1
|
Cai S, Wei X, Li Q, Jiang Z, Li L. Smart materials in pharmacological drug development: Neutrophils and its constituents for drug delivery and consequent antitumor effects. Mol Immunol 2025; 183:18-32. [PMID: 40318595 DOI: 10.1016/j.molimm.2025.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/17/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025]
Abstract
Neutrophil-based drug delivery systems for targeted therapy of cancer have been studied widely in the recent past. Chemotactic cytokines including colony-stimulating factors (CSFs) recruit various immune cells including the neutrophils to the tumor microenvironment (TME) leading to enhanced metastasis. These cytokines can be targeted effectively by immunotherapeutic agents such as checkpoint inhibitors and mAbs that can lead to systemic toxicity. To minimize the systemic adverse effects, camouflaged nanoparticles can be used significantly as alternative therapeutic agents. The neutrophil-interacting NPs and neutrophil membrane coated NPs have been exploited recently for their antitumor properties in vitro and pose limited systemic adverse effects in vivo. Neutrophil-derived exosomes derived from immune cells can efficiently escape immune-surveillance and pass through the blood-brain barrier. They possess several intrinsic properties in drug delivery as they are nano-sized, extremely biocompatible, non-immunogenic, biodegradable, stable and can carry targeting agents with limited toxicity and display antitumor properties. Also, neutrophil-based nanotherapy is dependent on factors such as neutrophil kinetics and the physicochemical properties of NPs such as size, shape, and surface chemistry. Therefore, neutrophil-based drug delivery for cancer therapy via the use of polymer nanoparticles is widely studied as their clinical appliance in nanomedicine is still at its infancy.
Collapse
Affiliation(s)
- Shengjie Cai
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Xuehan Wei
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Qian Li
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Department of Oncology, Jiangsu Integrated Traditional Chinese and Western Medicine Hospital, Nanjing 210028, China; Department of Oncology, Ganyu Hospital of Traditional Chinese Medicine, Lianyungang, Jiangsu 222000, China
| | - Ziyu Jiang
- Department of Oncology, Lianyungang Integrated Traditional Chinese and Western Medicine Clinical College, Nanjing University of Chinese Medicine, Nanjing 222002, China; Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, China.
| | - Lingchang Li
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Department of Oncology, Jiangsu Integrated Traditional Chinese and Western Medicine Hospital, Nanjing 210028, China.
| |
Collapse
|
2
|
Rapozzi V, Comuzzi C, Di Giorgio E, Xodo LE. KRAS and NRF2 drive metabolic reprogramming in pancreatic cancer cells: the influence of oxidative and nitrosatice stress. Front Cell Dev Biol 2025; 13:1547582. [PMID: 40567499 PMCID: PMC12187747 DOI: 10.3389/fcell.2025.1547582] [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: 12/18/2024] [Accepted: 05/26/2025] [Indexed: 06/28/2025] Open
Abstract
Cancer cells are subject to metabolic reprogramming, which leads to a sustained production of reactive oxygen species (ROS). Increased oxidative stress contributes to genomic instability and promotes malignant transformation. To counteract excessive ROS levels, cells activate nuclear factor erythroid 2-related factor 2 (NRF2), a key regulator of redox homeostasis that coordinates the transcription of a wide range of antioxidant and cytoprotective genes. This review examines the metabolic adaptations controlled by the KRAS-NRF2 axis under oxidative stress conditions. In addition, we highlight a novel function of NRF2 in regulating the expression of NOS2 by binding to a DNA enhancer element, thereby modulating the production of reactive nitrogen species (RNS). Finally, we discuss novel molecular strategies aimed at disrupting adaptive antioxidant responses in cancer cells and provide insights into combinatorial therapeutic approaches targeting redox balance in cancer.
