1
|
Deng Z, Mei S, Ouyang Z, Wang R, Wang L, Zou B, Dai J, Mao K, Li Q, Guo Q, Yi C, Meng F, Xie M, Zhang X, Wang R, Deng T, Wang Z, Li X, Wang Q, Liu B, Tian X. Dysregulation of gut microbiota stimulates NETs-driven HCC intrahepatic metastasis: therapeutic implications of healthy faecal microbiota transplantation. Gut Microbes 2025; 17:2476561. [PMID: 40099491 PMCID: PMC11925110 DOI: 10.1080/19490976.2025.2476561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 12/04/2024] [Accepted: 03/03/2025] [Indexed: 03/20/2025] Open
Abstract
The stringent regulation of intrahepatic metastases is essential for improving survival outcomes in patients with hepatocellular carcinoma (HCC). This study investigated the impact of gut microbiota on intrahepatic metastasis of HCC and evaluated the therapeutic potential of healthy fecal microbiota transplantation (FMT). Dysregulation of the gut microbiota, characterized by a significant reduction in the abundance of beneficial bacteria, such as Anaerotruncus colihominis and Dysosmobacter welbionis, was observed in patients with intrahepatic metastatic HCC. A human flora-associated (HFA) intrahepatic metastatic HCC mouse model was successfully established through consecutive 4 weeks of human-mouse FMT. Dysregulation of gut microbiota promoted intrahepatic metastasis in the mouse model, primarily by enhancing neutrophil-mediated inflammatory responses and lead to excessive formation of neutrophil extracellular traps (NETs). Consequently, it promoted tumor vascular growth and tissue necrosis, resulting in intrahepatic metastasis of HCC. Notably, FMT from healthy donors mitigated these pathological processes. This study elucidated the role and mechanism of dysregulated gut microbiota in promoting intrahepatic metastasis of HCC. Healthy FMT emerges as a promising novel therapeutic strategy for preventing and treating intrahepatic metastasis of HCC.
Collapse
Affiliation(s)
- Zhe Deng
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Si Mei
- Hunan Province University Key Laboratory of Oncology of Traditional Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Traditional Chinese Medicine for Mechanism of Tumor Prevention &Treatment, Changsha, Hunan, China
- Department of Physiology, Faculty of Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhaoguang Ouyang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, China
| | - Ruoyu Wang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lihuai Wang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Bo Zou
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jingjing Dai
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Kexin Mao
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Qian Li
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Qianqian Guo
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chun Yi
- Department of Pathology, Faculty of Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Fanying Meng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Mingxia Xie
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xue Zhang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Rongrong Wang
- Hunan Province Integrated Traditional Chinese and Western Medicine Hospital, Changsha, Hunan, China
| | - Tianhao Deng
- Hunan Province Integrated Traditional Chinese and Western Medicine Hospital, Changsha, Hunan, China
| | - Zhenyu Wang
- JCY Biotech Ltd., Pingshan Translational Medicine Center, Shenzhen Bay Laboratory, Shenzhen, China
| | - Xiaozheng Li
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, China
| | - Qing Wang
- Shanghai OE Biotech Co. Ltd, Shanghai, China
| | - Bin Liu
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xuefei Tian
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Province University Key Laboratory of Oncology of Traditional Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Traditional Chinese Medicine for Mechanism of Tumor Prevention &Treatment, Changsha, Hunan, China
| |
Collapse
|
2
|
Karaman G, Ipek V. Preliminary study of neutrophils and neutrophil extracellular traps (NETs) in canine mammary tumors. Res Vet Sci 2025; 186:105573. [PMID: 39965363 DOI: 10.1016/j.rvsc.2025.105573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 01/23/2025] [Accepted: 02/12/2025] [Indexed: 02/20/2025]
Abstract
Neutrophils play a complex role in cancer biology, can contributing to tumor progression and immune defense. Neutrophil extracellular traps (NETs) have emerged as key modulators within the tumor microenvironment. Herein, the association between molecular classification, histological grade, necrosis, tumor-infiltrating neutrophils, and NETs was assessed in 19 canine mammary malignant tumors. Immunohistochemistry using citrullinated histone-3 (cith3) and myeloperoxidase (MPO) antibodies were used to detect NETs. A fading and re-staining method was applied on the same sections. NETs were scored based on the presence of cith3 positive areas and compared with tumor grade. The neutrophil score numerically increased as the tumor grade increased. The NET score was slightly higher in grade I carcinomas compared to carcinomas with other grades. On contrary, the necrosis score was also higher in grade II and III tumors than grade I tumors. A low but non-significant negative correlation existed between tumor grade and NET score (r = -0.219). No statistically significant associations between the tumor markers (ER, PR, HER2) and molecular subtypes with tumor grade, NET score, neutrophil count, and necrosis. In this study, the presence of NETs in canine malignant mammary tumor of different histological subtypes and grades was reported. Preliminary evidence was gathered that NETs are negatively correlated with tumor grade, suggesting their potential role in prognostication.
Collapse
Affiliation(s)
- Gulsum Karaman
- Burdur Mehmet Akif Ersoy University, Health Sciences Institute, Burdur, Türkiye
| | - Volkan Ipek
- Burdur Mehmet Akif Ersoy University, Faculty of Veterinary Medicine, Department of Pathology, Burdur, Türkiye.
| |
Collapse
|
3
|
Boixareu C, Taha T, Venkadakrishnan VB, de Bono J, Beltran H. Targeting the tumour cell surface in advanced prostate cancer. Nat Rev Urol 2025:10.1038/s41585-025-01014-w. [PMID: 40169837 DOI: 10.1038/s41585-025-01014-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2025] [Indexed: 04/03/2025]
Abstract
Prostate cancer remains a substantial health challenge, with >375,000 annual deaths amongst men worldwide. Most prostate cancer-related deaths are attributable to the development of resistance to standard-of-care treatments. Characterization of the diverse and complex surfaceome of treatment-resistant prostate cancer, combined with advances in drug development that leverage cell-surface proteins to enhance drug delivery or activate the immune system, have provided novel therapeutic opportunities to target advanced prostate cancer. The prostate cancer surfaceome, including proteins such as prostate-specific membrane antigen (PSMA), B7-H3, six transmembrane epithelial antigen of the prostate 1 (STEAP1), delta-like ligand 3 (DLL3), trophoblastic cell-surface antigen 2 (TROP2), prostate stem cell antigen (PSCA), HER3, CD46 and CD36, can be exploited as therapeutic targets, as regulatory mechanisms might contribute to the heterogeneity of expression of these proteins and subsequently affect treatment response and resistance. Specific treatment strategies targeting the surfaceome are in clinical development, including radionuclides, antibody-drug conjugates, T cell engagers and chimeric antigen receptor (CAR) T cells. Ultimately, biomarker development and clinical implementation of these agents will be informed and refined by further understanding of the biology of various targets; the target specificity and sensitivity of different agents; and off-target and toxic effects associated with these agents. Understanding the dynamic nature of cell-surface targets and non-overlapping expression patterns might also lead to future combinational strategies.
Collapse
Affiliation(s)
- Cristina Boixareu
- The Institute of Cancer Research, The Royal Marsden Hospital, London, UK
| | - Tarek Taha
- The Institute of Cancer Research, The Royal Marsden Hospital, London, UK
| | | | - Johann de Bono
- The Institute of Cancer Research, The Royal Marsden Hospital, London, UK.
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Meier C, Brieger A. The role of IL-8 in cancer development and its impact on immunotherapy resistance. Eur J Cancer 2025; 218:115267. [PMID: 39899909 DOI: 10.1016/j.ejca.2025.115267] [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: 12/18/2024] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/05/2025]
Abstract
Tumors are structures of high complexity. Plurality of their structural and functional components - heterogeneity, diversity, directionality, interdependence and integration of signaling pathways - seem to follow isolated local rules, whereby a superordinate structure remains largely unknown. Understanding the complexity of cancer is the mainstay in finding determinants and developing effective therapies. Interleukin 8 (IL-8) is a potent pro-inflammatory chemokine that is significantly elevated in many different tumor entities. In contrast to its initially postulated anti-tumor properties, an increasing number of studies have been published in recent years linking this chemokine with tumor-promoting features and poor prognosis. This review summarizes the current state and diversity of the role of IL-8 in the development of cancer.
Collapse
Affiliation(s)
- Clara Meier
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Biomedical Research Laboratory, Frankfurt am Main, Germany
| | - Angela Brieger
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Biomedical Research Laboratory, Frankfurt am Main, Germany.
| |
Collapse
|
5
|
Lu HJ, Ren GC, Wang Y, Wang CQ, Zhang DH. Preoperative and Postoperative Neutrophil-Lymphocyte Ratio and Platelet-Lymphocyte Ratio Measured From the Peripheral Blood of Patients with Colorectal Cancer. Cancer Manag Res 2025; 17:527-540. [PMID: 40093570 PMCID: PMC11909476 DOI: 10.2147/cmar.s504532] [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: 11/14/2024] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Background The neutrophil-lymphocyte ratio (NLR) and platelet-lymphocyte ratio (PLR) have been confirmed to be related to the clinicopathological features and prognosis of colorectal cancer (CRC) patients. However, the results have been inconsistent, and few studies have focused on a specific point in time during surgery and dynamic changes prior to and after surgery. Methods We conducted a retrospective analysis of 349 CRC patients and explored the value of NLR, PLR and their dynamic changes in predicting clinicopathological variables and prognosis in CRC. Results Preoperative NLR (Pre-NLR) was correlated with CEA, CA199 levels, tumor location and tumor stage (P=0.041, P=0.002, P=0.001 and P=0.012, respectively), whereas postoperative NLR (post-NLR) was relevant to age, sex, CA125 levels and T stage significantly (P=0.032, P=0.002, P=0.026, P=0.019, respectively). When comparing post- and pre-NLR values, there was a positive connection between increases in NLR and BMI, tumor location, T stage, and tumor stage (P=0.034, P=0.005, P=0.023, P=0.023, respectively). In addition, Preoperative PLR (pre-PLR) was correlated with sex, smoke and drink history, CEA and CA199 levels, tumor location, T stage and tumor stage (P=0.006, P=0.037, P=0.040, P=0.006, P=0.005, P<0.001, P=0.007, P=0.003 respectively), while postoperativePLR (post-PLR) was only associated with tumor location (P=0.010). Increases in PLR were significantly related to sex, smoking history, tumor location and differentiation (P=0.001, P=0.002, P<0.001, P=0.034, respectively). Patients with CRC who had a high post-PLR experienced significantly shorter relapse-free survival (RFS) compared to other patients (HR 0.607 (0.381-0.968), P=0.036). Furthermore, this high post-PLR has tendency association with shorter overall survival (OS) (HR 0.596 (0.338-1.050), P=0.076). Conclusion These findings suggest that levels and changes in NLR/PLR are associated with several unfavorable clinicopathological features in CRC patients. Furthermore, patients with high levels of post-PLR exhibit a worse prognosis.
Collapse
Affiliation(s)
- Hua-Jun Lu
- Department of Oncological Radiotherapy, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, People's Republic of China
| | - Guo-Chao Ren
- Department of Oncological Radiotherapy, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, People's Republic of China
| | - Yan Wang
- Department of Medical Oncology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, People's Republic of China
| | - Chao-Qun Wang
- Department of Pathology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, People's Republic of China
| | - Da-Hai Zhang
- Department of Oncological Radiotherapy, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, People's Republic of China
| |
Collapse
|
6
|
Wen Y, Zhao G, Dai C. Cell-free DNA: plays an essential role in early diagnosis and immunotherapy of pancreatic cancer. Front Immunol 2025; 16:1546332. [PMID: 40124355 PMCID: PMC11925872 DOI: 10.3389/fimmu.2025.1546332] [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: 12/16/2024] [Accepted: 02/24/2025] [Indexed: 03/25/2025] Open
Abstract
Pancreatic cancer is renowned for its aggressive nature and dismal prognosis, with the majority of patients diagnosed at an advanced stage. The prognosis for patients with pancreatic cancer can be improved by early diagnosis and effective treatment. Circulating cell-free DNA (cfDNA) has emerged as a promising biomarker for the early diagnosis and monitoring of pancreatic cancer. This research presents a review of circulating cell-free DNA essential role in the early diagnosis and immunotherapy of pancreatic cancer. The detection methods of cfDNA, its potential as a diagnostic biomarker, and the latest research progress in cfDNA-based immunotherapy are discussed. The findings suggest that cfDNA plays a vital role in the early detection and personalised treatment of pancreatic cancer, holding great promise for improving patient outcomes.
Collapse
Affiliation(s)
- Yi Wen
- College of Outstanding Clinician, Jiangsu University, Zhenjiang, China
| | - Gengmin Zhao
- Graduate School, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Chunhua Dai
- Department of Thoracic Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
7
|
Qian YY, Xu M, Huang XK, Zhu B. Bioinformatic analysis indicated that LINC01150 might be a novel neutrophil extracellular traps-related biomarker of gastric cancer. Sci Rep 2025; 15:7875. [PMID: 40050656 PMCID: PMC11885803 DOI: 10.1038/s41598-025-92968-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 03/04/2025] [Indexed: 03/09/2025] Open
Abstract
Gastric cancer (GC) is a highly aggressive malignancy associated with poor prognosis, particularly in its advanced stages. Neutrophil extracellular traps (NETs) have been implicated in cancer progression and immune therapy responses; however, the role of NETs-related long non-coding RNAs (lncRNAs) in GC remains poorly understood. This study used data from the Cancer Genome Atlas (TCGA) and previous research to identify NETs-related lncRNAs in GC. A prognostic signature comprising four NETs-related lncRNAs (NlncSig) was developed and validated, serving as a predictor for patient survival and response to immunotherapy. The NlncSig was correlated with poorer outcomes in high-risk patients and demonstrated that those with lower risk scores exhibited more favorable responses to immunotherapy. In vitro experiments confirmed that LINC01150 enhances GC cell proliferation, migration, and invasion. This robust NlncSig provides a reliable tool for predicting survival and immune characteristics in GC, with the potential to guide personalized therapeutic approaches and improve patient care.
