1
|
Cai S, Wei X, Li Q, Jiang Z, Li L. Smart materials in pharmacological drug development: Neutrophils and its constituents for drug delivery and consequent antitumor effects. Mol Immunol 2025; 183:18-32. [PMID: 40318595 DOI: 10.1016/j.molimm.2025.04.010] [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: 12/09/2024] [Revised: 03/17/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025]
Abstract
Neutrophil-based drug delivery systems for targeted therapy of cancer have been studied widely in the recent past. Chemotactic cytokines including colony-stimulating factors (CSFs) recruit various immune cells including the neutrophils to the tumor microenvironment (TME) leading to enhanced metastasis. These cytokines can be targeted effectively by immunotherapeutic agents such as checkpoint inhibitors and mAbs that can lead to systemic toxicity. To minimize the systemic adverse effects, camouflaged nanoparticles can be used significantly as alternative therapeutic agents. The neutrophil-interacting NPs and neutrophil membrane coated NPs have been exploited recently for their antitumor properties in vitro and pose limited systemic adverse effects in vivo. Neutrophil-derived exosomes derived from immune cells can efficiently escape immune-surveillance and pass through the blood-brain barrier. They possess several intrinsic properties in drug delivery as they are nano-sized, extremely biocompatible, non-immunogenic, biodegradable, stable and can carry targeting agents with limited toxicity and display antitumor properties. Also, neutrophil-based nanotherapy is dependent on factors such as neutrophil kinetics and the physicochemical properties of NPs such as size, shape, and surface chemistry. Therefore, neutrophil-based drug delivery for cancer therapy via the use of polymer nanoparticles is widely studied as their clinical appliance in nanomedicine is still at its infancy.
Collapse
Affiliation(s)
- Shengjie Cai
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Xuehan Wei
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Qian Li
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Department of Oncology, Jiangsu Integrated Traditional Chinese and Western Medicine Hospital, Nanjing 210028, China; Department of Oncology, Ganyu Hospital of Traditional Chinese Medicine, Lianyungang, Jiangsu 222000, China
| | - Ziyu Jiang
- Department of Oncology, Lianyungang Integrated Traditional Chinese and Western Medicine Clinical College, Nanjing University of Chinese Medicine, Nanjing 222002, China; Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, China.
| | - Lingchang Li
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Department of Oncology, Jiangsu Integrated Traditional Chinese and Western Medicine Hospital, Nanjing 210028, China.
| |
Collapse
|
2
|
Marciniak M, Stachowicz-Suhs M, Wagner M. The role of innate immune cells in modulating vascular dynamics in skin malignancies. Biochim Biophys Acta Rev Cancer 2025; 1880:189331. [PMID: 40280501 DOI: 10.1016/j.bbcan.2025.189331] [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: 01/28/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
A developing tumor relies heavily on blood vessels to supply oxygen and nutrients. As a result, angiogenesis, the formation of new blood vessels, supports tumor growth and progression. Similarly, lymphangiogenesis, the formation of new lymphatic vessels, plays a critical role in metastatic dissemination by providing pathways for malignant cells to spread. The tumor microenvironment is crucial for establishing and maintaining these vascular networks, with innate immune cells playing a key regulatory role. Notably, immune cells are specifically enriched in barrier tissues, such as the skin, emphasizing their importance in skin malignancies. Therefore, understanding their role in regulating angiogenesis and lymphangiogenesis is essential for developing novel therapeutic strategies. This review article explores how innate immune cells influence tumor vasculature and highlights the therapeutic potential that may arise from this knowledge.
Collapse
Affiliation(s)
- Mateusz Marciniak
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland; Department of Biochemistry and Immunochemistry, Wrocław Medical University, Wrocław, Poland
| | - Martyna Stachowicz-Suhs
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland
| | - Marek Wagner
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland.
| |
Collapse
|
3
|
Issa H, Singh L, Lai KS, Parusheva-Borsitzky T, Ansari S. Dynamics of inflammatory signals within the tumor microenvironment. World J Exp Med 2025; 15:102285. [DOI: 10.5493/wjem.v15.i2.102285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/31/2024] [Accepted: 01/11/2025] [Indexed: 04/16/2025] Open
Abstract
Tumor stroma, or tumor microenvironment (TME), has been in the spotlight during recent years for its role in tumor development, growth, and metastasis. It consists of a myriad of elements, including tumor-associated macrophages, cancer-associated fibroblasts, a deregulated extracellular matrix, endothelial cells, and vascular vessels. The release of proinflammatory molecules, due to the inflamed microenvironment, such as cytokines and chemokines is found to play a pivotal role in progression of cancer and response to therapy. This review discusses the major key players and important chemical inflammatory signals released in the TME. Furthermore, the latest breakthroughs in cytokine-mediated crosstalk between immune cells and cancer cells have been highlighted. In addition, recent updates on alterations in cytokine signaling between chronic inflammation and malignant TME have also been reviewed.
Collapse
Affiliation(s)
- Hala Issa
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Lokjan Singh
- Department of Microbiology, Karnali Academy of Health Sciences, Jumla 21200, Karnali, Nepal
| | - Kok-Song Lai
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Tina Parusheva-Borsitzky
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Shamshul Ansari
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| |
Collapse
|
4
|
Sekar Y, Ishwar D, Tan B, Venkatakrishnan K. Nano biosensor unlocks tumor derived immune signals for the early detection of ovarian cancer. Biosens Bioelectron 2025; 278:117368. [PMID: 40088704 DOI: 10.1016/j.bios.2025.117368] [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: 01/08/2025] [Revised: 02/25/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
Ovarian cancer is a critical health issue for women nowadays. Its impact is significant because of its high mortality rate (324,603 worldwide), late-stage diagnosis and poor survival rate. Lack of screening tests, vague symptoms, misdiagnosis, and age factor makes it even more difficult to detect. Neutrophils, a subset of immune cells, undergo tumor-specific changes as ovarian cancer progresses inside ovarian tumour microenvironment. Therefore, monitoring the time-specific activity of neutrophils in circulation has the potential to aid in the diagnosis of ovarian cancer. Most ovarian tumor-specific antigens are unknown, making it difficult to identify neutrophils associated with ovarian tumor. We present ovarian tumor-associated circulating neutrophil cell profiling as a stand-alone cancer diagnostic method using a liquid biopsy. Using a SERS-functionalized nano probe, the metabolic profiles of neutrophils from ovarian tumor interaction are detected. We demonstrate that neutrophils associated with cancer stem cells have a distinct metabolic profile and are useful in the diagnosis of early ovarian cancer. Using 5 μL of peripheral blood and an artificial neural network, the characteristics of neutrophil profiles in patient blood could distinguish cancer cohort from non-cancer (healthy) with a 90 % sensitivity and 100 % specificity. Our results demonstrate the viability of using circulating neutrophils for non-invasive cancer diagnostics.
Collapse
Affiliation(s)
- Yuvaraj Sekar
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership Between Toronto Metropolitan University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada; Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada; Nano Characterization Laboratory, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada; Nano-Bio Interface Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada
| | - Deeptha Ishwar
- Department of Stomatology, Faculty of Dental Medicine, Universite de Montreal, Montreal, QC, H3C 3J7, Canada
| | - Bo Tan
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership Between Toronto Metropolitan University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada; Nano Characterization Laboratory, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada; Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
| | - Krishnan Venkatakrishnan
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership Between Toronto Metropolitan University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada; Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada; Nano-Bio Interface Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada; Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada.
| |
Collapse
|
5
|
Qu Y, Liang W, Yu M, Wang C, Luo M, Zhong L, Li Z, Wang F. MYO1F in neutrophils is required for the response to immune checkpoint blockade therapy. J Exp Med 2025; 222:e20241957. [PMID: 40202509 PMCID: PMC11980683 DOI: 10.1084/jem.20241957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/29/2025] [Accepted: 02/21/2025] [Indexed: 04/10/2025] Open
Abstract
Tumor-associated neutrophils (TANs) represent a significant barrier to the effectiveness of immune checkpoint blockade (ICB) therapy. A comprehensive understanding of TANs' regulatory mechanisms is therefore essential for predicting ICB efficacy and improving immunotherapy strategies. Our study reveals that MYO1F is selectively downregulated in neutrophils within both human cancers and murine tumor models, showing a negative correlation with ICB response. Mechanistically, MYO1F normally inhibits neutrophil immunosuppression and proliferation by restraining STAT3 activity. However, during tumorigenesis, tumor-derived TGF-β1 disrupts the binding of SPI1 to intron 8 of Myo1f via DNA methylation, thereby suppressing Myo1f transcription. The resultant decrease in MYO1F reprograms neutrophils into an immunosuppressive state through the STAT3-dependent signaling pathways. This immunosuppressive state further contributes to tumor microenvironment (TME) remodeling by inducing CTL exhaustion. These findings establish MYO1F as a critical regulator within TANs, highlighting its significant role in modulating ICB therapy efficacy.
Collapse
Affiliation(s)
- Yingying Qu
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Microbiota and Immunological Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenhua Liang
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingzhu Yu
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenhui Wang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Min Luo
- Institute of Pediatrics of Children’s Hospital of Fudan University, The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Lin Zhong
- Department of Liver Surgery and Organ Transplantation Center, Shenzhen Third People’s Hospital, Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhigang Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Wang
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Xiao L, Li J, Liao J, Wu M, Lu X, Li J, Zeng Y. BCL2A1‑ and G0S2‑driven neutrophil extracellular traps: A protective mechanism linking preeclampsia to reduced breast cancer risk. Oncol Rep 2025; 53:64. [PMID: 40242964 PMCID: PMC12030921 DOI: 10.3892/or.2025.8897] [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: 12/04/2024] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Preeclampsia has been associated with a reduced risk of breast cancer (BC), but the mechanisms underlying this relationship remain unclear. It has been suggested that neutrophil extracellular traps (NETs), which are released upon neutrophil activation, play a key role in both preeclampsia and BC. To investigate this link, the single‑cell RNA sequencing dataset GSE173193 was analyzed and upregulated genes BCL2A1 and G0/G1 switch gene 2 (G0S2) were identified in neutrophils from preeclamptic placentas. These findings were validated using reverse transcription‑quantitative PCR and western blotting. Combined analyses of preeclampsia and BC tissues, from Gene Expression Omnibus (GSE24129) and The Cancer Genome Atlas databases respectively, identified 2,040 upregulated differentially expressed genes, including BCL2A1 and G0S2. Furthermore, these genes showed clinical relevance to BC, as demonstrated by Receiver Operating Characteristic curve, survival analyses and weighted gene co‑expression network analysis. Functional experiments revealed that overexpression of BCL2A1 and G0S2 increased NET release and inhibited BC cell proliferation, invasion and migration. The present study provides novel insights into the shared molecular pathways of preeclampsia and BC, emphasizing NETs as a potential protective mechanism as increased NET production in preeclampsia may contribute to a reduced BC risk by influencing tumor progression and offer avenues for further research into therapeutic interventions.
Collapse
Affiliation(s)
- Lu Xiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jing Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jiahao Liao
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Min Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiujing Lu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jiehua Li
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yachang Zeng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
7
|
Cheng J, Gao J, Li J, Tian H. Neutrophils: a new target for postoperative cognitive dysfunction. Apoptosis 2025; 30:1117-1132. [PMID: 40042745 DOI: 10.1007/s10495-025-02098-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] [Accepted: 02/24/2025] [Indexed: 06/16/2025]
Abstract
Postoperative cognitive dysfunction (POCD) is a prevalent complication affecting the central nervous system after surgery, manifesting as a decline in cognitive abilities, particularly common among elderly patients. Surgical stress and anesthesia can activate systemic inflammation, prompting immune cells, including neutrophils, to infiltrate the brain, thereby triggering neuroinflammation and resulting in cognitive impairment. Neutrophils, as crucial effector cells in innate immunity, have been increasingly recognized in recent years for their significant role in the pathogenesis of POCD due to their vital function in inflammatory responses. They are not only rapidly activated in peripheral blood, secreting a range of cytokines, chemokines, and neutrophil extracellular traps (NETs), but also possess the capacity to alter the permeability of the blood-brain barrier (BBB), further facilitating the development of neuroinflammation. This paper systematically reviews the recent findings on the diverse functions of neutrophils and their role in POCD, aiming to provide novel theoretical foundations and cutting-edge perspectives for advancing foundational research and optimizing clinical intervention strategies for POCD.
Collapse
Affiliation(s)
- Jianxin Cheng
- Department of Anesthesiology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, Qingdao, 266035, China
| | - Jin Gao
- Department of Clinical Psychology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, Qingdao, 266035, China
| | - Jianjun Li
- Department of Anesthesiology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, Qingdao, 266035, China.
| | - Hao Tian
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| |
Collapse
|
8
|
Xiao Z, Puré E. The fibroinflammatory response in cancer. Nat Rev Cancer 2025; 25:399-425. [PMID: 40097577 DOI: 10.1038/s41568-025-00798-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/06/2025] [Indexed: 03/19/2025]
Abstract
Fibroinflammation refers to the highly integrated fibrogenic and inflammatory responses mediated by the concerted function of fibroblasts and innate immune cells in response to tissue perturbation. This process underlies the desmoplastic remodelling of the tumour microenvironment and thus plays an important role in tumour initiation, growth and metastasis. More specifically, fibroinflammation alters the biochemical and biomechanical signalling in malignant cells to promote their proliferation and survival and further supports an immunosuppressive microenvironment by polarizing the immune status of tumours. Additionally, the presence of fibroinflammation is often associated with therapeutic resistance. As such, there is increasing interest in targeting this process to normalize the tumour microenvironment and thus enhance the treatment of solid tumours. Herein, we review advances made in unravelling the complexity of cancer-associated fibroinflammation that can inform the rational design of therapies targeting this.