Collapse
Affiliation(s)
- Valentina Rapozzi
- Department of Medicine, Laboratory of Biochemistry, University of Udine, Udine, Italy
| | - Clara Comuzzi
- Department of Agricultural Food Environmental and Animal Science, University of Udine, Udine, Italy
| | - Eros Di Giorgio
- Department of Medicine, Laboratory of Biochemistry, University of Udine, Udine, Italy
| | - Luigi E. Xodo
- Department of Medicine, Laboratory of Biochemistry, University of Udine, Udine, Italy
| |
Collapse
|
3
|
Zhang T, Li Y, Zhai E, Zhao R, Qian Y, Huang Z, Liu Y, Zhao Z, Xu X, Liu J, Li Z, Liang Z, Wei R, Ye L, Ma J, Wu Q, Chen J, Cai S. Intratumoral Fusobacterium nucleatum Recruits Tumor-Associated Neutrophils to Promote Gastric Cancer Progression and Immune Evasion. Cancer Res 2025; 85:1819-1841. [PMID: 39992708 PMCID: PMC12079103 DOI: 10.1158/0008-5472.can-24-2580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/07/2024] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Intratumoral microbiota can affect the development and progression of many types of cancer, including gastric cancer. A better understanding of the precise mechanisms by which microbiota support gastric cancer could lead to improved therapeutic approaches. In this study, we investigated the effect of intratumoral microbiota on the tumor immune microenvironment during gastric cancer malignant progression. Analysis of human gastric cancer tissues with 16S rRNA amplicon sequencing revealed that Fusobacterium nucleatum was significantly enriched in gastric cancer tissues with lymph node metastasis and correlated with a poor prognosis. F. nucleatum infection spontaneously induced chronic gastritis and promoted gastric mucosa dysplasia in mice. Furthermore, gastric cancer cells infected with F. nucleatum showed accelerated growth in immunocompetent mice compared with immunodeficient mice. Single-cell RNA sequencing uncovered that F. nucleatum recruited tumor-associated neutrophils (TAN) to reshape the tumor immune microenvironment. Mechanistically, F. nucleatum invaded gastric cancer cells and activated IL17/NF-κB/RelB signaling, inducing TAN recruitment. F. nucleatum also stimulated TAN differentiation into the protumoral subtype and subsequent promotion of PD-L1 expression, further facilitating gastric cancer immune evasion while also enhancing the efficacy of anti-PD-L1 antibody therapy. Together, these data uncover mechanisms by which F. nucleatum affects gastric cancer immune evasion and immunotherapy efficacy, providing insights for developing effective treatment strategies. Significance: Intratumoral F. nucleatum activates NF-κB signaling to facilitate gastric cancer immune evasion by promoting tumor-associated neutrophil recruitment that sensitizes tumors to immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Tianhao Zhang
- Division of Gastrointestinal Surgery Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of General Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ying Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Ertao Zhai
- Division of Gastrointestinal Surgery Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Risheng Zhao
- Division of Gastrointestinal Surgery Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan Qian
- Division of Gastrointestinal Surgery Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhixin Huang
- Division of Gastrointestinal Surgery Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of General Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yinan Liu
- Division of Gastrointestinal Surgery Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of General Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zeyu Zhao
- Division of Gastrointestinal Surgery Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of General Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiang Xu
- Division of Gastrointestinal Surgery Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of General Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jianqiu Liu
- Division of Gastrointestinal Surgery Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of General Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zikang Li
- Division of Gastrointestinal Surgery Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of General Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhi Liang
- Division of Gastrointestinal Surgery Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of General Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ran Wei
- Division of Gastrointestinal Surgery Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of General Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Linying Ye
- Division of Gastrointestinal Surgery Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of General Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jinping Ma
- Division of Gastrointestinal Surgery Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Jianhui Chen
- Division of Gastrointestinal Surgery Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of General Surgery, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, Guangxi, China
| | - Shirong Cai