Collapse
Affiliation(s)
- Yang-Yang Qian
- Department of Central Laboratory, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng, China
- Department of General Surgery, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng, China
| | - Min Xu
- Department of Central Laboratory, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng, China
- Department of General Surgery, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng, China
| | - Xin-Kun Huang
- Department of General Surgery, Affiliated Tumor Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Bin Zhu
- Department of Central Laboratory, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng, China.
- Department of General Surgery, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng, China.
| |
Collapse
|
8
|
Jacome MA, Wu Q, Chen J, Mohamed ZS, Mokhtari S, Piña Y, Etame AB. Molecular Underpinnings of Brain Metastases. Int J Mol Sci 2025; 26:2307. [PMID: 40076927 PMCID: PMC11900073 DOI: 10.3390/ijms26052307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Brain metastases are the most commonly diagnosed type of central nervous system tumor, yet the mechanisms of their occurrence are still widely unknown. Lung cancer, breast cancer, and melanoma are the most common etiologies, but renal and colorectal cancers have also been described as metastasizing to the brain. Regardless of their origin, there are common mechanisms for progression to all types of brain metastases, such as the creation of a suitable tumor microenvironment in the brain, priming of tumor cells, adaptations to survive spreading in lymphatic and blood vessels, and development of mechanisms to penetrate the blood-brain barrier. However, there are complex genetic and molecular interactions that are specific to every type of primary tumor, making the understanding of the metastatic progression of tumors to the brain a challenging field of study. In this review, we aim to summarize current knowledge on the pathophysiology of brain metastases, from specific genetic characteristics of commonly metastatic tumors to the molecular and cellular mechanisms involved in progression to the central nervous system. We also briefly discuss current challenges in targeted therapies for brain metastases and how there is still a gap in knowledge that needs to be overcome to improve patient outcomes.
Collapse
Affiliation(s)
- Maria A. Jacome
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Qiong Wu
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (Q.W.); (J.C.); (S.M.); (Y.P.)
| | - Jianan Chen
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (Q.W.); (J.C.); (S.M.); (Y.P.)
| | | | - Sepideh Mokhtari
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (Q.W.); (J.C.); (S.M.); (Y.P.)
| | - Yolanda Piña
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (Q.W.); (J.C.); (S.M.); (Y.P.)
| | - Arnold B. Etame
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (Q.W.); (J.C.); (S.M.); (Y.P.)
| |
Collapse
|
9
|
Pranzini E, Ippolito L, Pardella E, Giannoni E, Chiarugi P. Adapt and shape: metabolic features within the metastatic niche. Trends Endocrinol Metab 2025; 36:205-218. [PMID: 39122599 DOI: 10.1016/j.tem.2024.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024]
Abstract
The success of disseminating cancer cells (DTCs) at specific metastatic sites is influenced by several metabolic factors. Even before DTCs arrival, metabolic conditioning from the primary tumor participates in creating a favorable premetastatic niche at distant organs. In addition, DTCs adjust their metabolism to better survive along the metastatic journey and successfully colonize their ultimate destination. However, the idea that the environment of the target organs may metabolically impact the metastatic fate is often underestimated. Here, we review the coexistence of two distinct strategies by which cancer cells shape and/or adapt to the metabolic profile of colonized tissues, ultimately creating a proper soil for their seeding and proliferation.
Collapse
Affiliation(s)
- Erica Pranzini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni, 50, 50134 Firenze, (FI), Italy
| | - Luigi Ippolito
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni, 50, 50134 Firenze, (FI), Italy
| | - Elisa Pardella
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni, 50, 50134 Firenze, (FI), Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni, 50, 50134 Firenze, (FI), Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni, 50, 50134 Firenze, (FI), Italy.
| |
Collapse
|
10
|
Huang S, Shi J, Shen J, Fan X. Metabolic reprogramming of neutrophils in the tumor microenvironment: Emerging therapeutic targets. Cancer Lett 2025; 612:217466. [PMID: 39862916 DOI: 10.1016/j.canlet.2025.217466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
Neutrophils are pivotal in the immune system and have been recognized as significant contributors to cancer development and progression. These cells undergo metabolic reprogramming in response to various stimulus, including infections, diseases, and the tumor microenvironment (TME). Under normal conditions, neutrophils primarily rely on aerobic glucose metabolism for energy production. However, within the TME featured by hypoxic and nutrient-deprived conditions, they shift to altered anaerobic glycolysis, lipid metabolism, mitochondrial metabolism and amino acid metabolism to perform their immunosuppressive functions and facilitate tumor progression. Targeting neutrophils within the TME is a promising therapeutic approach. Yet, focusing on their metabolic pathways presents a novel strategy to enhance cancer immunotherapy. This review synthesizes the current understanding of neutrophil metabolic reprogramming in the TME, with an emphasis on the underlying molecular mechanisms and signaling pathways. Studying neutrophil metabolism in the TME poses challenges, such as their short lifespan and the metabolic complexity of the environment, necessitating the development of advanced research methodologies. This review also discusses emerging solutions to these challenges. In conclusion, given their integral role in the TME, targeting the metabolic pathways of neutrophils could offer a promising avenue for cancer therapy.
Collapse
Affiliation(s)
- Shiyun Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China.
| | - Jiahao Shi
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China.
| | - Jianfeng Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China.
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China.
| |
Collapse
|
11
|
Wang X, He S, Gong X, Lei S, Zhang Q, Xiong J, Liu Y. Neutrophils in colorectal cancer: mechanisms, prognostic value, and therapeutic implications. Front Immunol 2025; 16:1538635. [PMID: 40092983 PMCID: PMC11906667 DOI: 10.3389/fimmu.2025.1538635] [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: 12/03/2024] [Accepted: 02/04/2025] [Indexed: 03/19/2025] Open
Abstract
Neutrophils, the most abundant myeloid cells in human peripheral blood, serve as the first defense line against infection and are also significantly involved in the initiation and progression of cancer. In colorectal cancer (CRC), neutrophils exhibit a dual function by promoting tumor events and exerting antitumor activity, which is related to the heterogeneity of neutrophils. The neutrophil extracellular traps (NETs), gut microbiota, and various cells within the tumor microenvironment (TME) are involved in shaping the heterogeneous function of neutrophils. This article provides an updated overview of the complex functions and underlying mechanisms of neutrophils in CRC and their pivotal role in guiding prognosis assessment and therapeutic strategies, aiming to offer novel insights into neutrophil-associated treatment approaches for CRC.
Collapse
Affiliation(s)
- Xingyue Wang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shukang He
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangmei Gong
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shijun Lei
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qianwen Zhang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junqi Xiong
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Liu
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Liu Y, Ma J, Ma Y, Wang BZ, Wang Y, Yuan J, Zhang F, Zhao X, Chen K, Zhang X, Wang H. Neutrophil extracellular traps impede cancer metastatic seeding via protease-activated receptor 2-mediated downregulation of phagocytic checkpoint CD24. J Immunother Cancer 2025; 13:e010813. [PMID: 40010762 DOI: 10.1136/jitc-2024-010813] [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] [Accepted: 02/10/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Phagocytic clearance by macrophages represents a critical immune surveillance mechanism in cancer liver metastasis. Neutrophils, the most abundant immune cells encountered by cancer cells in circulation, play key roles in metastasis through neutrophil extracellular traps (NETs). Although NETs promote macrophage phagocytosis during infection, whether they regulate phagocytosis during cancer metastasis is unknown. The present study aimed to explore the roles of NETs in regulating macrophage phagocytosis during the seeding process of liver metastasis and the mechanisms underlying the roles. METHODS A lipopolysaccharide-induced NET model was applied to study the role of NETs on colorectal cancer (CRC) liver metastasis. The neutrophils isolated from human peripheral blood were stimulated with PMA to release NETs, which were collected and added to the cultures of different CRC cell lines for in vitro studies. Macrophage phagocytosis was assessed with flow cytometry in vitro and in vivo. RNA-seq and microRNA array analyses were performed to identify key pathways regulated by NETs and downstream key molecules. The macrophage phenotypes were evaluated using immunohistochemistry, flow cytometry, and cytokine and chemokine arrays. RESULTS NETs promote macrophage phagocytosis both in vitro and in vivo. Neutrophil elastase (NE), which was able to inactivate the canonical signal of protease-activated receptor 2 (PAR2), downregulated the phagocytotic checkpoint CD24. Notably, PAR2 deficiency imitated the effect of NETs on phagocytosis and CD24. Mechanistic studies indicated that inhibiting PAR2 expression upregulated miR-34a and miR-146a and downregulated CD24 in cancer cells. In addition, PAR2 depletion enhanced the recruitment and M1 polarization of macrophages by upregulating CSF-1 and CXCL1. The correlation of NETs/NE and CD24 was corroborated using human CRC specimens. Furthermore, PAR2 blockade combined with an anti-EGFR antibody (cetuximab (CTX)) synergistically enhanced the phagocytic ability of macrophages and suppressed liver metastasis in vivo. CONCLUSIONS NET-derived elastase inactivated PAR2 canonical signaling and promoted phagocytosis by downregulating CD24, which functions as a phagocytotic checkpoint in CRC liver metastasis. Thus, PAR2 inhibitors combined with CTX may serve as a novel therapeutic strategy against advanced CRC.
Collapse
Affiliation(s)
- Yu Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Jianhui Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Yiming Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Bing-Zhi Wang
- Department of Pathology and Resident Training Base, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Yinong Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Junhu Yuan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Fanyu Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Xinhua Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| | - Kun Chen
- Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Liaoning, China
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Liaoning, China
| | - Xiaoli Zhang
- Department of Injury and Repair, Beijing Neurosurgical Institute, Capital Medical University, Beijing, Beijing, China
| | - Hongying Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, China
| |
Collapse
|
13
|
Toghraie FS, Bayat M, Hosseini MS, Ramezani A. Tumor-infiltrating myeloid cells; mechanisms, functional significance, and targeting in cancer therapy. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01051-y. [PMID: 39998754 DOI: 10.1007/s13402-025-01051-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
14
|
Li Y, Liu F, Cai Q, Deng L, Ouyang Q, Zhang XHF, Zheng J. Invasion and metastasis in cancer: molecular insights and therapeutic targets. Signal Transduct Target Ther 2025; 10:57. [PMID: 39979279 PMCID: PMC11842613 DOI: 10.1038/s41392-025-02148-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 12/24/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
The progression of malignant tumors leads to the development of secondary tumors in various organs, including bones, the brain, liver, and lungs. This metastatic process severely impacts the prognosis of patients, significantly affecting their quality of life and survival rates. Research efforts have consistently focused on the intricate mechanisms underlying this process and the corresponding clinical management strategies. Consequently, a comprehensive understanding of the biological foundations of tumor metastasis, identification of pivotal signaling pathways, and systematic evaluation of existing and emerging therapeutic strategies are paramount to enhancing the overall diagnostic and treatment capabilities for metastatic tumors. However, current research is primarily focused on metastasis within specific cancer types, leaving significant gaps in our understanding of the complex metastatic cascade, organ-specific tropism mechanisms, and the development of targeted treatments. In this study, we examine the sequential processes of tumor metastasis, elucidate the underlying mechanisms driving organ-tropic metastasis, and systematically analyze therapeutic strategies for metastatic tumors, including those tailored to specific organ involvement. Subsequently, we synthesize the most recent advances in emerging therapeutic technologies for tumor metastasis and analyze the challenges and opportunities encountered in clinical research pertaining to bone metastasis. Our objective is to offer insights that can inform future research and clinical practice in this crucial field.
Collapse
Affiliation(s)
- Yongxing Li
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA
- Graduate School of Biomedical Science, Cancer and Cell Biology Program, Baylor College of Medicine, Houston, TX, USA
| | - Qingjin Cai
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lijun Deng
- Department of Medicinal Chemistry, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qin Ouyang
- Department of Medicinal Chemistry, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA.
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
15
|
Wang Z, Liu Z, Lv M, Luan Z, Li T, Hu J. Novel histone modifications and liver cancer: emerging frontiers in epigenetic regulation. Clin Epigenetics 2025; 17:30. [PMID: 39980025 PMCID: PMC11841274 DOI: 10.1186/s13148-025-01838-8] [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: 11/06/2024] [Accepted: 02/08/2025] [Indexed: 02/22/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide, and its onset and progression are closely associated with epigenetic modifications, particularly post-translational modifications of histones (HPTMs). In recent years, advances in mass spectrometry (MS) have revealed a series of novel HPTMs, including succinylation (Ksuc), citrullination (Kcit), butyrylation (Kbhb), lactylation (Kla), crotonylation (Kcr), and 2-hydroxyisobutyrylation (Khib). These modifications not only expand the histone code but also play significant roles in key carcinogenic processes such as tumor proliferation, metastasis, and metabolic reprogramming in HCC. This review provides the first comprehensive analysis of the impact of novel HPTMs on gene expression, cellular metabolism, immune evasion, and the tumor microenvironment. It specifically focuses on their roles in promoting tumor stem cell characteristics, epithelial-mesenchymal transition (EMT), and therapeutic resistance. Additionally, the review highlights the dynamic regulation of these modifications by specific enzymes, including "writers," "readers," and "erasers."