Collapse
Affiliation(s)
- Zebin Xiao
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Ellen Puré
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Zheng L, Wang J, Jin X, Cheng Q, Zhang X, Li Y, Wang D, Song H, Zhu X, Lin L, Ma J, Gao J, Liang J, Tong J, Shi L. Erythroblastic island: the niche for erythroid terminal differentiation and beyond. BLOOD SCIENCE 2025; 7:e00228. [PMID: 40129604 PMCID: PMC11932602 DOI: 10.1097/bs9.0000000000000228] [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: 11/21/2024] [Accepted: 02/19/2025] [Indexed: 03/26/2025] Open
Abstract
The erythroblastic island (EBI) is a multicellular structure defined by the presence of 1 or 2 central macrophages surrounded by at least 3 erythroblasts. EBIs were initially proposed as a specialized microenvironment exclusively for erythroid terminal differentiation. Recent advancements in techniques such as lineage tracing mouse models, imaging flow cytometry, and single-cell RNA sequencing, accumulating evidence has provided novel insights that challenge this conventional view. Notably, the erythropoietin receptor has been identified as a novel marker for EBI macrophages. Additionally, neutrophils have been identified as novel cellular components of EBIs, raising the intriguing hypothesis that EBIs may support other hematopoietic lineage cells as well. Beyond the diverse cellular components of various hematopoietic lineages, even within the erythroid lineage, an immune-prone erythroblast subpopulation has been reported, although it remains unclear whether and how these immune-prone erythroblasts mature in EBIs. These observations indicate that EBIs are a heterogeneous population. In this review, we summarize the most recent findings on EBIs, discuss their potential immune functions, and provide a perspective for future investigations.
Collapse
Affiliation(s)
- Lingyue Zheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Jingwei Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Xu Jin
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Qimei Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Xiaoru Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Yue Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Di Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Haoze Song
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Xu Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Lexuan Lin
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Jinfa Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Jie Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Jing Liang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Jingyuan Tong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
| | - Lihong Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| |
Collapse
|
10
|
Toghraie FS, Bayat M, Hosseini MS, Ramezani A. Tumor-infiltrating myeloid cells; mechanisms, functional significance, and targeting in cancer therapy. Cell Oncol (Dordr) 2025; 48:559-590. [PMID: 39998754 PMCID: PMC12119771 DOI: 10.1007/s13402-025-01051-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 02/27/2025] Open
Abstract
Tumor-infiltrating myeloid cells (TIMs), which encompass tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), and tumor-associated dendritic cells (TADCs), are of great importance in tumor microenvironment (TME) and are integral to both pro- and anti-tumor immunity. Nevertheless, the phenotypic heterogeneity and functional plasticity of TIMs have posed challenges in fully understanding their complexity roles within the TME. Emerging evidence suggested that the presence of TIMs is frequently linked to prevention of cancer treatment and improvement of patient outcomes and survival. Given their pivotal function in the TME, TIMs have recently been recognized as critical targets for therapeutic approaches aimed at augmenting immunostimulatory myeloid cell populations while depleting or modifying those that are immunosuppressive. This review will explore the important properties of TIMs related to immunity, angiogenesis, and metastasis. We will also document the latest therapeutic strategies targeting TIMs in preclinical and clinical settings. Our objective is to illustrate the potential of TIMs as immunological targets that may improve the outcomes of existing cancer treatments.
Collapse
Affiliation(s)
- Fatemeh Sadat Toghraie
- Institute of Biotechnology, Faculty of the Environment and Natural Sciences, Brandenburg University of Technology, Cottbus, Germany
| | - Maryam Bayat
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Sadat Hosseini
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Amin Ramezani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
11
|
Gao F, Peng H, Gou R, Zhou Y, Ren S, Li F. Exploring neutrophil extracellular traps: mechanisms of immune regulation and future therapeutic potential. Exp Hematol Oncol 2025; 14:80. [PMID: 40442839 PMCID: PMC12123823 DOI: 10.1186/s40164-025-00670-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Accepted: 05/13/2025] [Indexed: 06/02/2025] Open
Abstract
Neutrophil extracellular traps (NETs) are complex, web-like structures consisting of DNA intertwined with antimicrobial proteins, which neutrophils release upon immune activation. These structures play a crucial role in pathogen elimination, particularly in infectious diseases. However, their involvement in various pathological conditions is multifaceted and context-dependent, while NETs contribute to host defense against infections, they can also exacerbate sterile inflammation, autoimmune disorders, and tumor progression. This review provides a comprehensive analysis of the molecular mechanisms governing NET formation and examines their interactions with immune cells, emphasizing how these interactions shape immune responses and drive disease dynamics. Furthermore, it explores ongoing clinical trials and emerging therapeutic strategies targeting NETs, offering critical insights into their potential translational applications in clinical practice.
Collapse
Affiliation(s)
- Fan Gao
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hongwei Peng
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ruixue Gou
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yulan Zhou
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Clinical Research Center for Hematologic Disease, Nanchang, China
- Institute of Lymphoma and Myeloma, Nanchang University, Nanchang, China
| | - Simei Ren
- National Center for Clinical Laboratories, Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
| | - Fei Li
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
- Jiangxi Clinical Research Center for Hematologic Disease, Nanchang, China.
- Institute of Lymphoma and Myeloma, Nanchang University, Nanchang, China.
| |
Collapse
|
12
|
Zhang J, Zhang Q, Lin G, Wang Y, Li J, Wang P, Qi J, Liang Y, He S, Gong Y, Feng N, Wang Y, Ma Y, Zhang M, Shi Y, Li X, Ci W, Zhou L. Single-Cell Analysis Reveals that Vitamin C Inhibits Bone Metastasis of Renal Cancer via Cell Cycle Arrest and Microenvironment Remodeling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e01011. [PMID: 40433925 DOI: 10.1002/advs.202501011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/24/2025] [Indexed: 05/29/2025]
Abstract
Bone metastasis is the second most common site of distant metastatic spread in renal cell carcinoma (RCC) patients, significantly contributing to cancer-related mortality. The metastatic process is driven by both intrinsic tumor cell properties, such as cancer stem cell-like characteristics, and the bone microenvironment. Understanding the complex interactions between cancer cells and their niche is crucial for identifying therapeutic targets to eliminate metastasis-initiating cells and prevent overt metastasis. In this study, a murine bone metastasis model is developed using renal cancer cells derived from fibrin gel-induced 3D tumor spheres, which exhibit stem-like phenotypes. It is found that a stable form of vitamin C, L-ascorbic acid 2-phosphate sesquimagnesium (APM), significantly inhibits the growth of renal cancer stem-like cells in vitro and the progression of RCC bone metastasis in vivo. Single-cell RNA sequencing revealed that APM induces cell cycle arrest and reduces the metastatic potential of cancer cells. Furthermore, APM remodels the tumor microenvironment by suppressing osteoclast differentiation and neutrophil recruitment. Combining APM with a CXCR2 antagonist, SB225002, further inhibits bone metastasis progression. This study provides a high-resolution profile of vitamin C's antitumor effects in the bone metastatic microenvironment and supports the rationale for clinical trials of vitamin C in bone metastatic RCC.
Collapse
Affiliation(s)
- Jianye Zhang
- Department of Urology, Peking University First Hospital, Beijing, 100034, P. R. China
- Institute of Urology, Peking University, Beijing, 100034, P. R. China
- National Urological Cancer Center, Beijing, 100034, P. R. China
| | - Qi Zhang
- China National Center for Bioinformation, Beijing, 100101, P. R. China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Gang Lin
- Department of Thoracic Surgery, Peking University First Hospital, Peking University, Beijing, 100034, P. R. China
| | - Ying Wang
- China National Center for Bioinformation, Beijing, 100101, P. R. China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Juan Li
- China National Center for Bioinformation, Beijing, 100101, P. R. China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Ping Wang
- China National Center for Bioinformation, Beijing, 100101, P. R. China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Jie Qi
- China National Center for Bioinformation, Beijing, 100101, P. R. China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Yuan Liang
- China National Center for Bioinformation, Beijing, 100101, P. R. China
| | - Shiming He
- Department of Urology, Peking University First Hospital, Beijing, 100034, P. R. China
- Institute of Urology, Peking University, Beijing, 100034, P. R. China
- National Urological Cancer Center, Beijing, 100034, P. R. China
| | - Yanqing Gong
- Department of Urology, Peking University First Hospital, Beijing, 100034, P. R. China
- Institute of Urology, Peking University, Beijing, 100034, P. R. China
- National Urological Cancer Center, Beijing, 100034, P. R. China
| | - Ninghan Feng
- Department of Urology, Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, 214002, P. R. China
| | - Yang Wang
- Department of Urology, Affiliated Wuxi No. 2 Hospital of Nanjing Medical University, Wuxi, 214002, P. R. China
| | - Yuanyuan Ma
- Animal Center, Peking University First Hospital, Beijing, 100034, P. R. China
| | - Mei Zhang
- China National Center for Bioinformation, Beijing, 100101, P. R. China
| | - Yue Shi
- China National Center for Bioinformation, Beijing, 100101, P. R. China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Xuesong Li
- Department of Urology, Peking University First Hospital, Beijing, 100034, P. R. China
- Institute of Urology, Peking University, Beijing, 100034, P. R. China
- National Urological Cancer Center, Beijing, 100034, P. R. China
| | - Weimin Ci
- China National Center for Bioinformation, Beijing, 100101, P. R. China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- Department of Urology, Chinese PLA General Hospital, Beijing, 100039, P. R. China
| | - Liqun Zhou
- Department of Urology, Peking University First Hospital, Beijing, 100034, P. R. China
- Institute of Urology, Peking University, Beijing, 100034, P. R. China
- National Urological Cancer Center, Beijing, 100034, P. R. China
- Department of Urology, The First Affiliated Hospital of Henan University, Kaifeng, 475001, P. R. China
| |
Collapse
|
13
|
Ding Y, Chen Y, Zhang J, Wang Q, Zhu S, Jiang J, He C, Wang J, Tou L, Zheng J, Chen B, Hu S, Yu X, Wang H, Lu Y, Kong M, Chen Y, Wang H, Zhang H, Xu H, Teng F, Shen X, Xu N, Ruan J, Zhou Z, Lu J, Teng L. Blood Biomarker-Based Predictive Indicator for Liver Metastasis in Alpha-Fetoprotein-Producing Gastric Cancer and Multi-Omics Tumor Microenvironment Insights. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e03499. [PMID: 40433893 DOI: 10.1002/advs.202503499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/22/2025] [Indexed: 05/29/2025]
Abstract
Alpha-fetoprotein-producing gastric cancer (AFPGC) is a rare but highly aggressive subtype of gastric cancer. Patients with AFPGC are at high risk of liver metastasis, and the tumor microenvironment (TME) is complex. A multicenter retrospective study is conducted from January 2011 to December 2021 and included 317 AFPGC patients. Using a multivariable logistic regression model, a nomogram for predicting liver metastasis is built. By combining AFP and the neutrophil-lymphocyte ratio (NLR), we developed a novel and easily applicable predictive indicator, termed ANLiM score, for liver metastasis in AFPGC. An integrated multi-omics analysis, including whole-exome sequencing and proteomic analysis, is conducted and revealed an immunosuppressive TME in AFPGC with liver metastasis. Single-cell RNA sequencing and multiplex immunofluorescence identified the potential roles of tumor-associated neutrophils and tertiary lymphoid structures in shaping the immune microenvironment. These findings are validated in a real-world cohort receiving anti-programmed cell death 1 (anti-PD-1) therapy, which showed concordant effectiveness. In addition, the ANLiM score is also identified as a promising biomarker for predicting immunotherapy efficacy. Overall, a blood biomarker-based predictive indicator is developed for liver metastasis and immunotherapy response in AFPGC. The findings on immune microenvironmental alterations for AFPGC with liver metastasis provide new insights for optimizing immunotherapy strategies.
