID: PMC11803937 DOI: 10.1186/s13046-025-03303-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
V-set and immunoglobulin domain-containing 4 (VSIG4) positive tumor-associated macrophage (VSIG4+ TAM) is an immunosuppressive subpopulation newly identified in aggressive cancers. However, the mechanism how VSIG4+ TAMs mediate immune evasion in aggressive cancers have not been fully elucidated. In our study, we found targeting VSIG4+ TAMs by VSIG4 deficiency or blockade remarkably limited tumor growth and metastasis, especially those derived from anaplastic thyroid cancer (ATC) and pancreatic cancer, two extremely aggressive types. Moreover, the combination of VSIG4 blockade with a BRAF inhibitor synergistically enhanced anti-tumor activity in ATC-tumor bearing mice. VSIG4 deficiency recovered the antigen presentation (B2m, H2-k1, H2-d1) of TAMs and activated antigen-specific CD8+ T cells by promoting their in vivo proliferation and intratumoral infiltration. Notably, loss of VSIG4 in TAMs significantly reduced the production of lactate and histone H3 lysine 18 lactylation, resulting the decreased transcription of SPP1 mediated by STAT3, which collectively disrupted the cell-cell interactions between TAMs and neutrophils. Further combination of VSIG4 with SPP1 blockade synergistically boosted anti-tumor activity. Overall, our studies demonstrate the epigenetic regulation function of VSIG4 confers on TAMs an alternative pattern, beyond the checkpoint role of VSIG4, to shape the immunosuppressive tumor microenvironment and impair antigen-specific immunity against aggressive cancers.
Collapse
Affiliation(s)
- Zongfu Pan
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | - Jinming Chen
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Tong Xu
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | - Anqi Cai
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Bing Han
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Ying Li
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Ziwen Fang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Dingyi Yu
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | - Shanshan Wang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Junyu Zhou
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Yingying Gong
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Yulu Che
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Xiaozhou Zou
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | - Lei Cheng
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Zhuo Tan
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | - Minghua Ge
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China.
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China.
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China.
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China.
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China.
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China.
| |
Collapse
|
86
|
Li Y, Zheng Y, Huang J, Nie RC, Wu QN, Zuo Z, Yuan S, Yu K, Liang CC, Pan YQ, Zhao BW, Xu Y, Zhang Q, Zheng Y, Chen J, Zeng ZL, Wei W, Liu ZX, Xu RH, Luo HY. CAF-macrophage crosstalk in tumour microenvironments governs the response to immune checkpoint blockade in gastric cancer peritoneal metastases. Gut 2025; 74:350-363. [PMID: 39537239 PMCID: PMC11874311 DOI: 10.1136/gutjnl-2024-333617] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Peritoneal metastasis is the most common metastasis pattern of gastric cancer. Patients with gastric cancer peritoneal metastasis (GCPM) have a poor prognosis and respond poorly to conventional treatments. Recently, immune checkpoint blockade (ICB) has demonstrated favourable efficacy in the treatment of GCPM. Stratification of best responders and elucidation of resistance mechanisms of ICB therapies are highly important and remain major clinical challenges. DESIGN We performed a phase II trial involving patients with GCPM treated with ICB (sintilimab) combined with chemotherapy. The samples of primary tumours, GCPMs and peripheral blood from patients were collected for single-cell sequencing to comprehensively interpret the tumour microenvironment of GCPM and its impacts on immunotherapy efficacy. RESULTS The GCPM ecosystem coordinates a unique immunosuppressive pattern distinct from that of primary GC, which is dominated by a stroma-myeloid niche composed of SPP1+tumour-associated macrophages (TAMs) and Thrombospondin 2 (THBS2)+matrix cancer-associated fibroblasts (mCAFs). Consequently, this stroma-myeloid crosstalk is the major mediator of ICB resistance in patients with GCPM. Mechanistically, the accumulated THBS2+mCAFs facilitate the recruitment of peritoneum-specific tissue-resident macrophages and their transformation into SPP1+TAMs via the complement C3 and its receptor C3a receptor 1 (C3AR1), thereby forming a protumoral stroma-myeloid niche. Blocking the C3-C3AR1 axis disrupts the stroma-myeloid crosstalk and thereby significantly improves the benefits of ICB in in vivo models. CONCLUSION Our findings provide a new molecular portrait of cell compositions associated with ICB resistance in patients with GCPM and aid in the prioritisation of therapeutic candidates to potentiate immunotherapy.
