1
|
Gao D. The role of non-malignant B cells in malignant hematologic diseases. Hematology 2025; 30:2466261. [PMID: 39964954 DOI: 10.1080/16078454.2025.2466261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 02/08/2025] [Indexed: 02/20/2025] Open
Abstract
The tumor microenvironment (TME) represents a heterogeneous, complicated ecosystem characterized by intricate interactions between tumor cells and immune cells. During the past decade, immune cells especially T cells were found to play an important role in the progression of tumor and many related immune checkpoints drugs were created. In recent years, more and more scientists revealed the critical role of B-cells within the TME, particularly various populations of non-malignant B cells. Some studies indicated that non-malignant B cells may exert a 'double-edged sword' role in solid tumors. However, there has been comparatively less focus on the role of non-malignant B cells in hematologic malignancies. In this review, we characterized the development of B cells and summarized its functions of antitumor immunity within TME, with an emphasis on elucidating the roles and potential mechanisms of non-malignant B cells in the progression of hematologic diseases including classical Hodgkin's lymphoma, non-Hodgkin's B-cell lymphoma, non-Hodgkin's T-cell lymphoma, leukemia and multiple myeloma.
Collapse
Affiliation(s)
- Daquan Gao
- Department of Hematology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, People's Republic of China
| |
Collapse
|
2
|
Wu Z, Shan Q, Jiang Y, Huang W, Wang Z, Zhuang Y, Liu J, Li T, Yang Z, Li C, Wei T, Wen C, Cui W, Qiu Z, Liu X, Wang Z. Irreversible electroporation combined with PD-L1/IL-6 dual blockade promotes anti-tumor immunity via cDC2/CD4 +T cell axis in MHC-I deficient pancreatic cancer. Cancer Lett 2025; 617:217620. [PMID: 40068706 DOI: 10.1016/j.canlet.2025.217620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 03/15/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a "cold" solid tumor with frequent Major Histocompatibility Complex I (MHC-I) deficiency, thereby making it resistant to type-1-conventional dendritic cell (cDC1)-CD8+T cell mediated anti-tumor immunity. Current studies have demonstrated the emerging compensatory role of MHC-II-mediated antigen presentation and CD4+T cell activation in anti-tumor immunity against MHC-I-deficient tumors. However, the underlying mechanism of the compensatory immune response by CD4+T cells in cancer ablation therapy remains to be elucidate. In clinical samples and murine models, we observed that irreversible electroporation (IRE) ablation therapy promoted immune infiltration and the conversion of CD4+T cells into anti-tumor IFN-γ+Th1 cells and Th17 cells in MHC-I low-expressed PDAC using scRNA-seq and flow-cytometry analyses. Furthermore, we found that PD-L1 blockade predominantly enhanced the activation of CD11b+CD103-type-2 conventional dendritic cells (cDC2s) and their antigen presentation to CD4+T cells after ablation, stimulating the anti-tumor immune response through the tumor antigen-specific IFN-γ+Th1-NK cell axis. Elevated plasma levels of IL-6 in pancreatic cancer patients receiving ablation therapy are significant indicators for impaired prognosis. IL-6 and PD-L1 dual blockade could significantly augment the ratio of IFN-γ+Th1 in CD4+T cells to boost the anti-tumor immunity of NK cells, leading to prolonged survival of mouse bearing pancreatic cancer. Collectively, we have elucidated that PD-L1 blockade activates the cDC2-CD4+T cell axis after IRE therapy, thereby playing a pivotal compensatory anti-tumor role in MHC-I low-expressed pancreatic cancer. Moreover, a combination strategy involving dual-target blockade of PD-L1/IL-6 along with ablation therapy could emerge as a novel therapeutic approach for MHC-I deficient tumors.
Collapse
Affiliation(s)
- Zhuozhuo Wu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Qungang Shan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Yuyue Jiang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Wei Huang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Ziyin Wang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Yaping Zhuang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Jingjing Liu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Tiankuan Li
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Ziyu Yang
- Faculty of Medical Imaging Technology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Chaojie Li
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China; Department of Radiology, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, No.149, South Chongqing Road, Shanghai, 200025, China
| | - Tao Wei
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, China
| | - Chenlei Wen
- Department of Pancreatic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Zilong Qiu
- Songjiang Research Institute, Institute of Autism & MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China, No.748, Middle Zhongshan Road, Shanghai, 200025, China
| | - Xiaoyu Liu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China; Faculty of Medical Imaging Technology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Zhongmin Wang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China; Faculty of Medical Imaging Technology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China; Department of Radiology, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, No.149, South Chongqing Road, Shanghai, 200025, China
| |
Collapse
|
3
|
Ackermann J, Bernard C, Sirven P, Salmon H, Fraldi M, Ben Amar MD. Mechanistic insight for T-cell exclusion by cancer-associated fibroblasts in human lung cancer. eLife 2025; 13:RP101885. [PMID: 40208246 PMCID: PMC11984955 DOI: 10.7554/elife.101885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025] Open
Abstract
The tumor stroma consists mainly of extracellular matrix, fibroblasts, immune cells, and vasculature. Its structure and functions are altered during malignancy: tumor cells transform fibroblasts into cancer-associated fibroblasts, which exhibit immunosuppressive activities on which growth and metastasis depend. These include exclusion of immune cells from the tumor nest, cancer progression, and inhibition of T-cell-based immunotherapy. To understand these complex interactions, we measure the density of different cell types in the stroma using immunohistochemistry techniques on tumor samples from lung cancer patients. We incorporate these data into a minimal dynamical system, explore the variety of outcomes, and finally establish a spatio-temporal model that explains the cell distribution. We reproduce that cancer-associated fibroblasts act as a barrier to tumor expansion, but also reduce the efficiency of the immune response. Our conclusion is that the final outcome depends on the parameter values for each patient and leads to either tumor invasion, persistence, or eradication as a result of the interplay between cancer cell growth, T-cell cytotoxicity, and fibroblast activity. However, despite the existence of a wide range of scenarios, distinct trajectories, and patterns allow quantitative predictions that may help in the selection of new therapies and personalized protocols.
Collapse
Affiliation(s)
- Joseph Ackermann
- Laboratoire Jean Perrin, Sorbonne UniversitéParisFrance
- Laboratoire de Physique de l’Ecole normale supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris CitéParisFrance
| | - Chiara Bernard
- Department of Structures for Engineering and Architecture, University of Naples "Federico II"NaplesItaly
| | | | - Helene Salmon
- Institut Curie, PSL Research University, INSERMParisFrance
| | - Massimiliano Fraldi
- Laboratoire de Physique de l’Ecole normale supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris CitéParisFrance
- Department of Structures for Engineering and Architecture, University of Naples "Federico II"NaplesItaly
| | - Martine D Ben Amar
- Laboratoire de Physique de l’Ecole normale supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris CitéParisFrance
- Institut Universitaire de Cancérologie, Faculté de médecine, Sorbonne UniversitéParisFrance
| |
Collapse
|
4
|
Lu Y, Liu Y, Zuo X, Li G, Wang J, Liu J, Wang X, Wang S, Zhang W, Zhang K, Lei X, Hao Q, Li W, Liu L, Li M, Zhang C, Zhang HM, Zhang Y, Gao Y. CXCL12 + tumor-associated endothelial cells promote immune resistance in hepatocellular carcinoma. J Hepatol 2025; 82:634-648. [PMID: 39393439 DOI: 10.1016/j.jhep.2024.09.044] [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: 03/25/2024] [Revised: 09/05/2024] [Accepted: 09/25/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND & AIMS The tumor microenvironment (TME) plays a crucial role in the limited efficacy of existing treatments for hepatocellular carcinoma (HCC), with tumor-associated endothelial cells (TECs) serving as fundamental TME components that substantially influence tumor progression and treatment efficacy. However, the precise roles and mechanisms of TECs in HCC remain inadequately understood. METHODS We employed a multi-omics profiling strategy to investigate the single-cell and spatiotemporal evolution of TECs within the microenvironment of HCC tumors, showcasing varied responses to immunotherapy. Through an analysis of a clinical cohort of patients with HCC, we explored the correlation between TEC subpopulations and immunotherapy outcomes. The influence of TEC subsets on the immune microenvironment was confirmed through comprehensive in vitro and in vivo studies. To further explore the mechanisms of distinct TEC subpopulations in microenvironmental modulation and their impact on immunotherapy, we utilized TEC subset-specific knockout mouse models as well as humanized mouse models. RESULTS In this study, we identified a new subset of CXCL12+ TECs that exert a crucial role in immune suppression within the HCC TME. Functionally, CXCL12+ TECs impede the differentiation of CD8+ naïve T cells into CD8+ cytotoxic T cells by secreting CXCL12. Furthermore, they attract myeloid-derived suppressor cells (MDSCs). A bispecific antibody was developed to target both CXCL12 and PD1 specifically, showing significant promise in bolstering anti-tumor immune responses and advancing HCC therapy. CONCLUSIONS CXCL12+ TECs are pivotal in mediating immunosuppression within the HCC microenvironment and targeting CXCL12+ TECs presents a promising approach to augment the efficacy of immunotherapies in patients with HCC. IMPACT AND IMPLICATIONS This investigation reveals a pivotal mechanism wherein CXCL12+ tumor-associated endothelial cells (TECs) emerge as crucial modulators of immune suppression in the tumor microenvironment of hepatocellular carcinoma (HCC). The discovery of CXCL12+ TECs as inhibitors of CD8+ naïve T cell activation and recruiters of myeloid-derived suppressor cells significantly advances our grasp of the dynamic between HCC and immune regulation. Moreover, the development and application of a bispecific antibody precisely targeting CXCL12 and PD1 has proven to enhance immune responses in a humanized mouse HCC model. This finding underscores a promising therapeutic direction for HCC, offering the potential to amplify the impact of current immunotherapies.
Collapse
Affiliation(s)
- Yajie Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China; The Department of Clinical Oncology, Xijing Hospital, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China; Innovation Research Institute, Xijing Hospital, Air Force Medical University, 710032 Xi'an, People's Republic of China
| | - Yunpeng Liu
- The Department of Clinical Oncology, Xijing Hospital, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China
| | - Xiaoshuang Zuo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China
| | - Guodong Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China
| | - Jianlin Wang
- The Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China
| | - Jianshan Liu
- The Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China
| | - Xiangxu Wang
- The Department of Clinical Oncology, Xijing Hospital, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China; Innovation Research Institute, Xijing Hospital, Air Force Medical University, 710032 Xi'an, People's Republic of China
| | - Shuning Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China
| | - Wangqian Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China
| | - Kuo Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China
| | - Xiaoying Lei
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China
| | - Qiang Hao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China
| | - Weina Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China
| | - Lei Liu
- Innovation Research Institute, Xijing Hospital, Air Force Medical University, 710032 Xi'an, People's Republic of China
| | - Meng Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China
| | - Cun Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China.
| | - Hong-Mei Zhang
- The Department of Clinical Oncology, Xijing Hospital, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China.
| | - Yingqi Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China.
| | - Yuan Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032 Xi'an, People's Republic of China; Innovation Research Institute, Xijing Hospital, Air Force Medical University, 710032 Xi'an, People's Republic of China.
| |
Collapse
|
5
|
Ghorbaninezhad F, Nour MA, Farzam OR, Saeedi H, Vanan AG, Bakhshivand M, Jafarlou M, Hatami-Sadr A, Baradaran B. The tumor microenvironment and dendritic cells: Developers of pioneering strategies in colorectal cancer immunotherapy? Biochim Biophys Acta Rev Cancer 2025; 1880:189281. [PMID: 39929377 DOI: 10.1016/j.bbcan.2025.189281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 01/25/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
Colorectal cancer (CRC) is the world's third most frequent cancer, and both its incidence and fatality rates are rising. Despite various therapeutic approaches, neither its mortality rate nor its recurrence frequency has decreased significantly. Additionally, conventional treatment approaches, such as chemotherapy and radiotherapy, have several side effects and risks for patients with CRC. Accordingly, the need for alternative and effective treatments for CRC patients is critical. Immunotherapy that utilizes dendritic cells (DCs) harnesses the patient's immune system to combat cancer cells effectively. DCs are the most potent antigen-presenting cells (APCs), which play a vital role in generating anti-cancer T cell responses. A significant barrier to the immune system's ability to eliminate CRC is the establishment of a potent immunosuppressive tumor milieu by malignant cells. Since DCs are frequently defective in this milieu, the tumor setting significantly reduces the effectiveness of DC-based therapy. Determining central mechanisms contributing to tumor growth by unraveling and comprehending the interaction between CRC tumor milieu and DCs may lead to new therapeutic approaches. This study aims to review DC biology and discuss its role in T-cell-mediated anti-tumor immunity, as well as to highlight the immunosuppressive effects of the CRC tumor milieu on the function of DCs. We will also highlight the tumor microenvironment (TME)-related factors that interfere with DC function as a possible therapeutic target to enhance DC-based cell therapy efficacy.
Collapse
Affiliation(s)
- Farid Ghorbaninezhad
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mina Afrashteh Nour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Omid Rahbar Farzam
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Saeedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Ghorbani Vanan
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Bakhshivand
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Jafarlou
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Behzad Baradaran
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Zhu X, Hu M, Huang X, Li L, Lin X, Shao X, Li J, Du X, Zhang X, Sun R, Tong T, Ma Y, Ning L, Jiang Y, Zhang Y, Shao Y, Wang Z, Zhou Y, Ding J, Zhao Y, Xuan B, Zhang H, Zhang Y, Hong J, Fang JY, Xiao X, Shen B, He S, Chen H. Interplay between gut microbial communities and metabolites modulates pan-cancer immunotherapy responses. Cell Metab 2025; 37:806-823.e6. [PMID: 39909032 DOI: 10.1016/j.cmet.2024.12.013] [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: 04/14/2024] [Revised: 10/20/2024] [Accepted: 12/21/2024] [Indexed: 02/07/2025]
Abstract
Immune checkpoint blockade (ICB) therapy has revolutionized cancer treatment but remains effective in only a subset of patients. Emerging evidence suggests that the gut microbiome and its metabolites critically influence ICB efficacy. In this study, we performed a multi-omics analysis of fecal microbiomes and metabolomes from 165 patients undergoing anti-programmed cell death protein 1 (PD-1)/programmed death ligand 1 (PD-L1) therapy, identifying microbial and metabolic entities associated with treatment response. Integration of data from four public metagenomic datasets (n = 568) uncovered cross-cohort microbial and metabolic signatures, validated in an independent cohort (n = 138). An integrated predictive model incorporating these features demonstrated robust performance. Notably, we characterized five response-associated enterotypes, each linked to specific bacterial taxa and metabolites. Among these, the metabolite phenylacetylglutamine (PAGln) was negatively correlated with response and shown to attenuate anti-PD-1 efficacy in vivo. This study sheds light on the interplay among the gut microbiome, the gut metabolome, and immunotherapy response, identifying potential biomarkers to improve treatment outcomes.