Collapse
Affiliation(s)
- Yongfeng Ding
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Yiran Chen
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Jing Zhang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Qingrui Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems & Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310000, China
| | - Songting Zhu
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Junjie Jiang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, 310000, China
| | - Chao He
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Jincheng Wang
- Department of Radiology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Laizhen Tou
- Department of Gastrointestinal Surgery, Lishui Central Hospital, the Fifth Hospital Affiliated to Wenzhou Medical University, Lishui, 323000, China
| | - Jingwei Zheng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Bicheng Chen
- Department of General Surgery, Jinyun People's Hospital, Lishui, 323000, China
| | - Sizhe Hu
- Department of Gastrointestinal Surgery, Dongyang Hospital Affiliated to Wenzhou Medical University, Dongyang People's Hospital, Jinhua, 321000, China
| | - Xiongfei Yu
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Haohao Wang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Yimin Lu
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Mei Kong
- Department of Pathology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Yanyan Chen
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Haiyong Wang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Haibin Zhang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Hongxia Xu
- Innovation Institute for Artificial Intelligence in Medicine and Liangzhu Laboratory, School of medicine, Zhejiang University, Hangzhou, 310000, China
| | - Fei Teng
- Department of Cell Biology, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Xian Shen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Nong Xu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Jian Ruan
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Zhan Zhou
- State Key Laboratory of Advanced Drug Delivery and Release Systems & Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310000, China
| | - Jun Lu
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Lisong Teng
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| |
Collapse
|
14
|
Zhu X, Zhang L, Yu X, Yan P, Zhang X, Zhao Y, Wang D, Yang XA. Elucidating the tumor microenvironment interactions in breast, cervical, and ovarian cancer through single-cell RNA sequencing. Sci Rep 2025; 15:17846. [PMID: 40404741 PMCID: PMC12098903 DOI: 10.1038/s41598-025-03017-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 05/19/2025] [Indexed: 05/24/2025] Open
Abstract
This study aimed to identify the key cell types and their interactions in gynecological oncology of breast cancer, cervical cancer, and ovarian cancer. Single-cell RNA sequencing was performed on tumor samples of gynecological oncology from the GEO database. Cell types were identified using SingleR and cell composition was analyzed to understand the tumor microenvironment (TME). CellChat was used to analyze cell interactions, and pseudotemporal analysis was conducted on cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs) to understand their differentiation status. Four CAF subtypes were identified: iCAF, myCAF, proCAF, and matCAF. The iCAF subpopulation secreted COL1A1 and promoted tumor cell migration, while myCAF was involved in angiogenesis. The matCAF subpopulation was present throughout tumor development. TAMs were found to promote angiogenesis through the VEGFA_VEGFR2 signaling pathway. CAFs and TAMs play pivotal roles in tumor progression through their interactions and signaling pathways.
Collapse
Affiliation(s)
- Xiaoyue Zhu
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
- Graduate School of Chengde Medical University, Chengde, 067000, China
| | - Liang Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Xiaomin Yu
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
| | - Pengxian Yan
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
| | - Xiaoyu Zhang
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
- Graduate School of Chengde Medical University, Chengde, 067000, China
| | - Yunlong Zhao
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
- Graduate School of Chengde Medical University, Chengde, 067000, China
| | - Dongze Wang
- Clinical and Basic Medical College, Shandong First Medical University, Jinan, 250000, China
| | - Xiu-An Yang
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China.
- Hebei Key Laboratory of Nerve Injury and Repair, Chengde Medical University, Chengde, 067000, China.
| |
Collapse
|
15
|
Bandyopadhyay A, Sinha S, Roy R, Biswas N. Autophagy mediated immune response regulation and drug resistance in cancer. Mol Biol Rep 2025; 52:492. [PMID: 40402380 DOI: 10.1007/s11033-025-10573-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 05/02/2025] [Indexed: 05/23/2025]
Abstract
Autophagy is a critical regulator of cellular homeostasis. The proteins involved in autophagy orchestrate the functions to strike the balance between cell survival and cell death in context-specific situations like aging, infections, inflammation and most importantly carcinogenesis. One of the major dead-locks in cancer treatment is the development of resistance to the available drugs (multi-drug resistance) as well as immune-suppressions in patients. Different studies over time have shown that autophagy is being involved in chemotherapy by working hand in hand with apoptosis or drug resistance through proliferative signals. Resistance to various drugs, such as, Cisplatin, Vincristine, Tamoxifen (TAM) occurs by epigenetic modifications, changed expression levels of microRNAs (miRNAs/miRs), and long non-coding RNAs (lncRNAs), which are regulated by the aberrant autophagy levels in lung, and breast cancers. More interestingly in the tumour microenvironment the immune suppressor cells also bring in autophagy in different pathway regulations either helping or opposing the whole carcinogenesis process. Macrophages, T cells, B cells, dendritic cells (DCs), neutrophils, and fibroblasts show involvement of autophagy in their differentiation and development in the tumor microenvironment (TME). Here, this extensive review for the first time tries to bring under a single canopy, several recent examples of autophagy-mediated immune regulations and autophagy-mediated epigenetically regulated drug resistance in different types of cancers.
Collapse
Affiliation(s)
- Anupriya Bandyopadhyay
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Samraj Sinha
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Rajdeep Roy
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Nabendu Biswas
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India.
| |
Collapse
|
16
|
Zhang D, Wang X, Li W, Wan D, Zhou Y, Ma C, Yang Z, Zhang Y, Li W, Li Z, Lin H, Jin Z, Wu W, Huang H. A Single-Cell Atlas-Inspired Hitchhiking Therapeutic Strategy for Acute Pancreatitis by Restricting ROS in Neutrophils. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2502200. [PMID: 40395143 DOI: 10.1002/adma.202502200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Neutrophils can undergo transcriptional and epigenetic reprogramming in disease, thus causing inflammation or modulating tissue repair and fibrosis. A thorough understanding of the neutrophil subpopulation composition and their polarization processes in acute pancreatitis (AP) is essential to open up design of treatments tailored to individual patients. Herein, this study distinct subgroups and two differentiation pathways associated with N1 and N2 polarization during AP by single-cell sequencing. Inspired by this, a hollow manganese dioxide (HMnO2)-based nanoreactor (Pyp@APHM) conjugated with neutrophil-binding Ly-6G antibody and loaded with porphyrin is developed for targeted and in situ modulation of neutrophil polarization. Pyp@APHM can enrich the AP site by hitchhiking on neutrophils and then degrade in response to a weakly acidic environment to simultaneously release manganese ions and porphyrin ligands, enabling in situ synthesis of manganese porphyrin antioxidants. Leveraging this strategy, Pyp@APHM can effectively eliminate reactive oxygen species (ROS) and broadly inhibit both N1 and N2 polarization, as well as enhance tissue oxygenation by generating O2, thereby further mitigating pancreatic inflammation. This study provides a comprehensive single-cell atlas of neutrophils in AP and proposes an innovative hitchhiking therapeutic strategy for AP by restricting ROS in neutrophils.
Collapse
Affiliation(s)
- Deyu Zhang
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Xinyue Wang
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Wanshun Li
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Dongling Wan
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Yuyan Zhou
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Congjia Ma
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Zhenghui Yang
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Yang Zhang
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Wenhao Li
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Zhaoshen Li
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Han Lin
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Zhendong Jin
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Wencheng Wu
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Haojie Huang
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
17
|
Blümke J, Schameitat M, Verma A, Limbecker C, Arlt E, Kessler SM, Kielstein H, Krug S, Bazwinsky-Wutschke I, Haemmerle M. Innate Immunity and Platelets: Unveiling Their Role in Chronic Pancreatitis and Pancreatic Cancer. Cancers (Basel) 2025; 17:1689. [PMID: 40427186 PMCID: PMC12110028 DOI: 10.3390/cancers17101689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 05/10/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal forms of cancer, characterized by a highly desmoplastic tumor microenvironment. One main risk factor is chronic pancreatitis (CP). Progression of CP to PDAC is greatly influenced by persistent inflammation promoting genomic instability, acinar-ductal metaplasia, and pancreatic intraepithelial neoplasia (PanIN) formation. Components of the extracellular matrix, including immune cells, can modulate this progression phase. This includes cells of the innate immune system, such as natural killer (NK) cells, macrophages, dendritic cells, mast cells, neutrophils, and myeloid-derived suppressor cells (MDSCs), either promoting or inhibiting tumor growth. On one hand, innate immune cells can trigger inflammatory responses that support tumor progression by releasing cytokines and growth factors, fostering tumor cell proliferation, invasion, and metastasis. On the other hand, they can also activate immune surveillance mechanisms, which can limit tumor development. For example, NK cells are cytotoxic innate lymphoid cells that are able to kill tumor cells, and active dendritic cells are crucial for a functioning anti-tumor immune response. In contrast, mast cells and MDSCs rather support a pro-tumorigenic tumor microenvironment that is additionally sustained by platelets. Once thought to play a role in hemostasis only, platelets are now recognized as key players in inflammation and cancer progression. By releasing cytokines, growth factors, and pro-angiogenic mediators, platelets help shape an immunosuppressive microenvironment that promotes fibrotic remodeling, tumor initiation, progression, metastasis, and immune evasion. Neutrophils and macrophages exist in different functional subtypes that can both act pro- and anti-tumorigenic. Understanding the complex interactions between innate immune cells, platelets, and early precursor lesions, as well as PDAC cells, is crucial for developing new therapeutic approaches that can harness the immune and potentially also the coagulation system to target and eliminate tumors, offering hope for improved patient outcomes.
Collapse
Affiliation(s)
- Juliane Blümke
- Institute of Pathology, Section of Experimental Pathology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany;
| | - Moritz Schameitat
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Atul Verma
- Department of Internal Medicine I, Medical Faculty, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (A.V.); (S.K.)
| | - Celina Limbecker
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Elise Arlt
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Sonja M. Kessler
- Institute of Pharmacy, Experimental Pharmacology for Natural Sciences, Faculty of Natural Sciences, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany;
| | - Heike Kielstein
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Sebastian Krug
- Department of Internal Medicine I, Medical Faculty, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (A.V.); (S.K.)
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Ivonne Bazwinsky-Wutschke
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Monika Haemmerle
- Institute of Pathology, Section of Experimental Pathology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany;
| |
Collapse
|
18
|
Gill GS, Kharb S, Goyal G, Das P, Kurdia KC, Dhar R, Karmakar S. Immune Checkpoint Inhibitors and Immunosuppressive Tumor Microenvironment: Current Challenges and Strategies to Overcome Resistance. Immunopharmacol Immunotoxicol 2025:1-45. [PMID: 40376861 DOI: 10.1080/08923973.2025.2504906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 05/06/2025] [Indexed: 05/18/2025]
Abstract
Immune checkpoint inhibitors (ICIs) are shown to improve cancer treatment effectiveness by boosting the immune system of the patient. Nevertheless, the unique and highly suppressive TME poses a significant challenge, causing heterogeneity of response or resistance in a considerable number of patients. This review focuses on the evasive attributes of the TME. Immune evasion mechanism in TME include immunosuppressive cells, cytokine and chemokine signaling, metabolic alterations and overexpression of immune checkpoint molecules such as PD-1, CTLA-4, LAG-3, TIM-3, TIGIT, BTLA and their interactions within the TME. In addition, this review focuses on the overcoming resistance by targeting immunosuppressive cells, normalizing tumor blood vessels, blocking two or three checkpoints simultaneously, combining vaccines, oncolytic viruses and metabolic inhibitors with ICIs or other therapies. This review also focuses on the necessity of finding predictive markers for the stratification of patients and to check response of ICIs treatment. It remains to be made certain by new research and intelligent innovations how these discoveries of the TME and its interplay facilitate ICI treatment and change the face of cancer treatment.
Collapse
Affiliation(s)
- Gurpreet Singh Gill
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Simmi Kharb
- Department of Biochemistry, Pt. B.D. Sharma Postgraduate Institute of Medical Sciences, Rohtak, India
| | - Gitanjali Goyal
- Department of Biochemistry, All India Institute of Medical Sciences, Bathinda, India
| | - Prasenjit Das
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Kailash Chand Kurdia
- Department of GI Surgery & Liver Transplantation, All India Institute of Medical Sciences, New Delhi, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
19
|
Kwak JW, Houghton AM. Targeting neutrophils for cancer therapy. Nat Rev Drug Discov 2025:10.1038/s41573-025-01210-8. [PMID: 40374764 DOI: 10.1038/s41573-025-01210-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2025] [Indexed: 05/18/2025]
Abstract
Neutrophils are among the most abundant immune cell types in the tumour microenvironment and have been associated with poor outcomes across multiple cancer types. Yet despite mounting evidence of their role in tumour progression, therapeutic strategies targeting neutrophils have only recently gained attention and remain limited in scope. This is probably due to the increasing number of distinct neutrophil subtypes identified in cancer and the limited understanding of the mechanisms by which these subsets influence tumour progression and immune evasion. In this Review, we discuss the spectrum of neutrophil subtypes - including those with antitumour activity - and their potential to polarize towards tumour-suppressive phenotypes. We explore the molecular pathways and effector functions by which neutrophils modulate cancer progression, with an emphasis on identifying tractable therapeutic targets. Finally, we examine emerging clinical trials aimed at modulating neutrophil lineages and consider their implications for patient outcomes.
Collapse
Affiliation(s)
- Jeff W Kwak
- Translational Science and Therapeutics Division and Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - A McGarry Houghton
- Translational Science and Therapeutics Division and Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
20
|
Nowaczewska-Kuchta A, Ksiazek-Winiarek D, Glabinski A. Interaction Between Neutrophils and Elements of the Blood-Brain Barrier in the Context of Multiple Sclerosis and Ischemic Stroke. Int J Mol Sci 2025; 26:4437. [PMID: 40362673 PMCID: PMC12072651 DOI: 10.3390/ijms26094437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 05/03/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
The blood-brain barrier (BBB) is a semi-permeable membrane in physiological conditions, but in pathologies like multiple sclerosis (MS) and ischemic stroke (IS), its permeability increases. In this review, we focus on neutrophils and their interaction with cellular components of the BBB: endothelial cells (EC), pericytes (PC), and astrocytes (AC). Nowadays, neutrophils receive more attention, mostly due to advanced research techniques that show the complexity of their population. Additionally, neutrophils have the ability to secrete extracellular vesicles (EVs), reactive oxygen species (ROS) and cytokines, which both destroy and restore the BBB. Astrocytes, PCs, and ECs also have dual roles in the pathogenesis of MS and IS. The interaction between neutrophils and cellular components of the BBB provides us with a wider insight into the pathogenesis of common diseases in the central nervous system. Further, we comprehensively review knowledge about the influence of neutrophils on the BBB in the context of MS and IS. Moreover, we describe new therapeutic strategies for patients with MS and IS like cell-based therapies and therapies that use the neutrophil function.