- Division of Gastrointestinal Surgery Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Ma Y, Qi J, Zhang X, Liu K, Liu Y, Yu X, Bu Y, Chen B. Development and application of an early warning model for predicting early mortality following stent placement in malignant biliary obstruction: A comparative analysis of logistic regression and artificial neural network approaches. Oncol Lett 2025; 29:237. [PMID: 40166368 PMCID: PMC11956143 DOI: 10.3892/ol.2025.14983] [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: 10/15/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Patients with malignant biliary obstruction (MBO) are often treated with endoscopic retrograde cholangiopancreatography (ERCP) combined with biliary stent placement for tumor progression. However, certain patients die within 30 days after the procedure, increasing healthcare resource consumption and patient burden. Therefore, the development of early mortality prediction models is important for optimizing treatment decisions. The present study retrospectively analyzed the clinical data of 285 patients with MBO, including demographic information, laboratory indicators and tumor-related factors. Logistic regression and artificial neural network (ANN) models were used to construct a prediction tool, and the model performance was evaluated using area under the curve (AUC), accuracy, sensitivity and specificity. The logistic regression model, which identified the cancer antigen 19-9 (CA19-9) level and a history of previous ERCP surgery as independent risk factors, had an AUC of 0.727 and an accuracy of 65.0%. The ANN model, which combined five variables, namely CA19-9, history of previous ERCP surgery, neutrophil-lymphocyte ratio (NLR), liver metastasis and carcinoembryonic antigen, demonstrated that NLR was the most weighted predictor. Furthermore, the ANN model had an AUC of 0.813, an accuracy of 88.2% and a specificity that was markedly higher than that of the logistic regression model (95.5 vs. 83.3%). However, the ANN model was revealed to be slightly less sensitive compared with the logistic regression model (61.1 vs. 61.2%). In conclusion, compared with logistic regression, the ANN model had a greater performance level in terms of predictive power and specificity, and is suitable for capturing complex non-linear relationships. However, its complexity and risk of overfitting need to be further optimized. The present study provides a new tool for the accurate prediction of the risk of early death after ERCP in patients with MBO, which could help improve individualized treatment strategies.
Collapse
Affiliation(s)
- Yongxin Ma
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
- Department of Hepatobiliary Surgery, First Clinical College of Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Jiaojiao Qi
- Department of Obstetrics Function Center Inspection, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Xusheng Zhang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
- Department of Hepatobiliary Surgery, First Clinical College of Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Kejun Liu
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
- Department of Hepatobiliary Surgery, First Clinical College of Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yimin Liu
- Department of Hepatobiliary Surgery, First Clinical College of Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Xuehai Yu
- Department of Pediatric Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yang Bu
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
- Department of Hepatobiliary Surgery, First Clinical College of Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Bendong Chen
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
- Department of Hepatobiliary Surgery, First Clinical College of Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
5
|
Wei Y, Mao D, Liu T, Wu W, Yang Z, Liu X. The predictive value of PIV, PLR, LMR, NPR, and NLR for the prognosis of transarterial chemoembolization in patients with hepatocellular carcinoma combined with liver cirrhosis. BMC Gastroenterol 2025; 25:315. [PMID: 40301803 PMCID: PMC12042468 DOI: 10.1186/s12876-025-03815-0] [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: 12/10/2024] [Accepted: 03/24/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Transarterial chemoembolization (TACE) is a primary treatment for hepatocellular carcinoma (HCC) in patients with liver cirrhosis. Prognostic markers that reliably predict outcomes in these patients post-TACE remain insufficiently defined. Systemic inflammatory markers such as the Pan-Immunological Value (PIV), Platelet-Lymphocyte Ratio (PLR), Lymphocyte-Monocyte Ratio (LMR), Neutrophil-Platelet Ratio (NPR), and Neutrophil-Lymphocyte Ratio (NLR) offer potential prognostic insights for various disease. This study aims to evaluate this markers to ascertain their predictive value in determining prognosis post-TACE. METHODS This retrospective study involved 216 patients with HCC and cirrhosis treated with TACE at a single hospital from May 2017 to May 2023. Patients were stratified into good (n = 92) and poor prognosis groups (n = 124) based on one-year post-operative outcomes using the Response Evaluation Criteria in Solid Tumors (RECIST). We evaluated preoperative inflammatory markers, biochemical and imaging data, and utilized univariate and multivariate logistic regression analyses to determine predictive factors for prognosis. RESULTS Patients in the poor prognosis group exhibited significantly higher PIV, PLR, NLR, and NPR, and lower LMR (P < 0.05). Multivariate analysis identified PIV and NPR as the strongest independent predictors of poor prognosis (OR: 1.021, P < 0.001 and OR: 2.909, P < 0.001, respectively). ROC analysis demonstrated that PIV had the greatest predictive accuracy (AUC = 0.803). CONCLUSION PIV, PLR, LMR, NPR, and NLR serve as significant prognostic markers for patients with HCC undergoing TACE in the context of liver cirrhosis.