Collapse
Affiliation(s)
- Xiaoqiang Zhu
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Department of Gastroenterology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Muni Hu
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaowen Huang
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lingxi Li
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaolin Lin
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyan Shao
- Department of Medical Oncology, Xuzhou Central Hospital, Clinical School of Xuzhou Medical University, Xuzhou, China
| | - Jiantao Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyue Du
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xinjia Zhang
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, China
| | - Rongrong Sun
- Department of Medical Oncology, Xuzhou Central Hospital, Clinical School of Xuzhou Medical University, Xuzhou, China
| | - Tianying Tong
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanru Ma
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lijun Ning
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Jiang
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Zhang
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuqi Shao
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenyu Wang
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yilu Zhou
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jinmei Ding
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Zhao
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Baoqin Xuan
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hongyang Zhang
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, China
| | - Youwei Zhang
- Department of Medical Oncology, Xuzhou Central Hospital, Clinical School of Xuzhou Medical University, Xuzhou, China
| | - Jie Hong
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing-Yuan Fang
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiuying Xiao
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Bo Shen
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China.
| | - Songbing He
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Haoyan Chen
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
7
|
Li CX, Huang C, Chen DS. scPANDA: PAN-Blood Data Annotator with a 10-Million Single-Cell Atlas. CHINESE MEDICAL SCIENCES JOURNAL = CHUNG-KUO I HSUEH K'O HSUEH TSA CHIH 2025; 0:1-21. [PMID: 40164519 DOI: 10.24920/004472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
OBJECTIVES Recent advancements in single-cell RNA sequencing (scRNA-seq) have revolutionized the study of cellular heterogeneity, particularly within the hematological system. However, accurately annotating cell types remains challenging due to the complexity of immune cells. To address this challenge, we develop a PAN-blood single-cell Data Annotator (scPANDA), which leverages a comprehensive 10-million-cell atlas to provide precise cell type annotation. METHODS The atlas, constructed from data collected in 16 studies, incorporated rigorous quality control, preprocessing, and integration steps to ensure a high-quality reference for annotation. scPANDA utilizes a three-layer inference approach, progressively refining cell types from broad compartments to specific clusters. Iterative clustering and harmonization processes were employed to maintain cell type purity throughout the analysis. Furthermore, the performance of scPANDA was evaluated in three external datasets. RESULTS The atlas was structured hierarchically, consisting of 16 compartments, 54 classes, 4,460 low-level clusters (pd_cc_cl_tfs), and 611 high-level clusters (pmid_cts). Robust performance of the tool was demonstrated in annotating diverse immune scRNA-seq datasets, analyzing immune-tumor coexisting clusters in renal cell carcinoma, and identifying conserved cell clusters across species. CONCLUSIONS scPANDA exemplifies effective reference mapping with a large-scale atlas, enhancing the accuracy and reliability of blood cell type identification.
Collapse
Affiliation(s)
- Chang-Xiao Li
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu Province, China
| | - Can Huang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu Province, China
| | - Dong-Sheng Chen
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu Province, China.
| |
Collapse
|
8
|
Qin J, Hu S, Chen Y, Xu M, Xiao Q, Lou J, Ding M, Sun H, Xu T, Pan Y, Wang S. Hypoxia Promotes Malignant Progression of Colorectal Cancer by Inducing POSTN + Cancer-Associated Fibroblast Formation. Mol Carcinog 2025; 64:716-732. [PMID: 39835715 DOI: 10.1002/mc.23882] [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: 09/19/2024] [Revised: 12/18/2024] [Accepted: 01/04/2025] [Indexed: 01/22/2025]
Abstract
Colorectal cancer (CRC) is one of the most common malignancies. Hypoxia can promote the occurrence and development of CRC. However, how hypoxia regulates the CRC immune microenvironment needs to be further explored. The bulk RNA sequencing data and clinicopathological information of CRC patients were enrolled from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. The single-cell RNA sequencing (scRNA-seq) datasets of CRC were collected from and analyzed from the GEO database and the ArrayExpress database. The score of the hypoxia gene set was estimated using the "ssGSEA" algorithm in the "GSVA" R package. The functional characteristics of CAF subtypes were studied by bioinformatics analysis and in vitro experiments, and a prognostic model was constructed based on machine learning correlation. Hypoxia is associated with poor prognosis in CRC patients. Periostin (POSTN) + Fib is a cancer-associated fibroblast (CAF) closely associated with hypoxia, and high infiltration of POSTN + Fib is associated with adverse outcomes in overall survival (OS) and relapse-free survival (RFS) in CRC patients. Hypoxia can induce POSTN expression and secretion in CAFs. Hypoxia-induced increase of POSTN expression in CAFs can significantly promote the migration and proliferation of CRC cells. Hypoxia-induced increase of POSTN expression in CAFs can significantly promote the proliferation and migration of CRC cells. The POSTN+Fib Hypoxia-Related Risk Model (PFHRM) can predict the survival and immunotherapy response of CRC patients. Our study identified a POSTN+Fib cell subpopulation closely associated with hypoxia, which promotes the malignant progression of CRC. The development of PFHRM provides a theoretical basis for improving patient survival and prognosis.
Collapse
Affiliation(s)
- Jian Qin
- School of Medicine, Southeast University, Nanjing, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shangshang Hu
- School of Medicine, Southeast University, Nanjing, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yuhan Chen
- School of Basic Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
| | - Mu Xu
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qianni Xiao
- School of Basic Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
| | - Jinwei Lou
- School of Basic Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
| | - Muzi Ding
- School of Basic Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
| | - Huiling Sun
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Tao Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yuqin Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- School of Basic Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
| | - Shukui Wang
- School of Medicine, Southeast University, Nanjing, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- School of Basic Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
- Jiangsu Collaborative Innovation Center on Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
9
|
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: 0] [Impact Index Per Article: 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
|
10
|
Caronni N, La Terza F, Frosio L, Ostuni R. IL-1β + macrophages and the control of pathogenic inflammation in cancer. Trends Immunol 2025:S1471-4906(25)00059-6. [PMID: 40169292 DOI: 10.1016/j.it.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 04/03/2025]
Abstract
While highlighting the complexity and heterogeneity of tumor immune microenvironments, the application of single-cell analyses in human cancers has identified recurrent subsets of tumor-associated macrophages (TAMs). Among these, interleukin (IL)-1β+ TAMs - cells with high levels of expression of inflammatory response and tissue repair genes, but with limited capacity to stimulate cytotoxic immunity - are emerging as key drivers of pathogenic inflammation in cancer. In this review we discuss recent literature defining the phenotypical, molecular, and functional properties of IL-1β+ TAMs, as well as their temporal dynamics and spatial organization. Elucidating the biology of these cells across tumor initiation, progression, metastasis, and therapy could inform the design and interpretation of clinical trials targeting IL-1β and/or other inflammatory factors in cancer immunotherapy.
Collapse
Affiliation(s)
- Nicoletta Caronni
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Federica La Terza
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Frosio
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Renato Ostuni
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
11
|
Hu X, Li H, Chen M, Qian J, Jiang H. Reference-informed evaluation of batch correction for single-cell omics data with overcorrection awareness. Commun Biol 2025; 8:521. [PMID: 40158033 PMCID: PMC11954866 DOI: 10.1038/s42003-025-07947-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
Batch effect correction (BEC) is fundamental to integrate multiple single-cell RNA sequencing datasets, and its success is critical to empower in-depth interrogation for biological insights. However, no simple metric is available to evaluate BEC performance with sensitivity to data overcorrection, which erases true biological variations and leads to false biological discoveries. Here, we propose RBET, a reference-informed statistical framework for evaluating the success of BEC. Using extensive simulations and six real data examples including scRNA-seq and scATAC-seq datasets with different numbers of batches, batch effect sizes and numbers of cell types, we demonstrate that RBET evaluates the performance of BEC methods more fairly with biologically meaningful insights from data, while other methods may lead to false results. Moreover, RBET is computationally efficient, sensitive to overcorrection and robust to large batch effect sizes. Thus, RBET provides a robust guideline on selecting case-specific BEC method, and the concept of RBET is extendable to other modalities.
Collapse
Affiliation(s)
- Xiaoyue Hu
- Center for Data Science, Zhejiang University, Hangzhou, China
- School of Mathematical Sciences, Zhejiang University, Hangzhou, China
| | - He Li
- Center for Data Science, Zhejiang University, Hangzhou, China
| | - Ming Chen
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Junbin Qian
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Child Health, Hangzhou, China.
| | - Hangjin Jiang
- Center for Data Science, Zhejiang University, Hangzhou, China.
| |
Collapse
|
12
|
Huijbers EJM, van Beijnum JR, van Loon K, Griffioen CJ, Volckmann R, Bassez A, Lambrechts D, Nunes Monteiro M, Jimenez CR, Hogendoorn PCW, Koster J, Griffioen AW. Embryonic reprogramming of the tumor vasculature reveals targets for cancer therapy. Proc Natl Acad Sci U S A 2025; 122:e2424730122. [PMID: 40096611 PMCID: PMC11962416 DOI: 10.1073/pnas.2424730122] [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: 11/28/2024] [Accepted: 01/27/2025] [Indexed: 03/19/2025] Open
Abstract
A sustained blood supply is critical for tumor growth, as it delivers the nutrients and oxygen required for development. Targeting of blood vessel formation via immunotherapies is an area of great importance. Knowing that certain embryonic genes, such as carcinoembryonic antigens (CEA) and oncofetal fibronectin, become reexpressed in malignant transformation, we hypothesized that a similar phenomenon holds true for tumor endothelial cells (TECs) as well. An approach for identification of highly selective tumor endothelial markers was conducted to develop targeted antiangiogenic immunotherapies. We first queried the transcriptome that is present during embryo development. We then performed a systematic search for genes selectively expressed in the mouse embryo at days E11 and E18, as compared to the transcriptome of the adult mouse. Subsequently, we queried for expression of these embryonic genes in sorted murine TECs. This approach identified among others the tumor endothelial antigens fibrillin-2 (Fbn2), elastin microfibril interface-located protein 2 (Emilin2) as well as the tumor endothelial antigens lysyl oxidase (Lox) and serine/cysteine protease inhibitor, clade E, member 1 (Serpine1; Pai-1). For these selected genes, functional involvement in angiogenesis was confirmed in in vitro bioassays. We subsequently used iBoost conjugate vaccine technology to develop vaccines against the selected targets. For all four targets, vaccination readily induced target-specific antibody responses in mice, resulting in inhibition of tumor growth. Access to highly specific tumor endothelial markers provides opportunities for direct targeting of the tumor vasculature with high specificity, without affecting healthy vasculature.
Collapse
Affiliation(s)
- Elisabeth J. M. Huijbers
- Department of Medical Oncology, Angiogenesis Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
- CimCure Besloten Vennootschap, Amsterdam1066 CX, The Netherlands
| | - Judy R. van Beijnum
- Department of Medical Oncology, Angiogenesis Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Karlijn van Loon
- Department of Medical Oncology, Angiogenesis Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Christian J. Griffioen
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1105 AZ, The Netherlands
| | - Richard Volckmann
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1105 AZ, The Netherlands
| | - Ayse Bassez
- Vlaams Instituut voor Biotechnologie-Katolieke Universiteit Leuven Center for Cancer Biology, Leuven3000, Belgium
| | - Diether Lambrechts
- Vlaams Instituut voor Biotechnologie-Katolieke Universiteit Leuven Center for Cancer Biology, Leuven3000, Belgium
| | - Madalena Nunes Monteiro
- Department of Medical Oncology, Oncoproteomics Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Connie R. Jimenez
- Department of Medical Oncology, Oncoproteomics Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
| | | | - Jan Koster
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1105 AZ, The Netherlands
| | - Arjan W. Griffioen
- Department of Medical Oncology, Angiogenesis Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
- CimCure Besloten Vennootschap, Amsterdam1066 CX, The Netherlands
| |
Collapse
|
13
|
Zhang H, Xu Q, Kan H, Yang Y, Cai Y. Exploration of the clinicopathological and prognostic significance of BRCA1 in gastric cancer. Discov Oncol 2025; 16:381. [PMID: 40126757 PMCID: PMC11933547 DOI: 10.1007/s12672-025-02159-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/17/2025] [Indexed: 03/26/2025] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies and is a highly heterogeneous disease; it is also a leading cause of cancer-related death. Owing to the complexity and late-stage diagnosis of GC, the prognosis remains poor. To explore potential biomarkers for GC, GC patient transcriptome data were subjected to a comprehensive approach involving machine learning, binary nomogram prediction model construction, the topological algorithm of CytoHubba, and Kaplan-Meier and Mendelian randomization (MR) analyses. First, gene expression data for normal and GC tissues were assessed via machine learning and the topological algorithm of CytoHubba, and a total of 792 differentially expressed genes (DEGs) and nine core genes were identified. Kaplan-Meier analysis and analysis of a nomogram binary prediction model for the core genes revealed that the expression level of BRCA1 was closely and significantly correlated with the survival time of GC patients, suggesting that BRCA1 may be considered a valuable biomarker for GC diagnosis. Furthermore, MR analysis revealed that BRCA1 promotes the transformation of normal cells into GC cells by regulating NADPH levels, leading to a continuous increase in oxidative stress. This is one of the initial comprehensive analysis involving MR and multidimensional approaches; it revealed the significant role of BRCA1 in GC, providing new ideas on drugs and targets for GC clinical treatment.
Collapse
Affiliation(s)
- Hongrong Zhang
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Qi Xu
- School of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Hongxing Kan
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Yinfeng Yang
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
| | - Yunquan Cai
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, 341000, Jiangxi, China.
| |
Collapse
|
14
|
Cen X, Lan Y, Zou J, Chen R, Hu C, Tong Y, Zhang C, Chen J, Wang Y, Zhou R, He W, Lu T, Dubee F, Jovic D, Dong W, Gao Q, Ma M, Lu Y, Xue Y, Cheng X, Li Y, Yang H. Pan-cancer analysis shapes the understanding of cancer biology and medicine. Cancer Commun (Lond) 2025. [PMID: 40120098 DOI: 10.1002/cac2.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 03/25/2025] Open
Abstract
Advances in multi-omics datasets and analytical methods have revolutionized cancer research, offering a comprehensive, pan-cancer perspective. Pan-cancer studies identify shared mechanisms and unique traits across different cancer types, which are reshaping diagnostic and treatment strategies. However, continued innovation is required to refine these approaches and deepen our understanding of cancer biology and medicine. This review summarized key findings from pan-cancer research and explored their potential to drive future advancements in oncology.