Collapse
Affiliation(s)
| | | | - Andrzej Glabinski
- Department of Neurology and Stroke, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (A.N.-K.); (D.K.-W.)
| |
Collapse
|
21
|
Garner H, Martinovic M, Liu NQ, Bakker NAM, Velilla IQ, Hau CS, Vrijland K, Kaldenbach D, Kok M, de Wit E, de Visser KE. Understanding and reversing mammary tumor-driven reprogramming of myelopoiesis to reduce metastatic spread. Cancer Cell 2025:S1535-6108(25)00166-7. [PMID: 40345190 DOI: 10.1016/j.ccell.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 10/11/2024] [Accepted: 04/15/2025] [Indexed: 05/11/2025]
Abstract
Tumor-induced systemic accumulation and polarization of neutrophils to an immunosuppressive phenotype is a potent driver of metastasis formation. Yet, how mammary tumors reprogram granulopoiesis at the molecular level and when tumor imprinting occurs during neutrophil development remains underexplored. Here, we combined single-cell, chromatin and functional analyses to unravel the tumor-driven reprogramming of granulopoiesis in the bone marrow, along with intervention studies aimed at reversing this process. We observe that mammary tumors accelerate commitment to the neutrophil lineage at the expense of lymphopoiesis and erythropoiesis without stimulating the development of a novel myeloid lineage. Moreover, tumor-directed immunosuppressive imprinting of neutrophils starts early in hematopoiesis. Treatment with anti-IL-1β normalizes tumor-induced granulopoiesis, reducing neutrophil immunosuppressive phenotype and mitigating metastatic spread. Together, these data provide molecular insights into the aberrant, tumor-driven neutrophil differentiation pathway leading to metastasis-promoting chronic inflammation and how it can be reversed to reduce metastatic spread.
Collapse
Affiliation(s)
- Hannah Garner
- Department of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands.
| | - Moreno Martinovic
- Division of Gene Regulation, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ning Qing Liu
- Division of Gene Regulation, Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Noor A M Bakker
- Department of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands; Department of Immunology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Irene Querol Velilla
- Department of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands; Department of Immunology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Cheei-Sing Hau
- Department of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Kim Vrijland
- Department of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Daphne Kaldenbach
- Department of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Marleen Kok
- Department of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Elzo de Wit
- Division of Gene Regulation, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Karin E de Visser
- Department of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands; Department of Immunology, Leiden University Medical Centre, Leiden, the Netherlands.
| |
Collapse
|
22
|
Chu X, Pu N, Yang X, Xie Y, Liu L, Jin Y. Subtypes of tumor-associated neutrophils and their roles in cancer immunotherapy. Crit Rev Oncol Hematol 2025; 212:104763. [PMID: 40334802 DOI: 10.1016/j.critrevonc.2025.104763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/09/2025] Open
Abstract
Neutrophils are essential components of the innate immune system. Tumor-associated neutrophils (TANs) are shaped by tumor microenvironment (TME), leading to significant heterogeneity in biological characteristics and functions. Recent advances in single-cell sequencing have revealed a wide array of TAN subtypes, while a comprehensive classification system is still lacking. This review aims to summarize the alterations observed in TAN subgroups following cancer immunotherapy, and identify the distinctions and commonalities between pro-tumor and anti-tumor subgroups. Current progress of preclinical and clinical studies is also highlighted, involving novel therapies targeting TANs.
Collapse
Affiliation(s)
- Xinyun Chu
- Department of Hepatobiliary & Pancreatic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650000, China; Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ning Pu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xue Yang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuqi Xie
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Yun Jin
- Department of Hepatobiliary & Pancreatic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650000, China.
| |
Collapse
|
23
|
León-Vega II, Oregon R, Schnoor M, Vadillo E. From Ulcerative Colitis to Metastatic Colorectal Cancer: The Neutrophil Contribution. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:814-830. [PMID: 39889826 DOI: 10.1016/j.ajpath.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 02/03/2025]
Abstract
Ulcerative colitis (UC) is an inflammatory colon and rectum disease affecting approximately 5 million people worldwide. There is no cure for UC, and approximately 8% of patients with UC develop colorectal cancer (CRC) by gradual acquisition of mutations driving the formation of adenomas and their progression to adenocarcinomas and metastatic disease. CRC constitutes 10% of total cancer cases worldwide and 9% of cancer deaths. Both UC and CRC have an increasing incidence worldwide. Although the epithelium has been well studied in UC and CRC, the contribution of neutrophils is less clear. Neutrophils are rapidly recruited in excessive amounts from peripheral blood to the colon during UC, and their overactivation in the proinflammatory UC tissue environment contributes to tissue damage. In CRC, the role of neutrophils is controversial, but emerging evidence suggests that their role depends on the evolution and context of the disease. The role of neutrophils in the transition from UC to CRC is even less clear. However, recent studies propose neutrophils as therapeutic targets for better clinical management of both diseases. This review summarizes the current knowledge on the roles of neutrophils in UC and CRC.
Collapse
Affiliation(s)
- Iliana I León-Vega
- Department of Molecular Biomedicine, Cinvestav-National Polytechnic Institute, Mexico City, Mexico
| | - Reyna Oregon
- Oncology Research Unit, Oncology Hospital, National Medical Center, Mexican Institute of Social Security, Mexico City, Mexico
| | - Michael Schnoor
- Department of Molecular Biomedicine, Cinvestav-National Polytechnic Institute, Mexico City, Mexico.
| | - Eduardo Vadillo
- Oncology Research Unit, Oncology Hospital, National Medical Center, Mexican Institute of Social Security, Mexico City, Mexico.
| |
Collapse
|
24
|
Li H, Zeng J, You Q, Zhang M, Shi Y, Yang X, Gu W, Liu Y, Hu N, Wang Y, Chen X, Mu J. X-ray-activated nanoscintillators integrated with tumor-associated neutrophils polarization for improved radiotherapy in metastatic colorectal cancer. Biomaterials 2025; 316:123031. [PMID: 39709848 DOI: 10.1016/j.biomaterials.2024.123031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/26/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024]
Abstract
Radiotherapy, employing high-energy rays to precisely target and eradicate tumor cells, plays a pivotal role in the treatment of various malignancies. Despite its therapeutic potential, the effectiveness of radiotherapy is hindered by the tumor's inherent low radiosensitivity and the immunosuppressive microenvironment. Here we present an innovative approach that integrates peroxynitrite (ONOO-)-mediated radiosensitization with the tumor-associated neutrophils (TANs) polarization for the reversal of immunosuppressive tumor microenvironment (TME), greatly amplifying the potency of radiotherapy. Our design employs X-ray-activated lanthanide-doped scintillators (LNS) in tandem with photosensitive NO precursor to achieve in-situ ONOO- generation. Concurrently, the co-loaded TGF-β inhibitor SB525334, released from the LNS-RS nanoplatform in response to the overexpressed GSH in tumor site, promotes the reprogramming of TANs from N2 phenotype toward N1 phenotype, effectively transforming the tumor-promoting microenvironment into a tumor-inhibiting state. This 'one-two punch' therapy efficiently trigger a robust anti-tumor immune response and exert potent therapeutic effects in orthotopic colorectal cancer and melanoma mouse model. Meanwhile, it also significantly prevents liver metastasis and recurrence in metastatic colorectal cancer. The development of X-ray-controlled platforms capable of activating multiple therapeutic modalities may accelerate the clinical application of radiotherapy-based collaborative therapy.
Collapse
Affiliation(s)
- Hui Li
- Institute of Precision Medicine, Peking University Shenzhen Hospital, 518036, Shenzhen, China; Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Junyi Zeng
- Institute of Precision Medicine, Peking University Shenzhen Hospital, 518036, Shenzhen, China; Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Qing You
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
| | - Miaomiao Zhang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, 518036, Shenzhen, China
| | - Yuanchao Shi
- Institute of Precision Medicine, Peking University Shenzhen Hospital, 518036, Shenzhen, China
| | - Xiaodong Yang
- Department of Radiation Oncology, Peking University Shenzhen Hospital, 518036, Shenzhen, China
| | - Wenxing Gu
- Institute of Precision Medicine, Peking University Shenzhen Hospital, 518036, Shenzhen, China; Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Yajie Liu
- Department of Radiation Oncology, Peking University Shenzhen Hospital, 518036, Shenzhen, China
| | - Ning Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Yu Wang
- Institute of Microscale Optoelectronics, Shenzhen University, 518060, Shenzhen, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
| | - Jing Mu
- Institute of Precision Medicine, Peking University Shenzhen Hospital, 518036, Shenzhen, China.
| |
Collapse
|
25
|
Zhong H, Zhou S, Yin S, Qiu Y, Liu B, Yu H. Tumor microenvironment as niche constructed by cancer stem cells: Breaking the ecosystem to combat cancer. J Adv Res 2025; 71:279-296. [PMID: 38866179 DOI: 10.1016/j.jare.2024.06.014] [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: 04/08/2024] [Revised: 05/27/2024] [Accepted: 06/09/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are a distinct subpopulation of cancer cells with the capacity to constantly self-renew and differentiate, and they are the main driver in the progression of cancer resistance and relapse. The tumor microenvironment (TME) constructed by CSCs is the "soil" adapted to tumor growth, helping CSCs evade immune killing, enhance their chemical resistance, and promote cancer progression. AIM OF REVIEW We aim to elaborate the tight connection between CSCs and immunosuppressive components of the TME. We attempt to summarize and provide a therapeutic strategy to eradicate CSCs based on the destruction of the tumor ecological niche. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three main key concepts. First, we highlight that CSCs recruit and transform normal cells to construct the TME, which further provides ecological niche support for CSCs. Second, we describe the main characteristics of the immunosuppressive components of the TME, targeting strategies and summarize the progress of corresponding drugs in clinical trials. Third, we explore the multilevel insights of the TME to serve as an ecological niche for CSCs.
Collapse
Affiliation(s)
- Hao Zhong
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Shiyue Zhou
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Shuangshuang Yin
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin, China.
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.
| | - Haiyang Yu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China.
| |
Collapse
|
26
|
Szlachetko JA, Hofmann-Vega F, Budeus B, Schröder LJ, Dumitru CA, Schmidt M, Deuss E, Vollmer S, Hanschmann EM, Busch M, Kehrmann J, Lang S, Dünker N, Hussain T, Brandau S. Tumor cells that resist neutrophil anticancer cytotoxicity acquire a prometastatic and innate immune escape phenotype. Cell Mol Immunol 2025; 22:527-540. [PMID: 40155451 PMCID: PMC12041228 DOI: 10.1038/s41423-025-01283-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: 09/23/2024] [Accepted: 03/16/2025] [Indexed: 04/01/2025] Open
Abstract
In the tumor host, neutrophils may exhibit protumor or antitumor activity. It is hypothesized that in response to host-derived or therapy-induced factors, neutrophils adopt diverse functional states to ultimately execute these differential functions. Here, we provide an alternative scenario in which the response of an individual tumor cell population determines the overall protumor versus antitumor outcome of neutrophil‒tumor interactions. Experimentally, we show that human neutrophils, which are sequentially stimulated with bacteria and secreted factors from tumor cells, kill a certain proportion of tumor target cells. However, the majority of the tumor cells remained resistant to this neutrophil-mediated killing and underwent a functional, phenotypic and transcriptomic switch that was reminiscent of partial epithelial‒to-mesenchymal transition. This cell biological switch was associated with physical escape from NK-mediated killing and resulted in enhanced metastasis to the lymph nodes in a preclinical orthotopic mouse model. Mechanistically, we identified the antimicrobial neutrophil granule proteins neutrophil elastase (NE) and matrix metalloprotease-9 (MMP-9) as the molecular mediators of this functional switch. We validated these data in patients with head and neck cancer and identified bacterially colonized intratumoral niches that were enriched for mesenchymal tumor cells and neutrophils expressing NE and MMP-9. Our data reveal the parallel execution of tumor cytotoxic and prometastatic activity by activated neutrophils and identify NE and MMP-9 as mediators of lymph node metastasis. The identified mechanism explains the functional dichotomy of tumor-associated neutrophils at the level of the tumor target cell response and has implications for superinfected cancers and the dysbiotic tumor microenvironment.
Collapse
Affiliation(s)
| | | | - Bettina Budeus
- Institute of Cell Biology, University Hospital Essen, Essen, 45147, Germany
| | - Lara-Jasmin Schröder
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany
- Institute of Pathology, Medical School Hannover, Hannover, 30625, Germany
| | - Claudia Alexandra Dumitru
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany
- Department of Neurosurgery, Otto-von-Guericke University, Magdeburg, 39106, Germany
| | - Mathias Schmidt
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany
| | - Eric Deuss
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany
| | - Sebastian Vollmer
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany
| | - Eva-Maria Hanschmann
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany
| | - Maike Busch
- Institute of Anatomy II, Department of Neuroanatomy, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University of Duisburg-Essen, Medical Faculty, Essen, 45147, Germany
| | - Jan Kehrmann
- Institute of Medical Microbiology, University Hospital Essen, Essen, 45147, Germany
| | - Stephan Lang
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany
| | - Nicole Dünker
- Institute of Anatomy II, Department of Neuroanatomy, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University of Duisburg-Essen, Medical Faculty, Essen, 45147, Germany
| | - Timon Hussain
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany
- Department of Otorhinolaryngology, Klinikum rechts der Isar, Technical University Munich, Munich, 81675, Germany
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany.