Collapse
Affiliation(s)
- Yanan Wei
- Department of Oncology, The First Clinical Medical College, Three Gorges University/Yichang City Central People's Hospital, Yichang, 443000, China
| | - Dongting Mao
- Department of Clinical Laboratory, The First Clinical Medical College, Three Gorges University, Yichang City Central People's Hospital, Yichang, 443000, China
| | - Tiantian Liu
- Department of Oncology, The First Clinical Medical College, Three Gorges University/Yichang City Central People's Hospital, Yichang, 443000, China
| | - Wanyan Wu
- Department of Oncology, The First Clinical Medical College, Three Gorges University/Yichang City Central People's Hospital, Yichang, 443000, China
| | - Ziwei Yang
- Department of Oncology, The First Clinical Medical College, Three Gorges University/Yichang City Central People's Hospital, Yichang, 443000, China
| | - Xiuli Liu
- Department of Oncology, The First Clinical Medical College, Three Gorges University/Yichang City Central People's Hospital, Yichang, 443000, China.
| |
Collapse
|
6
|
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
|
7
|
Belyaev IB, Griaznova OY, Yaremenko AV, Deyev SM, Zelepukin IV. Beyond the EPR effect: Intravital microscopy analysis of nanoparticle drug delivery to tumors. Adv Drug Deliv Rev 2025; 219:115550. [PMID: 40021012 DOI: 10.1016/j.addr.2025.115550] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
Delivery of nanoparticles (NPs) to solid tumors has long relied on enhanced permeability and retention (EPR) effect, involving permeation of NPs through a leaky vasculature with prolonged retention by reduced lymphatic drainage in tumor. Recent research studies and clinical data challenge EPR concept, revealing alternative pathways and approaches of NP delivery. The area was significantly impacted by the implementation of intravital optical microscopy, unraveling delivery mechanisms at cellular level in vivo. This review presents analysis of the reasons for EPR heterogeneity in tumors and describes non-EPR based concepts for drug delivery, which can supplement the current paradigm. One of the approaches is targeting tumor endothelium by NPs with subsequent intravascular drug release and gradient-driven drug transport to tumor interstitium. Others exploit various immune cells for tumor infiltration and breaking endothelial barriers. Finally, we discuss the involvement of active transcytosis through endothelial cells in NP delivery. This review aims to inspire further understanding of the process of NP extravasation in tumors and provide insights for developing next-generation nanomedicines with improved delivery.
Collapse
Affiliation(s)
- Iaroslav B Belyaev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Eindhoven University of Technology, Eindhoven 5600 MB, the Netherlands
| | - Olga Yu Griaznova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | | | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Ivan V Zelepukin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75123, Sweden.
| |
Collapse
|
8
|
Ghorbaninezhad F, Nour MA, Farzam OR, Saeedi H, Vanan AG, Bakhshivand M, Jafarlou M, Hatami-Sadr A, Baradaran B. The tumor microenvironment and dendritic cells: Developers of pioneering strategies in colorectal cancer immunotherapy? Biochim Biophys Acta Rev Cancer 2025; 1880:189281. [PMID: 39929377 DOI: 10.1016/j.bbcan.2025.189281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 01/25/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
Colorectal cancer (CRC) is the world's third most frequent cancer, and both its incidence and fatality rates are rising. Despite various therapeutic approaches, neither its mortality rate nor its recurrence frequency has decreased significantly. Additionally, conventional treatment approaches, such as chemotherapy and radiotherapy, have several side effects and risks for patients with CRC. Accordingly, the need for alternative and effective treatments for CRC patients is critical. Immunotherapy that utilizes dendritic cells (DCs) harnesses the patient's immune system to combat cancer cells effectively. DCs are the most potent antigen-presenting cells (APCs), which play a vital role in generating anti-cancer T cell responses. A significant barrier to the immune system's ability to eliminate CRC is the establishment of a potent immunosuppressive tumor milieu by malignant cells. Since DCs are frequently defective in this milieu, the tumor setting significantly reduces the effectiveness of DC-based therapy. Determining central mechanisms contributing to tumor growth by unraveling and comprehending the interaction between CRC tumor milieu and DCs may lead to new therapeutic approaches. This study aims to review DC biology and discuss its role in T-cell-mediated anti-tumor immunity, as well as to highlight the immunosuppressive effects of the CRC tumor milieu on the function of DCs. We will also highlight the tumor microenvironment (TME)-related factors that interfere with DC function as a possible therapeutic target to enhance DC-based cell therapy efficacy.