Collapse
Affiliation(s)
- Xiaoping Cen
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
- Guangzhou National Laboratory, Guangzhou, Guangdong, P. R. China
| | - Yuanyuan Lan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Jiansheng Zou
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. China
| | - Ruilin Chen
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. China
| | - Can Hu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yahan Tong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Chen Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Jingyue Chen
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yuanmei Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Run Zhou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Weiwei He
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Tianyu Lu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Fred Dubee
- BGI Research, Shenzhen, Guangdong, P. R. China
| | | | - Wei Dong
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- Clin Lab, BGI Genomics, Beijing, P. R. China
| | - Qingqing Gao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Man Ma
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Youyong Lu
- Laboratory of Molecular Oncology, Peking University Cancer Hospital and Institute, Beijing, P. R. China
| | - Yu Xue
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Xiangdong Cheng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yixue Li
- Guangzhou National Laboratory, Guangzhou, Guangdong, P. R. China
- GZMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Huanming Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- BGI, Shenzhen, Guangdong, P. R. China
- James D. Watson Institute of Genome Sciences, Hangzhou, Zhejiang, P. R. China
| |
Collapse
|
15
|
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] [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
|
16
|
Chen A, Kroehling L, Ennis CS, Denis GV, Monti S. A highly resolved integrated transcriptomic atlas of human breast cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.13.643025. [PMID: 40161579 PMCID: PMC11952505 DOI: 10.1101/2025.03.13.643025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
In this study, we developed an integrated single cell transcriptomic (scRNAseq) atlas of human breast cancer (BC), the largest resource of its kind, totaling > 600,000 cells across 138 patients. Rigorous integration and annotation of publicly available scRNAseq data enabled a highly resolved characterization of epithelial, immune, and stromal heterogeneity within the tumor microenvironment (TME). Within the immune compartment we were able to characterize heterogeneity of CD4, CD8 T cells and macrophage subpopulations. Within the stromal compartment, subpopulations of endothelial cells (ECs) and cancer associated fibroblasts (CAFs) were resolved. Within the cancer epithelial compartment, we characterized the functional heterogeneity of cells across the axes of stemness, epithelial-mesenchymal plasticity, and canonical cancer pathways. Across all subpopulations observed in the TME, we performed a multi-resolution survival analysis to identify epithelial cell states and immune cell types which conferred a survival advantage in both The Cancer Genome Atlas (TCGA) and METABRIC. We also identified robust associations between TME composition and clinical phenotypes such as tumor subtype and grade that were not discernible when the analysis was limited to individual datasets, highlighting the need for atlas-based analyses. This atlas represents a valuable resource for further high-resolution analyses of TME heterogeneity within BC.
Collapse
Affiliation(s)
- Andrew Chen
- Section of Computational Biomedicine, Boston University Chobanian and Avesidian School of Medicine, Boston, MA 02118, USA
- Bioinformatics Program, Center for Computing & Data Sciences, Boston University, Boston, MA 022158, USA
| | - Lina Kroehling
- Section of Computational Biomedicine, Boston University Chobanian and Avesidian School of Medicine, Boston, MA 02118, USA
- Bioinformatics Program, Center for Computing & Data Sciences, Boston University, Boston, MA 022158, USA
| | - Christina S. Ennis
- Boston University-Boston Medical Center Cancer Center, Boston University Chobanian and Avesidian School of Medicine, Boston, MA, 02118, USA
| | - Gerald V. Denis
- Boston University-Boston Medical Center Cancer Center, Boston University Chobanian and Avesidian School of Medicine, Boston, MA, 02118, USA
- Section of Hematology and Medical Oncology, Department of Medicine, Boston University Chobanian and Avesidian School of Medicine and Boston Medical Center, Boston, MA, 02118, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University Chobanian and Avesidian School of Medicine, Boston, MA, 02118, USA
| | - Stefano Monti
- Section of Computational Biomedicine, Boston University Chobanian and Avesidian School of Medicine, Boston, MA 02118, USA
- Bioinformatics Program, Center for Computing & Data Sciences, Boston University, Boston, MA 022158, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| |
Collapse
|
17
|
Li X, Zhao Z, Cheng Y, Yan J, Ren F, Jia Y, Li J, Wang B, Liu J, Wang C, Gao M, Gu H, Fan M, Shi H, Ji M, Zhao Q. Single-cell transcriptomic analyses reveal heterogeneity and key subsets associated with survival and response to PD-1 blockade in cervical squamous cell carcinoma. Cancer Cell Int 2025; 25:90. [PMID: 40082876 PMCID: PMC11907857 DOI: 10.1186/s12935-025-03725-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Understanding the intricate tumor microenvironment (TME) is crucial for elucidating the mechanisms underlying the progression of cervical squamous cell carcinoma (CSCC) and its response to anti-PD-1 therapy. METHODS In this study, we characterized 50,649 cells obtained from the CSCC for single-cell RNA sequencing and integrated bulk sequencing data from The Cancer Genome Atlas (TCGA) and clinical samples to explore their cell composition, metabolic processes, signaling pathways, specific transcription factors, lineage tracking and response to immunotherapy. In vivo experiments were performed to validate the function of key cell subsets. RESULTS We identified ten major cell type and 35 subsets of stromal and immune cells in TME and observed distinct patterns in the metabolic processes and signaling pathways of these cells between tumor and normal tissues. Furthermore, PCNA clamp-associated factor (PCLAF)+ tumor-associated epithelial cell (TAEpis) was negatively correlated with the number of C-X-C motif chemokine ligand 13 (CXCL13)+ CD8+ T cells, overall survival, and response to anti-programmed cell death-1(PD-1) therapy in patients with CSCC. Both in vivo and in vitro experiments demonstrated that PCLAF+ TAEpis promotes the apoptosis of CD8+ T and tumor growth, while also inhibiting T cell infiltration and function. CONCLUSION Our findings illuminate the heterogeneity of the complex TME in CSCC and offer evidence supporting PCLAF+ TAEpis as a promising therapeutic target.
Collapse
Affiliation(s)
- Xia Li
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China.
| | - Zhao Zhao
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Yanmei Cheng
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Jiaqin Yan
- Department of Oncology, the first affiliated hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Fang Ren
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Yanyan Jia
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Juanhua Li
- Department of Digestive System, Henan Electric Power Hospital, Zhengzhou, Henan Province, 450052, China
| | - Binhui Wang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Chenyin Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Meimei Gao
- Department of Pharmacy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hao Gu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Mingliang Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Huirong Shi
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China.
| | - Mei Ji
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China.
| | - Qitai Zhao
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
18
|
Wu B, Yang X, Kong N, Liang J, Li S, Wang H. Engineering Modular Peptide Nanoparticles for Ferroptosis-Enhanced Tumor Immunotherapy. Angew Chem Int Ed Engl 2025; 64:e202421703. [PMID: 39721975 DOI: 10.1002/anie.202421703] [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: 11/07/2024] [Revised: 12/14/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors are promising for treating tumors but have limited efficacy due to the immunosuppressive tumor microenvironment. In this study, we develop an orchestrated nanoparticle system using modular peptide assemblies, where the co-assembled sequences are designed for the specific binding to the hydrophobic and hydrophilic domains, guiding the assembly process and enabling the customization of nanoparticle properties. We exploit the modularity of this platform to integrate a hydrophobic ferroptosis precursor, an IDO1 inhibitor, and a hydrophilic peptidic PD-L1 antagonist for optimizing therapeutic outcomes through ferroptosis-enhanced tumor immunotherapy. The resulting nanoparticles induce immunogenic ferroptosis, enhance the intratumoral function of T lymphocytes, suppress regulatory T cells, and effectively modulate the immunosuppressive tumor microenvironment, thereby facilitating regression of tumor growth. This work provides a modular peptide-based nanoparticle engineering strategy and holds significant potential for advancing cancer treatment.
Collapse
Affiliation(s)
- Bihan Wu
- Department of Chemistry, School of Science, Westlake University Institution Institute of Natural Sciences, Westlake Institute for Advanced Study, No. 600 Dunyu Road, Hangzhou, 310024, Zhejiang Province, China
| | - Xuejiao Yang
- Department of Chemistry, School of Science, Westlake University Institution Institute of Natural Sciences, Westlake Institute for Advanced Study, No. 600 Dunyu Road, Hangzhou, 310024, Zhejiang Province, China
| | - Nan Kong
- Department of Chemistry, School of Science, Westlake University Institution Institute of Natural Sciences, Westlake Institute for Advanced Study, No. 600 Dunyu Road, Hangzhou, 310024, Zhejiang Province, China
| | - Juan Liang
- Department of Chemistry, School of Science, Westlake University Institution Institute of Natural Sciences, Westlake Institute for Advanced Study, No. 600 Dunyu Road, Hangzhou, 310024, Zhejiang Province, China
| | - Sangshuang Li
- Department of Chemistry, School of Science, Westlake University Institution Institute of Natural Sciences, Westlake Institute for Advanced Study, No. 600 Dunyu Road, Hangzhou, 310024, Zhejiang Province, China
| | - Huaimin Wang
- Department of Chemistry, School of Science, Westlake University Institution Institute of Natural Sciences, Westlake Institute for Advanced Study, No. 600 Dunyu Road, Hangzhou, 310024, Zhejiang Province, China
- Westlake Laboratory of Life Sciences and Biomedicine, 18 Shilongshan Road, Hangzhou, 310024, Zhejiang Province, China
| |
Collapse
|
19
|
Liu R, Tang L, Liu Y, Hu H, Liu J. Causal relationship between immune cell signatures and colorectal cancer: a bi-directional, two-sample mendelian randomization study. BMC Cancer 2025; 25:387. [PMID: 40033246 DOI: 10.1186/s12885-025-13576-4] [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: 04/24/2024] [Accepted: 01/21/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Prior studies have demonstrated the association between immune cells and colorectal cancer (CRC). However, the causal link to specific immunophenotypes is limited. This study intends to elucidate the causal relationship of immune cell signatures on CRC. METHODS We performed a bi-directional and two-sample mendelian randomization (MR) study, utilizing GWAS summary data of 731 immune cell traits (n = 3,757) and CRC statistics (n = 470,002). The primary MR methodology was inverse-variance weighted (IVW) method. Furthermore, heterogeneity was evaluated by Cochran's Q test. MR-PRESSO and MR-Egger were employed to assess horizontal and vertical pleiotropy respectively. Sensitivity analysis and FDR correction were conducted in our results. These results were validated in both the UK Biobank and FinnGen cohorts. We also extracted transcriptomic data of CRC and adjacent non-tumor tissues from TCGA, and used CIBERSORT to compare the infiltration patterns of 22 immune cell panels between normal tissues and the tumor microenvironment (TME). RESULTS Our study indicated nine immune cell signatures had significant causality with the risk of CRC after sensitivity analysis and FDR correction. The positive results covered four panels: B cell, CD8 + T cell, Treg, and monocyte. IgD- CD38br and IgD + CD38br B cell, CD8dim and CD28 + CD45RA- CD8dim T cell, and CD14 on CD14 + CD16- monocyte were the protective factors of CRC. However, CD39 + resting Treg, CX3CR1 on CD14- CD16 + monocyte, FSC-A on HLA DR + T cell, and BAFF-R on B cell increased the risk of CRC. The results were validated in the UK Biobank data and FinnGen cohorts. The data from the TCGA database also confirmed the infiltration of B cell, CD8 + T cell, Treg, and monocyte panels in the TME. CONCLUSION This study highlights the causal link between specific immune cell phenotypes and CRC, providing valuable insights into the immune microenvironment's role in CRC. The validation of our findings using large-scale datasets (UK Biobank, FinnGen) and TCGA underscores the robustness of our results, offering new potential therapeutic targets for CRC treatment.
Collapse
Affiliation(s)
- Ruizhi Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liansha Tang
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, Sichuan Province, 610041, China
| | - Yunjia Liu
- Mental Health Center and Psychiatric Laboratory, The State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Handan Hu
- Queen Mary College, Nanchang University, Nanchang, Jiangxi, China
| | - Jiyan Liu
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, Sichuan Province, 610041, China.
| |
Collapse
|
20
|
Dini A, Barker H, Piki E, Sharma S, Raivola J, Murumägi A, Ungureanu D. A multiplex single-cell RNA-Seq pharmacotranscriptomics pipeline for drug discovery. Nat Chem Biol 2025; 21:432-442. [PMID: 39482470 PMCID: PMC11867973 DOI: 10.1038/s41589-024-01761-8] [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: 08/23/2023] [Accepted: 09/22/2024] [Indexed: 11/03/2024]
Abstract
The gene-regulatory dynamics governing drug responses in cancer are yet to be fully understood. Here, we report a pipeline capable of producing high-throughput pharmacotranscriptomic profiling through live-cell barcoding using antibody-oligonucleotide conjugates. This pipeline combines drug screening with 96-plex single-cell RNA sequencing. We show the potential of this approach by exploring the heterogeneous transcriptional landscape of primary high-grade serous ovarian cancer (HGSOC) cells after treatment with 45 drugs, with 13 distinct classes of mechanisms of action. A subset of phosphatidylinositol 3-OH kinase (PI3K), protein kinase B (AKT) and mammalian target of rapamycin (mTOR) inhibitors induced the activation of receptor tyrosine kinases, such as the epithelial growth factor receptor (EGFR), and this was mediated by the upregulation of caveolin 1 (CAV1). This drug resistance feedback loop could be mitigated by the synergistic action of agents targeting PI3K-AKT-mTOR and EGFR for HGSOC with CAV1 and EGFR expression. Using this workflow could enable the personalized testing of patient-derived tumor samples at single-cell resolution.