Collapse
Affiliation(s)
- Yuanfang Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yongqiang Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jiaqian Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Run-Cong Nie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Qi-Nian Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zhijun Zuo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Shuqiang Yuan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Kai Yu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Cheng-Cai Liang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yi-Qian Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Bai-Wei Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yuhong Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Qihua Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yashang Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Junquan Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zhao-Lei Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Wei Wei
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Ze-Xian Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Rui-Hua Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Hui-Yan Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| |
Collapse
|
87
|
Chiu SL, Chiang TI, Chen CL. Identification and validation of SPP1 as a potential biomarker for COPD through comprehensive bioinformatics analysis. Respir Med 2025; 237:107953. [PMID: 39832670 DOI: 10.1016/j.rmed.2025.107953] [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] [Received: 11/12/2024] [Revised: 12/30/2024] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Chronic Obstructive Pulmonary Disease (COPD) is a challenging respiratory condition characterized by persistent airflow limitation and progressive lung function decline. The identification of robust biomarkers is crucial for early diagnosis, monitoring disease progression, and guiding therapeutic strategies. METHODS In this study, we employed a comprehensive bioinformatics approach utilizing multiple Gene Expression Omnibus (GEO) datasets to identify potential COPD biomarkers. Differentially expressed genes (DEGs) were identified from GSE38974 and GSE76925, and Weighted Gene Co-expression Network Analysis (WGCNA) on GSE76925 revealed significant gene modules associated with COPD traits. RESULTS Integrative analysis highlighted five candidate genes, with Secreted Phosphoprotein 1 (SPP1) emerging as a promising biomarker. SPP1 exhibited consistent negative correlations with lung function parameters in human datasets (GSE103174) and significant upregulation in COPD-relevant animal models (GSE36174 and GSE52509). Moreover, SPP1 levels were elevated across various respiratory samples, including alveolar epithelium, alveolar macrophages, sputum, and lung tissue, from COPD patients. CONCLUSION These findings highlight the potential of SPP1 as a diagnostic and prognostic biomarker for COPD, emphasizing the need for further investigation into its role in COPD pathogenesis and its effectiveness in clinical applications.
Collapse
Affiliation(s)
- Shin-Lin Chiu
- Department of Ophthalmology, Changhua Christian Hospital, Changhua City, Taiwan; Department of Optometry, Da-Yeh University, Changhua County, Taiwan; Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung City, Taiwan.
| | - Tsay-I Chiang
- Department of Nursing, Hungkuang University, Taichung City, Taiwan.
| | - Chiu-Liang Chen
- Department of Nursing, Hungkuang University, Taichung City, Taiwan; Department of Orthopedics, Changhua Christian Hospital, Changhua City, Taiwan.
| |
Collapse
|
88
|
Zuyin L, Zhao L, Qian C, Changkun Z, Delin M, Jialing H, Zhuomiaoyu C, Yuzi L, Jiaxi Z, Jie G, Jiye Z. Single-Cell and Spatial Transcriptomics Delineate the Microstructure and Immune Landscape of Intrahepatic Cholangiocarcinoma in the Leading-Edge Area. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412740. [PMID: 39716897 PMCID: PMC11831447 DOI: 10.1002/advs.202412740] [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: 10/11/2024] [Revised: 11/29/2024] [Indexed: 12/25/2024]
Abstract
Intrahepatic cholangiocarcinoma (ICC) tumor cells and their interactions with the immune microenvironment, particularly at the leading-edge area, have been underexplored. This study employs single-cell RNA sequencing (scRNA-seq) and spatial transcriptome (ST) analysis on samples from the tumor core, adjacent non-tumorous tissue, and the leading-edge area of nine ICC patients. These findings indicate that tumor cells at the leading-edge area demonstrate enhanced proliferation and are tightly associated with the stroma, including endothelial cells and POSTN+ FAP+ fibroblasts. Notably, CD8+ T cells in this region exhibit a naive phenotype with low cytotoxicity and signs of exhaustion, likely due to compromised antigen presentation by antigen-presenting cells (APCs). The predominant CD8+ T cell subset, mucosal-associated invariant T (MAIT) cells, recruits SPP1+ macrophages within the stroma. This interaction, along with the presence of POSTN+ cancer-associated fibroblasts (CAFs) and endothelial cells, forms a unique "triad structure" that fosters tumor growth and ICC progression. The research highlights the intricate characteristics and interactions of ICC tumor cells in the leading-edge area, offering insights into potential therapeutic targets for intervention.
Collapse
Affiliation(s)
- Li Zuyin
- Department of Hepatobiliary SurgeryPeking University Organ Transplantation InstitutePeking University People's HospitalBeijing100044China
- Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| | - Li Zhao
- Department of Hepatobiliary SurgeryPeking University Organ Transplantation InstitutePeking University People's HospitalBeijing100044China
- Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| | - Cheng Qian
- Department of Hepatobiliary SurgeryPeking University Organ Transplantation InstitutePeking University People's HospitalBeijing100044China
- Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| | - Zhang Changkun
- Department of Hepatobiliary SurgeryPeking University Organ Transplantation InstitutePeking University People's HospitalBeijing100044China
- Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| | - Ma Delin
- Department of Hepatobiliary SurgeryPeking University Organ Transplantation InstitutePeking University People's HospitalBeijing100044China
- Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| | - Hao Jialing
- Department of Hepatobiliary SurgeryPeking University Organ Transplantation InstitutePeking University People's HospitalBeijing100044China
- Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| | - Chen Zhuomiaoyu
- Department of Hepatobiliary SurgeryPeking University Organ Transplantation InstitutePeking University People's HospitalBeijing100044China
- Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| | - Li Yuzi
- Department of Hepatobiliary SurgeryPeking University Organ Transplantation InstitutePeking University People's HospitalBeijing100044China
- Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| | - Zheng Jiaxi
- Department of Hepatobiliary SurgeryPeking University Organ Transplantation InstitutePeking University People's HospitalBeijing100044China
- Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| | - Gao Jie
- Department of Hepatobiliary SurgeryPeking University Organ Transplantation InstitutePeking University People's HospitalBeijing100044China
- Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| | - Zhu Jiye
- Department of Hepatobiliary SurgeryPeking University Organ Transplantation InstitutePeking University People's HospitalBeijing100044China
- Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| |
Collapse
|
89
|
Ji S, Wang F, Wu Y, Hu H, Xing Z, Zhu J, Xu S, Han T, Liu G, Wu Z, Fei C, Kong L, Chen J, Ding Z, Huang Z, Zhang J. Large-scale transcript variants dictate neoepitopes for cancer immunotherapy. SCIENCE ADVANCES 2025; 11:eado5600. [PMID: 39888994 PMCID: PMC11784853 DOI: 10.1126/sciadv.ado5600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 01/02/2025] [Indexed: 02/02/2025]
Abstract
Precise neoepitope discovery is crucial for effective cancer therapeutic vaccines. Conventional approaches struggle to build a repertoire with sufficient immunogenic epitopes. We developed a workflow leveraging full-length ribosome-nascent chain complex-bound mRNA sequencing (FL-RNC seq) and artificial intelligence-based predictive models to accurately identify the neoepitope landscape, especially large-scale transcript variants (LSTVs) missed by short-read sequencing. In the MC38 mouse model, we identified 22 LSTV-derived neoepitopes encoded by a synthesized mRNA lipid nanoparticle vaccine. As a standalone therapy and combined with anti-PD-1 immunotherapy, the vaccine curbed tumor progression, induced robust T cell-specific immunity, and modulated the tumor microenvironment. This underscores the multifaceted potentials of LSTV-derived vaccines. Our approach expands the neoepitope source repertoire, offering a method for discovering personalized cancer vaccines applicable to a broader tumor range. The results highlight the importance of comprehensive neoepitope identification and the promise of LSTV-based vaccines for cancer immunotherapy.
Collapse
Affiliation(s)
- Shiliang Ji
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Feifan Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Yongjie Wu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Haoran Hu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Zhen Xing
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jie Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Shi Xu
- Nanjing Chengshi Biomedical Technology Co. Ltd., Nanjing 210031, China
| | - Tiyun Han
- Nanjing Chengshi Biomedical Technology Co. Ltd., Nanjing 210031, China
| | - Guilai Liu
- Nanjing Chengshi Biomedical Technology Co. Ltd., Nanjing 210031, China
| | - Zengding Wu
- Nanjing Chengshi Biomedical Technology Co. Ltd., Nanjing 210031, China
| | - Caiyi Fei
- Nanjing Chengshi Biomedical Technology Co. Ltd., Nanjing 210031, China
| | - Lingming Kong
- Nanjing Chengshi Biomedical Technology Co. Ltd., Nanjing 210031, China
| | - Jiangning Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Zhi Ding
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Zhen Huang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| |
Collapse
|
90
|
Zhao M, Jankovic D, Link VM, Souza COS, Hornick KM, Oyesola O, Belkaid Y, Lack J, Loke P. Genetic variation in IL-4 activated tissue resident macrophages determines strain-specific synergistic responses to LPS epigenetically. Nat Commun 2025; 16:1030. [PMID: 39863579 PMCID: PMC11762786 DOI: 10.1038/s41467-025-56379-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
How macrophages in the tissue environment integrate multiple stimuli depends on the genetic background of the host, but this is still poorly understood. We investigate IL-4 activation of male C57BL/6 and BALB/c strain specific in vivo tissue-resident macrophages (TRMs) from the peritoneal cavity. C57BL/6 TRMs are more transcriptionally responsive to IL-4 stimulation, with induced genes associated with more super enhancers, induced enhancers, and topologically associating domains (TAD) boundaries. IL-4-directed epigenomic remodeling reveals C57BL/6 specific enrichment of NF-κB, IRF, and STAT motifs. Additionally, IL-4-activated C57BL/6 TRMs demonstrate an augmented synergistic response upon in vitro lipopolysaccharide (LPS) exposure, despite naïve BALB/c TRMs displaying a more robust transcriptional response to LPS. Single-cell RNA sequencing (scRNA-seq) analysis of mixed bone marrow chimeras indicates that transcriptional differences and synergy are cell intrinsic within the same tissue environment. Hence, genetic variation alters IL-4-induced cell intrinsic epigenetic reprogramming resulting in strain specific synergistic responses to LPS exposure.