Collapse
Affiliation(s)
- Zhonghua Wang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Ziwen Liu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Mengxin Lv
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Zhou Luan
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Tao Li
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Jinhua Hu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, 250021, Shandong, People's Republic of China.
| |
Collapse
|
16
|
Bryzek D, Gasiorek A, Kowalczyk D, Santocki M, Ciaston I, Dobosz E, Kolaczkowska E, Kjøge K, Kantyka T, Lech M, Potempa B, Enghild JJ, Potempa J, Koziel J. Non-classical neutrophil extracellular traps induced by PAR2-signaling proteases. Cell Death Dis 2025; 16:109. [PMID: 39971938 PMCID: PMC11840154 DOI: 10.1038/s41419-025-07428-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/21/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025]
Abstract
Neutrophil extracellular traps (NETs) are associated with diseases linked to aberrant coagulation. The blood clotting cascade involves a series of proteases, some of which induce NET formation via a yet unknown mechanism. We hypothesized that this formation involves signaling via a factor Xa (FXa) activation of the protease-activated receptor 2 (PAR2). Our findings revealed that NETs can be triggered in vitro by enzymatically active proteases and PAR2 agonists. Intravital microscopy of the liver vasculature revealed that both FXa infusion and activation of endogenous FX promoted NET formation, effects that were prevented by the FXa inhibitor, apixaban. Unlike classical NETs, these protease-induced NETs lacked bactericidal activity and their proteomic signature indicates their role in inflammatory disorders, including autoimmune diseases and carcinogenesis. Our findings suggest a novel mechanism of NET formation under aseptic conditions, potentially contributing to a self-amplifying clotting and NET formation cycle. This mechanism may underlie the pathogenesis of disseminated intravascular coagulation and other aseptic conditions.
Collapse
Affiliation(s)
- Danuta Bryzek
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| | - Anna Gasiorek
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Dominik Kowalczyk
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Michal Santocki
- Department of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Izabela Ciaston
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Ewelina Dobosz
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Elzbieta Kolaczkowska
- Department of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Katarzyna Kjøge
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | - Maciej Lech
- LMU Hospital, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians University, Munich, Germany
| | - Barbara Potempa
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, Kentucky, USA
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan Potempa
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, Kentucky, USA
| | - Joanna Koziel
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
17
|
Zhao JL, Wang MY, Lv YZ, Zhou YJ. Prognostic value of inflammatory markers in predicting recurrence-free survival in gastrointestinal stromal tumor patients: A nomogram-based approach. World J Gastrointest Oncol 2025; 17:94956. [PMID: 39958548 PMCID: PMC11755990 DOI: 10.4251/wjgo.v17.i2.94956] [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: 03/28/2024] [Revised: 10/02/2024] [Accepted: 10/25/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND There are currently no relevant studies at home or abroad that combine inflammatory indicators and nomograms to predict the prognosis of gastrointestinal stromal tumor (GIST) patients after surgery. The purpose of this study was to investigate the predictive value of related inflammatory indicators [systemic immune-inflammation index (SII), neutrophil/lymphocyte ratio (NLR), platelet/lymphocyte ratio (PLR) and monocyte/Lymphocyte ratio (MLR)] in patients undergoing GIST surgery, incorporating relevant risk factors to establish a nomogram prediction model, with the aim of better predicting the prognosis of GIST patients. AIM To explore the relationships between the SII, NLR, PLR, and MLR and postoperative recurrence in patients with GIST. METHODS This study retrospectively included patients who underwent GIST surgery from January 2014 to January 2017 and analyzed the potential relationships between the preoperative SII, NLR, PLR, and MLR and clinicopathological features. The independent risk factors influencing the prognosis of GIST patients were obtained via multivariate regression analysis, and a nomogram model based on the independent risk factors was established. RESULTS Among the 124 GIST patients included in the present study, 31 (25%) experienced recurrence within 5 years. Kaplan-Meier survival analysis revealed a correlation between the MLR and PLR and tumor size (P = 0.016 and P = 0.002, respectively). The preoperative SII, MLR, NLR, and PLR were significantly associated with recurrence-free survival (RFS) (P < 0.05). The multivariate analysis results identified the PLR, MLR, and targeted therapy as independent prognostic factors for patient outcomes. CONCLUSION Preoperative MLR and PLR, which are independent risk factors for GIST recurrence, were correlated with RFS. Nomograms based on the PLR, MLR and targeted therapy can be used for clinical treatment.
Collapse
Affiliation(s)
- Jin-Long Zhao
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Mao-Ying Wang
- Department of Anesthesiology, People’s Hospital of Qingbaijiang District, Chengdu 610300, Sichuan Province, China
| | - Yan-Zhi Lv
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Ye-Jiang Zhou
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| |
Collapse
|
18
|
Aierken Y, Tan K, Liu T, Lv Z. Prognosis and immune infiltration prediction in neuroblastoma based on neutrophil extracellular traps-related gene signature. Sci Rep 2025; 15:5343. [PMID: 39948114 PMCID: PMC11825912 DOI: 10.1038/s41598-025-88608-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
Neuroblastoma (NB) is a malignant tumor originating from the peripheral sympathetic nervous system and high-risk NB patients have a dismal prognosis. Recent studies have underscored the pivotal role of neutrophil extracellular traps (NETs) in the proliferation, metastasis and immune evasion of cancer. To explore the effect of NETs on NB, we have carried out a systematic analysis and showed several findings in the present work. First, expression profiles along with clinical data were analyzed using the training dataset GSE62564 and 36 NETs-related genes were identified to be significantly associated with overall survival. Following LASSO regression analysis, 11 genes were enrolled to construct the NETs signature, which exhibited a robust predictive capability for overall survival with exhibiting high AUC values within the training set. Validation cohorts confirmed a similar predictive efficacy. Next, NB patients were classified into subgroups based on median risk scores and differentially expressed genes were analyzed. Furthermore, the study performed comprehensive analyses encompassing functional enrichment, immune infiltration and drug sensitivity. Enrichment analysis revealed that the high-risk NBs with high-risk score displayed characteristics of oncogenic malignancy, poor prognosis and immunosuppression. Notably, the risk score exhibited a strong correlation with infiltration levels of various immune cells and the sensitivity to anti-cancer drugs, and was further recognized as an independent prognostic factor for NB patients. In summary, our study elucidates a novel NETs-related gene signature comprising 11 genes, which serves a reliable predictor for NB prognosis.
Collapse
Affiliation(s)
- Yeerfan Aierken
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Kezhe Tan
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Tao Liu
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Zhibao Lv
- Department of General Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China.
| |
Collapse
|
19
|
Dong Y, Wang F, Deng J, Tong T, Chen X, Wu L, Wang Y, Kang M, Xu Y, Shi G, Zhu L. Prognostic value of serum inflammatory markers in patients with esophageal squamous cell carcinoma undergoing surgery: a two-center retrospective cohort study. BMC Cancer 2025; 25:213. [PMID: 39920653 PMCID: PMC11804429 DOI: 10.1186/s12885-025-13600-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 01/28/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND AND PURPOSE The combined assessment of the neutrophil-platelet score and prognostic nutritional index is yet to be applied in evaluating the prognosis of patients following an operation for esophageal cancer. This study aimed to identify independent prognostic factors for patients undergoing operation for esophageal cancer and to construct a nomogram model for predicting median overall survival (mOS). METHODS A cohort of 660 patients with esophageal cancer from two clinical centers in China was recruited, comprising a training cohort (n = 511) and a validation cohort (n = 149). Survival rates were compared using Kaplan-Meier curves and the log-rank test. The Cox regression model was used to identify independent prognostic factors. RESULTS The Multi-variable Cox regression analysis revealed that tumor, node, metastasis stage, smoking status, neutrophil-platelet score, and prognostic nutritional index were independent postoperative prognostic factors for patients with esophageal cancer (P < 0.05). The mOS (129.0 months, 95% confidence interval (CI) not reached) was significantly higher in patients with a low neutrophil-platelet score than those with a high score (43.0 months, 28.1-57.9; P < 0.05). Conversely, the mOS (42.0 months, 95% CI 6.7-55.2) was lower in the low prognostic nutritional index score group than the high score group (125.0 months, 95% CI 96.4-153.6; P < 0.05). Evaluation metrics, including the concordance index (0.69 in the training group and 0.70 in the validation group), receiver operator characteristic curve analysis (In the training cohort, the AUCs for predicting 1-, 3-, 5- and 10-year overall survival rates using the nomogram were 76.19%, 69.07%, 70.12% and 68.17%, respectively; in the validation cohort, the corresponding predicted values were 76.66%, 77.18%, 75.04% and 79.57%, respectively), calibration curve, and decision curve analysis, demonstrated the clinical utility of the constructed prediction model. CONCLUSIONS Serving as systemic inflammatory biomarkers, the neutrophil-platelet score and prognostic nutritional index offer valuable prognostic insights for patients with esophageal cancer, augmenting predictive capabilities.
Collapse
Affiliation(s)
- Yaqin Dong
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Hefei, Anhui, 230022, People's Republic of China
| | - Fan Wang
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Hefei, Anhui, 230022, People's Republic of China.
| | - Jiaying Deng
- Department of Radiation Oncology, the Fudan University Shanghai Cancer Center, Shanghai, China
| | - Tong Tong
- Department of Radiology, the Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiangxun Chen
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Hefei, Anhui, 230022, People's Republic of China
| | - Liming Wu
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Hefei, Anhui, 230022, People's Republic of China
| | - Yichun Wang
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Hefei, Anhui, 230022, People's Republic of China
| | - Mei Kang
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Hefei, Anhui, 230022, People's Republic of China
| | - Yutong Xu
- Department of Clinical Medicine, the First College of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Guangjie Shi
- Department of Medical Imaging, First College of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Liyang Zhu
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Hefei, Anhui, 230022, People's Republic of China.
| |
Collapse
|
20
|
Desharnais L, Sorin M, Rezanejad M, Liu B, Karimi E, Atallah A, Swaby AM, Yu MW, Doré S, Hartner S, Fiset B, Wei Y, Kadang B, Rayes R, Joubert P, Camilleri-Broët S, Fiset PO, Quail DF, Spicer JD, Walsh LA. Spatially mapping the tumour immune microenvironments of non-small cell lung cancer. Nat Commun 2025; 16:1345. [PMID: 39905080 PMCID: PMC11794701 DOI: 10.1038/s41467-025-56546-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/20/2025] [Indexed: 02/06/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths. An enhanced understanding of the immune microenvironments within these tumours may foster more precise and efficient treatment, particularly for immune-targeted therapies. The spatial architectural differences between lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) are relatively unexplored. Here, we applied imaging mass cytometry to a balanced cohort of LUAD and LUSC patients, matched for clinical factors such as age, sex, and smoking history, to analyze 204 histopathology images of tumours from 102 individuals with non-small cell lung cancer (NSCLC). By analyzing interactions and broader cellular networks, we interrogate the tumour microenvironment to understand how immune cells are spatially organized in clinically matched adenocarcinoma and squamous cell carcinoma subsets. This spatial analysis revealed distinct patterns of immune cell aggregation, particularly among macrophage populations, that correlated with patient prognosis differentially in adenocarcinoma and squamous cell carcinoma, suggesting potential new strategies for therapeutic intervention. Our findings underscore the importance of analyzing NSCLC histological subtypes separately when investigating the spatial immune landscape, as microenvironmental characteristics and cellular interactions differed by subtype. Recognizing these distinctions is essential for designing precision therapies tailored to each subtype's unique immune architecture, ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Lysanne Desharnais
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Mark Sorin
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Morteza Rezanejad
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Bridget Liu
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Elham Karimi
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Aline Atallah
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Anikka M Swaby
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Miranda W Yu
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Samuel Doré
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Saskia Hartner
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Benoit Fiset
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Yuhong Wei
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Baharak Kadang
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Roni Rayes
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Philippe Joubert
- Département de biologie moléculaire, biochimie médicale et de pathologie, Laval University, Québec City, QC, Canada
| | | | | | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Jonathan D Spicer
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.
- Department of Surgery, McGill University Health Center, Montreal, QC, Canada.
| | - Logan A Walsh
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.