Collapse
Affiliation(s)
- Alice Dini
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Harlan Barker
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
- Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Emilia Piki
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Subodh Sharma
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Juuli Raivola
- Applied Tumor Genomics, Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Astrid Murumägi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Daniela Ungureanu
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
- Applied Tumor Genomics, Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
21
|
Zhao F, An R, Ma Y, Yu S, Gao Y, Wang Y, Yu H, Xie X, Zhang J. Integrated spatial multi-omics profiling of Fusobacterium nucleatum in breast cancer unveils its role in tumour microenvironment modulation and cancer progression. Clin Transl Med 2025; 15:e70273. [PMID: 40070022 PMCID: PMC11897063 DOI: 10.1002/ctm2.70273] [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: 12/01/2024] [Revised: 02/17/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
Tumour-associated microbiota are integral components of the tumour microenvironment (TME). However, previous studies on intratumoral microbiota primarily rely on bulk tissue analysis, which may obscure their spatial distribution and localized effects. In this study, we applied in situ spatial-profiling technology to investigate the spatial distribution of intratumoral microbiota in breast cancer and their interactions with the local TME. Using 5R 16S rRNA gene sequencing and RNAscope FISH/CISH on patients' tissue, we identified significant spatial heterogeneity in intratumoral microbiota, with Fusobacterium nucleatum (F. nucleatum) predominantly localized in tumour cell-rich areas. GeoMx digital spatial profiling (DSP) revealed that regions colonized by F. nucleatum exhibit significant influence on the expression of RNAs and proteins involved in proliferation, migration and invasion. In vitro studies indicated that co-culture with F. nucleatum significantly stimulates the proliferation and migration of breast cancer cells. Integrative spatial multi-omics and co-culture transcriptomic analyses highlighted the MAPK signalling pathways as key altered pathways. By intersecting these datasets, VEGFD and PAK1 emerged as critical upregulated proteins in F. nucleatum-positive regions, showing strong positive correlations with MAPK pathway proteins. Moreover, the upregulation of VEGFD and PAK1 by F. nucleatum was confirmed in co-culture experiments, and their knockdown significantly reduced F. nucleatum-induced proliferation and migration. In conclusion, intratumoral microbiota in breast cancer exhibit significant spatial heterogeneity, with F. nucleatum colonization markedly altering tumour cell protein expression to promote progression and migration. These findings provide novel perspectives on the role of microbiota in breast cancer, identify potential therapeutic targets, and lay the foundation for future cancer treatments. KEY POINTS: Intratumoral Fusobacterium nucleatum exhibits significant spatial heterogeneity within breast cancer tissues. F. nucleatum colonization alters the expression of key proteins involved in tumour progression and migration. The MAPK signalling pathway is a critical mediator of F. nucleatum-induced breast cancer cell proliferation and migration. VEGFD and PAK1 are potential therapeutic targets to mitigate F. nucleatum-induced tumour progression.
Collapse
Affiliation(s)
- Feng Zhao
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Rui An
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Yilei Ma
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Shaobo Yu
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Yuzhen Gao
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Yanzhong Wang
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Haitao Yu
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Xinyou Xie
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Jun Zhang
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| |
Collapse
|
22
|
Johri S, Bi K, Titchen BM, Fu J, Conway J, Crowdis JP, Vokes NI, Fan Z, Fong L, Park J, Liu D, He MX, Van Allen EM. Dissecting tumor cell programs through group biology estimation in clinical single-cell transcriptomics. Nat Commun 2025; 16:2090. [PMID: 40025015 PMCID: PMC11873288 DOI: 10.1038/s41467-025-57377-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
With the growth of clinical cancer single-cell RNA sequencing studies, robust differential expression methods for case/control analyses (e.g., treatment responders vs. non-responders) using gene signatures are pivotal to nominate hypotheses for further investigation. However, many commonly used methods produce a large number of false positives, do not adequately represent the patient-specific hierarchical structure of clinical single-cell RNA sequencing data, or account for sample-driven confounders. Here, we present a nonparametric statistical method, BEANIE, for differential expression of gene signatures between clinically relevant groups that addresses these issues. We demonstrate its use in simulated and real-world clinical datasets in breast cancer, lung cancer and melanoma. BEANIE outperforms existing methods in specificity while maintaining sensitivity, as demonstrated in simulations. Overall, BEANIE provides a methodological strategy to inform biological insights into unique and shared differentially expressed gene signatures across different tumor states, with utility in single-study, meta-analysis, and cross-validation across cell types.
Collapse
Affiliation(s)
- Shreya Johri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Kevin Bi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Breanna M Titchen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard Graduate Program in Biological and Biomedical Sciences, Boston, MA, USA
| | - Jingxin Fu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jake Conway
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard Graduate Program in Bioinformatics and Integrative Genomics, Boston, MA, USA
| | - Jett P Crowdis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Natalie I Vokes
- Department of Thoracic and Head and Neck Oncology, MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Zenghua Fan
- Division of Hematology/Oncology, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, CA, USA
| | - Lawrence Fong
- Division of Hematology/Oncology, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, CA, USA
| | - Jihye Park
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - David Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Meng Xiao He
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
23
|
Luo H, She X, Zhang Y, Xie B, Zhang S, Li Q, Zhou Y, Guo S, Zhang S, Jiang Y, Dong Y, He J, Wang L, Zhang Q, Zhuang Y, Deng P, Wang F, Liu J, Chen X, Nie H, He H. PLIN2 Promotes Lipid Accumulation in Ascites-Associated Macrophages and Ovarian Cancer Progression by HIF1α/SPP1 Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411314. [PMID: 39921309 PMCID: PMC11948008 DOI: 10.1002/advs.202411314] [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: 09/14/2024] [Revised: 01/21/2025] [Indexed: 02/10/2025]
Abstract
A major characteristic of ovarian cancer (OC) is its unique route of metastasis via ascites. The immune microenvironment in ascites remains understudied, leaving the mechanism of ascites-mediated abdominal metastasis obscure. Here, a single-cell transcriptomic landscape of CD45+ immune cells across multiple anatomical sites is depicted, including primary tumors, metastatic lesions, and ascites, from patients diagnosed with high-grade serous ovarian carcinoma (HGSOC). A novel subset of perilipin 2 high (PLIN2hi) macrophages are identified that are enriched in ascites and positively correlated with OC progression, hence being designated as "ascites-associated macrophages (AAMs)". AAMs are lipid-loaded with overexpression of the lipid droplet protein PLIN2. Overexpression or suppression of PLIN2 can enhance or inhibit tumor cell migration, invasion, and vascular permeability in vitro, which is also confirmed in vivo. Mechanistically, it is demonstrated that PLIN2 boosts HIF1α/SPP1 signaling in macrophages, thereby exerting pro-tumor functions. Finally, a PLIN2-targeting liposome is designed to efficiently suppress ascites production and tumor metastasis. Taken together, this work provides a comprehensive characterization of the cancer-promoting function and lipid-rich property of ascites-enriched PLIN2hi macrophages, establishes a link between lipid metabolism and hypoxia within the context of the ascites microenvironment, and elucidates the pivotal role of ascites in trans-coelomic metastasis of OC.
Collapse
Affiliation(s)
- Hui Luo
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Xiaolu She
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Yubo Zhang
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Bingfan Xie
- Department of GynecologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Shibo Zhang
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Qianqian Li
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Yangyang Zhou
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Shuang Guo
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Shushan Zhang
- Department of UltrasoundThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Yanhui Jiang
- Cancer CenterThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Yingying Dong
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Jianzhong He
- Cancer CenterThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Lijie Wang
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Qianqian Zhang
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Yuan Zhuang
- Department of GynecologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Panxia Deng
- Department of GynecologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Feng Wang
- Department of GynecologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Jihong Liu
- Department of Gynecology OncologyState Key Laboratory of Oncology in South ChinaSun Yat‐Sen University Cancer CenterGuangzhouGuangdong510060China
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauMacau999078China
| | - Huilong Nie
- Department of GynecologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Huanhuan He
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| |
Collapse
|
24
|
Teng F, Wei H, Che D, Miao K, Dong X. Identifying macrophage-associated subtypes in patients with serous ovarian cancer and exploring potential personalized therapeutic drugs using combined single-cell and bulk RNA sequencing omics. Heliyon 2025; 11:e42429. [PMID: 40028569 PMCID: PMC11870195 DOI: 10.1016/j.heliyon.2025.e42429] [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: 05/29/2024] [Revised: 01/14/2025] [Accepted: 01/31/2025] [Indexed: 03/05/2025] Open
Abstract
Purpose We aimed to analyze the sensitivity of patients to chemotherapy drugs and actively explore potential new intervention targets, providing an essential reference for personalized treatment. Methods Candidate markers with significant differential expression in macrophages were identified by analyzing gene expression at the single-cell level. A weighted gene co-expression network (WGCN) was constructed on the GSE26712 dataset to explore the modules most relevant to macrophages. Differentially expressed genes for specific markers were identified. A multi-factor regulatory network was constructed based on single-cell dataset markers screening, differentially expressed genes, and genes commonly present in WGCNA modules. Different macrophage subtypes were identified using this network. Machine learning was used to filter and predict the markers' drug sensitivity, and the potential therapeutic compounds for specific markers were screened. Results We identified 14 and 17 of M1 and M2 macrophage candidate markers, respectively. In the multi-factor regulatory network of M1 macrophages, 6 out of 14 markers recognized 159 transcription factors (TFs) and 48 micro RNAs (miRNAs), whereas 13 of 17 markers recognized 191 TFs and 182 miRNAs in the multi-factor regulatory network of M2 macrophages. Filtering of the identified differentially expressed genes using random forests yielded 15 M1 and M2 macrophage-specific markers. Drug sensitivity prediction analysis and in vitro experiments revealed the close association of these markers with common chemotherapy drug sensitivity. Conclusion We identified specific M1 and M2 macrophage markers and found potential therapeutic compounds (dasatinib and afatinib) in these specific markers. These potential therapeutic compounds provide insight into the underlying mechanisms of serous ovarian cancer (OC) and inspire more effective treatment methods.
Collapse
Affiliation(s)
- Fei Teng
- In-Patient Ultrasound Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Ultrasound Department, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hong Wei
- In-Patient Ultrasound Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dehong Che
- Ultrasound Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kuo Miao
- Ultrasound Department, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoqiu Dong
- Ultrasound Department, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
25
|
Ortega-Batista A, Jaén-Alvarado Y, Moreno-Labrador D, Gómez N, García G, Guerrero EN. Single-Cell Sequencing: Genomic and Transcriptomic Approaches in Cancer Cell Biology. Int J Mol Sci 2025; 26:2074. [PMID: 40076700 PMCID: PMC11901077 DOI: 10.3390/ijms26052074] [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: 10/30/2024] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
This article reviews the impact of single-cell sequencing (SCS) on cancer biology research. SCS has revolutionized our understanding of cancer and tumor heterogeneity, clonal evolution, and the complex interplay between cancer cells and tumor microenvironment. SCS provides high-resolution profiling of individual cells in genomic, transcriptomic, and epigenomic landscapes, facilitating the detection of rare mutations, the characterization of cellular diversity, and the integration of molecular data with phenotypic traits. The integration of SCS with multi-omics has provided a multidimensional view of cellular states and regulatory mechanisms in cancer, uncovering novel regulatory mechanisms and therapeutic targets. Advances in computational tools, artificial intelligence (AI), and machine learning have been crucial in interpreting the vast amounts of data generated, leading to the identification of new biomarkers and the development of predictive models for patient stratification. Furthermore, there have been emerging technologies such as spatial transcriptomics and in situ sequencing, which promise to further enhance our understanding of tumor microenvironment organization and cellular interactions. As SCS and its related technologies continue to advance, they are expected to drive significant advances in personalized cancer diagnostics, prognosis, and therapy, ultimately improving patient outcomes in the era of precision oncology.
Collapse
Affiliation(s)
- Ana Ortega-Batista
- Faculty of Science and Technology, Technological University of Panama, Ave Justo Arosemena, Entre Calle 35 y 36, Corregimiento de Calidonia, Panama City, Panama; (A.O.-B.)
| | - Yanelys Jaén-Alvarado
- Faculty of Science and Technology, Technological University of Panama, Ave Justo Arosemena, Entre Calle 35 y 36, Corregimiento de Calidonia, Panama City, Panama; (A.O.-B.)
- Gorgas Memorial Institute for Health Studies, Ave Justo Arosemena, Entre Calle 35 y 36, Corregimiento de Calidonia, Panama City, Panama
| | - Dilan Moreno-Labrador
- Faculty of Science and Technology, Technological University of Panama, Ave Justo Arosemena, Entre Calle 35 y 36, Corregimiento de Calidonia, Panama City, Panama; (A.O.-B.)
| | - Natasha Gómez
- Faculty of Science and Technology, Technological University of Panama, Ave Justo Arosemena, Entre Calle 35 y 36, Corregimiento de Calidonia, Panama City, Panama; (A.O.-B.)
| | - Gabriela García
- Faculty of Science and Technology, Technological University of Panama, Ave Justo Arosemena, Entre Calle 35 y 36, Corregimiento de Calidonia, Panama City, Panama; (A.O.-B.)
| | - Erika N. Guerrero
- Gorgas Memorial Institute for Health Studies, Ave Justo Arosemena, Entre Calle 35 y 36, Corregimiento de Calidonia, Panama City, Panama
- Sistema Nacional de Investigación, Secretaria Nacional de Ciencia y Tecnología, Edificio 205, Ciudad del Saber, Panama City, Panama
| |
Collapse
|
26
|
An Q, Duan L, Wang Y, Wang F, Liu X, Liu C, Hu Q. Role of CD4 + T cells in cancer immunity: a single-cell sequencing exploration of tumor microenvironment. J Transl Med 2025; 23:179. [PMID: 39953548 PMCID: PMC11829416 DOI: 10.1186/s12967-025-06167-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/22/2025] [Indexed: 02/17/2025] Open
Abstract
Recent oncological research has intensely focused on the tumor immune microenvironment (TME), particularly the functions of CD4 + T lymphocytes. CD4+ T lymphocytes have been implicated in antigen presentation, cytokine release, and cytotoxicity, suggesting their contribution to the dynamics of the TME. Furthermore, the application of single-cell sequencing has yielded profound insights into the phenotypic diversity and functional specificity of CD4+ T cells in the TME. In this review, we discuss the current findings from single-cell analyses, emphasizing the heterogeneity of CD4+ T cell subsets and their implications in tumor immunology. In addition, we review the critical signaling pathways and molecular networks underpinning CD4+ T cell activities, thereby offering novel perspectives on therapeutic targets and strategies for cancer treatment and prognosis.