- German Cancer Consortium, DKTK, Partner Site Essen-Düsseldorf, Essen, 45147, Germany.
| |
Collapse
|
27
|
Rys RN, Calcinotto A. Senescent neutrophils: a hidden role in cancer progression. Trends Cell Biol 2025; 35:399-411. [PMID: 39362804 DOI: 10.1016/j.tcb.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/05/2024]
Abstract
Neutrophils have recently received increased attention in cancer because they contribute to all stages of cancer. Neutrophils are so far considered to have a short half-life. However, a growing body of literature has shown that tumor-associated neutrophils (TANs) acquire a prolonged lifespan. This review discusses recent work surrounding the mechanisms by which neutrophils can persist in the tumor microenvironment (TME). It also highlights different scenarios for therapeutic targeting of protumorigenic neutrophils, supporting the idea that, in tumors, inhibition of neutrophil recruitment is not sufficient because these cells can persist and remain hidden from current interventions. Hence, the elimination of long-lived neutrophils should be pursued to increase the efficacy of standard therapy.
Collapse
Affiliation(s)
- Ryan N Rys
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Arianna Calcinotto
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland.
| |
Collapse
|
28
|
Chaves MM. Neutrophils and purinergic signaling: Partners in the crime against Leishmania parasites? Biochimie 2025; 232:43-53. [PMID: 39855456 DOI: 10.1016/j.biochi.2025.01.004] [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: 06/30/2024] [Revised: 12/18/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
The parasite of the genus Leishmania is the causative agent of diseases that affect humans called leishmaniasis. These diseases affect millions of people worldwide and the currently existing drugs are either very toxic or the parasites acquire resistance. Therefore, new elimination mechanisms need to be elucidated so that new therapeutic strategies can be developed. Much has already been discussed about the role of neutrophils in Leishmania infection, and their participation is still controversial. A recent study showed that receptors present in the neutrophil membrane, the purinergic receptors, can control the infection when activated, but the triggering mechanism has not been elucidated. In this review, we will address the possible participation of purinergic receptors expressed in the neutrophil extracellular membrane that may be participating in the detection of Leishmania infection and their possible effects during parasitism.
Collapse
Affiliation(s)
- Mariana M Chaves
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Bio-Manguinhos, Oswaldo Cruz Foundation, Brazilian Ministry of Health, Rio de Janeiro, Brazil.
| |
Collapse
|
29
|
Yang L, Shi F, Cao F, Wang L, She J, He B, Xu X, Kong L, Cai B. Neutrophils in Tissue Injury and Repair: Molecular Mechanisms and Therapeutic Targets. MedComm (Beijing) 2025; 6:e70184. [PMID: 40260014 PMCID: PMC12010766 DOI: 10.1002/mco2.70184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/23/2025] Open
Abstract
Tissue repair represents a highly intricate and ordered dynamic process, critically reliant on the orchestration of immune cells. Among these, neutrophils, the most abundant leukocytes in the body, emerge as the initial immune responders at injury sites. Traditionally recognized for their antimicrobial functions in innate immunity, neutrophils now garner attention for their indispensable roles in tissue repair. This review delves into their novel functions during the early stages of tissue injury. We elucidate the mechanisms underlying neutrophil recruitment and activation following tissue damage and explore their contributions to vascular network formation. Furthermore, we investigate the pivotal role of neutrophils during the initial phase of repair across different tissue types. Of particular interest is the investigation into how the fate of neutrophils influences overall tissue healing outcomes. By shedding light on these emerging aspects of neutrophil function in tissue repair, this review aims to pave the way for novel strategies and approaches in future organ defect repair, regeneration studies, and advancements in tissue engineering. The insights provided here have the potential to significantly impact the field of tissue repair and regeneration.
Collapse
Affiliation(s)
- Luying Yang
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Fan Shi
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Feng Cao
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Le Wang
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Jianzhen She
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Boling He
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Xiaoying Xu
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Liang Kong
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Bolei Cai
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
30
|
Xiong B, Zhang Y, Liu S, Liao S, Zhou Z, He Q, Zhou Y. NOX Family: Regulators of Reactive Oxygen Species Balance in Tumor Cells. FASEB J 2025; 39:e70565. [PMID: 40266050 PMCID: PMC12017260 DOI: 10.1096/fj.202500238rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
Cancer cells are capable of surviving, proliferating, and invading or migrating within hypoxic environments by regulating various adaptive mechanisms. Due to the activation of oncogenes and the inactivation of tumor suppressor genes, and relative deficiencies in oxygen and nutrients, cancer cells demonstrate elevated production of reactive oxygen species (ROS), primarily sourced from NADPH oxidases (NOX family). A key aspect of the reorientation of tumor cell metabolism is the combating of cellular oxidative stress through the promotion of antioxidant molecule synthesis to counteract ROS production. Given that most cancers experience hypoxia and that NOX is closely linked to numerous redox-dependent signaling pathways, the expression and function of NOX are altered in various malignancies. Therefore, this review summarizes the characteristics of NOX family members, their influence on tumor proliferation, invasion, and migration, the role of NOX in promoting tumor angiogenesis, the impact of NOX on the function of immune cells within the tumor microenvironment, and the potential of targeting NOX in tumor therapy. This aims to offer a fresh viewpoint on a comprehensive understanding of the functions of NOX family members.
Collapse
Affiliation(s)
- Bin Xiong
- Department of Radiation OncologyThe Affiliated Cancer Hospital of Xiangya School of Medicine Central South University/Hunan Cancer HospitalChangshaHunanChina
- Cancer Research Institute, Basic School of MedicineCentral South UniversityChangshaHunanChina
| | - Yang Zhang
- Cancer Research Institute, Basic School of MedicineCentral South UniversityChangshaHunanChina
| | - Siyi Liu
- Cancer Research Institute, Basic School of MedicineCentral South UniversityChangshaHunanChina
| | - Shan Liao
- Department of PathologyThe Third Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Zihua Zhou
- Department of OncologyLoudi Central HospitalLoudiHunanChina
| | - Qian He
- Department of Radiation OncologyThe Affiliated Cancer Hospital of Xiangya School of Medicine Central South University/Hunan Cancer HospitalChangshaHunanChina
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of MedicineCentral South UniversityChangshaHunanChina
| |
Collapse
|
31
|
Cai W, Fan T, Xiao C, Deng Z, Liu Y, Li C, He J. Neutrophils in cancer: At the crucial crossroads of anti-tumor and pro-tumor. Cancer Commun (Lond) 2025. [PMID: 40296668 DOI: 10.1002/cac2.70027] [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: 09/19/2024] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025] Open
Abstract
Neutrophils are important components of the immune system and play a key role in defending against pathogenic infections and responding to inflammatory cues, including cancer. Their dysregulation indicates potential disease risk factors. However, their functional importance in disease progression has often been underestimated due to their short half-life, especially as there is limited information on the role of intratumoral neutrophils. Recent studies on their prominent role in cancer have led to a paradigm shift in our understanding of the functional diversity of neutrophils. These studies highlight that neutrophils have emerged as key components of the tumor microenvironment, where they can play a dual role in promoting and suppressing cancer. Moreover, several approaches to therapeutically target neutrophils have emerged, and clinical trials are investigating their efficacy. In this review, we discussed the involvement of neutrophils in cancer initiation and progression. We summarized recent advances in therapeutic strategies targeting neutrophils and, most importantly, suggested future research directions that could facilitate the manipulation of neutrophils for therapeutic purposes in cancer patients.
Collapse
Affiliation(s)
- Wenpeng Cai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Yixiao Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| |
Collapse
|
32
|
Gao Z, Wang X, Mei Q, Shen T, Wang J, Liu C. Sulfated chitosan directs the recovery of ischemic stroke by attenuating the inflammatory cascade. Theranostics 2025; 15:5870-5889. [PMID: 40365292 PMCID: PMC12068298 DOI: 10.7150/thno.111681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/03/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Ischemic stroke is considered a fatal ischemic disease with high mortality and morbidity. Acute ischemic stroke is a cascade of inflammatory reactions, which not only causes vascular degeneration but also leads to neurological disorders. During this period, the rapid response of neutrophil-dominated granulocytes releases cytokines and chemokines to affect tissue repair. Thus, effective regulation of neutrophils appears to be the key in treating major organ injuries associated with inflammation. Methods: This study developed a semisynthetic sulfated chitosan (SCS) associated with the functional sulfated groups. The immunoregulatory effects of SCS on neutrophils were tested by Real-Time Quantitative Reverse Transcription (RT-PCR), ELISA and immunofluorescence staining at gene and protein levels in vitro. Flow cytometry, WB and PCR were used to study the effect of neutrophils on macrophages, indicating the regulation of the inflammatory cascade by SCS. Acute ischemic stroke model was established to verify the effectiveness and the regulation of inflammatory cascade of SCS. Finally, the lower limb ischemia model was used to verify the universality of SCS in the treatment of ischemic diseases, especially with regard to acute inflammatory-related major organ damage. Results: SCS can not only promote neutrophil apoptosis, but also enable neutrophils to produce vascular-related subsets to regulate immunity and promote angiogenesis. Neutrophil stimulated by SCS mediated macrophage polarization via IL-10-induced Stat3 signaling pathway to weaken the inflammatory cascade. In animal models of ischemic hind limb and ischemic stroke, SCS had demonstrated its ability to shorten the acute inflammatory period, as indicated by neutrophil, and accelerate the subsequent repair period characterized by the presence of M2 macrophages. Additionally, SCS effectively inhibits the expression of MMP-9 to provide a favorable environment for rapid extracellular matrix reconstruction. Encouragingly, treatment with SCS had been shown to reduce the expansion of the infarct volume by approximately 20% in our experiments. Conclusion: This study underscores the effect of SCS in regulating the heterogeneity of neutrophils in order to suppress the initiation of inflammation to treat ischemic stroke. Crucially, our approach relies on non-exogenous growth factors and cells, highlighting its remarkable potential for clinical translatability in the treatment of major organ injuries.
Collapse
Affiliation(s)
- Zehua Gao
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| | - Xuanlin Wang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| | - Qiyong Mei
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Tong Shen
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| | - Jing Wang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
33
|
Bai M, Jin Y, Jin Z, Xie Y, Chen J, Zhong Q, Wang Z, Zhang Q, Cai Y, Qun F, Yuki N, Xin C, Shen X, Zhu J. Distinct immunophenotypic profiles and neutrophil heterogeneity in colorectal cancer. Cancer Lett 2025; 616:217570. [PMID: 39993650 DOI: 10.1016/j.canlet.2025.217570] [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: 01/07/2025] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/26/2025]
Abstract
Colorectal cancer (CRC) exhibits significant molecular and immunological heterogeneity. Neutrophil infiltration patterns play a crucial yet poorly understood role in tumor progression and patient outcomes. This study presents a comprehensive single-cell atlas of the CRC tumor microenvironment (TME), integrating transcriptomic data from 388,511 cells across 98 samples from 63 patients. Employing advanced computational methods, we stratified patients based on their immune cell infiltration profiles, revealing distinct immunophenotypes with potential therapeutic implications. Our analysis focused on tissue-resident neutrophils (TRNs) and uncovered previously uncharacterized subpopulations with diverse functional states. Trajectory inference analysis revealed a dynamic differentiation path from normal-associated neutrophils to tumor-associated neutrophils, highlighting the remarkable plasticity of these cells within the tumor environment. By integrating single-cell data with bulk transcriptomic and clinical information, we identified specific neutrophil-derived gene signatures associated with poor prognosis in CRC, suggesting their potential as novel prognostic biomarkers. This study not only provides unprecedented insights into neutrophil heterogeneity in CRC but also identifies potential targets for immunomodulatory therapies. Our findings lay the groundwork for developing more nuanced, personalized immunotherapeutic strategies for CRC, potentially improving treatment efficacy for patients who currently show a limited response to existing immunotherapies.
Collapse
Affiliation(s)
- Minghua Bai
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Yuzhao Jin
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China; Wenzhou Medical University, Wenzhou, China
| | - Zihao Jin
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, China
| | - Yuhao Xie
- College of Pharmacy and Health Sciences, St. John's University, Queens, New York, USA
| | - Jinggang Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, China
| | - Qingping Zhong
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China; Wenzhou Medical University, Wenzhou, China
| | | | - Qian Zhang
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yibo Cai
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - FangYa Qun
- National Institutes for Quantum Science and Technology(QST), Chiba, Japan
| | - Nitta Yuki
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Cheng Xin
- Department of Colorectal Surgery, Changhai Hospital, Naval Mdical University, Shanghai, China.
| | - Xiaohui Shen
- Department of General Surgery, Department of General Practice, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China.
| | - Ji Zhu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China; Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
34
|
Lee J, Jin BR, Cho J. Spatiotemporal regulation of neutrophil heterogeneity in health and disease. Hum Mol Genet 2025:ddaf008. [PMID: 40287830 DOI: 10.1093/hmg/ddaf008] [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: 09/14/2024] [Revised: 11/05/2024] [Accepted: 01/08/2025] [Indexed: 04/29/2025] Open
Abstract
Neutrophils are the most abundant leukocytes in humans and are indispensable for innate immunity. They are short-lived, terminally differentiated cells. However, mounting evidence indicates that neutrophils are heterogeneous in health and disease: they are young or aged in a steady state, while their heterogeneity becomes more diverse in disease conditions, such as cancer, sepsis, and thromboinflammation. Although the presence of distinct neutrophil subsets is well recognized, it is not fully understood how neutrophils have functional and phenotypic heterogeneity and what mechanisms control it. This review will focus on our current understanding of the molecular basis for neutrophil heterogeneity in pathophysiological conditions. In addition, we will discuss the possibility of targeting a specific subset of neutrophils to attenuate inflammation and tissue damage without compromising innate immune responses.