Collapse
Affiliation(s)
- Farid Ghorbaninezhad
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mina Afrashteh Nour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Omid Rahbar Farzam
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Saeedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Ghorbani Vanan
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Bakhshivand
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Jafarlou
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Behzad Baradaran
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
9
|
Varjú I, Tanka-Salamon A, Kolev K. Neutrophil Extracellular Traps: At the Interface of Thrombosis and Comorbidities. Semin Thromb Hemost 2025. [PMID: 40020757 DOI: 10.1055/a-2548-0805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Abstract
Since their discovery in 2004, neutrophil extracellular traps (NETs) have been at the center of multidisciplinary attention. Although a key tool in neutrophil-mediated immunity, these filamentous, enzyme-enriched DNA-histone complexes can be detrimental to tissues and have been identified as an underlying factor in a range of pathological conditions. Building on more than 20 years of research into NETs, this review places thrombosis, the pathological formation of blood clots, in the spotlight. From this point of view, we discuss the structure and formation of NETs, as well as the interaction of their components with the hemostatic system, dissecting the pathways through which NETs exert their marked effect on formation and the dissolution of thrombi. We pay distinct attention to the latest developments in the research of a key player in NET formation, peptidyl-arginine-deiminase (PAD) enzymes: their types, sources, and potential cross-play with the hemostatic machinery. Besides these molecular details, we elaborate on the link between pathological thrombosis, NETs, and widespread conditions that represent a debilitating public health burden worldwide, such as sepsis and neoplasms. Finally, future implications on the treatment of thrombosis-related conditions will be discussed.
Collapse
Affiliation(s)
- Imre Varjú
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Anna Tanka-Salamon
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Krasimir Kolev
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
10
|
Saxena R, Gottlin EB, Campa MJ, He YW, Patz EF. Complement regulators as novel targets for anti-cancer therapy: A comprehensive review. Semin Immunol 2025; 77:101931. [PMID: 39826189 DOI: 10.1016/j.smim.2025.101931] [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: 10/25/2024] [Revised: 01/04/2025] [Accepted: 01/04/2025] [Indexed: 01/22/2025]
Abstract
Cancer remains a formidable global health challenge requiring the continued exploration of innovative therapeutic approaches. While traditional treatment strategies including surgery, chemotherapy, and radiation therapy have had some success, primarily in early-stage disease, the quest for more targeted, personalized, safer, and effective therapies remains an ongoing pursuit. Over the past decade, significant advances in the field of tumor immunology have dramatically shifted a focus towards immunotherapy, although the ability to harness and coopt the immune system to treat cancer is still just beginning to be realized. One important area that has yet to be fully explored is the complement system, an integral part of innate immunity that has gathered attention recently as a source of potential targets for anti-cancer therapy. The complement system has a complex and context dependent role in cancer biology in that it not only contributes to immune surveillance but also may promote tumor progression. Complement regulators, including CD46, CD55, CD59, and complement factor H, exercise defined control over complement activation, and have also been acknowledged for their role in the tumor microenvironment. This review explores the intricate role of complement regulators in cancer development and progression, examining their potential as therapeutic targets, current strategies, challenges, and the evolving landscape of clinical research.