Collapse
Affiliation(s)
- Qi An
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Li Duan
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yuanyuan Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Fuxin Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiang Liu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Gannan Medical University, Jiangxi, 341000, China.
| | - Chao Liu
- Department of Radiation Oncology, Peking University First Hospital, Beijing, 100034, China.
| | - Qinyong Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
27
|
Wang C, Liu ZP. Diffusion-based generation of gene regulatory networks from scRNA-seq data with DigNet. Genome Res 2025; 35:340-354. [PMID: 39694856 PMCID: PMC11874984 DOI: 10.1101/gr.279551.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024]
Abstract
A gene regulatory network (GRN) intricately encodes the interconnectedness of identities and functionalities of genes within cells, ultimately shaping cellular specificity. Despite decades of endeavors, reverse engineering of GRNs from gene expression profiling data remains a profound challenge, particularly when it comes to reconstructing cell-specific GRNs that are tailored to precise cellular and genetic contexts. Here, we propose a discrete diffusion generation model, called DigNet, capable of generating corresponding GRNs from high-throughput single-cell RNA sequencing (scRNA-seq) data. DigNet embeds the network generation process into a multistep recovery procedure with Markov properties. Each intermediate step has a specific model to recover a portion of the gene regulatory architectures. It thus can ensure compatibility between global network structures and regulatory modules through the unique multistep diffusion procedure. Furthermore, through iMetacell integration and non-Euclidean discrete space modeling, DigNet is robust to the presence of noise in scRNA-seq data and the sparsity of GRNs. Benchmark evaluation results against more than a dozen state-of-the-art network inference methods demonstrate that DigNet achieves superior performance across various single-cell GRN reconstruction experiments. Furthermore, DigNet provides unique insights into the immune response in breast cancer, derived from differential gene regulation identified in T cells. As an open-source software, DigNet offers a powerful and effective tool for generating cell-specific GRNs from scRNA-seq data.
Collapse
Affiliation(s)
- Chuanyuan Wang
- Department of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Zhi-Ping Liu
- Department of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| |
Collapse
|
28
|
Xu S, Han C, Zhou J, Yang D, Dong H, Zhang Y, Zhao T, Tian Y, Wu Y. Distinct maturity and spatial distribution of tertiary lymphoid structures in head and neck squamous cell carcinoma: implications for tumor immunity and clinical outcomes. Cancer Immunol Immunother 2025; 74:107. [PMID: 39932546 PMCID: PMC11813844 DOI: 10.1007/s00262-025-03952-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/20/2025] [Indexed: 02/14/2025]
Abstract
The influence of tertiary lymphoid structures (TLSs) on disease progression and the response to immunotherapy in head and neck squamous cell carcinoma (HNSCC) is well established, yet the heterogeneity among these structures remains largely unexplored. We utilized digital spatial profiling technology to perform in situ transcriptomic sequencing of TLSs across varying levels of maturation and distinct tumor regions within HNSCC. We assessed the prognostic significance of TLS maturation and spatial distribution in 260 patients with HNSCC through hematoxylin and eosin staining and multiplex immunohistochemistry. Furthermore, we established a TLS scoring system to predict survival in patients with HNSCC. Our study revealed that mature TLSs in the intratumor region (Intra-TLSs) of HNSCC, enriched with memory B cells, plasma cells, and CD4+ T cells, presented increased B-cell activity gene expression. Conversely, early TLSs (E-TLSs), abundant in endothelial cells, fibroblasts, and regulatory T cells, express epithelial‒mesenchymal transition (EMT)-related genes, potentially fostering tumor growth. Compared with mature TLSs within the peritumoral region (Peri-TLSs), mature Intra-TLSs have greater memory B-cell and macrophage densities and upregulate genes involved in B-cell receptor signaling and immune effector processes. Mature Peri-TLSs, characterized by endothelial cell enrichment and EMT receptor interaction genes, may contribute to tumor progression and immune evasion. Patients with mature Intra-TLSs or invasive margin TLSs (Invas-TLSs) have improved 5-year survival, whereas those with mature Peri-TLSs have poorer prognoses. By integrating TLS maturity and distribution in HNSCC, we developed a TLS scoring system to guide personalized treatments, which is crucial for predicting outcomes.
Collapse
Affiliation(s)
- Shuai Xu
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Department of Head and Neck Surgery, The Second Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, People's Republic of China
| | - Chao Han
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
| | - Jian Zhou
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
| | - Di Yang
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
| | - Hui Dong
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
| | - Yiwei Zhang
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
| | - Tingting Zhao
- Chongqing International Institute for Immunology, Chongqing, 400030, People's Republic of China
| | - Yi Tian
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China.
| | - Yuzhang Wu
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China.
- Chongqing International Institute for Immunology, Chongqing, 400030, People's Republic of China.
| |
Collapse
|
29
|
Samiea A, Celis G, Yadav R, Rodda LB, Moreau JM. B cells in non-lymphoid tissues. Nat Rev Immunol 2025:10.1038/s41577-025-01137-6. [PMID: 39910240 DOI: 10.1038/s41577-025-01137-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2025] [Indexed: 02/07/2025]
Abstract
B cells have long been understood to be drivers of both humoral and cellular immunity. Recent advances underscore this importance but also indicate that in infection, inflammatory disease and cancer, B cells function directly at sites of inflammation and form tissue-resident memory populations. The spatial organization and cellular niches of tissue B cells have profound effects on their function and on disease outcome, as well as on patient response to therapy. Here we review the role of B cells in peripheral tissues in homeostasis and disease, and discuss the newly identified cellular and molecular signals that are involved in regulating their activity. We integrate emerging data from multi-omic human studies with experimental models to propose a framework for B cell function in tissue inflammation and homeostasis.
Collapse
Affiliation(s)
- Abrar Samiea
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA
| | - George Celis
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Rashi Yadav
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Lauren B Rodda
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA.
| | - Joshua M Moreau
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA.
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA.
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA.
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
30
|
Zhang P, Wang X, Cen X, Zhang Q, Fu Y, Mei Y, Wang X, Wang R, Wang J, Ouyang H, Liang T, Xia H, Han X, Guo G. A deep learning framework for in silico screening of anticancer drugs at the single-cell level. Natl Sci Rev 2025; 12:nwae451. [PMID: 39872221 PMCID: PMC11771446 DOI: 10.1093/nsr/nwae451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 01/30/2025] Open
Abstract
Tumor heterogeneity plays a pivotal role in tumor progression and resistance to clinical treatment. Single-cell RNA sequencing (scRNA-seq) enables us to explore heterogeneity within a cell population and identify rare cell types, thereby improving our design of targeted therapeutic strategies. Here, we use a pan-cancer and pan-tissue single-cell transcriptional landscape to reveal heterogeneous expression patterns within malignant cells, precancerous cells, as well as cancer-associated stromal and endothelial cells. We introduce a deep learning framework named Shennong for in silico screening of anticancer drugs for targeting each of the landscape cell clusters. Utilizing Shennong, we could predict individual cell responses to pharmacologic compounds, evaluate drug candidates' tissue damaging effects, and investigate their corresponding action mechanisms. Prioritized compounds in Shennong's prediction results include FDA-approved drugs currently undergoing clinical trials for new indications, as well as drug candidates reporting anti-tumor activity. Furthermore, the tissue damaging effect prediction aligns with documented injuries and terminated discovery events. This robust and explainable framework has the potential to accelerate the drug discovery process and enhance the accuracy and efficiency of drug screening.
Collapse
Affiliation(s)
- Peijing Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xueyi Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Xufeng Cen
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
- Research Center of Clinical Pharmacy of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310058, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Yuqing Mei
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Xinru Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Renying Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Hongwei Ouyang
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310058, China
| | - Hongguang Xia
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
- Research Center of Clinical Pharmacy of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaoping Han
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou 310000, China
| |
Collapse
|
31
|
Liu C, Chen Y, Li X, Bai Z, Jiang M, Sheng D, Zou W, Huang R, Huang Q, Wang F, Zhu J, Sun H, Liu B, Li Z, Sun B. Pre-immunotherapy alters stereotactic ablative radiotherapy-induced systemic T cell responses in early-stage NSCLC. Cancer Immunol Immunother 2025; 74:80. [PMID: 39891774 PMCID: PMC11787101 DOI: 10.1007/s00262-024-03935-8] [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: 07/08/2024] [Accepted: 12/27/2024] [Indexed: 02/03/2025]
Abstract
BACKGROUND Stereotactic ablative radiotherapy (SABR) is thought to activate T cell responses in patients with cancer, leading to its combination with immunotherapy and chemotherapy for treatment of non-small-cell lung cancer (NSCLC). Here, we aimed to provide a high-resolution transcriptomic profiling of the systemic T cell response following SABR, with or without preceding immunotherapy/chemotherapy. METHODS We conducted single-cell RNA and T cell receptor (TCR) sequencing of T cells from peripheral blood of seven patients with early-stage NSCLC taken pre- and post-SABR without or with prior immunotherapy and chemotherapy (icSABR). Other flow cytometry, single-cell RNA-seq data and bulk RNA-seq data were used to validate the results. RESULTS We uncovered distinct T cell response patterns induced by these treatments: while terminal effector CD8+ T cells showed increased cytotoxic and inhibitory scores, and upregulated immune-activated pathways post-SABR, the reverse responses occurred post-icSABR. Furthermore, the proportion of large T cell clones increased and single clone decreased post-SABR, while the opposite was seen post-icSABR. Of note, both SABR and icSABR largely changed TCR clonotypes, which were mainly large clones post-SABR but single clone post-icSABR, and predominantly from terminal effector CD8+ T cells and T helper cells, respectively. CONCLUSIONS These findings reveal a complex interplay between SABR and immunotherapy, with potentially valuable implications for treatment strategies involving SABR and immunotherapy to induce systemic T cell responses for tumor eradication in patients with NSCLC.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/radiotherapy
- Lung Neoplasms/immunology
- Lung Neoplasms/therapy
- Lung Neoplasms/pathology
- Radiosurgery/methods
- Immunotherapy/methods
- Male
- Female
- Aged
- Middle Aged
- Neoplasm Staging
- T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Combined Modality Therapy
Collapse
Affiliation(s)
- Chao Liu
- Department of Radiation Oncology, Peking University First Hospital, Beijing, 100034, China
| | - Yanjuan Chen
- Department of Geriatrics and Division of Rheumatology and Research, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China
| | - Xiaohui Li
- Department of Medical Oncology, Peking University First Hospital, Beijing, 100034, China
| | - Zhijie Bai
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Meilin Jiang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Dongsheng Sheng
- Department of Thoracic Surgery, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Wenxue Zou
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Rui Huang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Qingyu Huang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Fuhao Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Jingyang Zhu
- Department of Radiation Oncology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Huiru Sun
- Department of Radiation Oncology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Bing Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China.
| | - Zongcheng Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China.
| | - Bing Sun
- Department of Radiation Oncology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China.
| |
Collapse
|
32
|
Supplitt S, Karpinski P, Sasiadek M, Laczmanski L, Kujawa D, Matkowski R, Kasprzak P, Abrahamowska M, Maciejczyk A, Iwaneczko E, Laczmanska I. The analysis of transcriptomic signature of TNBC-searching for the potential RNA-based predictive biomarkers to determine the chemotherapy sensitivity. J Appl Genet 2025; 66:171-182. [PMID: 38722458 PMCID: PMC11761126 DOI: 10.1007/s13353-024-00876-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 08/17/2024]
Abstract
Neoadjuvant chemotherapy is the foundation treatment for triple-negative breast cancer (TNBC) and frequently results in pathological complete response (pCR). However, there are large differences in clinical response and survival after neoadjuvant chemotherapy of TNBC patients. The aim was to identify genes whose expression significantly associates with the efficacy of neoadjuvant chemotherapy in patients with TNBC. Transcriptomes of 46 formalin-fixed paraffin-embedded (FFPE) tumor samples from TNBC patients were analyzed by RNA-seq by comparing 26 TNBCs with pCR versus 20 TNBCs with pathological partial remission (pPR). Subsequently, we narrowed down the list of genes to those that strongly correlated with drug sensitivity of 63 breast cancer cell lines based on Dependency Map Consortium data re-analysis. Furthermore, the list of genes was limited to those presenting specific expression in breast tumor cells as revealed in three large published single-cell RNA-seq breast cancer datasets. Finally, we analyzed which of the selected genes were significantly associated with overall survival (OS) in TNBC TCGA dataset. A total of 105 genes were significantly differentially expressed in comparison between pPR versus pCR. As revealed by PLSR analysis in breast cancer cell lines, out of 105 deregulated genes, 42 were associated with sensitivity to docetaxel, doxorubicin, paclitaxel, and/or cyclophosphamide. We found that 24 out of 42 sensitivity-associated genes displayed intermediate or strong expression in breast malignant cells using single-cell RNAseq re-analysis. Finally, 10 out of 24 genes were significantly associated with overall survival in TNBC TCGA dataset. Our RNA-seq-based findings suggest that there might be transcriptomic signature consisted of 24 genes specifically expressed in tumor malignant cells for predicting neoadjuvant response in FFPE samples from TNBC patients prior to treatment initiation. Additionally, nine out of 24 genes were potential survival predictors in TNBC. This group of 24 genes should be further investigated for its potential to be translated into a predictive test(s).
Collapse
Affiliation(s)
- Stanislaw Supplitt
- Lower Silesian Oncology, Pulmonology and Hematology Center, Hirszfelda Sq. 12, 53-413, Wroclaw, Poland
- Department of Genetics, Wroclaw Medical University, Marcinkowskiego 1, 50-368, Wroclaw, Poland
| | - Pawel Karpinski
- Department of Genetics, Wroclaw Medical University, Marcinkowskiego 1, 50-368, Wroclaw, Poland
| | - Maria Sasiadek
- Department of Genetics, Wroclaw Medical University, Marcinkowskiego 1, 50-368, Wroclaw, Poland
| | - Lukasz Laczmanski
- Laboratory of Genomics and Bioinformatics, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Dorota Kujawa
- Laboratory of Genomics and Bioinformatics, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Rafal Matkowski
- Lower Silesian Oncology, Pulmonology and Hematology Center, Hirszfelda Sq. 12, 53-413, Wroclaw, Poland
- Department of Oncology, Wroclaw Medical University, Hirszfelda 12, 53-413, Wroclaw, Poland
| | - Piotr Kasprzak
- Lower Silesian Oncology, Pulmonology and Hematology Center, Hirszfelda Sq. 12, 53-413, Wroclaw, Poland
| | - Mariola Abrahamowska
- Lower Silesian Oncology, Pulmonology and Hematology Center, Hirszfelda Sq. 12, 53-413, Wroclaw, Poland
- Department of Oncology, Wroclaw Medical University, Hirszfelda 12, 53-413, Wroclaw, Poland
| | - Adam Maciejczyk
- Lower Silesian Oncology, Pulmonology and Hematology Center, Hirszfelda Sq. 12, 53-413, Wroclaw, Poland
- Department of Oncology, Wroclaw Medical University, Hirszfelda 12, 53-413, Wroclaw, Poland
| | - Ewelina Iwaneczko
- Lower Silesian Oncology, Pulmonology and Hematology Center, Hirszfelda Sq. 12, 53-413, Wroclaw, Poland
| | - Izabela Laczmanska
- Lower Silesian Oncology, Pulmonology and Hematology Center, Hirszfelda Sq. 12, 53-413, Wroclaw, Poland.