Collapse
Affiliation(s)
- Mingming Zhao
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Dragana Jankovic
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Verena M Link
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Camila Oliveira Silva Souza
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Katherine M Hornick
- NIAID Collaborative Bioinformatics Resource, Integrated Data Sciences Section, Research Technology Branch, Division of Intramural Research, NIAID, NIH, Bethesda, MD, USA
| | - Oyebola Oyesola
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Justin Lack
- NIAID Collaborative Bioinformatics Resource, Integrated Data Sciences Section, Research Technology Branch, Division of Intramural Research, NIAID, NIH, Bethesda, MD, USA
| | - Png Loke
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
91
|
Zotta A, Toller-Kawahisa J, Palsson-McDermott EM, O’Carroll SM, Henry ÓC, Day EA, McGettrick AF, Ward RW, Ryan DG, Watson MA, Brand MD, Runtsch MC, Maitz K, Lueger A, Kargl J, Miljkovic JL, Lavelle EC, O’Neill LAJ. Mitochondrial respiratory complex III sustains IL-10 production in activated macrophages and promotes tumor-mediated immune evasion. SCIENCE ADVANCES 2025; 11:eadq7307. [PMID: 39841829 PMCID: PMC11789823 DOI: 10.1126/sciadv.adq7307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025]
Abstract
The cytokine interleukin-10 (IL-10) limits the immune response and promotes resolution of acute inflammation. Because of its immunosuppressive effects, IL-10 up-regulation is a common feature of tumor progression and metastasis. Recently, IL-10 regulation has been shown to depend on mitochondria and redox-sensitive signals. We have found that Suppressor of site IIIQo Electron Leak 1.2 (S3QEL 1.2), a specific inhibitor of reactive oxygen species (ROS) production from mitochondrial complex III, and myxothiazol, a complex III inhibitor, decrease IL-10 in lipopolysaccharide (LPS)-activated macrophages. IL-10 down-regulation is likely to be mediated by suppression of c-Fos, which is a subunit of activator protein 1 (AP1), a transcription factor required for IL-10 gene expression. S3QEL 1.2 impairs IL-10 production in vivo after LPS challenge and promotes the survival of mice bearing B16F10 melanoma by lowering tumor growth. Our data identify a link between complex III-dependent ROS generation and IL-10 production in macrophages, the targeting of which could have potential in boosting antitumor immunity.
Collapse
Affiliation(s)
- Alessia Zotta
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Juliana Toller-Kawahisa
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Eva M. Palsson-McDermott
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Shane M. O’Carroll
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Órlaith C. Henry
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Emily A. Day
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Anne F. McGettrick
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Ross W. Ward
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Dylan G. Ryan
- Mitochondria Biology Unit, University of Cambridge, Cambridge, UK
| | | | | | - Marah C. Runtsch
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Kathrin Maitz
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Anna Lueger
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Julia Kargl
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Jan L. Miljkovic
- Mitochondria Biology Unit, University of Cambridge, Cambridge, UK
| | - Ed C. Lavelle
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Luke A. J. O’Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
92
|
Zhao H, Park YM, Zheng Y, Mao Q, Collet C, Hu B, Zhou T, Lin L, Wong S, Pan Y, Monreal AV, Sinha UK, Sedghizadeh P, Soragni A, Lin DC. Genetically Defined Organoid Models Reveal Mechanisms Driving Squamous Cell Neoplastic Evolution and Identify Potential Therapeutic Vulnerabilities. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.18.631624. [PMID: 39896470 PMCID: PMC11785044 DOI: 10.1101/2025.01.18.631624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Upper aerodigestive squamous cell carcinoma (UASCC) is an aggressive and lethal neoplasm, with its early neoplastic transformation mechanisms remaining poorly understood. Here, we characterize over 25 genetically-defined organoid models derived from murine and human oral/esophageal tissues harboring key driver mutations. Double knockout of TP53 and CDKN2A induced morphological dysplasia, hyperproliferation, loss of squamous differentiation, and tumorigenicity, which were further exacerbated by additional driver mutations (e.g., PIK3CA, NOTCH1, KMT2C). Single-cell analysis revealed an expansion of quiescent basal cells and proliferative squamous cells, alongside a loss of differentiated squamous cells during malignant transformation. A distinct senescence program, regulated by ANXA1, was markedly diminished during early neoplastic evolution. Mechanistically, the ANXA1-SMAD3-p27KIP1 pathway was identified as a critical regulator of this senescence program, acting to suppress neoplastic features in organoid models. Lastly, our high-throughput, single-organoid-resolution drug screens unexpectedly revealed PIK3CA-driven organoids exhibited sensitivity to Mitomycin C and Onalespib. This study provides novel mechanistic insights into early neoplastic evolution and underscores the value of genetically-defined organoid models for investigating cancer biology and identifying targeted therapies.
Collapse
Affiliation(s)
- Hua Zhao
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, USA
| | - Young Min Park
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
| | - Yueyuan Zheng
- Clinical Big Data Research Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, P.R. China
| | - Qiong Mao
- Clinical Big Data Research Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, P.R. China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, P.R. China
| | - Casey Collet
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, USA
| | - Boyan Hu
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, USA
| | - Tianming Zhou
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, USA
| | - Luda Lin
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Stephanie Wong
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, USA
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Yuhao Pan
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, USA
| | - Anette Vistoro Monreal
- Department of Diagnostic Sciences, Anesthesia & Emergency Medicine, Infection and Immunity Laboratory, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, USA
| | - Uttam K. Sinha
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Parish Sedghizadeh
- Department of Diagnostic Sciences, Anesthesia & Emergency Medicine, Infection and Immunity Laboratory, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, USA
| | - Alice Soragni
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - De-Chen Lin
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, USA
| |
Collapse
|
93
|
Guo H, Wang M, Ni C, Yang C, Fu C, Zhang X, Chen X, Wu X, Hou J, Wang L. TREM2 promotes the formation of a tumor-supportive microenvironment in hepatocellular carcinoma. J Exp Clin Cancer Res 2025; 44:20. [PMID: 39838454 PMCID: PMC11748316 DOI: 10.1186/s13046-025-03287-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/13/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Triggering receptor expressed on myeloid cells 2 (TREM2), a surface receptor predominantly expressed on myeloid cells, is a major hub gene in pathology-induced immune signaling. However, its function in hepatocellular carcinoma (HCC) remains controversial. This study aimed to evaluate the role of TREM2 in the tumor microenvironment in the context of HCC progression. METHODS HCC was experimentally induced in wild-type (WT) and Trem2-deficient (Trem2-/-) mice, and clinical sample analysis and in vitro studies on macrophages were conducted. HCC cells were treated with conditioned medium from WT or Trem2-/- macrophages, and their malignant phenotypes and underlying mechanisms were analyzed. RESULTS TREM2 deficiency reduced liver tumor burden in orthotopic and subcutaneous HCC models by altering CD8+ T cell infiltration. Trem2-deficient macrophages presented increased chemokine secretion. TGF-β1 was found to be positively correlated with TREM2 expression in HCC, and TGF-β blockade reversed TREM2 induction. On the other hand, TREM2+ macrophages were found to be associated with glycolysis and PKM2 expression in HCC cells; this association may be related to the secretion of IL-1β, which enhances the malignant phenotypes of HCC cells. CONCLUSIONS These results reveal that TREM2+ macrophages play a driving role in HCC progression by suppressing CD8+ T cell infiltration and promoting tumor cell glycolysis, providing a new therapeutic target for HCC.