- Department of Human Genetics, McGill University, Montreal, QC, Canada.
| |
Collapse
|
21
|
Tao L, Guo Y, Zhu M, Wang H, Liu Y, Wang W. Cell Type-Preferential Expression of Peptidylarginine Deiminase 4 and p53-Dependent Therapeutic Vulnerabilities in Gastric Cancer. Dig Dis Sci 2025; 70:566-580. [PMID: 39661279 DOI: 10.1007/s10620-024-08734-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND AND AIMS Previous studies have demonstrated that peptidylarginine deiminase 4 (PAD4) functions as a suppressor, promoter, or both in cancer pathogenesis and therapeutic outcomes. Although PAD4 expression has been proposed to be one of the molecular features of gastric cancer (GC), the biological basis of PAD4 in GC progression and chemotherapy has not been formally established. METHODS Cell type-preferential expression of PAD4 was analyzed in both preclinical and clinical models. The colocalization of PAD4 expression and tumor-infiltrating neutrophils in GC patients was evaluated by immunofluorescence assay. The effects of forced expression of PAD4 on GC cell proliferation were evaluated both in vitro and in vivo. The effects of forced expression of PAD4 on the cytotoxicity of 5-fluorouracil and oxaliplatin were performed by EdU, Annexin V/PI, and comet assays. Co-immunoprecipitation assay was used to investigate the endogenous and exogenous interaction between PAD4 and p53. To investigate whether p53 participated in the chemopotentiating effects of PAD4, small interfering RNA (siRNA)-mediated knockdown of p53 was conducted in PAD4-overexpressing GC cells. RESULTS Contrary to the previous report, we initially observed that PAD4 was underexpressed in GC patients and presented as a favorable prognostic factor across the TCGA cohort. Interestingly, the normal gastric epithelial cell line GES-1 exhibited low-level expression of PAD4. In comparison, PAD4 was not detected in GC cell lines, including AGS, HGC-27, and MKN-45. Using an orthotopic mouse model of GC, we found that PAD4 was surprisingly abundant in HGC-27 cell line-derived xenografts. Immunofluorescence staining for PAD4 and neutrophil elastase indicated that PAD4 was mainly expressed in neutrophils but not in GC cells. PAD4 expression was upregulated in all-trans retinoic acid-induced neutrophil-like dHL-60 cells, which were used as a positive control for PAD4 expression. Surprisingly, the enforced expression of PAD4 suppressed the proliferation of GC cells in vitro and in vivo. In addition, cells harboring high PAD4 expression were more susceptible to G1/S boundary arrest, apoptotic death, and DNA damage by regulating p53 target proteins. Mechanistically, PAD4 might affect p53 function through physical interaction with p53, and the chemopotentiating effects of PAD4 could be compromised by p53 knockdown. CONCLUSIONS PAD4 appeared to be constitutively expressed in tumor-infiltrating neutrophils but not in GC cells. Our findings highlight unique roles of PAD4, in which origin-dependent PAD4 might work complementarily on the progression and treatment vulnerability of GC.
Collapse
Affiliation(s)
- Li Tao
- Department of Pharmacy, School of Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China
- The State Administration of Traditional Chinese Medicine Key Laboratory of Toxic Pathogens-Based Therapeutic Approaches of Gastric Cancer, Yangzhou University, 136 Jiangyang Avenue, Building 41, Room 301, Yangzhou, 225009, Jiangsu, China
| | - Yajie Guo
- Department of Pharmacy, School of Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China
- The State Administration of Traditional Chinese Medicine Key Laboratory of Toxic Pathogens-Based Therapeutic Approaches of Gastric Cancer, Yangzhou University, 136 Jiangyang Avenue, Building 41, Room 301, Yangzhou, 225009, Jiangsu, China
| | - Miao Zhu
- The State Administration of Traditional Chinese Medicine Key Laboratory of Toxic Pathogens-Based Therapeutic Approaches of Gastric Cancer, Yangzhou University, 136 Jiangyang Avenue, Building 41, Room 301, Yangzhou, 225009, Jiangsu, China
| | - Haibo Wang
- The State Administration of Traditional Chinese Medicine Key Laboratory of Toxic Pathogens-Based Therapeutic Approaches of Gastric Cancer, Yangzhou University, 136 Jiangyang Avenue, Building 41, Room 301, Yangzhou, 225009, Jiangsu, China
| | - Yanqing Liu
- The State Administration of Traditional Chinese Medicine Key Laboratory of Toxic Pathogens-Based Therapeutic Approaches of Gastric Cancer, Yangzhou University, 136 Jiangyang Avenue, Building 41, Room 301, Yangzhou, 225009, Jiangsu, China.
| | - Weimin Wang
- The State Administration of Traditional Chinese Medicine Key Laboratory of Toxic Pathogens-Based Therapeutic Approaches of Gastric Cancer, Yangzhou University, 136 Jiangyang Avenue, Building 41, Room 301, Yangzhou, 225009, Jiangsu, China
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, 75 Tongzhenguan Avenue, Yixing, 214200, Jiangsu, China
| |
Collapse
|
22
|
Melzer YF, Fergen NL, Mess C, Stadler JC, Geidel G, Schwietzer YA, Kött J, Pantel K, Schneider SW, Utikal J, Wladykowski E, Vidal-Y-Sy S, Bauer AT, Gebhardt C. Evaluation of S100A8/A9 and neutrophils as prognostic markers in metastatic melanoma patients under immune-checkpoint inhibition. Transl Oncol 2025; 52:102224. [PMID: 39700646 PMCID: PMC11718343 DOI: 10.1016/j.tranon.2024.102224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 12/21/2024] Open
Abstract
Immune-checkpoint inhibitors (ICIs) have revolutionized melanoma treatment, yet approximately half of patients do not respond to these therapies. Identifying prognostic biomarkers is crucial for treatment decisions. Our retrospective study assessed liquid biopsies and tumor tissue analyses for two potential biomarkers: danger-associated molecular pattern (DAMP) S100A8/A9 and its source, neutrophils. In 43 metastatic unresected stage III/IV melanoma patients, elevated serum levels of S100A8/A9 and neutrophils before and during ICI treatment correlated with worse outcomes. Furthermore, in 113 melanoma patients, neutrophil expression in the tumor microenvironment (TME) was associated with relapse and reduced survival. Measuring S100A8/A9 and neutrophils could enhance immunotherapy monitoring by predicting impaired clinical outcomes and non-response to ICIs. Serum S100A8/A9 levels and neutrophil counts at baseline (T0) and during treatment (T3) correlated with reduced progression-free survival (PFS). Elevated S100A8/A9 levels at T0 and T3 negatively impacted overall survival (OS). Notably, neutrophil infiltration was more prevalent in primary melanomas than in nevi and metastases, and its presence in primary melanomas was linked to poorer survival. S100A8/A9 serum levels, neutrophil counts, and tumor-associated neutrophil infiltration represent promising biomarkers for predicting treatment response and clinical outcomes in melanoma patients receiving ICIs. SIGNIFICANCE: These findings underscore the critical need for reliable biomarkers in melanoma research, particularly for predicting responses to immune-checkpoint inhibitors (ICIs). Identifying S100A8/A9 levels and neutrophil infiltration as potential indicators of treatment outcomes offers valuable insights for personalized therapy decisions. By enhancing monitoring and prognosis assessment, these biomarkers contribute to refining treatment strategies, ultimately improving patient care and outcomes. This research bridges gaps in understanding melanoma response mechanisms and highlights avenues for further investigation into immune-related markers, fostering advancements in precision medicine for melanoma patients.
Collapse
Affiliation(s)
- Yasmin F Melzer
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Nadine L Fergen
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Christian Mess
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Julia-Christina Stadler
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Glenn Geidel
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Ysabel A Schwietzer
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Julian Kött
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Klaus Pantel
- Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Department of Tumor Biology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Stefan W Schneider
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.
| | - Ewa Wladykowski
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Sabine Vidal-Y-Sy
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Alexander T Bauer
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.
| | - Christoffer Gebhardt
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| |
Collapse
|
23
|
Zhu M, Jia R, Zhang X, Xu P. The success of the tumor immunotherapy: neutrophils from bench to beside. Front Immunol 2025; 16:1524038. [PMID: 39925807 PMCID: PMC11802522 DOI: 10.3389/fimmu.2025.1524038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/07/2025] [Indexed: 02/11/2025] Open
Abstract
The present immune therapy was focused on the immune checkpoint blockade or Chimeric Antigen Receptor T-Cell Immunotherapy (CART) transfer, but how to activate the innate immune system to antitumor still lags out. Neutrophils are the most abundant circulating leukocytes in human, and heterogeneous neutrophils have been increasingly recognized as important players in tumor progression. They play double "edge-sward" by either supporting or suppressing the tumor growth, including driving angiogenesis, extracellular matrix remodeling to promote tumor growth, participating in antitumor adaptive immunity, or killing tumor cells directly to inhibit the tumor growth. The complex role of neutrophils in various tumors depends on the tumor microenvironment (TME) they are located, and emerging evidence has suggested that neutrophils may determine the success of tumor immunotherapy in the context of the immune checkpoint blockade, innate immune training, or drug-loaded extracellular microvesicles therapy, which makes them become an exciting target for tumor immunotherapy, but still with challenges. Here, we summarize the latest insights on how to activate neutrophils in antitumor immunity and discuss the advances of neutrophil-targeted immunotherapy strategies.
Collapse
Affiliation(s)
- Meng Zhu
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ru Jia
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaojie Zhang
- Department of Obstetrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Pingwei Xu
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
24
|
Wang WY, Chen Y, Chen Q, Sun HW, Niu NX, Li HH, Cao YD, Bai YX, Li X. Nomogram-derived immune-inflammation-nutrition score could act as a novel prognostic indicator for patients with head and neck squamous cell carcinoma. Front Immunol 2025; 15:1500525. [PMID: 39877368 PMCID: PMC11772279 DOI: 10.3389/fimmu.2024.1500525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025] Open
Abstract
Aim This study aims to create and validate a novel systematic immune-inflammation-nutrition (SIIN) score to provide a non-invasive and accurate prognostic tool for head and neck squamous cell carcinoma (HNSCC) patients. Methods 259 participants diagnosed with HNSCC from the First Affiliated Hospital of Xi'an Jiaotong University between 2008 and 2017 was included in this retrospective study. Patients were assigned to training (n=181) and validation (n=78) sets. A LASSO Cox regression model was employed to identify significant biomarkers for constructing a SIIN nomogram and to create SIIN score from this nomogram. The prognostic accuracy of the SIIN score was assessed by exploiting receiver operating characteristic (ROC) analysis, Kaplan-Meier survival analysis, Cox proportional hazard regression models, calibration and DCA curves. Results The SIIN score was formulated based on six biomarkers-platelet-lymphocyte ratio (PLR), prognostic nutritional index (PNI), systemic immune-inflammation index (SII), albumin-bilirubin index (ALBI), fibrinogen (FIB) and monocyte count-identified by LASSO regression analysis. (1)The SIIN score demonstrated superior predictive value, achieving area under the ROC curve (AUC) values of 0.736 and 0.700 for 3- and 5-year OS. For recurrence-free survival (RFS), the AUC values were 0.752 for 3-year and 0.701 5-year RFS, as assessed in the training set. Validated as an independent prognostic factor in both cohorts, the SIIN score showed strong correlation with adverse clinicopathological outcomes. Conclusion The SIIN score is a promising prognostic tool that integrates immune, inflammatory, and nutritional factors for predicting clinical outcomes in HNSCC patients. It offers enhanced predictive accuracy compared to existing markers and has the potential to guide personalized treatment strategies and clinical decision-making.
Collapse
Affiliation(s)
- Wen-Yan Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yue Chen
- Center for Gut Microbiome Research, Med-X Institute Centre, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Qian Chen
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Hong-Wei Sun
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Nuo-Xuan Niu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Hong-Hui Li
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yu-Dan Cao
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yan-Xia Bai
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xiang Li
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Center for Gut Microbiome Research, Med-X Institute Centre, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
25
|
Ng M, Cerezo-Wallis D, Ng LG, Hidalgo A. Adaptations of neutrophils in cancer. Immunity 2025; 58:40-58. [PMID: 39813993 DOI: 10.1016/j.immuni.2024.12.009] [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: 09/02/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
There is a renewed interest in neutrophil biology, largely instigated by their prominence in cancer. From an immunologist's perspective, a conceptual breakthrough is the realization that prototypical inflammatory, cytotoxic leukocytes can be tamed to promote the survival and growth of other cells. This has sparked interest in defining the biological principles and molecular mechanisms driving the adaptation of neutrophils to cancer. Yet, many questions remain: is this adaptation mediated by reprogramming mature neutrophils inside the tumoral mass, or rather by rewiring granulopoiesis in the bone marrow? Why, in some instances, are neutrophils beneficial and in others detrimental to cancer? How many different functional programs can be induced in neutrophils by tumors, and is this dependent on the type of tumor? This review summarizes what we know about these questions and discusses therapeutic strategies based on our incipient knowledge of how neutrophils adapt to cancer.
Collapse
Affiliation(s)
- Melissa Ng
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore, Singapore.
| | - Daniela Cerezo-Wallis
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Lai Guan Ng
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Andres Hidalgo
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
26
|
Lee W, Ko SY, Akasaka H, Weigert M, Lengyel E, Naora H. Neutrophil extracellular traps promote pre-metastatic niche formation in the omentum by expanding innate-like B cells that express IL-10. Cancer Cell 2025; 43:69-85.e11. [PMID: 39753138 PMCID: PMC11732717 DOI: 10.1016/j.ccell.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 08/31/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025]
Abstract
Disseminated cancer cells in the peritoneal fluid often colonize omental fat-associated lymphoid clusters but the mechanisms are unclear. Here, we identify that innate-like B cells accumulate in the omentum of mice and women with early-stage ovarian cancer concomitantly with the extrusion of chromatin fibers by neutrophils called neutrophil extracellular traps (NETs). Studies using genetically modified NET-deficient mice, pharmacologic inhibition of NETs, and adoptive B cell transfer show that NETs induce expression of the chemoattractant CXCL13 in the pre-metastatic omentum, stimulating recruitment of peritoneal innate-like B cells that in turn promote expansion of regulatory T cells and omental metastasis through producing interleukin (IL)-10. Ex vivo studies show that NETs elicit IL-10 production in innate-like B cells by inactivating SHP-1, a phosphatase that inhibits B cell activation pathways, and by generating reactive oxygen species. These findings reveal that NETs alter immune cell dynamics in the pre-metastatic omentum, rendering this niche conducive for colonization.