Collapse
Affiliation(s)
- Ruchi Saxena
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Elizabeth B Gottlin
- Department of Radiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Michael J Campa
- Department of Radiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - You-Wen He
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Edward F Patz
- Department of Radiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
11
|
Chu B, Chen Y, Pan J. Prognostic significance of systemic immune inflammation index for ovarian cancer: An updated systematic review and meta-analysis. J Ovarian Res 2025; 18:41. [PMID: 40016853 PMCID: PMC11869409 DOI: 10.1186/s13048-025-01626-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/14/2025] [Indexed: 03/01/2025] Open
Abstract
OBJECTIVE Several inflammatory indices have been used to assess the prognosis of ovarian cancer, with variable results. This review assessed whether the systemic immune inflammation index (SII) can predict outcomes in patients with ovarian cancer. METHODS Embase, PubMed, CENTRAL, Web of Science, and Scopus databases were searched by the two reviewers from inception to 15th October 2024 for studies assessing the relationship between SII and overall survival (OS) or disease-free survival (DFS). RESULTS Ten studies with eleven cohorts were included. Pooled analysis showed that higher SII was a significant predictor of poor OS (HR: 2.35 95% CI: 1.56, 3.55 I2 = 88%) and worse DFS (HR: 2.51 95% CI: 1.71, 3.67 I2 = 80%) after ovarian cancer. Sensitivity analysis failed to change the significance of the results. No publication bias was noted. Most results remained significant on subgroup analyses based on location, sample size, FIGO stage, treatment, adjusted outcomes, cut-off of SII, method of determining cut-off, and quality score. CONCLUSIONS SII can be a potential predictor of OS and DFS after ovarian cancer. Further studies are required to improve the evidence.
Collapse
Affiliation(s)
- Boliang Chu
- Department of Gynecology, Huzhou Maternity & Child Health Care Hospital, No. 2, East Street, Wuxing District, Huzhou, Zhejiang Province, 313000, China
| | - Yingying Chen
- Department of Gynecology, Huzhou Maternity & Child Health Care Hospital, No. 2, East Street, Wuxing District, Huzhou, Zhejiang Province, 313000, China
| | - Jiewei Pan
- Department of Gynecology, Huzhou Maternity & Child Health Care Hospital, No. 2, East Street, Wuxing District, Huzhou, Zhejiang Province, 313000, China.
| |
Collapse
|
12
|
Wang T, Qi L, Zhao Y, Ma X, Li T. Inflammatory biomarker correlations and prognosis in high-risk gastrointestinal stromal tumor patients: a multicenter retrospective analysis. BMC Gastroenterol 2025; 25:119. [PMID: 40011800 PMCID: PMC11863871 DOI: 10.1186/s12876-025-03710-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 02/18/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND The accurate prognosis of gastrointestinal stromal tumors (GISTs) has garnered substantial attention, yet a gap persists in understanding the influence of inflammatory markers on the prognosis of high-risk GIST patients. This study investigated the relationship between various factors and the prognosis of high-risk GIST patients, with a specific focus on first recurrence-free survival (RFS) and overall survival (OS) as crucial prognostic indicators. METHODS A comprehensive collection of clinical data was conducted on 145 high-risk GIST patients meeting specific inclusion and exclusion criteria at 17 medical centers in Ningxia Hui Autonomous Region, China, covering the period from January 2013 to December 2019. Single-factor analysis and survival curves were used to analyze the variables, while the Cox regression model evaluated independent prognostic factors. RESULTS Within the cohort, a balanced male-to-female ratio of 1:1.1 was observed. Univariate analysis revealed compelling associations between RFS and age, preoperative neutrophil-to-lymphocyte ratio (NLR), preoperative platelet-to-lymphocyte ratio (PLR), preoperative systemic immune-inflammatory index (SII), preoperative prognostic nutritional index (PNI), mitotic index, and whether or not imatinib (IM) was taken regularly in high-risk GIST patients (P < 0.05). Except age, these other variables were also significantly correlated with OS (P < 0.05). Cox regression analysis showed that age, preoperative PNI, mitotic index and postoperative IM adjuvant therapy independently affected RFS (P < 0.05). In addition, preoperative PNI and postoperative IM adjuvant therapy were also independent factors of OS, with statistical significance (P < 0.05). Age was negatively correlated with RFS, and early routine IM treatment after operation significantly reduced the risk of recurrence and death. Higher mitotic index is closely related to poor RFS, and higher preoperative PNI indicates a better prognosis. CONCLUSION A close correlation between young age, low preoperative PNI, high mitotic index, and lack of IM treatment had an unfavorable prognosis in high-risk GIST patients. Notably, the PNI was identified as a potential additional prognostic factor, enhancing the accuracy of predicting treatment efficacy and patient outcomes in high-risk patients with GISTs. Therefore, we advocate for the serious consideration of the PNI as a valuable addition to standard clinical practice for managing high-risk GIST patients.