Collapse
Affiliation(s)
- Hanrui Guo
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Department of Clinical Laboratory, Jinan Maternity and Child Care Hospital, Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Meiling Wang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Department of Pathology, Jinan Maternity and Child Care Hospital, Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Caiya Ni
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Chun Yang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Department of Pathology, Affiliated Tianfu Hospital of Southwest Medical University (Meishan Tianfu New Area People's Hospital), Meishan, Sichuan, China
| | - Chunxue Fu
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xiaoman Zhang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xueling Chen
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xiangwei Wu
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Jun Hou
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
| | - Lianghai Wang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
| |
Collapse
|
94
|
Li S, Xia S, Lawas M, Kulshreshtha A, Garb BF, Perera AAC, Li C, Qin T, Welch JD, D’Silva NJ, Rozek LS, Sartor MA. HPV integration in head and neck cancer: downstream splicing events and expression ratios linked with poor outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633627. [PMID: 39896613 PMCID: PMC11785119 DOI: 10.1101/2025.01.17.633627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
HPV integration (HPVint) is associated with carcinogenesis and tumor progression in HPV-associated cancers, including head and neck squamous cell carcinomas (HNSCC). While its impact on human DNA has been well characterized, its relationship with clinical outcomes remains unconfirmed. Here we investigate the consequences of HPVint both with respect to human and HPV characteristics by analyzing 261 HPV-associated HNSCC bulk and single-cell RNA-seq samples from five cohorts, and DNA HPVint events from 102 HPV+ participants in two of the cohorts. By leveraging this large meta-cohort, we first reveal an oncogenic network based on the recurrent HPV integration locations in HNSCC. We then classify HPVint-positive (HPVint(+)) participants by HPV RNA features, specifically based on spliced HPV-human fusion transcripts and ratios of HPV gene transcripts, showing that subsets of participants have worse clinical outcomes. Our analyses, focused mainly on RNA instead of DNA, expand our understanding of the carcinogenic mechanisms of HPVint, partially addressing the conflicting findings of whether HPVint is associated with aggressive phenotypes and worse clinical consequences, and provide potential biomarkers to advance precision oncology in HPV-associated HNSCC.
Collapse
Affiliation(s)
- Shiting Li
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Shaomiao Xia
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Maria Lawas
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Aishani Kulshreshtha
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Bailey F. Garb
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - AA Chamila Perera
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Chen Li
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Tingting Qin
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Joshua D. Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Nisha J. D’Silva
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Laura S. Rozek
- Georgetown University, Oncology Department, School of Medicine, Washington DC, USA
| | - Maureen A. Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
95
|
Yu XJ, Zou P, Li TQ, Bai XF, Wang SX, Guan JB, Zhao YT, Li MW, Wang X, Wang YG, Hao DJ. Deciphering SPP1-related macrophage signaling in the pathogenesis of intervertebral disc degeneration. Cell Biol Toxicol 2025; 41:33. [PMID: 39825191 PMCID: PMC11748470 DOI: 10.1007/s10565-024-09948-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 11/20/2024] [Indexed: 01/20/2025]
Abstract
This study delved into the molecular mechanisms underlying mechanical stress-induced intervertebral disc degeneration (msi-IDD) through single-cell and high-throughput transcriptome sequencing in mouse models and patient samples. Results exhibited an upsurge in macrophage presence in msi-IDD intervertebral disc (IVD) tissues, with secreted phosphoprotein 1 (SPP1) identified as a pivotal driver exacerbating degeneration via the protein kinase RNA-like endoplasmic reticulum kinase/ activating transcription factor 4/ interleukin-10 (PERK/ATF4/IL-10) signaling axis. Inhibition of SPP1 demonstrated promising outcomes in mitigating msi-IDD progression in both in vitro and in vivo models. These findings underscore the therapeutic promise associated with the modulation of the PERK signaling pathway in IDD, shedding light on the pathogenesis of msi-IDD and proposing a promising avenue for intervention strategies.