Collapse
Affiliation(s)
- WonJae Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Song Yi Ko
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hironari Akasaka
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Melanie Weigert
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, IL 60637, USA
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, IL 60637, USA
| | - Honami Naora
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
27
|
Ma C, Li Y, Li M, Lv C, Tian Y. Targeting immune checkpoints on myeloid cells: current status and future directions. Cancer Immunol Immunother 2025; 74:40. [PMID: 39751898 PMCID: PMC11699031 DOI: 10.1007/s00262-024-03856-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/07/2024] [Indexed: 01/04/2025]
Abstract
Myeloid cells accumulate extensively in most tumors and play a critical role in immunosuppression of the tumor microenvironment (TME). Like T cells, myeloid cells also express immune checkpoint molecules, which induce the immunosuppressive phenotype of these cells. In this review, we summarize the tumor-promoting function and immune checkpoint expression of four types of myeloid cells: macrophages, neutrophils, dendritic cells, and myeloid-derived suppressor cells, which are the main components of the TME. By summarizing the research status of myeloid checkpoints, we propose that blocking immune checkpoints on myeloid cells might be an effective strategy to reverse the immunosuppressive status of the TME. Moreover, combining nanotechnology, cellular therapy, and bispecific antibodies to achieve precise targeting of myeloid immune checkpoints can help to avoid the adverse effects of systemic administration, ultimately achieving a balance between efficacy and safety in cancer therapy.
Collapse
Affiliation(s)
- Chuhan Ma
- Department of General Surgery, Shengjing Hospital of China Medical University, ShenyangLiaoning Province, 110004, China
| | - Yang Li
- Department of General Surgery, Shengjing Hospital of China Medical University, ShenyangLiaoning Province, 110004, China
| | - Min Li
- Department of Mammary Gland, Dalian Women and Children's Medical Center (Group), DalianLiaoning Province, 116000, China
| | - Chao Lv
- Department of General Surgery, Shengjing Hospital of China Medical University, ShenyangLiaoning Province, 110004, China.
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, ShenyangLiaoning Province, 110004, China.
| |
Collapse
|
28
|
Rivas CH, Liu F, Zhang XHF. The Roles of Myeloid Cells in Breast Cancer Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:397-412. [PMID: 39821035 DOI: 10.1007/978-3-031-70875-6_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
This chapter reviews tumor-associated myeloid cells, including macrophages, neutrophils, and other innate immune cells, and their multifaceted roles in supporting breast cancer progression and metastasis. In primary tumors, myeloid cells play key roles in promoting tumor epithelial-mesenchymal transition (EMT) and invasion. They can facilitate intravasation (entry into the bloodstream) and colonization, disrupting the endothelial cell layer and reshaping the extracellular matrix. They can also stimulate angiogenesis, suppress immune cell responses, and enhance cancer cell adaptability. In the bloodstream, circulating myeloid cells enable the survival of disseminated tumor cells via immunosuppressive effects and physical shielding. At the metastatic sites, they prime the premetastatic niche, facilitate tumor cell extravasation, and support successful colonization and outgrowth. Mechanistically, myeloid cells enhance cancer cell survival, dormancy escape, proliferation, and mesenchymal-epithelial transition (MET). Nonetheless, substantial gaps in our understanding persist regarding the functional and spatiotemporal diversity, as well as the evolutionary patterns, of myeloid cells during metastatic progression. Myeloid cell plasticity and differential responses to therapies present key barriers to successful treatments. Identifying specific pro-tumoral myeloid cell subpopulations and disrupting their interactions with cancer cells represent promising therapeutic opportunities. Emerging evidence suggests combining immunomodulators or stromal normalizers with conventional therapies could help overcome therapy-induced immunosuppression and improve patient outcomes. Overall, further elucidating myeloid cell heterogeneity and function throughout the process of breast cancer progression and metastasis will enable more effective therapeutic targeting of these critical stromal cells.
Collapse
Affiliation(s)
- Charlotte Helena Rivas
- Cancer and Cell Biology Program, Graduate School of Biomedical Sciences, San Antonio, TX, USA
| | - Fengshuo Liu
- Cancer and Cell Biology Program, Graduate School of Biomedical Sciences, San Antonio, TX, USA
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Berkeley, CA, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
29
|
Ferkel SAM, Holman EA, Sojwal RS, Rubin SJS, Rogalla S. Tumor-Infiltrating Immune Cells in Colorectal Cancer. Neoplasia 2025; 59:101091. [PMID: 39642846 DOI: 10.1016/j.neo.2024.101091] [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: 11/07/2024] [Accepted: 11/18/2024] [Indexed: 12/09/2024]
Abstract
Colorectal cancer encompasses a heterogeneous group of malignancies that differ in pathophysiological mechanisms, immune response and infiltration, therapeutic response, and clinical prognosis. Numerous studies have highlighted the clinical relevance of tumor-infiltrating immune cells among different types of colorectal tumors yet vary in cell type definitions and cell identification strategies. The distinction of immune signatures is particularly challenging when several immune subtypes are involved but crucial to identify novel intercellular mechanisms within the tumor microenvironment. In this review, we compile human and non-human studies on tumor-infiltrating immune cells and provide an overview of immune subtypes, their pathophysiological functions, and their prognostic role in colorectal cancer. We discuss how differentiating immune signatures can guide the development of immunotherapeutic targets and personalized treatment regimens. We analyzed comprehensive human protein biomarker profiles across the entire immune spectrum to improve interpretability and application of tumor studies and to ultimately enhance immunotherapy and advance precision medicine for colorectal cancer patients.
Collapse
Affiliation(s)
- Sonia A M Ferkel
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Elizabeth A Holman
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Raoul S Sojwal
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Samuel J S Rubin
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Stephan Rogalla
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA.
| |
Collapse
|
30
|
Andraska EA, Denorme F, Kaltenmeier C, Arivudainabi A, Mihalko EP, Dyer M, Annarapu GK, Zarisfi M, Loughran P, Ozel M, Williamson K, Mota Alvidrez RI, Thomas K, Shiva S, Shea SM, Steinman RA, Campbell RA, Rosengart MR, Neal MD. Alterations in visible light exposure modulate platelet function and regulate thrombus formation. J Thromb Haemost 2025; 23:123-138. [PMID: 39299611 DOI: 10.1016/j.jtha.2024.08.020] [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: 05/05/2024] [Revised: 08/20/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Variations in light exposure are associated with changes in inflammation and coagulation. The impact of light spectra on venous thrombosis (VT) and arterial thrombosis is largely unexplored. OBJECTIVES To investigate the impact of altering light spectrum on platelet function in thrombosis. METHODS Wild-type C57BL/6J mice were exposed to ambient (micewhite, 400 lux), blue (miceblue, 442 nm, 1400 lux), or red light (micered, 617 nm, 1400 lux) with 12:12 hour light:dark cycle for 72 hours. After 72 hours of light exposure, platelet aggregation, activation, transcriptomic, and metabolomic changes were measured. The ability of released products of platelet activation to induce thrombosis-generating neutrophil extracellular trap formation was quantified. Subsequent thrombosis was measured using murine models of VT and stroke. To translate our findings to human patients, light-filtering cataract patients were evaluated over an 8-year period for rate of venous thromboembolism with multivariable logistic regression clustered by hospital. RESULTS Exposure to long-wavelength red light resulted in reduced platelet aggregation and activation. RNA-seq analysis demonstrated no significant transcriptomic changes between micered and micewhite. However, there were global metabolomic changes in platelets from micered compared with micewhite. Releasate from activated platelets resulted in reduced neutrophil extracellular trap formation. Micered also had reduced VT weight and brain infarct size following stroke. On subgroup analysis of cataract patients, patients with a history of cancer had a lower lifetime risk of venous thromboembolism after implantation with lenses that filter low-wavelength light. CONCLUSION Light therapy may be a promising approach to thrombus prophylaxis by specifically targeting the intersection between innate immune function and coagulation.
Collapse
Affiliation(s)
- Elizabeth A Andraska
- Department of Surgery, Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA.
| | - Frederik Denorme
- Department of Emergency Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Christof Kaltenmeier
- University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA; MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital, Washington, DC, USA
| | | | - Emily P Mihalko
- Department of Surgery, Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mitchell Dyer
- Division of Vascular and Endovascular Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Gowtham K Annarapu
- Pittsburgh Heart, Lung, Blood, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mohammadreza Zarisfi
- Department of Surgery, Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Patricia Loughran
- Department of Surgery, Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA
| | - Mehves Ozel
- Department of Surgery, Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kelly Williamson
- Department of Surgery, Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Kimberly Thomas
- Vitalant Research Institute, Denver, Colorado, USA; Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sruti Shiva
- Division of Classical Hematology, Department of Medicine, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Susan M Shea
- Department of Surgery, Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Richard A Steinman
- University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA; Division of Hematology and Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert A Campbell
- Department of Emergency Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Matthew R Rosengart
- Division of Acute and Critical Care Surgery, Department of Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Matthew D Neal
- Department of Surgery, Trauma and Transfusion Medicine Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
31
|
Wang Y, Liang S, Hong Q, Mu J, Wu Y, Li K, Li Y, Wu Y, Lou X, Xu D, Cui W. Construction of a neutrophil extracellular trap formation-related gene model for predicting the survival of lung adenocarcinoma patients and their response to immunotherapy. Transl Lung Cancer Res 2024; 13:3407-3425. [PMID: 39830760 PMCID: PMC11736607 DOI: 10.21037/tlcr-24-463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 11/22/2024] [Indexed: 01/22/2025]
Abstract
Background Lung adenocarcinoma (LUAD) is associated with high morbidity and mortality rates. Increasing evidence indicates that neutrophil extracellular traps (NETs) play a critical role in tumor progression, metastasis and immunosuppression in the LUAD tumor microenvironment (TME). Nevertheless, the use of NET formation-related genes (NFRGs) to predict LUAD patient survival and response to immunotherapy has not been explored. Therefore, this study aimed to construct a NFRGs-based prognostic signature for stratifying LUAD patients and informing individualized management strategies. Methods The cell composition of the LUAD TME was investigated using the single-cell sequencing data in Single-Cell Lung Cancer Atlas (LuCA). NFRGs were identified to construct a prognostic signature based on The Cancer Genome Atlas (TCGA) cohort which was validated in the Gene Expression Omnibus (GEO) dataset. The univariate Cox and least absolute shrinkage and selection operator (LASSO) Cox regression models, receiver operating characteristic (ROC) and Brier Score were applied to assess the prognostic model. A nomogram was established to facilitate the clinical application of the risk score. The Estimation of STromal and Immune cells in MAlignant Tumor tissues (ESTIMATE) and Tumor Immune Dysfunction and Exclusion (TIDE) algorithm were utilized to assess the TME and predict immunotherapy response. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was applied to quantify the expression levels of four NFRGs in LUAD paired tissue samples. Results Single‑cell RNA sequence analysis showed the importance of neutrophils in LUAD TME. We developed and validated a 4-NFRG (CAT, CTSG, ENO1, TLR2) prognostic signature based on TCGA and GEO cohorts, which stratified patients into high-risk and low-risk groups. Univariate and multivariate analyses showed that our risk model could independently predict the survival of LUAD patients. Patients in the low-risk group exhibited a more active immune microenvironment, lower TIDE scores, lower half-maximal inhibitory concentration (IC50) values and higher immune checkpoint molecule expression. Our risk signature could serve as a biomarker for predicting immunotherapeutic benefits. Conclusions We developed a novel prognostic signature for LUAD patients based on NFRGs and emphasized the critical role of this signature in predicting LUAD patient survival and immunotherapy response.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuang Liang
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian Hong
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Juwei Mu
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuxin Wu
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kexin Li
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiling Li
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Wu
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoying Lou
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Danfei Xu
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Cui
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
32
|
Cao X, Lan Q, Xu H, Liu W, Cheng H, Hu X, He J, Yang Q, Lai W, Chu Z. Granulocyte-like myeloid-derived suppressor cells: The culprits of neutrophil extracellular traps formation in the pre-metastatic niche. Int Immunopharmacol 2024; 143:113500. [PMID: 39510029 DOI: 10.1016/j.intimp.2024.113500] [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: 08/10/2024] [Revised: 10/05/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024]
Abstract
Neutrophil extracellular traps (NETs) have been shown to exhibit chemotactic effects on circulating tumor cells at metastatic sites, promoting the progression of colorectal cancer liver metastasis (CRLM). However, the origin and factors contributing to the formation of NETs (NETosis) in the pre-metastatic niche (PMN) of target organs remain unclear. In this study, we investigated the relationship between phosphatase of regenerating liver-3 (PRL-3), myeloid-derived suppressor cells (MDSCs), neutrophils, and NETs through a retrospective clinical cohort study and a mouse model of CRLM. Our clinical findings revealed associations between PRL-3 expression and the infiltration of neutrophils and MDSCs in CRLM patients. Moreover, NETosis emerged as a robust indicator of poor prognosis for overall survival in these patients. In CRLM models, PRL-3 overexpression enhanced both primary tumor growth and liver metastasis. We found that the first week after tumor cell implantation appeared to be a crucial period for PMN formation, with notable infiltration of neutrophils, MDSCs, and NETosis during this time. Notably, co-culturing neutrophils with MDSCs induced NETosis in vitro, particularly with granulocyte-like MDSCs (Gr-MDSCs), the predominant subtype of infiltrating MDSCs. In summary, our findings elucidate the likely origin of NETs in the PMN during CRLM development and underscore the significant influence of PRL-3. These findings may offer potential immunotherapeutic targets for patients at risk of developing CRLM, warranting further investigation in clinical settings.