Collapse
Affiliation(s)
- Jingu Lee
- Division of Hematology, Department of Medicine, Washington University School of Medicine, 660 S Euclid Ave, St. Louis, MO 63110, United States
| | - Bo-Ram Jin
- Division of Hematology, Department of Medicine, Washington University School of Medicine, 660 S Euclid Ave, St. Louis, MO 63110, United States
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine, Washington University School of Medicine, 660 S Euclid Ave, St. Louis, MO 63110, United States
- Department of Pathology and Immunology, Washington University School of Medicine, 660 S Euclid Ave, St. Louis, MO 63110, United States
| |
Collapse
|
35
|
Yao Y, Yin Y, Shuai F, Lam W, Zhou T, Xie Y, He X, Han X. M2 Macrophage-Derived Extracellular Vesicles Reprogram Immature Neutrophils into Anxa1 hi Neutrophils to Enhance Inflamed Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2416159. [PMID: 40277454 DOI: 10.1002/advs.202416159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/22/2025] [Indexed: 04/26/2025]
Abstract
Periodontitis is a microbiome-related inflammation that can lead to irreversible bone reduction and even tooth loss. This study reveals that macrophage polarization states significantly influence periodontal homeostasis, with M2 macrophage-derived extracellular vesicles (M2-EVs) playing a pivotal role in mitigating periodontitis-induced bone loss. Single-cell RNA sequencing of periodontal tissues treated with M2-EVs uncovered a unique Anxa1hi neutrophil subpopulation exhibiting pro-reparative properties. This subpopulation is characterized by immaturity and demonstrated osteogenic and angiogenic capabilities in vivo, partially mediated through the secretion of oncostatin M (OSM) signals. The findings suggest that this functional heterogeneity arises from M2-EVs disrupting the neutrophil maturation trajectory, with pivotal reprogramming genes, such as Acvrl1 and Fpr2, driving the differentiation of the Anxa1hi reparative subpopulation. This work underscores the potential of targeting M2 macrophage-neutrophil interactions to promote the regeneration of inflamed bone tissues.
Collapse
Affiliation(s)
- Yufei Yao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yijia Yin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Fangyuan Shuai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Waishan Lam
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Tao Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yaxin Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xuesong He
- The ADA Forsyth Institute, 100 Chestnut Street, Somerville, MA, 02143, USA
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, 02115, USA
| | - Xianglong Han
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
36
|
Zhang X, Huang X, Zhang X, Lai L, Zhu B, Lin P, Kang Z, Yin D, Tian D, Chen Z, Gao J. The miR-941/FOXN4/TGF-β feedback loop induces N2 polarization of neutrophils and enhances tumor progression of lung adenocarcinoma. Front Immunol 2025; 16:1561081. [PMID: 40352924 PMCID: PMC12061992 DOI: 10.3389/fimmu.2025.1561081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/31/2025] [Indexed: 05/14/2025] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a major subtype of lung cancer and one of the deadliest cancers in humans. Dysregulation of miRNA activity in tumor-associated neutrophils (TANs) in the tumor microenvironment plays an important role in the occurrence and development of LUAD. Method In this study, the miReact algorithm was used to analyze the single-cell RNA sequencing data of LUAD samples to reveal the miRNA profile characteristics of TANs in LUAD patients. The function of miR-941 was investigated in vivo and in vitro. The target gene and underlying signaling pathway of miR-941 were predicted and validated with qPCR, luciferase assay, WB and ELISA assay. Results The results indicated the crucial role of TANs, especially N2-TANs in LUAD and miR-941 activity was significantly upregulated in TANs of LUAD patients. MiR-941 overexpression promoted the proliferation, invasion, migration and anti-apoptosis of A549 and H1299. In vivo xenograft mouse model confirmed that miR-941 overexpression enhanced the growth of tumors formed by H1299 cells. Bioinformatics analysis showed that miR-941 targeted the tumor suppressor gene FOXN4, and we confirmed that FOXN4 overexpression could counteract the malignant effects of miR-941. In addition, miR-941 may drive LUAD progression through the FOXN4/TGF-β feedback signaling loop and participate in the N2-TAN polarization. Conclusion In summary, these findings reveal the key role of N2-TANs and the miR-941/FOXN4/TGF-β signaling loop in LUAD progression and provide potential therapeutic targets for future interventions.
Collapse
Affiliation(s)
- Xiaojing Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Xitong Huang
- Department of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing, China
| | - Xianying Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Lichang Lai
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Baoyi Zhu
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Peibin Lin
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Zhanfang Kang
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Dazhong Yin
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Dongbo Tian
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Zisheng Chen
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| | - Jun Gao
- Department of Respiratory and Critical Care Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, China
| |
Collapse
|
37
|
Lin J, He XL, Zhang WW, Mo CF. Metabolic reprogramming of tumor-associated neutrophils in tumor treatment and therapeutic resistance. Front Cell Dev Biol 2025; 13:1584987. [PMID: 40342932 PMCID: PMC12058717 DOI: 10.3389/fcell.2025.1584987] [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: 02/28/2025] [Accepted: 04/07/2025] [Indexed: 05/11/2025] Open
Abstract
Tumor-associated neutrophils (TANs), pivotal immune cells within the tumor microenvironment (TME), exhibit dual potential in both pro- and anti-tumorigenic effects. These cells display remarkable heterogeneity and plasticity within the TME, adapting to hypoxic and nutrient-deprived conditions through metabolic reprogramming while critically influencing tumor progression, metastasis, and immune evasion. The metabolic reprogramming of TANs not only modulates their functional phenotypes but also reshapes tumor biological behaviors and therapeutic responses by regulating metabolic intermediates and cellular interactions within the TME. Therefore, elucidating the mechanisms underlying TANs metabolic reprogramming has significant implications for deciphering the molecular basis of tumorigenesis, identifying novel therapeutic targets, and optimizing immunotherapeutic strategies. This review systematically summarizes current knowledge regarding metabolic reprogramming mechanisms of TANs in the TME and their impact on tumor progression. We particularly focus on: 1) TAN-specific alterations in glucose, lipid, and amino acid metabolism within the TME; 2) Emerging immunotherapeutic strategies targeting TANs metabolic pathways; 3) Recent advances in understanding TAN-mediated immune evasion and therapy resistance. Furthermore, this review discusses potential challenges and corresponding solutions in targeting TANs metabolic reprogramming for therapeutic intervention, aiming to provide novel insights for advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Jun Lin
- Department of General Surgery, Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Xian-Lu He
- Department of General Surgery, Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Wei-Wei Zhang
- School of Public Health, Chengdu Medical College, Chengdu, China
| | - Chun-Fen Mo
- Department of General Surgery, Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| |
Collapse
|
38
|
Zhou X, Li R, Lai M, Lai C. Exploring molecular and cellular mechanisms of Pre-Metastatic niche in renal cell carcinoma. Mol Cancer 2025; 24:121. [PMID: 40264130 PMCID: PMC12012986 DOI: 10.1186/s12943-025-02315-9] [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: 12/21/2024] [Accepted: 03/25/2025] [Indexed: 04/24/2025] Open
Abstract
Renal cell carcinoma (RCC) is among the most frequently occurring types of cancer, and its metastasis is a major contributor to its elevated mortality. Before the primary tumor metastasizes to secondary or distant organs, it remodels the microenvironment of these sites, creating a pre-metastatic niche (PMN) conducive to the colonization and growth of metastatic tumors. RCC releases a variety of biomolecules that induce angiogenesis, alter vascular permeability, modulate immune cells to create an immunosuppressive microenvironment, affect extracellular matrix remodeling and metabolic reprogramming, and determine the organotropism of metastasis through different signaling pathways. This review summarizes the principal processes and mechanisms underlying the formation of the premetastatic niche in RCC. Additionally, we emphasize the significance and potential of targeting PMNs for the prevention and treatment of tumor metastasis in future therapeutic approaches. Finally, we summarized the currently potential targeted strategies for detecting and treating PMN in RCC and provide a roadmap for further in-depth studies on PMN in RCC.
Collapse
Affiliation(s)
- Xiao Zhou
- Department of Pathology, and Department of Pathology Sir Run Run Shaw Hospital, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Science (2019RU042), Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Ruirui Li
- Institute of Immunology, Department of Respiratory Disease of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Maode Lai
- Department of Pathology, and Department of Pathology Sir Run Run Shaw Hospital, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Science (2019RU042), Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
| | - Chong Lai
- Department of Urology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|
39
|
Franco-da-Silva MM, Aubin MR, de Vasconcellos AA, Sirena DH, Marchaki GB, Ruggeri LR, Hennigen AF, Muradás T, da Silveira ABT, Braganhol E, Schuh RS, Baldo G, Araújo AB, Paz AH. Effects of chorionic mesenchymal stromal cells, their conditioned medium, and membrane particles on neutrophil functionality. Cell Tissue Res 2025:10.1007/s00441-025-03970-6. [PMID: 40261417 DOI: 10.1007/s00441-025-03970-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 04/08/2025] [Indexed: 04/24/2025]
Abstract
Mesenchymal stromal cells (MSC) are multipotent cells that can modulate immune cells, affecting macrophages, monocytes, and lymphocytes. Neutrophils are circulating leucocytes responsible for the first line of defense and can assume different phenotypes depending on their environment: N0, the naïve form, N1 (inflammatory), N2 (anti-inflammatory). This study explores the potentially protective roles of chorionic membrane MSCs and their products-conditioned medium and pre-conditioned cMSC-derived membrane microparticles (MP-cMSC)-on neutrophils. Conditioned medium treatment reduced the rate of apoptosis and enhanced the immunosuppressive potential consistent with an anti-inflammatory profile. MP-cMSC are a noteworthy cell-free therapy, consisting of artificially generated circular lipid bilayer structures with no cargo and approximately 200 nm in size. When added to neutrophil culture, MPs increased neutral red uptake, suggesting an enhanced phagocytic activity. In the MSC co-culture group, a reduced rate of apoptosis, increased neutral red uptake, and elevated programed death-ligand 1 (PD-L1) expression were observed. These findings suggest that the distinct effects elicited by conditioned media, microparticles, and co-culture are likely influenced by the specific nature of the interactions involved-whether purely paracrine, mediated through direct cell-to-cell contact, or a combination of both.
Collapse
Affiliation(s)
- Monique Maria Franco-da-Silva
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Graduate Program in Physiology, Basic Health Sciences Institute, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Mariana Rauback Aubin
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Graduate Program in Physiology, Basic Health Sciences Institute, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Alessandra Amaral de Vasconcellos
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Graduate Program in Physiology, Basic Health Sciences Institute, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Dienifer Hermann Sirena
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Graduate Program in Physiology, Basic Health Sciences Institute, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Giovana Bangel Marchaki
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Laíza Rief Ruggeri
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - André Ferreira Hennigen
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Thaís Muradás
- Pharmaceutical Sciences - Graduate Program in Pharmaceutical Sciences, Pharmacy Faculty, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Ana Beatriz Tittoni da Silveira
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Elizandra Braganhol
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Roselena Silvestri Schuh
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Pharmaceutical Sciences - Graduate Program in Pharmaceutical Sciences, Pharmacy Faculty, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Guilherme Baldo
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Graduate Program in Physiology, Basic Health Sciences Institute, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Anelise Bergmann Araújo
- Cell Processing Center, Hemotherapy Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Ana Helena Paz
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
- Graduate Program in Physiology, Basic Health Sciences Institute, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil.
| |
Collapse
|
40
|
Chung MW, Tzeng CC, Huang YC, Wei KC, Hsu PW, Chuang CC, Lin YJ, Chen KT, Lee CC. Neutrophil-to-lymphocyte ratio dynamics: prognostic value and potential for surveilling glioblastoma recurrence. BMC Cancer 2025; 25:709. [PMID: 40241016 PMCID: PMC12004828 DOI: 10.1186/s12885-025-14118-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025] Open
Abstract
PURPOSE Glioblastoma (GBM) is a challenging malignancy with a poor prognosis. While the neutrophil-to-lymphocyte ratio (NLR) is reported to correlate with the prognosis, the significance of changes in the NLR and its prognostic value in GBM remain unclear. This study aims to evaluate changes in the NLR and its predictive value for GBM prognosis and recurrence. METHODS The cohort included 69 newly-diagnosed GBM patients undergoing a standard treatment protocol. NLR was assessed at multiple time points. The dynamic change in NLR (dNLR), defined as the NLR at the point of interest (post-CCRT or post-Stupp) divided by the preoperative NLR, also was assessed. Univariate and multivariate COX regression analyses were conducted to assess the association between the NLR, dNLR and overall survival (OS) and progression-free survival (PFS). RESULTS Univariate analysis revealed that age at diagnosis ≥ 70 (p = 0.019) and post-Stupp dNLR ≥ 1.3 (p = 0.006) were significantly associated with shorter OS. Significant correlations were found between pre-operative KPS ≥ 60 (p = 0.017), gross total resection (p = 0.042), post-Stupp dNLR ≥ 1.3 (p = 0.043) and PFS. Multivariate analysis showed age at diagnosis ≥ 70, pre-operative KPS ≥ 60, post-Stupp NLR ≥ 5 and dNLR ≥ 1.3 were significantly associated with a shorter OS. Significant correlation was found between pre-operative KPS ≥ 60 and PFS. CONCLUSION This study revealed that post-Stupp NLR ≥ 5 and dNLR ≥ 1.3 correlated significantly with a worse glioblastoma prognosis in OS, and dNLR might be more reliable. These two parameters are potentially surveilling markers for glioblastoma recurrence, however further studies are warranted.