Collapse
Affiliation(s)
- Xiao-Jun Yu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, 710054, Shaanxi, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Peng Zou
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, 710054, Shaanxi, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Tian-Qi Li
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, 710054, Shaanxi, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Xiao-Fan Bai
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, 710054, Shaanxi, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Shan-Xi Wang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, 710054, Shaanxi, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Jian-Bin Guan
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, 710054, Shaanxi, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Yuan-Ting Zhao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, 710054, Shaanxi, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Meng-Wei Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaodong Wang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, 710054, Shaanxi, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Ying-Guang Wang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, 710054, Shaanxi, China.
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China.
| | - Ding-Jun Hao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, 710054, Shaanxi, China.
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China.
| |
Collapse
|
96
|
Safaee Talkhoncheh M, Sjölund J, Bolivar P, Kurzejamska E, Cordero E, Vallès Pagès T, Larsson S, Lehn S, Frimannsson G, Ingesson V, Braun S, Pantaleo J, Oudenaarden C, Lauss M, Pearsall RS, Jönsson G, Rolny C, Bocci M, Pietras K. An activin receptor-like kinase 1-governed monocytic lineage shapes an immunosuppressive landscape in breast cancer metastases. J Clin Invest 2025; 135:e183086. [PMID: 39808498 PMCID: PMC11870737 DOI: 10.1172/jci183086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025] Open
Abstract
The biology centered around the TGF-β type I receptor activin receptor-like kinase (ALK) 1 (encoded by ACVRL1) has been almost exclusively based on its reported endothelial expression pattern since its first functional characterization more than 2 decades ago. Here, in efforts to better define the therapeutic context in which to use ALK1 inhibitors, we uncover a population of tumor-associated macrophages (TAMs) that, by virtue of their unanticipated Acvrl1 expression, are effector targets for adjuvant antiangiogenic immunotherapy in mouse models of metastatic breast cancer. The combinatorial benefit depended on ALK1-mediated modulation of the differentiation potential of bone marrow-derived granulocyte-macrophage progenitors, the release of CD14+ monocytes into circulation, and their eventual extravasation. Notably, ACVRL1+ TAMs coincided with an immunosuppressive phenotype and were overrepresented in human cancers progressing on therapy. Accordingly, breast cancer patients with a prominent ACVRL1hi TAM signature exhibited a significantly shorter survival. In conclusion, we shed light on an unexpected multimodal regulation of tumorigenic phenotypes by ALK1 and demonstrate its utility as a target for antiangiogenic immunotherapy.
Collapse
Affiliation(s)
- Mehrnaz Safaee Talkhoncheh
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
| | - Jonas Sjölund
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
| | - Paulina Bolivar
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
| | - Ewa Kurzejamska
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, Solna, Sweden
| | - Eugenia Cordero
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
- Lund University Diabetes Centre, Clinical Research Center, Lund University, Lund, Sweden
| | - Teia Vallès Pagès
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
| | - Sara Larsson
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
| | - Sophie Lehn
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
| | - Gustav Frimannsson
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
| | - Viktor Ingesson
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
| | - Sebastian Braun
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
| | - Jessica Pantaleo
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
| | - Clara Oudenaarden
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
- Biotech Research and Innovation Center, University of Copenhagen, Copenhagen, Denmark
| | - Martin Lauss
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University Cancer Centre, Lund University, Lund, Sweden
| | | | - Göran Jönsson
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University Cancer Centre, Lund University, Lund, Sweden
| | - Charlotte Rolny
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Solna, Sweden
| | - Matteo Bocci
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
- IO Biotech ApS, Copenhagen, Denmark
| | - Kristian Pietras
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
| |
Collapse
|
97
|
Panda VK, Mishra B, Mahapatra S, Swain B, Malhotra D, Saha S, Khanra S, Mishra P, Majhi S, Kumari K, Nath AN, Saha S, Jena S, Kundu GC. Molecular Insights on Signaling Cascades in Breast Cancer: A Comprehensive Review. Cancers (Basel) 2025; 17:234. [PMID: 39858015 PMCID: PMC11763662 DOI: 10.3390/cancers17020234] [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: 11/22/2024] [Revised: 12/27/2024] [Accepted: 01/01/2025] [Indexed: 01/27/2025] Open
Abstract
The complex signaling network within the breast tumor microenvironment is crucial for its growth, metastasis, angiogenesis, therapy escape, stem cell maintenance, and immunomodulation. An array of secretory factors and their receptors activate downstream signaling cascades regulating breast cancer progression and metastasis. Among various signaling pathways, the EGFR, ER, Notch, and Hedgehog signaling pathways have recently been identified as crucial in terms of breast cancer proliferation, survival, differentiation, maintenance of CSCs, and therapy failure. These receptors mediate various downstream signaling pathways such as MAPK, including MEK/ERK signaling pathways that promote common pro-oncogenic signaling, whereas dysregulation of PI3K/Akt, Wnt/β-catenin, and JAK/STAT activates key oncogenic events such as drug resistance, CSC enrichment, and metabolic reprogramming. Additionally, these cascades orchestrate an intricate interplay between stromal cells, immune cells, and tumor cells. Metabolic reprogramming and adaptations contribute to aggressive breast cancer and are unresponsive to therapy. Herein, recent insights into the novel signaling pathways operating within the breast TME that aid in their advancement are emphasized and current developments in practices targeting the breast TME to enhance treatment efficacy are reviewed.