Collapse
Affiliation(s)
- Xintong Cao
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong Province, China
| | - Qiusheng Lan
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong Province, China
| | - Heyang Xu
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong Province, China
| | - Wentao Liu
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
| | - Huaxi Cheng
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
| | - Xinwen Hu
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong Province, China
| | - Jiehua He
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong Province, China
| | - Qiong Yang
- Department of Medical Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China.
| | - Wei Lai
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China.
| | - Zhonghua Chu
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China.
| |
Collapse
|
33
|
Zeng F, Shao Y, Wu J, Luo J, Yue Y, Shen Y, Wang Y, Shi Y, Wu D, Cata JP, Yang S, Zhang H, Miao C. Tumor metastasis and recurrence: The role of perioperative NETosis. Cancer Lett 2024; 611:217413. [PMID: 39725150 DOI: 10.1016/j.canlet.2024.217413] [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: 04/29/2024] [Revised: 12/11/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Although surgical resection of tumor mass remains the mainstay of curative therapeutic management for solid tumors, accumulating studies suggest that these procedures promote tumor recurrence and metastasis. Regarded as the first immune cells to fight against infectious or inflammatory insults from surgery, neutrophils along with their ability of neutrophil extracellular traps (NETs) production has attracted much attention. A growing body of evidence suggests that NETs promote cancer metastasis by stimulating various stages, including local invasion, colonization, and growth. Therefore, we discussed the mechanism of NETosis induced by surgical stress and tumor cells, and the contribution of NETs on tumor metastasis: aid in the tumor cell migration and proliferation, evasion of immune surveillance, circulating tumor cell adhesion and establishment of a metastatic niche. Lastly, we summarized existing NET-targeting interventions, offering recent insights into potential targets for clinical intervention.
Collapse
Affiliation(s)
- Fu Zeng
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yuwen Shao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jingyi Wu
- Department of Anesthesiology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Jingwen Luo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ying Yue
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yang Shen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yanghanzhao Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yuxin Shi
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Dan Wu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Juan P Cata
- Department of Anesthesiology and Perioperative Medicine, The University of Texas-MD Anderson Cancer Center, Houston, TX, USA; Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA
| | - Shuofei Yang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Pujian Road 160, Shanghai, 200127, China.
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| |
Collapse
|
34
|
Grinat J, Shriever NP, Christophorou MA. Fantastic proteins and where to find them - histones, in the nucleus and beyond. J Cell Sci 2024; 137:jcs262071. [PMID: 39704565 PMCID: PMC11827605 DOI: 10.1242/jcs.262071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
Animal genomes are packaged into chromatin, a highly dynamic macromolecular structure of DNA and histone proteins organised into nucleosomes. This accommodates packaging of lengthy genomic sequences within the physical confines of the nucleus while also enabling precise regulation of access to genetic information. However, histones existed before chromatin and have lesser-known functions beyond genome regulation. Most notably, histones are potent antimicrobial agents, and the release of chromatin to the extracellular space is a defence mechanism nearly as ancient and widespread as chromatin itself. Histone sequences have changed very little throughout evolution, suggesting the possibility that some of their 'non-canonical' functions are at play in parallel or in concert with their genome regulatory functions. In this Review, we take an evolutionary perspective of histone, nuclear chromatin and extracellular chromatin biology and describe the known extranuclear and extracellular functions of histones. We detail molecular mechanisms of chromatin release and extracellular chromatin sensing, and we discuss their roles in physiology and disease. Finally, we present evidence and give a perspective on the potential of extracellular histones to act as bioactive, cell modulatory factors.
Collapse
|
35
|
Hussen BM, Rasul MF, Faraj GSH, Abdullah SR, Sulaiman SH, Pourmoshtagh H, Taheri M. Role of microRNAs in neutrophil extracellular trap formation and prevention: Systematic narrative review. Mol Cell Probes 2024; 78:101986. [PMID: 39389272 DOI: 10.1016/j.mcp.2024.101986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/06/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
Active neutrophils play a variety of roles in both innate and adaptive immune responses, and one of the most vital roles is the formation and release of neutrophil extracellular traps (NETs). NETs are created when neutrophils release their chromatin contents to get and eradicate pathogenic organisms essentially. While NET helps fight bacteria, viruses, parasites, and infections, it is also linked to asthma, atherosclerosis, and cancer metastasis. Thus, understanding the molecular mechanisms behind NETosis formation and its inhibition is crucial for developing safe and effective therapies. This systematic review aims to identify the list of miRNAs that are associated with the formation of NETosis and illustrate the mechanism of action by classifying them based on their expression site. Moreover, it summarizes the list of miRNAs that can be targeted therapeutically to reduce NETosis in various disorders. The current study entailed the searching of PubMed and Google Scholar for articles related to the research topic role of miRNAs in NETosis in all types of disorders. The search terms and phrases included "NETs," "neutrophil extracellular traps," "NETosis," "miRNA," "miR," and "micro-RNA." The search was limited to articles published in English since October 2024 in both databases. Following a review of 23 papers, 19 of them met the inclusion and exclusion criteria of this study. Four papers have been removed as they are duplicated or do not meet our criteria. According to the published articles till October 2024, there are 14 miRNAs involved in the molecular pathway of NETosis which are miR-155, miR-1696, miR-7, miR-223, miR-146a, miR-142a-3p, miR-3146, miR-505, miR-4512, miR-15b-5p, miR-16-5p, miR-26b-5p, miR-125a-3p and miR-378a-3p. Moreover, eight miRNAs have been identified as possible therapeutic targets for the suppression of NETosis based on in-vivo studies carried out in various organisms, which are miR-155, miR-146a, miR-1696, miR-223, miR-142a-3p, miR-3146, miR-4512, miR-16-5p. Different miRNAs that are expressed inside or outside of neutrophils can regulate and influence NETosis. Eight miRNAs have also been identified as potential therapeutic targets, which can be utilized to inhibit the molecular pathways associated with NETosis and prevent its negative effects, such as asthma, atherosclerosis, cancer metastasis, and cancer recurrence. However, further human-based research is necessary to completely understand the role of miRNAs in the development of NETosis in humans.
Collapse
Affiliation(s)
- Bashdar Mahmud Hussen
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq; Department of Biomedical Sciences, College of Science, Cihan University-Erbil, Kurdistan Region, Iraq
| | - Mohammed Fatih Rasul
- Department of Pharmaceutical Basic Science, Faculty of Pharmacy, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Goran Sedeeq Hama Faraj
- Department of Medical Laboratory Science, Komar University of Science and Technology, Sulaymaniyah, 46001, Iraq
| | - Snur Rasool Abdullah
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Seerwan Hamadameen Sulaiman
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Hasan Pourmoshtagh
- Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany.
| |
Collapse
|
36
|
Ekstedt S, Piersiala K, Kolev A, Farrajota Neves da Silva P, Margolin G, Kumlien Georén S, Cardell LO. Phenotypical differences of neutrophils patrolling tumour-draining lymph nodes in head and neck cancer. Br J Cancer 2024; 131:1893-1900. [PMID: 39543389 PMCID: PMC11628601 DOI: 10.1038/s41416-024-02891-5] [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: 05/06/2024] [Revised: 10/11/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND The complexity and heterogeneity of neutrophils are recognized, especially their roles in modulating inflammation and cancer immune responses. The detailed functions of neutrophils in human tumour-draining lymph nodes (TDLNs), specifically in the context of head and neck cancer, remain inadequately characterized. AIM This study aims to delineate the phenotypic diversity of neutrophils in TDLNs, non-tumour-draining lymph nodes (nTDLNs) from patients with oral squamous cell carcinoma (OSCC), and to evaluate their correlation with clinical outcomes. METHODS A flow cytometry-based investigation. RESULTS Neutrophils manifest a tissue-specific heterogeneity with significant phenotypic differences between compartments. A substantial fraction of neutrophils displayed an activated CD16highCD62Ldim profile in TDLNs, more prominent in patients with advanced T stages, implicating their involvement in the disease's progression. Notably, the presence of this activated neutrophil phenotype in TDLNs was strongly associated with poorer patient prognosis. CONCLUSIONS The study confirms the heterogeneity of neutrophils in human TDLNs, aligning with findings from animal models but extending them to show clinical relevance in human disease. The correlation of neutrophil phenotypes with cancer progression and prognosis emphasizes the importance of these cells in the tumour-microenvironment. The data suggests a future possibility to develop targeted therapies that modulate the neutrophilic response in OSCC.
Collapse
Affiliation(s)
- Sandra Ekstedt
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Krzysztof Piersiala
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Otorhinolaryngology, Karolinska University Hospital, Stockholm, Sweden
| | - Aeneas Kolev
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Otorhinolaryngology, Karolinska University Hospital, Stockholm, Sweden
- Medical unit Head Neck, Lung and Skin Cancer, Karolinska University Hospital, Stockholm, Sweden
| | | | - Gregori Margolin
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Otorhinolaryngology, Karolinska University Hospital, Stockholm, Sweden
- Medical unit Head Neck, Lung and Skin Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Susanna Kumlien Georén
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Otorhinolaryngology, Karolinska University Hospital, Stockholm, Sweden
| | - Lars-Olaf Cardell
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
- Department of Otorhinolaryngology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
37
|
Zhang Y, Wang C, Cheng S, Xu Y, Gu S, Zhao Y, Yang J, Wang Y. A Neutrophil Extracellular Traps-Related Signature Predicts Clinical Outcomes and Identifies Immune Landscape in Ovarian Cancer. J Cell Mol Med 2024; 28:e70302. [PMID: 39730971 DOI: 10.1111/jcmm.70302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 12/01/2024] [Accepted: 12/10/2024] [Indexed: 12/29/2024] Open
Abstract
Ovarian cancer (OvCa) is the most lethal gynaecology malignancies worldwide. Neutrophil extracellular traps (NETs), net-like protein structures produced by activated neutrophils and DNA-histone complexes, have a central role in tumours, though haven't been fully explored in OvCa. We obtained transcriptome data from TCGA-OvCa database (n = 376) as training, ICGC-OvCa database (n = 111) as validation and GTEx database (n = 180) as controls. Through LASSO-COX Regression analysis, we identified an eight-gene signature among 87 NETs-related genes, which was significantly related to poor prognosis in both TCGA-OvCa and ICGC-OvCa cohorts (Log-rank p-value = 0.0003 and 0.0014). Next, we constructed and validated a prognostic nomogram, consist of NETs-related signature and clinical features (C-index = 0.82). We evaluated 22 typical immune cell infiltration through CIBERSORT analysis, which implied upregulation of memory CD4 + T cells, follicular helper T cells and neutrophils in high-risk group. Additionally, we predicted therapy sensitivity through TIDE algorithm, indicating that high NETs-riskscore exhibited more sensitivity towards Sorafenib and less sensitivity towards immunotherapy. We initially reported that RAC2 upregulation was associated with NETs formation and poor prognosis (p-value < 0.05) through IHC analysis of tissue microarrays (n = 125). Conclusively, NETs-related signature was reliable for OvCa prognosis prediction and therapy assessment. Especially, RAC2 was predominantly related to NETs formation, thus providing hints towards anti-tumour mechanism of NETs in OvCa.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Gynecology, School of Medicine, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, School of Medicine, Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, China
| | - Chao Wang
- Department of Gynecology, School of Medicine, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, School of Medicine, Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, China
| | - Shanshan Cheng
- Department of Gynecology, School of Medicine, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, School of Medicine, Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, China
| | - Yanna Xu
- Department of Gynecology, School of Medicine, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, School of Medicine, Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, China
| | - Sijia Gu
- Department of Obstetrics and Gynecology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yaqian Zhao
- Department of Gynecology, School of Medicine, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, School of Medicine, Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, China
| | - Jiani Yang
- Department of Gynecology, School of Medicine, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, School of Medicine, Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, China
| | - Yu Wang
- Department of Gynecology, School of Medicine, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, School of Medicine, Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, China
| |
Collapse
|
38
|
Song C, Tong T, Dai B, Zhu Y, Chen E, Zhang M, Zhang W. Osteoimmunology in bone malignancies: a symphony with evil. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:354-368. [PMID: 39735445 PMCID: PMC11674455 DOI: 10.1016/j.jncc.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 12/31/2024] Open
Abstract
Bone marrow is pivotal for normal hematopoiesis and immune responses, yet it is often compromised by malignancies. The bone microenvironment (BME), composed of bone and immune cells, maintains skeletal integrity and blood production. The emergence of primary or metastatic tumors in the skeletal system results in severe complications and contributes significantly to cancer-related mortality. These tumors set off a series of interactions among cancer, bone, and immune cells, and disrupt the BME locally or distantly. However, the drivers, participants, and underlying molecules of these interactions are not fully understood. This review explores the crosstalk between bone metabolism and immune responses, synthesizing current knowledge on the intersection of cancer and osteoimmune biology. It outlines how bone marrow immune cells can either facilitate or hinder tumor progression by interacting with bone cells and pinpoints the molecules responsible for immunosuppression within bone tumors. Moreover, it discusses how primary tumors remotely alter the BME, leading to systemic immune suppression in cancer patients. This knowledge provides critical rationales for emerging immunotherapies in the treatment of bone-related tumors. Taken together, by summarizing the intricate relationship between tumor cells and the BME, this review aims to deepen the understanding of the diversity, complexity, and dynamics at play during bone tumor progression. Ultimately, it highlights the potential of targeting bone-tumor interactions to correct aberrant immune functions, thereby inhibiting tumor growth and metastasis.