Collapse
Affiliation(s)
- Meng-Wu Chung
- Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fuxing St., Guishan Dist, 33305, Taoyuan, Taiwan
| | - Ching-Chieh Tzeng
- Department of Medical Education, Chang Gung Memorial Hospital, 33305, Taoyuan, Taiwan
| | - Yin-Cheng Huang
- Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fuxing St., Guishan Dist, 33305, Taoyuan, Taiwan
- Chang Gung University, 33302, Taoyuan, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fuxing St., Guishan Dist, 33305, Taoyuan, Taiwan
- Chang Gung University, 33302, Taoyuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, 33305, Taoyuan, Taiwan
- Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, 236017, New Taipei, Taiwan
| | - Peng-Wei Hsu
- Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fuxing St., Guishan Dist, 33305, Taoyuan, Taiwan
- School of Medicine, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Chi-Cheng Chuang
- Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fuxing St., Guishan Dist, 33305, Taoyuan, Taiwan
- Chang Gung University, 33302, Taoyuan, Taiwan
| | - Ya-Jui Lin
- Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fuxing St., Guishan Dist, 33305, Taoyuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, 33305, Taoyuan, Taiwan
- Division of Natural Product, Graduate Institute of Biomedical Sciences, Chang Gung University, 33302, Taoyuan, Taiwan
| | - Ko-Ting Chen
- Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fuxing St., Guishan Dist, 33305, Taoyuan, Taiwan
- Chang Gung University, 33302, Taoyuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, 33305, Taoyuan, Taiwan
| | - Cheng-Chi Lee
- Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fuxing St., Guishan Dist, 33305, Taoyuan, Taiwan.
- Chang Gung University, 33302, Taoyuan, Taiwan.
| |
Collapse
|
41
|
Tan X, Wang Y, Yu Y, Zheng R, Li J, Chen S, Xie Q, Guo S, Zhang C, Deng X, Liu Z, Tang Y, Li H, Wu W, Chen J, Zhou Q, Wei W, Yao K, Wu Z. Neutrophil-to-lymphocyte ratio predicts a poor prognosis for penile cancer with an immunosuppressive tumor microenvironment. Front Immunol 2025; 16:1568825. [PMID: 40308599 PMCID: PMC12041217 DOI: 10.3389/fimmu.2025.1568825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/02/2025] [Indexed: 05/02/2025] Open
Abstract
Background Chronic inflammation related to poor genital hygiene is a well-recognized pathogenic trigger for penile cancer (PC). The neutrophil-to-lymphocyte ratio (NLR) is a simple, reproducible systemic inflammatory marker and has been reported to indicate unfavorable outcomes. However, previous studies were limited by small sample sizes, confounding prognostic factors and a lack of high-quality evidence demonstrating the significance of the NLR in PC. Methods A large multicenter cohort of 582 PC patients who underwent radical inguinal lymphadenectomy with definitive pN stage information was assessed. Univariate and multivariate Cox regression analyses were performed to investigate the prognostic value of inflammation-related markers. Propensity score matching (PSM) was used to minimize confounding prognostic clinicopathological features. Immunofluorescence was used to assess the immunosuppressive tumor microenvironment (TME). Results A high preoperative NLR (≥ 3.0) was associated with advanced pT, pN, and pathological grade and lymphovascular invasion in PC patients. After PSM to eliminate interference from clinical factors, pN and the NLR were found to be independent prognostic indicators (both p<0.001). PC patients with high NLRs had shorter progression-free survival (PFS) and poorer cisplatin-based chemotherapy and PD-1 immunotherapy response. We also found that the NLR is associated with proinflammatory cytokine secretion and increased N2 tumor-associated neutrophils (TANs) infiltration and CD8+ T-cell exhaustion in TME. N2 TANs induced neutrophil extracellular trap formation might contribute to tumor progression and resistance in high-NLR PC patients. Conclusions The NLR is an effective, simple and independent prognostic indicator for PC. A high NLR is associated with an immunosuppressive TME and poor outcomes.
Collapse
Affiliation(s)
- Xingliang Tan
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Yanjun Wang
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Yiqi Yu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Runhao Zheng
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Jing Li
- Department of Urology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Shaohua Chen
- Department of Urology, Guangxi Medical University Cancer Center, Nanning, China
| | - Qingling Xie
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Shengjie Guo
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Chichen Zhang
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Xinpei Deng
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Zhicheng Liu
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Yi Tang
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Hang Li
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Weicheng Wu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Juexiao Chen
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Qianghua Zhou
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Wensu Wei
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Kai Yao
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Zhiming Wu
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| |
Collapse
|
42
|
Yu J, Fu L, Wu R, Che L, Liu G, Ran Q, Xia Z, Liang X, Zhao G. Immunocytes in the tumor microenvironment: recent updates and interconnections. Front Immunol 2025; 16:1517959. [PMID: 40297580 PMCID: PMC12034658 DOI: 10.3389/fimmu.2025.1517959] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 03/11/2025] [Indexed: 04/30/2025] Open
Abstract
The tumor microenvironment (TME) is a complex, dynamic ecosystem where tumor cells interact with diverse immune and stromal cell types. This review provides an overview of the TME's evolving composition, emphasizing its transition from an early pro-inflammatory, immune-promoting state to a later immunosuppressive milieu characterized by metabolic reprogramming and hypoxia. It highlights the dual roles of key immunocytes-including T lymphocytes, natural killer cells, macrophages, dendritic cells, and myeloid-derived suppressor cells-which can either inhibit or support tumor progression based on their phenotypic polarization and local metabolic conditions. The article further elucidates mechanisms of immune cell plasticity, such as the M1/M2 macrophage switch and the balance between effector T cells and regulatory T cells, underscoring their impact on tumor growth and metastasis. Additionally, emerging therapeutic strategies, including checkpoint inhibitors and chimeric antigen receptor (CAR) T and NK cell therapies, as well as approaches targeting metabolic pathways, are discussed as promising avenues to reinvigorate antitumor immunity. By integrating recent molecular insights and clinical advancements, the review underscores the importance of deciphering the interplay between immunocytes and the TME to develop more effective cancer immunotherapies.
Collapse
Affiliation(s)
- Jiyao Yu
- Department of Ultrasound, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Li Fu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Gastroenterology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Rui Wu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Neurosurgery, Jiangyou People’s Hospital, Mianyang, China
| | - Linyi Che
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Qinwen Ran
- General Practice Department, Wufu Town Hospital, Chongqing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China
| | - Xisong Liang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Guanjian Zhao
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
43
|
Kaisar-Iluz N, Shaul ME, Deri O, Huszti E, Peled M, Bezalel Y, Fridlender ZG, Levy L. Dynamic Neutrophil Subsets and Function in Lung Transplant Recipients: Insights from a One-Year Longitudinal Pilot Study. J Clin Med 2025; 14:2660. [PMID: 40283490 PMCID: PMC12028280 DOI: 10.3390/jcm14082660] [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: 01/24/2025] [Revised: 03/14/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Neutrophils are key innate immune cells in peripheral blood. In recent years, sub-populations of neutrophils have been identified. In addition to the normal-density neutrophils (NDNs) in both healthy subjects and patients, low-density neutrophils (LDNs) were described in chronic inflammation and cancer. In lung transplants (LTx), neutrophils play crucial roles in reperfusion injury, acute rejection, and chronic lung allograft dysfunction. Our pilot study examines neutrophil subsets and function in LTx recipients during the first post-transplant year. Methods: We collected blood from 11 LTx recipients at various intervals. LDNs and normal-density neutrophils (NDNs) were isolated. The production of reactive oxygen species (ROS) by NDNs was measured after PMA activation using a Luminol-HRP assay. Neutrophil phenotypic markers were analyzed with flow cytometry. Results: The LDN-to-NDN ratio increased at 3 and 6 months post-transplant. Expression levels of CD62-L (aging marker), PDL-1 (immune checkpoint), CD15 (maturation), and CXCR4 (homeostasis regulator) showed modulation. Interestingly, ROS production by NDNs was mildly elevated at baseline, reduced at 6 months, and returned to baseline levels by 9 months post-transplant. Conclusions: Neutrophils exhibit dynamic changes in the first post-LTx year. Investigating neutrophil plasticity could reveal clinically relevant biomarkers and facilitate the development of diagnostic and therapeutic tools in LTx.
Collapse
Affiliation(s)
- Naomi Kaisar-Iluz
- Hadassah Medical Center, Institute of Pulmonary Medicine, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (N.K.-I.); (M.E.S.); (Z.G.F.)
| | - Merav E. Shaul
- Hadassah Medical Center, Institute of Pulmonary Medicine, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (N.K.-I.); (M.E.S.); (Z.G.F.)
| | - Ofir Deri
- Sheba Lung Transplant Program, Sheba Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 6997801, Israel; (O.D.); (M.P.); (Y.B.)
| | - Ella Huszti
- Biostatistics Research Unit, University Health Network, University of Toronto, Toronto, ON M5S 1A1, Canada;
| | - Michael Peled
- Sheba Lung Transplant Program, Sheba Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 6997801, Israel; (O.D.); (M.P.); (Y.B.)
| | - Yael Bezalel
- Sheba Lung Transplant Program, Sheba Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 6997801, Israel; (O.D.); (M.P.); (Y.B.)
| | - Zvi G. Fridlender
- Hadassah Medical Center, Institute of Pulmonary Medicine, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel; (N.K.-I.); (M.E.S.); (Z.G.F.)
| | - Liran Levy
- Sheba Lung Transplant Program, Sheba Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 6997801, Israel; (O.D.); (M.P.); (Y.B.)
| |
Collapse
|
44
|
Chen X, Chen B, Zhao H. Role of Neutrophils in Anti-Tumor Activity: Characteristics and Mechanisms of Action. Cancers (Basel) 2025; 17:1298. [PMID: 40282474 PMCID: PMC12025517 DOI: 10.3390/cancers17081298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
As one of the leading components in the immune system, neutrophils in the tumor microenvironment (TME) have received considerable attention in recent years. The tumor-killing effects of neutrophils in a variety of tumors have been reported. However, the functions of neutrophils in tumors remain to be completely elucidated, and both anti-tumor and tumor-promotion activities have been reported. This review focuses on the characteristics of neutrophils and their mechanisms of action in the TME, with an emphasis on their anti-tumor activity, including reactive oxygen species (ROS)-induced tumor killing, cytotoxic T lymphocytes (CTLs)-induced tumor killing, trogocytosis, cytotoxic enzymes, and trained immunity. Furthermore, the possible targets and methods of tumor treatment regimens for neutrophils are explored, with the aim of exploring the use of neutrophils in the future as a potential anti-tumor treatment strategy.
Collapse
Affiliation(s)
- Xin Chen
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi’an 710032, China;
| | - Bingdi Chen
- The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai 200092, China
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi’an 710032, China;
| |
Collapse
|
45
|
Öztürk Y, Kocabaş M, Karaköse M, Kulaksizoğlu M, Karakurt F. Prognostic significance of the pan-immune-inflammation value (PIV) in patients with differentiated thyroid carcinoma. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2025; 68:e240217. [PMID: 40215352 PMCID: PMC11967184 DOI: 10.20945/2359-4292-2024-0217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/15/2024] [Indexed: 04/15/2025]
Abstract
OBJECTIVE The pan-immune-inflammation value (PIV) is a novel inflammatory biomarker for evaluating inflammatory status in patients with cancer. The aim of this study was to evaluate the prognostic value of PIV in patients with differentiated thyroid cancers (DTCs). SUBJECTS AND METHODS The study included 376 patients with DTC who underwent curative resection. The PIV was calculated using the formula (neutrophils × monocytes × platelets)/lymphocytes. The TNM stages 3-4 were considered advanced. Patients were also categorized into low-, intermediate-, and high-risk groups according to the AmericanThyroid Association (ATA) risk classification system. Demographic, laboratory, and clinicopathological data were obtained from the patients' files. The predictive value of PIV on TNM advanced stage, ATA high-risk category, and distant metastases was evaluated using receiver operating characteristic (ROC) curve analysis. RESULTS The optimal PIV values for predicting TNM advanced stage, ATA high-risk category, and distant metastases were, respectively, 331.62 (area under the curve [AUC] 0.730, sensitivity 66.7%, specificity 64.8%), 365.52 (AUC 0.822, sensitivity 79.5%, specificity 76.9%), and 357.65 (AUC 0.774, sensitivity 75.2%, specificity 72%). On regression analysis, PIV ≥ 365.52 (odds ratio [OR] 29.150, 95% confidence interval [CI] 8.148-104.290, p < 0.001) and PIV ≥ 357.65 (OR 7.224, 95% CI 1.700-30.693, p = 0.007) emerged as independent risk factors for ATA high-risk category and distant metastases, respectively. CONCLUSION Among patients with DTC, PIV is an independent risk factor for distant metastases and ATA high-risk category. While this finding must be supported by more comprehensive studies, PIV has the potential to be used as a prognostic biomarker in these patients.