Collapse
Affiliation(s)
- Venketesh K. Panda
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
- School of Applied Sciences, KIIT Deemed to Be University, Bhubaneswar 751024, India
| | - Barnalee Mishra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Samikshya Mahapatra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Biswajit Swain
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Diksha Malhotra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Suryendu Saha
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sinjan Khanra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Priyanka Mishra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sambhunath Majhi
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Kavita Kumari
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Angitha N. Nath
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Swarnali Saha
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sarmistha Jena
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Gopal C. Kundu
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
- School of Applied Sciences, KIIT Deemed to Be University, Bhubaneswar 751024, India
- Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to Be University, Bhubaneswar 751024, India
| |
Collapse
|
98
|
Zhang H, Dai J, Mu Q, Zhao X, Lin Z, Wang K, Wang M, Sun D. Macrophage heterogeneity and oncogenic mechanisms in lung adenocarcinoma: insights from scRNA-seq analysis and predictive modeling. Front Immunol 2025; 15:1491872. [PMID: 39850883 PMCID: PMC11754191 DOI: 10.3389/fimmu.2024.1491872] [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: 09/05/2024] [Accepted: 12/03/2024] [Indexed: 01/25/2025] Open
Abstract
Background Macrophages play a dual role in the tumor microenvironment(TME), capable of secreting pro-inflammatory factors to combat tumors while also promoting tumor growth through angiogenesis and immune suppression. This study aims to explore the characteristics of macrophages in lung adenocarcinoma (LUAD) and establish a prognostic model based on macrophage-related genes. Method We performed scRNA-seq analysis to investigate macrophage heterogeneity and their potential pseudotime evolutionary processes. Specifically, we used scRNA-seq data processing, intercellular communication analysis, pseudotime trajectory analysis, and transcription factor regulatory analysis to reveal the complexity of macrophage subpopulations. Data from The Cancer Genome Atlas (TCGA) was used to assess the impact of various macrophage subtypes on LUAD prognosis. Univariate Cox regression was applied to select prognostic-related genes from macrophage markers. We constructed a prognostic model using Lasso regression and multivariate Cox regression, categorizing LUAD patients into high and low-risk groups based on the median risk score. The model's performance was validated across multiple external datasets. We also examined differences between high and low-risk groups in terms of pathway enrichment, mutation information, tumor microenvironment(TME), and immunotherapy efficacy. Finally, RT-PCR confirmed the expression of model genes in LUAD, and cellular experiments explored the carcinogenic mechanism of COL5A1. Results We found that signals such as SPP1 and MIF were more active in tumor tissues, indicating potential oncogenic roles of macrophages. Using macrophage marker genes, we developed a robust prognostic model for LUAD that effectively predicts prognosis and immunotherapy efficacy. A nomogram was constructed to predict LUAD prognosis based on the model's risk score and other clinical features. Differences between high and low-risk groups in terms of TME, enrichment analysis, mutational landscape, and immunotherapy efficacy were systematically analyzed. RT-PCR and cellular experiments supported the oncogenic role of COL5A1. Conclusion Our study identified potential oncogenic mechanisms of macrophages and their impact on LUAD prognosis. We developed a prognostic model based on macrophage marker genes, demonstrating strong performance in predicting prognosis and immunotherapy efficacy. Finally, cellular experiments suggested COL5A1 as a potential therapeutic target for LUAD.
Collapse
Affiliation(s)
- Han Zhang
- Tianjin Chest Hospital, Tianjin University, Tianjin, China
| | | | - Qiuqiao Mu
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| | - Xiaojiang Zhao
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| | - Ziao Lin
- OmixScience Research Institute, OmixScience Co., Ltd., Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Kai Wang
- Tianjin Chest Hospital, Tianjin University, Tianjin, China
| | - Meng Wang
- Tianjin Chest Hospital, Tianjin University, Tianjin, China
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| | - Daqiang Sun
- Tianjin Chest Hospital, Tianjin University, Tianjin, China
| |
Collapse
|
99
|
Ye L, Long C, Xu B, Yao X, Yu J, Luo Y, Xu Y, Jiang Z, Nian Z, Zheng Y, Cai Y, Xue X, Guo G. Multi‑omics identification of a novel signature for serous ovarian carcinoma in the context of 3P medicine and based on twelve programmed cell death patterns: a multi-cohort machine learning study. Mol Med 2025; 31:5. [PMID: 39773329 PMCID: PMC11707953 DOI: 10.1186/s10020-024-01036-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Predictive, preventive, and personalized medicine (PPPM/3PM) is a strategy aimed at improving the prognosis of cancer, and programmed cell death (PCD) is increasingly recognized as a potential target in cancer therapy and prognosis. However, a PCD-based predictive model for serous ovarian carcinoma (SOC) is lacking. In the present study, we aimed to establish a cell death index (CDI)-based model using PCD-related genes. METHODS We included 1254 genes from 12 PCD patterns in our analysis. Differentially expressed genes (DEGs) from the Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) were screened. Subsequently, 14 PCD-related genes were included in the PCD-gene-based CDI model. Genomics, single-cell transcriptomes, bulk transcriptomes, spatial transcriptomes, and clinical information from TCGA-OV, GSE26193, GSE63885, and GSE140082 were collected and analyzed to verify the prediction model. RESULTS The CDI was recognized as an independent prognostic risk factor for patients with SOC. Patients with SOC and a high CDI had lower survival rates and poorer prognoses than those with a low CDI. Specific clinical parameters and the CDI were combined to establish a nomogram that accurately assessed patient survival. We used the PCD-genes model to observe differences between high and low CDI groups. The results showed that patients with SOC and a high CDI showed immunosuppression and hardly benefited from immunotherapy; therefore, trametinib_1372 and BMS-754807 may be potential therapeutic agents for these patients. CONCLUSIONS The CDI-based model, which was established using 14 PCD-related genes, accurately predicted the tumor microenvironment, immunotherapy response, and drug sensitivity of patients with SOC. Thus this model may help improve the diagnostic and therapeutic efficacy of PPPM.