Collapse
Affiliation(s)
- Churui Song
- Department of Breast Surgery and Oncology, Cancer Institute, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tie Tong
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Biqi Dai
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yue Zhu
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Elina Chen
- College of Natural Sciences, University of Texas at Austin, 110 Inner Campus Drive, Austin, USA
| | - Min Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weijie Zhang
- Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, and Department of Orthopaedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
39
|
Zhang X, Zhang B, He Y, Xiong W, Du Y, Shang P. A nomogram based on preoperative NLR predicts distant metastasis of urothelial carcinoma of the bladder. Cancer Biomark 2024; 41:18758592241296279. [PMID: 40095506 DOI: 10.1177/18758592241296279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
BackgroundDistant metastasis (DM) remains the most commonly reported cause of death in patients with urothelial carcinoma of the bladder (UCB).ObjectiveWe aimed to develop a robust prognostic model to assess the risk of DM in patients with UCB.MethodsWe collected clinical data of 206 UCB patients treated with RC. Patients treated with RC between 2011-2015 that were enrolled as the training cohort (n = 105), while the patients between 2016-2019 were enrolled as the validation cohort (n = 101). Univariate and multivariate Cox regression models were used to identify independent risk factors associated with DM. We identified the variables by stepwise regression and established nomogram. We evaluated the nomograms using C-index, calibration and ROC curves. Decision curve analysis was performed to compare the net benefits between the nomogram and TNM staging. We divided the patients into high and low risk groups according to the nomogram and compared the DM between the groups.ResultsThe neutrophil-lymphocyte ratio (NLR) was an independent predictor of DM. We established nomogram by T-stage, N-stage and NLR. The C-index of the nomogram was 0.766 and 0.739 respectively in the two cohorts. In the training cohort, AUC for the nomogram at 1, 2 and 3 years was 0.816, 0.812 and 0.812, respectively. In the validation cohort, the AUC for the nomogram at 1, 2 and 3 years was 0.751, 0.757 and 0.716, respectively. The calibration curve was satisfactory. The nomogram has a higher clinical benefit compared to the TNM staging system. Kaplan-Meier curves showed that patients from the high-risk group had a higher probability of DM than patients from the low-risk group.ConclusionsNomograms established by NLR, T-stage and N-stage can accurately predict distant metastases in patients with UCB.
Collapse
Affiliation(s)
- Xingxing Zhang
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Bin Zhang
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Yang He
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Wei Xiong
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Yuelin Du
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Panfeng Shang
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
40
|
Chernosky NM, Tamagno I, Polak KL, Chan ER, Yuan X, Jackson MW. Toll-Like receptor 3-mediated interferon-β production is suppressed by oncostatin m and a broader epithelial-mesenchymal transition program. Breast Cancer Res 2024; 26:167. [PMID: 39593161 PMCID: PMC11590466 DOI: 10.1186/s13058-024-01918-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Patients with Triple Negative Breast Cancer (TNBC) currently lack targeted therapies, and consequently face higher mortality rates when compared to patients with other breast cancer subtypes. The tumor microenvironment (TME) cytokine Oncostatin M (OSM) reprograms TNBC cells to a more stem-like/mesenchymal state, conferring aggressive cancer cell properties such as enhanced migration and invasion, increased tumor-initiating capacity, and intrinsic resistance to the current standards of care. In contrast to OSM, Interferon-β (IFN-β) promotes a more differentiated, epithelial cell phenotype in addition to its role as an activator of anti-tumor immunity. Importantly, OSM suppresses the production of IFN-β, although the mechanism of IFN-β suppression has not yet been elucidated. METHODS IFN-β production and downstream autocrine signaling were assessed via quantitative real-time PCR (qRT-PCR) and Western blotting in TNBC cells following exposure to OSM. RNA-sequencing (RNA-seq) was used to assess an IFN-β metagene signature, and to assess the expression of innate immune sensors, which are upstream activators of IFN-β. Cell migration was assessed using an in vitro chemotaxis assay. Additionally, TNBC cells were exposed to TGF-β1, Snail, and Zeb1, and IFN-β production and downstream autocrine signaling were assessed via RNA-seq, qRT-PCR, and Western blotting. RESULTS Here, we identify the repression of Toll-like Receptor 3 (TLR3), an innate immune sensor, as the key molecular event linking OSM signaling and the repression of IFN-β transcription, production, and autocrine IFN signaling. Moreover, we demonstrate that additional epithelial-mesenchymal transition-inducing factors, such as TGF-β1, Snail, and Zeb1, similarly suppress TLR3-mediated IFN-β production and signaling. CONCLUSIONS Our findings provide a novel insight into the regulation of TLR3 and IFN-β production in TNBC cells, which are known indicators of treatment responses to DNA-damaging therapies. Furthermore, strategies to stimulate TLR3 in order to increase IFN-β within the TME may be ineffective in stem-like/mesenchymal cells, as TLR3 is strongly repressed. Rather, we propose that therapies targeting OSM or OSM receptor would reverse the stem-like/mesenchymal program and restore TLR3-mediated IFN-β production within the TME, facilitating improved responses to current therapies.
Collapse
Affiliation(s)
- Noah M Chernosky
- Department of Pathology Case, Western Reserve University, Cleveland, OH, 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
| | - Ilaria Tamagno
- Department of Pathology Case, Western Reserve University, Cleveland, OH, 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
| | - Kelsey L Polak
- Department of Pathology Case, Western Reserve University, Cleveland, OH, 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
| | - E Ricky Chan
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
- Cleveland Institute for Computational Biology, Cleveland, OH, 44106, USA
| | - Xueer Yuan
- Department of Pathology Case, Western Reserve University, Cleveland, OH, 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
| | - Mark W Jackson
- Department of Pathology Case, Western Reserve University, Cleveland, OH, 44106, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA.
| |
Collapse
|
41
|
Song G, Wei Z, Pei Y, Liu Z, Min Y, Li H, Gao K, Ge J, Qing Y, Wei Y, Chen Y, Peng X. Harnessing the Systemic Immunoinflammatory Index as a Potential Predictive Tool for Recurrent or Metastatic Nasopharyngeal Carcinoma Undergoing PD-L1 Inhibitor. J Inflamm Res 2024; 17:9169-9180. [PMID: 39600680 PMCID: PMC11589775 DOI: 10.2147/jir.s474162] [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: 06/19/2024] [Accepted: 11/10/2024] [Indexed: 11/29/2024] Open
Abstract
Purpose Immunotherapy has become the primary option for recurrent and metastatic nasopharyngeal cancer (R/M NPC) after failure of chemotherapy, but without good prognostic indicators. Our study aimed to assess the potential of the systemic immune-inflammation index (SII) in predicting the effectiveness of PD-L1 inhibitor therapy for R/M NPC. Patients and Methods The study cohort comprises of a prospective Phase 2 clinical trial population undergoing PD-L1 inhibitor for R/M NPC at 42 hospitals in China between 2019 and 2021. The SII is classified into high and low states based on the optimal threshold determined by the ROC curve. We assessed the relationship between SII status and objective remission rate (ORR), disease control rate (DCR), progression-free survival (PFS), overall survival (OS) using regression analyses and Kaplan-Meier method. We performed sensitivity analyses to confirm the results. Results Our study analyzed 153 patients from one of the largest cohorts to date of R/M NPC treated with PD-L1 inhibitor and found that SII showed a significant association with prognosis. We found higher ORR and DCR in the SII-Low group. Univariate analyses demonstrated that SII independently predicted DCR (OR, 0.43; 95% CI, 0.22-0.84; p = 0.001), PFS (HR, 1.85; 95% CI, 1.31-2.62; p < 0.001) and OS (HR, 1.92; 95% CI, 1.29-2.85; p < 0.001). After adjusting for covariates, multivariate analysis remains relevant. [DCR (OR, 0.47; 95% CI, 0.22-0.99; p = 0.048), PFS (HR, 1.72; 95% CI, 1.2-2.47; p =0.003); OS (HR, 2.08; 95% CI, 1.38-3.13; p < 0.001)]. Sensitivity analyses also support this conclusion. Conclusion SII may well provide predictive value for the efficacy and prognosis of patients with R/M NPC treated with PD-L1 inhibitor. Patients with high status of SII may have a poorer therapeutic effect and survival.
Collapse
Affiliation(s)
- Ge Song
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Zhigong Wei
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Yiyan Pei
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Zheran Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Yu Min
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Huilin Li
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Kun Gao
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Junyou Ge
- Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd., Chengdu, People’s Republic of China
| | - Yan Qing
- Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd., Chengdu, People’s Republic of China
| | - Youneng Wei
- Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd., Chengdu, People’s Republic of China
| | - Ye Chen
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
- Division of Abdominal Tumor Multimodality Treatment, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| |
Collapse
|
42
|
Zhu C, Liao JY, Liu YY, Chen ZY, Chang RZ, Chen XP, Zhang BX, Liang JN. Immune dynamics shaping pre-metastatic and metastatic niches in liver metastases: from molecular mechanisms to therapeutic strategies. Mol Cancer 2024; 23:254. [PMID: 39543660 PMCID: PMC11562679 DOI: 10.1186/s12943-024-02171-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
Liver metastases are commonly detected in the advanced stages of various malignant tumors, representing a significant clinical challenge. Throughout the process of liver metastases formation, immune cells play a pivotal role, particularly in the pre-metastatic and metastatic niches within the liver. Immune cells establish extensive and intricate interactions with tumor cells and other components in the liver, collectively promoting and sustaining the growth of liver metastases. Despite the limited efficacy of existing therapeutic modalities against some advanced liver metastases, novel immune-based treatment approaches are continuously being explored and validated. Building on the systematic elucidation of the immunosuppressive characteristics of liver metastases, we explored the potential of novel immunotherapies applicable to patients with liver metastases from multiple dimensions.
Collapse
Affiliation(s)
- Chang Zhu
- Hepatic Surgery Center, and Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Jing-Yu Liao
- Hepatic Surgery Center, and Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Yi-Yang Liu
- Hepatic Surgery Center, and Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Ze-Yu Chen
- Hepatic Surgery Center, and Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Rui-Zhi Chang
- Hepatic Surgery Center, and Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Xiao-Ping Chen
- Hepatic Surgery Center, and Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Bi-Xiang Zhang
- Hepatic Surgery Center, and Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China.
| | - Jun-Nan Liang
- Hepatic Surgery Center, and Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China.
| |
Collapse
|
43
|
Lin C, Herlihy SE, Li M, Deng H, Kim R, Bernabei L, Rosenwasser M, Gabrilovich DI, Vogl DT, Nefedova Y. Neutrophil extracellular traps promote tumor chemoresistance to anthracyclines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.07.622533. [PMID: 39605505 PMCID: PMC11601256 DOI: 10.1101/2024.11.07.622533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The microenvironment plays an important role in promoting tumor cell chemoresistance, but the mechanisms responsible for this effect are not clear. Here, using models of multiple myeloma (MM) and solid cancers, we demonstrate a novel mechanism mediated by neutrophils, a major cell population in the bone marrow (BM), that protects cancer cells from chemotherapeutics. We show that in response to tumor-derived soluble factors, BM neutrophils release their DNA in the form of neutrophil extracellular traps (NETs). Cell-free DNA derived from NETs is then taken up by tumor cells via endocytosis and localizes to the cytoplasm. We found that both NETs and cell-free DNA taken up by tumor cells can bind anthracyclines, leading to tumor cell resistance to this class of chemotherapeutic agents. Targeting cell-free DNA with Pulmozyme or blocking NET formation with a PAD4 inhibitor abrogates the chemoprotective effect of neutrophils and restores sensitivity of tumor cells to anthracyclines.
Collapse
|
44
|
Xia M, Han Y, Sun L, Li D, Zhu C, Li D. The role of neutrophils in osteosarcoma: insights from laboratory to clinic. Front Immunol 2024; 15:1490712. [PMID: 39582869 PMCID: PMC11582048 DOI: 10.3389/fimmu.2024.1490712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/21/2024] [Indexed: 11/26/2024] Open
Abstract
Osteosarcoma, a highly aggressive malignant bone tumor, is significantly influenced by the intricate interactions within its tumor microenvironment (TME), particularly involving neutrophils. This review delineates the multifaceted roles of neutrophils, including tumor-associated neutrophils (TANs) and neutrophil extracellular traps (NETs), in osteosarcoma's pathogenesis. TANs exhibit both pro- and anti-tumor phenotypes, modulating tumor growth and immune evasion, while NETs facilitate tumor cell adhesion, migration, and immunosuppression. Clinically, neutrophil-related markers such as the neutrophil-to-lymphocyte ratio (NLR) predict patient outcomes, highlighting the potential for neutrophil-targeted therapies. Unraveling these complex interactions is crucial for developing novel treatment strategies that harness the TME to improve osteosarcoma management.