- Department of Genetics, Wroclaw Medical University, Marcinkowskiego 1, 50-368, Wroclaw, Poland.
| |
Collapse
|
33
|
Belabed M, Park MD, Blouin CM, Balan S, Moon CY, Freed G, Quijada-Álamo M, Peros A, Mattiuz R, Reid AM, Yatim N, Boumelha J, Azimi CS, LaMarche NM, Troncoso L, Amabile A, Le Berichel J, Chen ST, Wilk CM, Brown BD, Radford KJ, Ghosh S, Rothlin CV, Yvan-Charvet L, Marron TU, Puleston DJ, Wagenblast E, Bhardwaj N, Lamaze C, Merad M. Cholesterol mobilization regulates dendritic cell maturation and the immunogenic response to cancer. Nat Immunol 2025; 26:188-199. [PMID: 39838105 DOI: 10.1038/s41590-024-02065-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 12/10/2024] [Indexed: 01/23/2025]
Abstract
Maturation of conventional dendritic cells (cDCs) is crucial for maintaining tolerogenic safeguards against auto-immunity and for promoting immunogenic responses to pathogens and cancer. The subcellular mechanism for cDC maturation remains poorly defined. We show that cDCs mature by leveraging an internal reservoir of cholesterol (harnessed from extracellular cell debris and generated by de novo synthesis) to assemble lipid nanodomains on cell surfaces of maturing cDCs, enhance expression of maturation markers and stabilize immune receptor signaling. This process is dependent on cholesterol transport through Niemann-Pick disease type C1 (NPC1) and mediates homeostatic and Toll-like receptor (TLR)-induced maturation. Importantly, we identified the receptor tyrosine kinase AXL as a regulator of the NPC1-dependent construction of lipid nanodomains. Deleting AXL from cDCs enhances their maturation, thus improving anti-tumor immunity. Altogether, our study presents new insights into cholesterol mobilization as a fundamental basis for cDC maturation and highlights AXL as a therapeutic target for modulating cDCs.
Collapse
Affiliation(s)
- 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 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cédric M Blouin
- Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, Centre de Recherche, Institut Curie, PSL Research University, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Centre National de la Recherche Scientifique, Paris, France
| | - Sreekumar Balan
- 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chang Y 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Grace Freed
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miguel Quijada-Álamo
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ante Peros
- 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 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amanda M Reid
- 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nader Yatim
- 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jesse Boumelha
- 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Camillia S Azimi
- 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nelson M LaMarche
- 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leanna Troncoso
- 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Angelo Amabile
- 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Le Berichel
- 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven T Chen
- 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - C Matthias Wilk
- 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian D Brown
- 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristen J Radford
- Mater Research Institute, the University of Queensland, Brisbane, Queensland, Australia
| | - Sourav Ghosh
- Department of Immunobiology and Department of Pharmacology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Carla V Rothlin
- Department of Neurology and Department of Pharmacology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Laurent Yvan-Charvet
- INSERM U1065, ATIP-Avenir, Fédération Hospitalo-Universitaire OncoAge, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Thomas U Marron
- 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel J 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elvin Wagenblast
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nina Bhardwaj
- 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christophe Lamaze
- Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, Centre de Recherche, Institut Curie, PSL Research University, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Centre National de la Recherche Scientifique, Paris, France
| | - 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 Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
34
|
Sebastião AI, Simões G, Oliveira F, Mateus D, Falcão A, Carrascal MA, Gomes C, Neves B, Cruz MT. Dendritic cells in triple-negative breast cancer: From pathophysiology to therapeutic applications. Cancer Treat Rev 2025; 133:102884. [PMID: 39837068 DOI: 10.1016/j.ctrv.2025.102884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/28/2024] [Accepted: 01/11/2025] [Indexed: 01/23/2025]
Abstract
Breast cancer is the second most commonly diagnosed cancer in women and the fifth leading cause of cancer-related deaths worldwide. It is a highly heterogeneous disease, consisting of multiple subtypes that vary significantly in clinical characteristics and survival outcomes. Triple-negative breast cancer (TNBC) is a particularly aggressive and challenging subtype of breast cancer. Several immunotherapeutic approaches have been tested in patients with TNBC to improve disease outcomes, including the administration of dendritic cell (DC)-based vaccines. DCs are a heterogeneous cell population that play a crucial role in bridging the innate and adaptive immune systems. Therefore, DCs have been increasingly used in cancer vaccines due to their ability to prime and boost antigen specific T-cell immune responses. This review aims to provide a comprehensive overview of TNBC, including potential targets and pharmacological strategies, as well as an overview of DCs and their relevance in TNBC. In addition, we review ongoing clinical trials and shed light on the evolving landscape of DC-based immunotherapy for TNBC.
Collapse
Affiliation(s)
- Ana Isabel Sebastião
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Coimbra, 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Gonçalo Simões
- Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Coimbra, 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Filomena Oliveira
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Daniela Mateus
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Coimbra, 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; BioMark@UC/CEB-LABBELS, Department of Chemical Engineering, Faculty of Sciences and Technology, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548 Coimbra, Portugal
| | | | - Célia Gomes
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research - iCBR, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Bruno Neves
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal; Institute of Biomedicine - iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Coimbra, 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal.
| |
Collapse
|
35
|
Yu J, Cha J, Koh G, Lee I. HCNetlas: A reference database of human cell type-specific gene networks to aid disease genetic analyses. PLoS Biol 2025; 23:e3002702. [PMID: 39908239 DOI: 10.1371/journal.pbio.3002702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 12/20/2024] [Indexed: 02/07/2025] Open
Abstract
Cell type-specific actions of disease genes add a significant layer of complexity to the genetic architecture underlying diseases, obscuring our understanding of disease mechanisms. Single-cell omics have revealed the functional roles of genes at the cellular level, identifying cell types critical for disease progression. Often, a gene impact on disease through its altered network within specific cell types, rather than mere changes in expression levels. To explore the cell type-specific roles of disease genes, we developed HCNetlas (human cell network atlas), a resource cataloging cell type-specific gene networks (CGNs) for various healthy tissue cells. We also devised 3 network analysis methods to investigate cell type-specific functions of disease genes. These methods involve comparing HCNetlas CGNs with those derived from disease-affected tissue samples. These methods find that systemic lupus erythematosus genes predominantly function in myeloid cells, and Alzheimer's disease genes mainly play roles in inhibitory and excitatory neurons. Additionally, they suggest that many lung cancer-related genes may exert their roles in immune cells. These findings suggest that HCNetlas has the potential to link disease-associated genes to cell types of action, facilitating development of cell type-resolved diagnostics and therapeutic strategies for complex human diseases.
Collapse
Affiliation(s)
- Jiwon Yu
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Junha Cha
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Geon Koh
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
- POSTECH Biotech Center, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| |
Collapse
|
36
|
Li Y, Li H, Lin Y, Zhang D, Peng D, Liu X, Xie J, Hu P, Chen L, Luo H, Peng X. MetaQ: fast, scalable and accurate metacell inference via single-cell quantization. Nat Commun 2025; 16:1205. [PMID: 39885131 PMCID: PMC11782697 DOI: 10.1038/s41467-025-56424-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 01/14/2025] [Indexed: 02/01/2025] Open
Abstract
To overcome the computational barriers of analyzing large-scale single-cell sequencing data, we introduce MetaQ, a metacell algorithm that scales to arbitrarily large datasets with linear runtime and constant memory usage. Inspired by cellular development, MetaQ conceptualizes each metacell as a collective ancestor of biologically similar cells. By quantizing cells into a discrete codebook, where each entry represents a metacell capable of reconstructing the original cells it quantizes, MetaQ identifies homogeneous cell subsets for efficient and accurate metacell inference. This approach reduces computational complexity from exponential to linear while maintaining or surpassing the performance of existing metacell algorithms. Extensive experiments demonstrate that MetaQ excels in downstream tasks such as cell type annotation, developmental trajectory inference, batch integration, and differential expression analysis. Thanks to its superior efficiency and effectiveness, MetaQ makes analyzing datasets with millions of cells practical, offering a powerful solution for single-cell studies in the era of high-throughput profiling.
Collapse
Affiliation(s)
- Yunfan Li
- School of Computer Science, Sichuan University, Chengdu, Sichuan, China
| | - Hancong Li
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan, China
| | - Yijie Lin
- School of Computer Science, Sichuan University, Chengdu, Sichuan, China
| | - Dan Zhang
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dezhong Peng
- School of Computer Science, Sichuan University, Chengdu, Sichuan, China
| | - Xiting Liu
- School of Computer Science, Georgia Insitute of Technology, Atlanta, GA, USA
| | - Jie Xie
- College of Life Science, Sichuan Normal University, Chengdu, Sichuan, China
| | - Peng Hu
- School of Computer Science, Sichuan University, Chengdu, Sichuan, China
| | - Lu Chen
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Han Luo
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan, China
| | - Xi Peng
- School of Computer Science, Sichuan University, Chengdu, Sichuan, China.
- State Key Laboratory of Hydraulics and Mountain River Engineering, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
37
|
Cheng Y, Chen X, Feng L, Yang Z, Xiao L, Xiang B, Wang X, Liu D, Lin P, Shi J, Song G, Qian W, Zhang B, Xu Y, Gao Z, Chen L, Wu Y, Ma J, Lin Y, Zhao H, Peng L, Mao X, Liu Y, Hou H, Yang M, Ji Y, Wang X, Zhou J, Xu X, Liu X, Wei W, Zhang X, Gao Q, Zhou H, Sun Y, Wu K, Fan J. Stromal architecture and fibroblast subpopulations with opposing effects on outcomes in hepatocellular carcinoma. Cell Discov 2025; 11:1. [PMID: 39870619 PMCID: PMC11772884 DOI: 10.1038/s41421-024-00747-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 10/29/2024] [Indexed: 01/29/2025] Open
Abstract
Dissecting the spatial heterogeneity of cancer-associated fibroblasts (CAFs) is vital for understanding tumor biology and therapeutic design. By combining pathological image analysis with spatial proteomics, we revealed two stromal archetypes in hepatocellular carcinoma (HCC) with different biological functions and extracellular matrix compositions. Using paired single-cell RNA and epigenomic sequencing with Stereo-seq, we revealed two fibroblast subsets CAF-FAP and CAF-C7, whose spatial enrichment strongly correlated with the two stromal archetypes and opposing patient prognosis. We discovered two functional units, one is the intratumor inflammatory hub featured by CAF-FAP plus CD8_PDCD1 proximity and the other is the marginal wound-healing hub with CAF-C7 plus Macrophage_SPP1 co-localization. Inhibiting CAF-FAP combined with anti-PD-1 in orthotopic HCC models led to improved tumor regression than either monotherapy. Collectively, our findings suggest stroma-targeted strategies for HCC based on defined stromal archetypes, raising the concept that CAFs change their transcriptional program and intercellular crosstalk according to the spatial context.
Collapse
Affiliation(s)
- Yifei Cheng
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaofang Chen
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, Zhejiang, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen, Guangdong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Li Feng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zhicheng Yang
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Liyun Xiao
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, Zhejiang, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen, Guangdong, China
| | - Bin Xiang
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Shanghai, China
| | - Xiaodong Wang
- School of Computer Science and Technology, Xidian University, Xi'an, Shaanxi, China
| | - Dongbin Liu
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, Zhejiang, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen, Guangdong, China
| | - Penghui Lin
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, Zhejiang, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen, Guangdong, China
| | - Jieyi Shi
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guohe Song
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wulei Qian
- School of Computer Science and Technology, Xidian University, Xi'an, Shaanxi, China
| | - Boan Zhang
- School of Computer Science and Technology, Xidian University, Xi'an, Shaanxi, China
| | - Yanan Xu
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zheng Gao
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lv Chen
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yingcheng Wu
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiaqiang Ma
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Youpei Lin
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haichao Zhao
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lihua Peng
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, Zhejiang, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen, Guangdong, China
| | | | - Yang Liu
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, Zhejiang, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen, Guangdong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Hao Hou
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, Zhejiang, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen, Guangdong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Mingyu Yang
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, Zhejiang, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen, Guangdong, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoying Wang
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xun Xu
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, Guangdong, China
| | - Xiyang Liu
- School of Computer Science and Technology, Xidian University, Xi'an, Shaanxi, China
| | - Wu Wei
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Shanghai, China
| | - Xiaoming Zhang
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology & Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Hu Zhou
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Yidi Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| | - Kui Wu
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, Zhejiang, China.
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen, Guangdong, China.
- Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen, Guangdong, China.
| | - Jia Fan
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
38
|
Zhou Y, Zhao L, Cai M, Luo D, Pang Y, Chen J, Luo Q, Lin Q. Utilizing sc-linker to integrate single-cell RNA sequencing and human genetics to identify cell types and driver genes associated with non-small cell lung cancer. BMC Cancer 2025; 25:130. [PMID: 39849454 PMCID: PMC11755902 DOI: 10.1186/s12885-025-13525-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 01/15/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) provide a powerful method for identifying the loci and genes that contribute to disease. However, in many cases, the specific cell types and states that confer disease risk through these genes remain unknown. Determining this relationship is crucial for identifying pathogenic processes and developing therapeutic strategies. METHODS In this study, we utilized the sc-linker framework developed by Jagadeesh, which is an integrated framework that combines single-cell RNA sequencing (scRNA-seq), epigenomic single nucleotide polymorphism (SNP)-to-gene mapping, and GWAS summary statistics to infer potential cell types and diseases affected by genetic variations. RESULTS Using normal cell type programs in the sc-linker, we identified type 2 alveolar cells in normal lung tissues that are closely associated with non-small cell lung cancer (NSCLC). Additionally, we identified cancer-associated fibroblasts (CAFs) associated with lung cancer using disease-dependent programs. By integrating extensive single-cell data from NSCLC, we discerned heterogeneity among CAFs subgroups. Finally, using MAGMA, we identified RAB31 as a driver gene in disease-related fibroblasts. Proteins from the RAB family are involved in the dynamic regulation of cell membrane compartments and are dysregulated in various tumor types, potentially altering biological properties such as the proliferation, migration, and invasion of cancer cells. We found that the Ras-related protein Rab-31 (RAB31) was significantly overexpressed in tumor-associated fibroblasts compared to that in normal fibroblasts and was closely associated with poor prognosis in patients with NSCLC. CONCLUSIONS By integrating scRNA-seq, epigenomic, and GWAS datasets, we found that ACT2 and CAFs have specific disease heritability in lung cancer and identified the driver gene RAB31 as a potential therapeutic target.