Collapse
Affiliation(s)
- Lele Ye
- Department of Gynecology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chunhao Long
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Binbing Xu
- First Clinical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuyang Yao
- First Clinical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiaye Yu
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yunhui Luo
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuan Xu
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhuofeng Jiang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zekai Nian
- Second Clinical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yawen Zheng
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yaoyao Cai
- Department of Obstetrics, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangyang Xue
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Gangqiang Guo
- Department of Gynecology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
100
|
Ray A, Hu KH, Kersten K, Courau T, Kuhn NF, Zaleta-Linares I, Samad B, Combes AJ, Krummel MF. Targeting CD206+ macrophages disrupts the establishment of a key antitumor immune axis. J Exp Med 2025; 222:e20240957. [PMID: 39601781 PMCID: PMC11602655 DOI: 10.1084/jem.20240957] [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: 06/03/2024] [Revised: 09/01/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
CD206 is a common marker of a putative immunosuppressive "M2" state in tumor-associated macrophages (TAMs). We made a novel conditional CD206 (Mrc1) knock-in mouse to specifically visualize and/or deplete CD206+ TAMs. Early depletion of CD206+ macrophages and monocytes (Mono/Macs) led to the indirect loss of conventional type I dendritic cells (cDC1), CD8 T cells, and NK cells in tumors. CD206+ TAMs robustly expressed CXCL9, contrasting with stress-responsive Spp1-expressing TAMs and immature monocytes, which became prominent with early depletion. CD206+ TAMs differentially attracted activated CD8 T cells, and the NK and CD8 T cells in CD206-depleted tumors were deficient in Cxcr3 and cDC1-supportive Xcl1 and Flt3l expressions. Disrupting this key antitumor axis decreased tumor control by antigen-specific T cells in mice. In human cancers, a CD206Replete, but not a CD206Depleted Mono/Mac gene signature correlated robustly with CD8 T cell, cDC1, and NK signatures and was associated with better survival. These findings negate the unqualified classification of CD206+ "M2-like" macrophages as immunosuppressive.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Mice
- Killer Cells, Natural/immunology
- Lectins, C-Type/metabolism
- Lectins, C-Type/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/genetics
- Macrophages/immunology
- Macrophages/metabolism
- Humans
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Mannose Receptor
- Mice, Inbred C57BL
- Mannose-Binding Lectins/metabolism
- Receptors, CXCR3/metabolism
- Receptors, CXCR3/genetics
- Chemokine CXCL9/metabolism
- Chemokine CXCL9/genetics
- Tumor-Associated Macrophages/immunology
- Tumor-Associated Macrophages/metabolism
- Membrane Glycoproteins/metabolism
- Membrane Glycoproteins/genetics
- Neoplasms/immunology
- Neoplasms/genetics
- Gene Knock-In Techniques
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/genetics
- Monocytes/immunology
- Monocytes/metabolism
- Receptors, Chemokine
Collapse
Affiliation(s)
- Arja Ray
- Department of Pathology, University of California, San Francisco, CA, USA
- ImmunoX Initiative, University of California, San Francisco, CA, USA
| | - Kenneth H. Hu
- Department of Pathology, University of California, San Francisco, CA, USA
- ImmunoX Initiative, University of California, San Francisco, CA, USA
| | - Kelly Kersten
- Department of Pathology, University of California, San Francisco, CA, USA
- ImmunoX Initiative, University of California, San Francisco, CA, USA
| | - Tristan Courau
- Department of Pathology, University of California, San Francisco, CA, USA
- ImmunoX Initiative, University of California, San Francisco, CA, USA
| | - Nicholas F. Kuhn
- Department of Pathology, University of California, San Francisco, CA, USA
- ImmunoX Initiative, University of California, San Francisco, CA, USA
| | - Itzia Zaleta-Linares
- Department of Pathology, University of California, San Francisco, CA, USA
- ImmunoX Initiative, University of California, San Francisco, CA, USA
| | - Bushra Samad
- ImmunoX Initiative, University of California, San Francisco, CA, USA
- UCSF CoLabs, University of California, San Francisco, CA, USA
| | - Alexis J. Combes
- Department of Pathology, University of California, San Francisco, CA, USA
- ImmunoX Initiative, University of California, San Francisco, CA, USA
- UCSF CoLabs, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Matthew F. Krummel
- Department of Pathology, University of California, San Francisco, CA, USA
- ImmunoX Initiative, University of California, San Francisco, CA, USA
- UCSF CoLabs, University of California, San Francisco, CA, USA
| |
Collapse
|