Collapse
Affiliation(s)
| | | | | | | | | | - Dongsong Li
- Department of Orthopedics, The First Hospital of Jilin University,
Changchun, Jilin, China
| |
Collapse
|
45
|
Zhu Y, Liang X, Zhi M, Li L, Zhang G, Chen C, Wang L, Wang P, Zhong N, Feng Q, Li Z. Succession of the multi-site microbiome along pancreatic ductal adenocarcinoma tumorigenesis. Front Immunol 2024; 15:1487242. [PMID: 39575247 PMCID: PMC11580624 DOI: 10.3389/fimmu.2024.1487242] [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: 08/27/2024] [Accepted: 10/14/2024] [Indexed: 11/24/2024] Open
Abstract
Background To investigate microbial characteristics across multibody sites from chronic pancreatitis (CP), through pancreatic benign tumors, to pancreatic ductal adenocarcinoma (PDAC) at different stages. Methods 16S ribosomal RNA (rRNA) amplicon sequencing was conducted on saliva, duodenal fluid, and pancreatic tissue obtained via endoscopic ultrasound-guided fine needle aspiration (EUS-FNA) of patients with CP, pancreatic benign tumors, PDAC in stage I/II, III, and IV. The neutral community model (NCM) assessed the microbial assembly contribution and MaAslin2 identified the differential microbes. Results From CP to stage IV PDAC patients, there was a marked surge in influence of salivary and duodenal microbiota on constitution of pancreatic microbial communities. Our observations revealed a successive alteration in microbial species across various bodily sites during PDAC tumorigenesis. Notably, Porphyromonas gingivalis, Treponema denticola, Peptoanaerobacter stomatis, Propionibacterium acidifaciens, Porphyromonas endodontalis, Filifactor alocis, etc., sequentially increased along PDAC progression in pancreatic tissue, whereas bacteria commonly used as probiotics Bifidobacterium breve, Lactiplantibacillus plantarum, etc., declined. Furthermore, the sequentially escalating trends of Peptoanaerobacter stomatis and Propionibacterium acidifaciens during PDAC tumorigenesis were mirrored in duodenal fluid and saliva. Porphyromonas gingivalis, Porphyromonas endodontalis, and Filifactor alocis, which intensified from CP to stage IV PDAC in pancreatic tissue, were also found to be enriched in saliva of patients with short-term survival (STS) compared with those with long-term survival (LTS). Conclusions Salivary and duodenal microorganisms were prominent factors in shaping pancreatic microbial landscape in PDAC context. Further exploration of these microbial entities is imperative to unravel their specific roles in PDAC pathogenesis, potentially yielding insights for future therapeutic strategies.
Collapse
Affiliation(s)
- Yiqing Zhu
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal (GI) Tumor, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiao Liang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal (GI) Tumor, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Mengfan Zhi
- Shandong Key Laboratory of Oral Tissue Regeneration, Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, Shandong, China
| | - Lixiang Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal (GI) Tumor, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Guoming Zhang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal (GI) Tumor, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Changxu Chen
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal (GI) Tumor, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Limei Wang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal (GI) Tumor, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Peng Wang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal (GI) Tumor, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ning Zhong
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal (GI) Tumor, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qiang Feng
- Shandong Key Laboratory of Oral Tissue Regeneration, Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, Shandong, China
| | - Zhen Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal (GI) Tumor, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
46
|
Inoue M, Takayama K, Hashimoto R, Enomoto M, Date N, Ohsumi A, Mizowaki T. Hyponatremia unleashes neutrophil extracellular traps elevating life-threatening pulmonary embolism risk. Proc Natl Acad Sci U S A 2024; 121:e2404947121. [PMID: 39475645 PMCID: PMC11551416 DOI: 10.1073/pnas.2404947121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 09/27/2024] [Indexed: 11/13/2024] Open
Abstract
Neutrophil extracellular traps (NETs), essential for controlling infections, can induce various pathologies when dysregulated. Known triggers for infection-independent NETs release exist, yet a comprehensive understanding of the conditions prompting such responses is lacking. In this study, we identify hyponatremia as an independent inducer of NETs release, a common clinical condition that disrupts sodium/calcium exchange within neutrophils. This disruption leads to an excess of intracellular calcium, subsequent elevation of reactive oxygen species (ROS), and the citrullination of histone H3, culminating in the activation of NETs-release pathways. Notably, under hyponatremic conditions, this mechanism is exacerbated during infectious states, leading to the deposition of NETs in the lungs and increasing the risk of life-threatening pulmonary embolism. Our findings underscore the critical role of sodium and calcium homeostasis in neutrophil functionality and provide insights into the pathogenesis of hyponatremia-associated diseases, highlighting potential therapeutic interventions targeting NETs dynamics.
Collapse
Affiliation(s)
- Minoru Inoue
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Radiation and Proton Therapy Center, Shizuoka Cancer Center Hospital, Shizuoka 411-8777, Japan
| | - Kazuo Takayama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Rina Hashimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Masahiro Enomoto
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Naoki Date
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto 606-8507, Japan
| | - Akihiro Ohsumi
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto 606-8507, Japan
| | - Takashi Mizowaki
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| |
Collapse
|
47
|
Seo Y, Kim SI, Song SH, Kim JG, Gu JY, Jeon HW, Lee M, Kim HK. Elevation of circulating neutrophil extracellular traps in endometrial cancer: Poor prognostic value of cell-free double-stranded DNA. Transl Oncol 2024; 49:102072. [PMID: 39128260 PMCID: PMC11366898 DOI: 10.1016/j.tranon.2024.102072] [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: 02/08/2024] [Revised: 07/02/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024] Open
Abstract
OBJECTIVE Neutrophils produce neutrophil extracellular traps (NETs) by releasing nuclear contents into the extracellular environment. NETs are associated with systemic inflammation and cancer development and progression. We aimed to investigate whether NET markers are associated with the prognosis of endometrial cancer. METHODS Circulating levels of three NET markers (histone-DNA complex, cell-free double-stranded DNA (dsDNA), and neutrophil elastase) were measured in 98 patients with endometrial cancer who underwent surgery as primary treatment between January 2015 and June 2018 and 45 healthy women. Area under the receiver operating characteristic curve (AUC) analyses were conducted to investigate the diagnostic and prognostic utility of the markers for endometrial cancer. RESULTS Patients with endometrial cancer showed significantly higher levels of the three NET markers than those in healthy controls. In discriminating endometrial cancer patients from healthy controls, the three NET markers showed AUC values in the following order: cell-free dsDNA (0.832; 95 % CI, 0.760-0.889), histone-DNA complex (0.740; 95 % CI, 0.660-0.809), and neutrophil elastase (0.689; 95 % CI, 0.607-0.764), comparable to those of CA-125 (0.741; 95 % CI, 0.659-0.813). Multivariate analysis adjusting for FIGO stage, histology, and lymphovascular space invasion, and lymph node involvement revealed that cell-free dsDNA level (cutoff: 95.2 ng/mL) was an independent prognostic marker for poor progression-free (adjusted HR, 2.75; 95 % CI, 1.096.92; P = 0.032) and overall survival (adjusted HR, 11.51; 95 % CI, 2.0664.22; P = 0.005) for patients with endometrial cancer. CONCLUSION High levels of circulating NET markers were observed in patients with endometrial cancer. Cell-free dsDNA levels may play a role as prognostic markers for endometrial cancer.
Collapse
Affiliation(s)
- Yeonju Seo
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Se Ik Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang Hoon Song
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Laboratory Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Jisoo G Kim
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Ja-Yoon Gu
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Hye Won Jeon
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea; Department of Obstetrics and Gynecology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, South Korea
| | - Maria Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea; Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, South Korea.
| | - Hyun Kyung Kim
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea; Department of Laboratory Medicine, Seoul National University Hospital, Seoul, South Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
48
|
Chen H, Zhou Y, Tang Y, Lan J, Lin C, Chen Q, Kuang H. Neutrophil extracellular traps in tumor progression of gynecologic cancers. Front Immunol 2024; 15:1421889. [PMID: 39555072 PMCID: PMC11563837 DOI: 10.3389/fimmu.2024.1421889] [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: 04/23/2024] [Accepted: 09/30/2024] [Indexed: 11/19/2024] Open
Abstract
This article delves into the intricate interplay between tumors, particularly gynecologic malignancies, and neutrophil extracellular traps (NETs). The relationship between tumors, specifically gynecologic malignancies, and NETs is a multifaceted and pivotal area of study. Neutrophils, pivotal components of the immune system, are tasked with combating foreign invaders. NETs, intricate structures released by neutrophils, play a vital role in combating systemic infections but also play a role in non-infectious conditions such as inflammation, autoimmune diseases, and cancer. Cancer cells have the ability to attract neutrophils, creating tumor-associated neutrophils, which then stimulate the release of NETs into the tumor microenvironment. The impact of NETs within the tumor microenvironment is profound and intricate. They play a significant role in influencing cancer development and metastasis, as well as modulating tumor immune responses. Through the release of proteases and pro-inflammatory cytokines, NETs directly alter the behavior of tumor cells, increasing invasiveness and metastatic potential. Additionally, NETs can trigger epithelial-mesenchymal transition in tumor cells, a process associated with increased invasion and metastasis. The interaction between tumors and NETs is particularly critical in gynecologic malignancies such as ovarian, cervical, and endometrial cancer. Understanding the mechanisms through which NETs operate in these tumors can offer valuable insights for the development of targeted therapeutic interventions. Researchers are actively working towards harnessing this interaction to impede tumor progression and metastasis, opening up new avenues for future treatment modalities. As our understanding of the interplay between tumors and NETs deepens, it is anticipated that novel treatment strategies will emerge, potentially leading to improved outcomes for patients with gynecologic malignancies. This article provides a comprehensive overview of the latest research findings on the interaction between NETs and cancer, particularly in gynecologic tumors, serving as a valuable resource for future exploration in this field.
Collapse
Affiliation(s)
- Hong Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yaling Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jianfa Lan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Chao Lin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Qionghua Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Hongying Kuang
- The Second Department of Gynecology, The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
49
|
Wang K, Wang X, Song L. Unraveling the complex role of neutrophils in lymphoma: From pathogenesis to therapeutic approaches (Review). Mol Clin Oncol 2024; 21:85. [PMID: 39347476 PMCID: PMC11428085 DOI: 10.3892/mco.2024.2783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/21/2024] [Indexed: 10/01/2024] Open
Abstract
Lymphoma, a malignancy of the lymphatic system, which is critical for maintaining the body's immune defenses, has become a focal point in recent research due to its intricate interplay with neutrophils-white blood cells essential for combating infections and inflammation. Unlike prior perceptions associating neutrophils only with tumor support, contemporary studies underscore their intricate and multifaceted involvement in the immune response to lymphoma. Recognizing the nuanced participation of neutrophils in lymphoma is crucial for developing innovative treatments to improve patient outcomes.
Collapse
Affiliation(s)
- Ke Wang
- Department of Cell Engineering, School of Life Sciences and Biotechnology, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Xiao Wang
- Reproduction Medicine Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, Guangdong 524002, P.R. China
| | - Li Song
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
50
|
Hu C, Long L, Lou J, Leng M, Yang Q, Xu X, Zhou X. CTC-neutrophil interaction: A key driver and therapeutic target of cancer metastasis. Biomed Pharmacother 2024; 180:117474. [PMID: 39316968 DOI: 10.1016/j.biopha.2024.117474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024] Open
Abstract
Circulating tumor cells (CTCs) are cancer cells that detach from the primary tumor and enter the bloodstream, where they can seed new metastatic lesions in distant organs. CTCs are often associated with white blood cells (WBCs), especially neutrophils, the most abundant and versatile immune cells in the blood. Neutrophils can interact with CTCs through various mechanisms, such as cell-cell adhesion, cytokine secretion, protease release, and neutrophil extracellular traps (NETs) formation. These interactions can promote the survival, proliferation, invasion, and extravasation of CTCs, as well as modulate the pre-metastatic niche and the tumor microenvironment. Therefore, inhibiting CTC-neutrophils interaction could be a potential strategy to reduce tumor metastasis and improve the prognosis of cancer patients. In this review, we summarize the current literature on CTC-neutrophils interaction' role in tumor metastasis and discuss the possible therapeutic approaches to target this interaction.
Collapse
Affiliation(s)
- Chengyi Hu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China; Yunnan Key Laboratory of Stem Cell and Regenerative Medicine & School of Rehabilitation, Kunming Medical University, Kunming 650500, PR China
| | - Ling Long
- School of Pharmacy, Kunming Medical University, Kunming 650500, PR China; Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing 400054, PR China
| | - Jie Lou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Mingjing Leng
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Qingqing Yang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Xiang Xu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine & School of Rehabilitation, Kunming Medical University, Kunming 650500, PR China; Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing 400042, PR China.
| | - Xing Zhou
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine & School of Rehabilitation, Kunming Medical University, Kunming 650500, PR China.
| |
Collapse
|