Collapse
Affiliation(s)
- Yusuf Öztürk
- Department of Endocrinology and Metabolism, Necmettin Erbakan University
Faculty of Medicine, Konya, Türkiye
| | - Muhammet Kocabaş
- Department of Endocrinology and Metabolism, Necmettin Erbakan University
Faculty of Medicine, Konya, Türkiye
| | - Melia Karaköse
- Department of Endocrinology and Metabolism, Necmettin Erbakan University
Faculty of Medicine, Konya, Türkiye
| | - Mustafa Kulaksizoğlu
- Department of Endocrinology and Metabolism, Necmettin Erbakan University
Faculty of Medicine, Konya, Türkiye
| | - Feridun Karakurt
- Department of Endocrinology and Metabolism, Necmettin Erbakan University
Faculty of Medicine, Konya, Türkiye
| |
Collapse
|
46
|
Parveen S, Konde DV, Paikray SK, Tripathy NS, Sahoo L, Samal HB, Dilnawaz F. Nanoimmunotherapy: the smart trooper for cancer therapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002308. [PMID: 40230883 PMCID: PMC11996242 DOI: 10.37349/etat.2025.1002308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/20/2025] [Indexed: 04/16/2025] Open
Abstract
Immunotherapy has gathered significant attention and is now a widely used cancer treatment that uses the body's immune system to fight cancer. Despite initial successes, its broader clinical application is hindered by limitations such as heterogeneity in patient response and challenges associated with the tumor immune microenvironment. Recent advancements in nanotechnology have offered innovative solutions to these barriers, providing significant enhancements to cancer immunotherapy. Nanotechnology-based approaches exhibit multifaceted mechanisms, including effective anti-tumor immune responses during tumorigenesis and overcoming immune suppression mechanisms to improve immune defense capacity. Nanomedicines, including nanoparticle-based vaccines, liposomes, immune modulators, and gene delivery systems, have demonstrated the ability to activate immune responses, modulate tumor microenvironments, and target specific immune cells. Success metrics in preclinical and early clinical studies, such as improved survival rates, enhanced tumor regression, and elevated immune activation indices, highlight the promise of these technologies. Despite these achievements, several challenges remain, including scaling up manufacturing, addressing off-target effects, and navigating regulatory complexities. The review emphasizes the need for interdisciplinary approaches to address these barriers, ensuring broader clinical adoption. It also provides insights into interdisciplinary approaches, advancements, and the transformative potential of nano-immunotherapy and promising results in checkpoint inhibitor delivery, nanoparticle-mediated photothermal therapy, immunomodulation as well as inhibition by nanoparticles and cancer vaccines.
Collapse
Affiliation(s)
- Suphiya Parveen
- Department of Biotechnology and Genetics, School of Sciences, Jain (Deemed-to-be-University), Bengaluru 560027, Karnataka, India
| | - Dhanshree Vikrant Konde
- Department of Biotechnology and Genetics, School of Sciences, Jain (Deemed-to-be-University), Bengaluru 560027, Karnataka, India
| | - Safal Kumar Paikray
- School of Biotechnology, Centurion University of Technology and Management, Jatni 752050, Odisha, India
| | - Nigam Sekhar Tripathy
- School of Biotechnology, Centurion University of Technology and Management, Jatni 752050, Odisha, India
| | - Liza Sahoo
- School of Biotechnology, Centurion University of Technology and Management, Jatni 752050, Odisha, India
| | - Himansu Bhusan Samal
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Jatni 752050, Odisha, India
| | - Fahima Dilnawaz
- School of Biotechnology, Centurion University of Technology and Management, Jatni 752050, Odisha, India
| |
Collapse
|
47
|
Zhao H, Niu M, Guo Y, Li Q, Wang Y, Jiang Q, Song Q, Zhang Y, Wang L. A lipid starvation strategy-synergized neutrophil activation for postoperative melanoma immunotherapy. J Control Release 2025; 380:860-874. [PMID: 39952297 DOI: 10.1016/j.jconrel.2025.02.027] [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: 12/02/2024] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Abnormal metabolism of melanoma cells on lipids reveals that breaking their lipid addiction provides a starvation strategy to enhance immunotherapy effects and reduce resistance. Herein, we propose an extracellular matrix-inspired scaffold fabricated by multiple cross-linking of collagen and elastin encapsulated with fatty acid transporter proteins (FATP) inhibitor lipofermata (Lipo) to close the "valve" of lipid transported into both melanoma cells and pro-tumor neutrophils. Meanwhile, model TGF-β inhibitor loaded in scaffold synergized with Lipo to polarize postoperative locally enriched neutrophils towards cytotoxic N1 phenotypes after blocking their energy supply and modulate postsurgical immunosuppressive tumor microenvironment. These N1 neutrophils induced tumor pyroptosis through a reactive oxygen species (ROS)-dependent pathway under melanoma cells suffered starvation, and the intracellular contents released from dead melanoma cells stimulated macrophages into producing proinflammatory cytokines, which recruited a secondary wave of neutrophils to the tumor site. Benefiting from the N1 neutrophil induced tumor pyroptosis feedback loop in situ, adaptive and memory antitumor immunity is activated for suppressing aggressive melanoma recurrence and metastasis. Altogether, this lipid starvation strategy synergized with neutrophil activation for amplification of tumor-specific immunotherapy provides a new paradigm for pyroptosis-mediated postsurgical melanoma therapy.
Collapse
Affiliation(s)
- Hongjuan Zhao
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 science avenue, Zhengzhou 450001, China; Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, 100 science avenue, Zhengzhou 450001, China
| | - Mengya Niu
- School of Pharmaceutical Sciences, Zhengzhou University, 100 science avenue, Zhengzhou 450001, China
| | - Yuxin Guo
- School of Pharmaceutical Sciences, Zhengzhou University, 100 science avenue, Zhengzhou 450001, China
| | - Qing Li
- School of Pharmaceutical Sciences, Zhengzhou University, 100 science avenue, Zhengzhou 450001, China
| | - Yinke Wang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 science avenue, Zhengzhou 450001, China
| | - Qianqian Jiang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 science avenue, Zhengzhou 450001, China
| | - Qingling Song
- School of Pharmaceutical Sciences, Zhengzhou University, 100 science avenue, Zhengzhou 450001, China; Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, 100 science avenue, Zhengzhou 450001, China
| | - Yi Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Lei Wang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 science avenue, Zhengzhou 450001, China; Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, 100 science avenue, Zhengzhou 450001, China.
| |
Collapse
|
48
|
Masmoudi D, Villalba M, Alix-Panabières C. Natural killer cells: the immune frontline against circulating tumor cells. J Exp Clin Cancer Res 2025; 44:118. [PMID: 40211394 PMCID: PMC11983744 DOI: 10.1186/s13046-025-03375-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Natural killer (NK) play a key role in controlling tumor dissemination by mediating cytotoxicity towards cancer cells without the need of education. These cells are pivotal in eliminating circulating tumor cells (CTCs) from the bloodstream, thus limiting cancer spread and metastasis. However, aggressive CTCs can evade NK cell surveillance, facilitating tumor growth at distant sites. In this review, we first discuss the biology of NK cells, focusing on their functions within the tumor microenvironment (TME), the lymphatic system, and circulation. We then examine the immune evasion mechanisms employed by cancer cells to inhibit NK cell activity, including the upregulation of inhibitory receptors. Finally, we explore the clinical implications of monitoring circulating biomarkers, such as NK cells and CTCs, for therapeutic decision-making and emphasize the need to enhance NK cell-based therapies by overcoming immune escape mechanisms.
Collapse
Affiliation(s)
- Doryan Masmoudi
- Laboratory of Rare Circulating Human Cells, University Medical Center of Montpellier, Montpellier, France
| | - Martin Villalba
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Circulating Human Cells, University Medical Center of Montpellier, Montpellier, France.
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, Montpellier, IRD, France.
- European Liquid Biopsy Society (ELBS), Hamburg, Germany.
- LCCRH, Site Unique de Biologie (SUB), 641, Avenue du Doyen Gaston Giraud, Montpellier, 34093, France.
| |
Collapse
|
49
|
Yu L, Liebenberg K, Shen Y, Liu F, Xu Z, Hao X, Wu L, Zhang W, Chan HL, Wei B, Lorenzi PL, Gao Y, Bado I, Becerra-Dominguez L, Rivas CH, Aguirre S, Pingel BC, Wu YH, Ding Y, Liu J, Edwards DG, Eberlin LS, Zhang XHF. Tumor-derived arachidonic acid reprograms neutrophils to promote immune suppression and therapy resistance in triple-negative breast cancer. Immunity 2025; 58:909-925.e7. [PMID: 40157359 PMCID: PMC11981829 DOI: 10.1016/j.immuni.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/08/2024] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
The combination of immune checkpoint blockade and chemotherapies is the standard of care for triple-negative breast cancer (TNBC). However, initially, responsive tumors can still develop recurrences, suggesting acquired resistance mechanisms that remain poorly understood. Herein, we discover that TNBC cells surviving anti-programmed cell death protein-1 (anti-PD-1) and chemotherapy treatment accumulate neutral lipids. Disrupting lipid droplet formation in cancer cells reverses resistance and mitigates the immunosuppressive microenvironment. Single-cell RNA sequencing reveals a subset of neutrophils exhibiting a lipid-laden phenotype similar to adjacent tumor cells. Mechanistically, tumor-derived extracellular vesicles carrying lipids, including arachidonic acid (AA), mediate neutrophil reprogramming. Blocking dietary intake of omega-6 fatty acids or inhibiting fatty acid elongation for AA synthesis restores anti-tumor immunity and re-sensitizes the resistant tumors to anti-PD-1 and chemotherapy treatment. In human patients, AA metabolism-related pathways correlates with neutrophil enrichment. Overall, we demonstrate how lipid accumulation in TNBC cells leads to immune suppression and therapy resistance.
Collapse
Affiliation(s)
- Liqun Yu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Keziah Liebenberg
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yichao Shen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Zhan Xu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiaoxin Hao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ling Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Weijie Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hilda L Chan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bo Wei
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Philip L Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yang Gao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Igor Bado
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Luis Becerra-Dominguez
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Immunology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Charlotte Helena Rivas
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Sergio Aguirre
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bradley C Pingel
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Immunology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yi-Hsuan Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yunfeng Ding
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jun Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - David G Edwards
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Livia S Eberlin
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; McNair Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
50
|
Yang Y, Liu Z, Wang Z, Fu X, Li Z, Li J, Xu Z, Cen B. Large-scale bulk and single-cell RNA sequencing combined with machine learning reveals glioblastoma-associated neutrophil heterogeneity and establishes a VEGFA + neutrophil prognostic model. Biol Direct 2025; 20:45. [PMID: 40188324 PMCID: PMC11972500 DOI: 10.1186/s13062-025-00640-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/22/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Neutrophils play a key role in the tumor microenvironment (TME); however, their functions in glioblastoma (GBM) are overlooked and insufficiently studied. A detailed analysis of GBM-associated neutrophil (GBMAN) subpopulations may offer new insights and opportunities for GBM immunotherapy. METHODS We analyzed single-cell RNA sequencing (scRNA-seq) data from 127 isocitrate dehydrogenase (IDH) wild-type GBM samples to characterize the GBMAN subgroups, emphasizing developmental trajectories, cellular communication, and transcriptional networks. We implemented 117 machine learning combinations to develop a novel risk model and compared its performance to existing glioma models. Furthermore, we assessed the biological and molecular features of the GBMAN subgroups in patients. RESULTS From integrated large-scale scRNA-seq data (498,747 cells), we identified 5,032 neutrophils and classified them into four distinct subtypes. VEGFA+GBMAN exhibited reduced inflammatory response characteristics and a tendency to interact with stromal cells. Furthermore, these subpopulations exhibited significant differences in transcriptional regulation. We also developed a risk model termed the "VEGFA+neutrophil-related signature" (VNRS) using machine learning methods. The VNRS model showed higher accuracy than previously published risk models and was an independent prognostic factor. Additionally, we observed significant differences in immunotherapy responses, TME interactions, and chemotherapy efficacy between high-risk and low-risk VNRS score groups. CONCLUSION Our study highlights the critical role of neutrophils in the TME of GBM, allowing for a better understanding of the composition and characteristics of GBMAN. The developed VNRS model serves as an effective tool for evaluating the risk and guiding clinical treatment strategies for GBM. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Yufan Yang
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong- Macao, Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Ziyuan Liu
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
- National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong- Macao, Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhongliang Wang
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Xiang Fu
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong- Macao, Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhiyong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jianlong Li
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
- Departments of Pediatrics, Weill Cornell Medicine, New York, NY, USA.
| | - Zhongyuan Xu
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
- National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong- Macao, Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Bohong Cen
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
- National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong- Macao, Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|