Collapse
Affiliation(s)
- Yangfan Zhou
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Liang Zhao
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Meimei Cai
- Department of Rheumatology and Clinical Immunology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Doudou Luo
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Yizhen Pang
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Jianhao Chen
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Qicong Luo
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China.
| | - Qin Lin
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China.
| |
Collapse
|
39
|
Huang YA, Li YC, You ZH, Hu L, Hu PW, Wang L, Peng Y, Huang ZA. Consensus representation of multiple cell-cell graphs from gene signaling pathways for cell type annotation. BMC Biol 2025; 23:23. [PMID: 39849579 PMCID: PMC11756145 DOI: 10.1186/s12915-025-02128-8] [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: 08/07/2024] [Accepted: 01/13/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Recent advancements in single-cell RNA sequencing have greatly expanded our knowledge of the heterogeneous nature of tissues. However, robust and accurate cell type annotation continues to be a major challenge, hindered by issues such as marker specificity, batch effects, and a lack of comprehensive spatial and interaction data. Traditional annotation methods often fail to adequately address the complexity of cellular interactions and gene regulatory networks. RESULTS We proposed scMCGraph, a comprehensive computational framework that integrates gene expression with pathway activity to accurately annotate cell types within diverse scRNA-seq datasets. Initially, our model constructs multiple pathway-specific views using various pathway databases, which reflect both gene expression and pathway activities. These pathway-specific views are then integrated into a consensus graph. The consensus graph is subsequently utilized to reconstruct the multiple pathway views. Our model demonstrated exceptional robustness and accuracy across various analyses, including cross-platform, cross-time, cross-sample, and clinical dataset evaluations. CONCLUSIONS scMCGraph represents a significant advance in cell type annotation. The experiments have demonstrated that introducing pathway information significantly improves the learning of cell-cell graphs, with their resulting consensus graph enhancing the predictive performance of cell type prediction. Different pathway databases provide complementary data, and an increase in the number of pathways can also boost model performance. Extensive testing shows that in various cross-dataset application scenarios, scMCGraph consistently exhibits both accuracy and robustness.
Collapse
Affiliation(s)
- Yu-An Huang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710000, China.
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518063, China.
| | - Yue-Chao Li
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710000, China
| | - Zhu-Hong You
- School of Electronic Information, Xijing University, Xi'an, 710000, China.
| | - Lun Hu
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Science, Urumqi, 830011, China
| | - Peng-Wei Hu
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Science, Urumqi, 830011, China
| | - Lei Wang
- Guangxi Key Lab of Human-Machine Interaction and Intelligent Decision, Guangxi Academy of Sciences, Nanning, 530001, China
| | - Yuzhong Peng
- Guangxi Key Lab of Human-Machine Interaction and Intelligent Decision, Nanning Normal University, Nanning, 530001, China
| | - Zhi-An Huang
- Research Office, City University of Hong Kong (Dongguan), Dongguan, 523000, China.
| |
Collapse
|
40
|
Wang Z, Dai R, Wang M, Lei L, Zhang Z, Han K, Wang Z, Guo Q. KanCell: dissecting cellular heterogeneity in biological tissues through integrated single-cell and spatial transcriptomics. J Genet Genomics 2025:S1673-8527(24)00310-2. [PMID: 39577768 DOI: 10.1016/j.jgg.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/07/2024] [Accepted: 11/10/2024] [Indexed: 11/24/2024]
Abstract
KanCell is a deep learning model based on Kolmogorov-Arnold networks (KAN) designed to enhance cellular heterogeneity analysis by integrating single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST) data. ST technologies provide insights into gene expression within tissue context, revealing cellular interactions and microenvironments. To fully leverage this potential, effective computational models are crucial. We evaluate KanCell on both simulated and real datasets from technologies such as STARmap, Slide-seq, Visium, and Spatial Transcriptomics. Our results demonstrate that KanCell outperforms existing methods across metrics like PCC, SSIM, COSSIM, RMSE, JSD, ARS, and ROC, with robust performance under varying cell numbers and background noise. Real-world applications on human lymph nodes, hearts, melanoma, breast cancer, dorsolateral prefrontal cortex, and mouse embryo brains confirmed its reliability. Compared with traditional approaches, KanCell effectively captures non-linear relationships and optimizes computational efficiency through KAN, providing an accurate and efficient tool for ST. By improving data accuracy and resolving cell type composition, KanCell reveals cellular heterogeneity, clarifies disease microenvironments, and identifies therapeutic targets, addressing complex biological challenges.
Collapse
Affiliation(s)
- Zhenghui Wang
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Ruoyan Dai
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Mengqiu Wang
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Lixin Lei
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Zhiwei Zhang
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Kaitai Han
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Zijun Wang
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Qianjin Guo
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China.
| |
Collapse
|
41
|
Glaviano A, Lau HSH, Carter LM, Lee EHC, Lam HY, Okina E, Tan DJJ, Tan W, Ang HL, Carbone D, Yee MYH, Shanmugam MK, Huang XZ, Sethi G, Tan TZ, Lim LHK, Huang RYJ, Ungefroren H, Giovannetti E, Tang DG, Bruno TC, Luo P, Andersen MH, Qian BZ, Ishihara J, Radisky DC, Elias S, Yadav S, Kim M, Robert C, Diana P, Schalper KA, Shi T, Merghoub T, Krebs S, Kusumbe AP, Davids MS, Brown JR, Kumar AP. Harnessing the tumor microenvironment: targeted cancer therapies through modulation of epithelial-mesenchymal transition. J Hematol Oncol 2025; 18:6. [PMID: 39806516 PMCID: PMC11733683 DOI: 10.1186/s13045-024-01634-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025] Open
Abstract
The tumor microenvironment (TME) is integral to cancer progression, impacting metastasis and treatment response. It consists of diverse cell types, extracellular matrix components, and signaling molecules that interact to promote tumor growth and therapeutic resistance. Elucidating the intricate interactions between cancer cells and the TME is crucial in understanding cancer progression and therapeutic challenges. A critical process induced by TME signaling is the epithelial-mesenchymal transition (EMT), wherein epithelial cells acquire mesenchymal traits, which enhance their motility and invasiveness and promote metastasis and cancer progression. By targeting various components of the TME, novel investigational strategies aim to disrupt the TME's contribution to the EMT, thereby improving treatment efficacy, addressing therapeutic resistance, and offering a nuanced approach to cancer therapy. This review scrutinizes the key players in the TME and the TME's contribution to the EMT, emphasizing avenues to therapeutically disrupt the interactions between the various TME components. Moreover, the article discusses the TME's implications for resistance mechanisms and highlights the current therapeutic strategies toward TME modulation along with potential caveats.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Hannah Si-Hui Lau
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lukas M Carter
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Donavan Jia Jie Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Wency Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Michelle Yi-Hui Yee
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Xiao Zi Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Lina H K Lim
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Ruby Yun-Ju Huang
- School of Medicine and Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, 23538, Lübeck, Germany
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Experimental Therapeutics (ET) Graduate Program, University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Tullia C Bruno
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Bin-Zhi Qian
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, W12 0BZ, UK
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Salem Elias
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Saurabh Yadav
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Minah Kim
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Caroline Robert
- Department of Cancer Medicine, Inserm U981, Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif, France
- Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, Paris, France
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Tao Shi
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, NY, USA
| | - Simone Krebs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironment Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Matthew S Davids
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jennifer R Brown
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| |
Collapse
|
42
|
Kobayashi H, Iida T, Ochiai Y, Malagola E, Zhi X, White RA, Qian J, Wu F, Waterbury QT, Tu R, Zheng B, LaBella JS, Zamechek LB, Ogura A, Woods SL, Worthley DL, Enomoto A, Wang TC. Neuro-Mesenchymal Interaction Mediated by a β2-Adrenergic Nerve Growth Factor Feedforward Loop Promotes Colorectal Cancer Progression. Cancer Discov 2025; 15:202-226. [PMID: 39137067 PMCID: PMC11729495 DOI: 10.1158/2159-8290.cd-24-0287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/25/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024]
Abstract
SIGNIFICANCE Our work demonstrates that the bidirectional interplay between sympathetic nerves and NGF-expressing CAFs drives colorectal tumorigenesis. This study also offers novel mechanistic insights into catecholamine action in colorectal cancer. Inhibiting the neuro-mesenchymal interaction by TRK blockade could be a potential strategy for treating colorectal cancer.
Collapse
Affiliation(s)
- Hiroki Kobayashi
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY, 10032, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY 10032, USA
| | - Tadashi Iida
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Yosuke Ochiai
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY, 10032, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY 10032, USA
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Ermanno Malagola
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY, 10032, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY 10032, USA
| | - Xiaofei Zhi
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY, 10032, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY 10032, USA
| | - Ruth A. White
- Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jin Qian
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY, 10032, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY 10032, USA
| | - Feijing Wu
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY, 10032, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY 10032, USA
| | - Quin T. Waterbury
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY, 10032, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY 10032, USA
| | - Ruhong Tu
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY, 10032, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY 10032, USA
| | - Biyun Zheng
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY, 10032, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY 10032, USA
| | - Jonathan S. LaBella
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY, 10032, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY 10032, USA
| | - Leah B. Zamechek
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY, 10032, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY 10032, USA
| | - Atsushi Ogura
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Susan L. Woods
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| | - Daniel L. Worthley
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
- Colonoscopy Clinic, Lutwyche, QLD, 4030, Australia
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Division of Molecular Pathology, Center for Neurological Disease and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Timothy C. Wang
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY, 10032, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY 10032, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA
| |
Collapse
|
43
|
Guinn S, Perez B, Tandurella JA, Ramani M, Lee JW, Zabransky DJ, Kartalia E, Patel J, Zlomke H, Nicolson N, Shin S, Barrett B, Sun N, Hernandez A, Coyne E, Cannon C, Gross NE, Charmsaz S, Cho Y, Leatherman J, Lyman M, Mitchell J, Kagohara LT, Goggins MG, Lafaro KJ, He J, Shubert C, Burns W, Zheng L, Fertig EJ, Jaffee EM, Burkhart RA, Ho WJ, Zimmerman JW. Cancer associated fibroblasts drive epithelial to mesenchymal transition and classical to basal change in pancreatic ductal adenocarcinoma cells with loss of IL-8 expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631784. [PMID: 39829906 PMCID: PMC11741337 DOI: 10.1101/2025.01.07.631784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) carries an extremely poor prognosis, in part resulting from cellular heterogeneity that supports overall tumorigenicity. Cancer associated fibroblasts (CAF) are key determinants of PDAC biology and response to systemic therapy. While CAF subtypes have been defined, the effects of patient-specific CAF heterogeneity and plasticity on tumor cell behavior remain unclear. Here, multi-omics was used to characterize the tumor microenvironment (TME) in tumors from patients undergoing curative-intent surgery for PDAC. In these same patients, matched tumor organoid and CAF lines were established to functionally validate the impact of CAFs on the tumor cells. CAFs were found to drive epithelial-mesenchymal transition (EMT) and a switch in tumor cell classificiaton from classical to basal subtype. Furthermore, we identified CAF-specific interleukin 8 (IL-8) as an important modulator of tumor cell subtype. Finally, we defined neighborhood relationships between tumor cell and T cell subsets.
Collapse
Affiliation(s)
- Samantha Guinn
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Brayan Perez
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joseph A Tandurella
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mili Ramani
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jae W Lee
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Daniel J Zabransky
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Emma Kartalia
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jignasha Patel
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Haley Zlomke
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Norman Nicolson
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sarah Shin
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Benjamin Barrett
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nicholas Sun
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alexei Hernandez
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Erin Coyne
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Courtney Cannon
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nicole E Gross
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Soren Charmsaz
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yeonju Cho
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - James Leatherman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Melissa Lyman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jacob Mitchell
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Luciane T Kagohara
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael G Goggins
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Kelly J Lafaro
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jin He
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christopher Shubert
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - William Burns
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lei Zheng
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elana J Fertig
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Applied Mathematics and Statistics, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland
- Institute for Genome Sciences, Department of Medicine, and Greenbaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Elizabeth M Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Richard A Burkhart
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Won Jin Ho
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jacquelyn W Zimmerman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- BloombergKimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
44
|
Wang X, Li Y, Lin Z, Pla I, Gajjela R, Mattamana BB, Joshi M, Liu Y, Wang H, Zun AB, Wang H, Wai CM, Agrawal V, Dunton CL, Duan C, Jiang B, Backman V, He TC, Reid RR, Luo Y, Ameer GA. Micropillar-induced changes in cell nucleus morphology enhance bone regeneration by modulating the secretome. RESEARCH SQUARE 2025:rs.3.rs-5530535. [PMID: 39866882 PMCID: PMC11760244 DOI: 10.21203/rs.3.rs-5530535/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Nuclear morphology, which modulates chromatin architecture, plays a critical role in regulating gene expression and cell functions. While most research has focused on the direct effects of nuclear morphology on cell fate, its impact on the cell secretome and surrounding cells remains largely unexplored, yet is especially crucial for cell-based therapies. In this study, we fabricated implants with a micropillar topography using methacrylated poly(octamethylene citrate)/hydroxyapatite (mPOC/HA) composites to investigate how micropillar-induced nuclear deformation influences cell paracrine signaling for osteogenesis and cranial bone regeneration. In vitro, cells with deformed nuclei showed enhanced secretion of proteins that support extracellular matrix (ECM) organization, which promoted osteogenic differentiation in neighboring human mesenchymal stromal cells (hMSCs). In a mouse model with critical-size cranial defects, nuclear-deformed hMSCs on micropillar mPOC/HA implants elevated Col1a2 expression, contributing to bone matrix formation, and drove cell differentiation toward osteogenic progenitor cells. These findings indicate that micropillars not only enhance the osteogenic differentiation of human mesenchymal stromal cells (hMSCs) but also modulate the secretome, thereby influencing the fate of surrounding cells through paracrine effects.
Collapse
Affiliation(s)
- Xinlong Wang
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Yiming Li
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Zitong Lin
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Indira Pla
- Proteomics Center of Excellence, Northwestern University, Evanston, IL 60208, USA
| | - Raju Gajjela
- Proteomics Center of Excellence, Northwestern University, Evanston, IL 60208, USA
| | - Basil Baby Mattamana
- Proteomics Center of Excellence, Northwestern University, Evanston, IL 60208, USA
| | - Maya Joshi
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Yugang Liu
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Huifeng Wang
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Amy B Zun
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Hao Wang
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Ching-Man Wai
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Vasundhara Agrawal
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Cody L Dunton
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Chongwen Duan
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Bin Jiang
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Vadim Backman
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Chemistry of Life Process Institute, Northwestern University, Evanston, IL 60208, USA
| | - Tong-Chuan He
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R Reid
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yuan Luo
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Northwestern University Clinical and Translational Sciences Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Center for Collaborative AI in Healthcare, Institute for AI in Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Guillermo A Ameer
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Northwestern University Clinical and Translational Sciences Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208, USA
- Simpson Querrey Institute for Bionanotechnology, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
45
|
Lee H, Ko DS, Heo HJ, Baek SE, Kim EK, Kwon EJ, Kang J, Yu Y, Baryawno N, Kim K, Lee D, Kim YH. Uncovering NK cell sabotage in gut diseases via single cell transcriptomics. PLoS One 2025; 20:e0315981. [PMID: 39752457 PMCID: PMC11698320 DOI: 10.1371/journal.pone.0315981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 12/03/2024] [Indexed: 01/06/2025] Open
Abstract
The identification of immune environments and cellular interactions in the colon microenvironment is essential for understanding the mechanisms of chronic inflammatory disease. Despite occurring in the same organ, there is a significant gap in understanding the pathophysiology of ulcerative colitis (UC) and colorectal cancer (CRC). Our study aims to address the distinct immunopathological response of UC and CRC. Using single-cell RNA sequencing datasets, we analyzed the profiles of immune cells in colorectal tissues obtained from healthy donors, UC patients, and CRC patients. The colon tissues from patients and healthy participants were visualized by immunostaining followed by laser confocal microscopy for select targets. Natural killer (NK) cells from UC patients on medication showed reduced cytotoxicity compared to those from healthy individuals. Nonetheless, a UC-specific pathway called the BAG6-NCR3 axis led to higher levels of inflammatory cytokines and increased the cytotoxicity of NCR3+ NK cells, thereby contributing to the persistence of colitis. In the context of colorectal cancer (CRC), both NK cells and CD8+ T cells exhibited significant changes in cytotoxicity and exhaustion. The GALECTIN-9 (LGALS9)-HAVCR2 axis was identified as one of the CRC-specific pathways. Within this pathway, NK cells solely communicated with myeloid cells under CRC conditions. HAVCR2+ NK cells from CRC patients suppressed NK cell-mediated cytotoxicity, indicating a reduction in immune surveillance. Overall, we elucidated the comprehensive UC and CRC immune microenvironments and NK cell-mediated immune responses. Our findings can aid in selecting therapeutic targets that increase the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Hansong Lee
- Medical Research Institute, Pusan National University, Yangsan, Republic of Korea
| | - Dai Sik Ko
- Division of Vascular Surgery, Department of General Surgery, Gachon University College of Medicine, Gil Medical Center, Incheon, Republic of Korea
| | - Hye Jin Heo
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Seung Eun Baek
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Eun Kyoung Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Eun Jung Kwon
- Medical Research Institute, Pusan National University, Yangsan, Republic of Korea
| | - Junho Kang
- Department of Research, Keimyung University Dongsan Medical Center, Daegu, Republic of Korea
| | - Yeuni Yu
- Medical Research Institute, Pusan National University, Yangsan, Republic of Korea
| | - Ninib Baryawno
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Kihun Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Republic of Korea
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan, Republic of Korea
- Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Yun Hak Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Republic of Korea
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
46
|
Tian Y, Zhang L, Ping Y, Zhang Z, Yao C, Shen C, Li F, Wen C, Zhang Y. CCR5 and IL-12 co-expression in CAR T cells improves antitumor efficacy by reprogramming tumor microenvironment in solid tumors. Cancer Immunol Immunother 2025; 74:55. [PMID: 39751840 PMCID: PMC11699016 DOI: 10.1007/s00262-024-03909-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025]
Abstract
Chimeric antigen receptor (CAR) T cell therapy for solid tumors faces significant challenges, including inadequate infiltration, limited proliferation, diminished effector function of CAR T cells, and an immunosuppressive tumor microenvironment (TME). In this study, we utilized The Cancer Genome Atlas database to identify key chemokines (CCL4, CCL5, and CCR5) associated with T cell infiltration across various solid tumor types. The CCL4/CCL5-CCR5 axis emerged as significantly correlated with the presence of T cells within tumors, and enhancing the expression of CCR5 in CAR T cells bolstered their migratory capacity. Furthermore, single-cell immunoprofiling of tumor tissues revealed that macrophages within the TME primarily interact with CD8+ T cells, impeding their tumor response. However, CAR T cells engineered to secrete Interleukin (IL)-12 can counteract macrophage-mediated immunosuppression and augment T cell functionality. To address these obstacles, we employed esophageal carcinoma as a model to develop mesothelin-targeted CAR T cells co-expressing CCR5 and IL-12 (CARTmeso-5-12), subsequently assessing their antitumor capabilities in vitro and in vivo. The CARTmeso-5-12 cells demonstrated enhanced tumor infiltration due to overexpression of CCR5, and IL-12 secretion further amplified CAR T cell efficacy by attenuating the suppressive influence of tumor-infiltrating macrophages, thus improving tumor eradication.
Collapse
Affiliation(s)
- Yonggui Tian
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Laboratory Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liubo Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yu Ping
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zhen Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Chang Yao
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Chunyi Shen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Feng Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Chunli Wen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- School of Public Health, Zhengzhou University, Zhengzhou, Henan, China.
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
47
|
Liu X, Tang G, Chen Y, Li Y, Li H, Wang X. SpatialDeX Is a Reference-Free Method for Cell-Type Deconvolution of Spatial Transcriptomics Data in Solid Tumors. Cancer Res 2025; 85:171-182. [PMID: 39387817 DOI: 10.1158/0008-5472.can-24-1472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/06/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024]
Abstract
The rapid development of spatial transcriptomics (ST) technologies has enabled transcriptome-wide profiling of gene expression in tissue sections. Despite the emergence of single-cell resolution platforms, most ST sequencing studies still operate at a multicell resolution. Consequently, deconvolution of cell identities within the spatial spots has become imperative for characterizing cell-type-specific spatial organization. To this end, we developed Spatial Deconvolution Explorer (SpatialDeX), a regression model-based method for estimating cell-type proportions in tumor ST spots. SpatialDeX exhibited comparable performance to reference-based methods and outperformed other reference-free methods with simulated ST data. Using experimental ST data, SpatialDeX demonstrated superior performance compared with both reference-based and reference-free approaches. Additionally, a pan-cancer clustering analysis on tumor spots identified by SpatialDeX unveiled distinct tumor progression mechanisms both within and across diverse cancer types. Overall, SpatialDeX is a valuable tool for unraveling the spatial cellular organization of tissues from ST data without requiring single-cell RNA-seq references. Significance: The development of a reference-free method for deconvolving the identity of cells in spatial transcriptomics datasets enables exploration of tumor architecture to gain deeper insights into the dynamics of the tumor microenvironment.
Collapse
Affiliation(s)
- Xinyi Liu
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Gongyu Tang
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, Illinois
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri
| | - Yuhao Chen
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Yuanxiang Li
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Hua Li
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, Missouri
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Xiaowei Wang
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, Illinois
- University of Illinois Cancer Center, Chicago, Illinois
| |
Collapse
|
48
|
Wang J, Hua D, Li M, Liu N, Zhang Y, Zhao Y, Jiang S, Hu X, Wang Y, Zhu H. The Role of Zuo Jin Wan in Modulating the Tumor Microenvironment of Colorectal Cancer. Comb Chem High Throughput Screen 2025; 28:523-532. [PMID: 38284730 DOI: 10.2174/0113862073281374231228041841] [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: 09/11/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/30/2024]
Abstract
INTRODUCTION Traditional Chinese medicine (TCM) can modulate the immune function of tumor patients in various ways. Zuojin Wan (ZJW, a 6:1 ratio of Huang Lian and Wu Zhu Yu) can modulate the microenvironment of ulcerative colitis, but its role in regulating the colorectal cancer (CRC) microenvironment remains unclear. Exploring the role of ZJW in CRC immunomodulation may improve the antitumor effect of existing immunotherapeutic strategies. MATERIAL AND METHODS The active compounds of each herb in ZJW were obtained from the HIT2.0 database with literature evidence. Single-cell RNA sequencing data of CRC were obtained from published studies (PMID: 32451460, 32103181, and 32561858). Pathway enrichment was analyzed using the reactome database, and intergenic correlation analysis was performed using the corrplot R software package. ZJW-regulated gene expression was verified by RT-qPCR. RESULTS Huang Lian and Wu Zhu Yu contain 19 and 4 compounds, respectively. Huang Lian targets 146 proteins, and Wu Zhu Yu targets 28 proteins based on evidence from the literature. ZJW regulates a range of biological processes associated with immune function, including cytokine signaling and Toll-Like Receptor 4 (TLR4) cascade. ZJW regulates malignant CRC cells, immune cells (including T-cells, B-cells, mast cells, NK/NKT cells, and myeloid cells), and other nonimmune cells (including endothelial cells, enteric glial cells, and pericytes). We confirmed that ZJW significantly downregulated the expression of TIMP1 and MTDHin CRC cell lines. CONCLUSIONS ZJW regulates a range of cells in the CRC microenvironment, including malignant CRC, immune cells, and stromal cells. In CRC cell lines, downregulation of TIMP1 and MTDH by ZJW may play an important role in the immunomodulation in CRC.
Collapse
Affiliation(s)
- Jiajia Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dongming Hua
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Mengyao Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ningning Liu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yingru Zhang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yiyang Zhao
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shasha Jiang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xueqing Hu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yan Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Huirong Zhu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
49
|
Stoler-Barak L, Sarusi-Portuguez A, Shulman Z. Repertoires and Tumor-Reactivity Analysis of B Cell Immunoglobulins Derived from Cancer Patients. Methods Mol Biol 2025; 2864:263-279. [PMID: 39527227 DOI: 10.1007/978-1-0716-4184-2_14] [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] [Indexed: 11/16/2024]
Abstract
Tumor-infiltrating B cells have emerged in recent years as key markers of patient prognosis and responsiveness to immunotherapy. Recent technical advances, such as single-cell RNA sequencing and B cell receptor immune profiling, revealed diverse subsets and the immunoglobulin landscape of B cells located within human tumors. Secreted antibodies in solid tumors exhibit multiple effector functions, with the potential to significantly impact distinct immune responses, clinical outcomes, and patient survival. Nonetheless, a few studies examine the tumor reactivity and specificity of these immunoglobulins. Here we describe our current methodology for retrieving single B cells from human primary solid tumors for single-cell RNA sequencing followed by computational analysis to identify B cell subpopulations and immunoglobulin receptor repertoires. Furthermore, we provide a technique for evaluating and quantifying the tumor-binding capabilities of expressed antibodies. This approach holds promise for future immunotherapies and enhances our understanding of their potential clinical applications.
Collapse
Affiliation(s)
- Liat Stoler-Barak
- Department of Systems Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Avital Sarusi-Portuguez
- Mantoux Bioinformatics Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Ziv Shulman
- Department of Systems Immunology, The Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
50
|
Hu S, Qin J, Ding M, Gao R, Xiao Q, Lou J, Chen Y, Wang S, Pan Y. Bulk integrated single-cell-spatial transcriptomics reveals the impact of preoperative chemotherapy on cancer-associated fibroblasts and tumor cells in colorectal cancer, and construction of related predictive models using machine learning. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167535. [PMID: 39374811 DOI: 10.1016/j.bbadis.2024.167535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 09/08/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024]
Abstract
BACKGROUND Preoperative chemotherapy (PC) is an important component of Colorectal cancer (CRC) treatment, but its effects on the biological functions of fibroblasts and epithelial cells in CRC are unclear. METHODS This study utilized bulk, single-cell, and spatial transcriptomic sequencing data from 22 independent cohorts of CRC. Through bioinformatics analysis and in vitro experiments, the research investigated the impact of PC on fibroblast and epithelial cells in CRC. Subpopulations associated with PC and CRC prognosis were identified, and a predictive model was constructed using machine learning. RESULTS PC significantly attenuated the pathways related to tumor progression in fibroblasts and epithelial cells. NOTCH3 + Fibroblast (NOTCH3 + Fib), TNNT1 + Epithelial (TNNT1 + Epi), and HSPA1A + Epithelial (HSPA1A + Epi) subpopulations were identified in the adjacent spatial region and were associated with poor prognosis in CRC. PC effectively diminished the presence of these subpopulations, concurrently inhibiting pathway activity and intercellular crosstalk. A risk signature model, named the Preoperative Chemotherapy Risk Signature Model (PCRSM), was constructed using machine learning. PCRSM emerged as an independent prognostic indicator for CRC, impacting both overall survival (OS) and recurrence-free survival (RFS), surpassing the performance of 89 previously published CRC risk signatures. Additionally, patients with a high PCRSM risk score showed sensitivity to fluorouracil-based adjuvant chemotherapy (FOLFOX) but resistance to single chemotherapy drugs (such as Bevacizumab and Oxaliplatin). Furthermore, this study predicted that patients with high PCRSM were resistant to anti-PD1therapy. CONCLUSION In conclusion, this study identified three cell subpopulations (NOTCH3 + Fib, TNNT1 + Epi, and HSPA1A + Epi) associated with PC, which can be targeted to improve the prognosis of CRC patients. The PCRSM model shows promise in enhancing the survival and treatment of CRC patients.
Collapse
Affiliation(s)
- Shangshang Hu
- School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Jian Qin
- School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Muzi Ding
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211122, Jiangsu, China
| | - Rui Gao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211122, Jiangsu, China
| | - QianNi Xiao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211122, Jiangsu, China
| | - Jinwei Lou
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211122, Jiangsu, China
| | - Yuhan Chen
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211122, Jiangsu, China
| | - Shukui Wang
- School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China; Jiangsu Collaborative Innovation Center on Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211100, Jiangsu, China.
| | - Yuqin Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China; Jiangsu Collaborative Innovation Center on Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211100, Jiangsu, China.
| |
Collapse
|