Collapse
Affiliation(s)
- Tao Wang
- Department of Surgical Oncology II, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Lihua Qi
- Department of Surgical Oncology II, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Yang Zhao
- Department of Surgical Oncology II, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China.
| | - Xiaolan Ma
- Department of Surgical Oncology II, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Tao Li
- Department of Surgical Oncology II, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China.
| |
Collapse
|
13
|
Zeng L, Tang Y, Huang X, Pei W, Liao Y, Liu J. Combined impact of prognostic nutritional index, fibrinogen-to-albumin ratio, and neutrophil-to-lymphocyte ratio on surgical outcomes and prognosis in hepatocellular carcinoma. Am J Cancer Res 2025; 15:439-451. [PMID: 40084351 PMCID: PMC11897630 DOI: 10.62347/rtmf3105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/07/2025] [Indexed: 03/16/2025] Open
Abstract
This study evaluated the predictive value of the prognostic nutritional index (PNI), fibrinogen-to-albumin ratio (FAR), and neutrophil-to-lymphocyte ratio (NLR) for overall survival in hepatocellular carcinoma (HCC) patients. A total of 283 HCC cases from Hunan Provincial People's Hospital were included in the analysis, with 45 additional patients as external validation. The relationship between these indices and patient prognosis was further evaluated using the Kaplan-Meier method and Cox regression analysis. Receiver operating characteristic (ROC) curve analysis was performed to assess the predictive performance of these indices for overall survival (OS) and to determine the optimal cutoff values. ROC curve analysis revealed that the area under the curve (AUC) for PNI, FAR, and NLR was 0.723, 0.857, and 0.872, respectively. Multivariate analysis identified hepatitis history, intraoperative blood transfusion, FAR, NLR, and PNI as independent prognostic factors (all P<0.05). The resulting prediction model demonstrated strong performance in both the training (C-index =0.917) and external validation (C-index =0.853) cohorts, with AUCs of 0.889 and 0.931 for 6-month and 1-year prediction in the validation set, respectively. These findings suggest that preoperative levels of peripheral blood PNI, FAR, and NLR are closely associated with the surgical prognosis of HCC patients. The prognostic prediction model developed based on these indices exhibits good predictive efficacy.
Collapse
Affiliation(s)
- Liuhaonan Zeng
- Department of Anesthesiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital) Changsha 410000, Hunan, China
| | - Yixun Tang
- Department of Anesthesiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital) Changsha 410000, Hunan, China
| | - Xiaoling Huang
- Department of Anesthesiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital) Changsha 410000, Hunan, China
| | - Wanmin Pei
- Department of Anesthesiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital) Changsha 410000, Hunan, China
| | - Yongqiong Liao
- Department of Anesthesiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital) Changsha 410000, Hunan, China
| | - Jitong Liu
- Department of Anesthesiology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital) Changsha 410000, Hunan, China
| |
Collapse
|
14
|
Zhang Y, Zhang F, Liu Z, Li M, Wu G, Li H. P2RX1-blocked neutrophils induce CD8 + T cell dysfunction and affect the immune escape of gastric cancer cells. Cell Immunol 2025; 408:104901. [PMID: 39675308 DOI: 10.1016/j.cellimm.2024.104901] [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: 03/19/2024] [Revised: 11/28/2024] [Accepted: 11/30/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Gastric cancer (GC) is one of the deadly malignancies of the gastrointestinal tract. Research has confirmed the linkage of P2RX1 with immune cell activation and tumor progression. This project focused on the impact of P2RX1 level in neutrophils on the efficacy of immune checkpoint inhibitor (ICI) treatment in GC. METHODS Blood samples from 23 GC patients eligible for camrelizumab treatment were collected. Flow cytometry was carried out to analyze the proportion of P2RX1 in neutrophils. IHC was utilized to detect the expression level of PD-L1. We also evaluated the chemotaxis ability of neutrophils using a Transwell system, assessed the viability and apoptosis rate of GC cells using CCK-8 and flow cytometry, measured the proportions of CD8+PD-1+ and CD8+GZMB+ cells, determined the expression levels of IL-6, TNFα, IFN-γ, IL-8, IL-12, IL-1β, and GZMB by utilizing enzyme-linked immunosorbent assay (ELISA), and examined the expression levels of P2RX1 and PD-L1 using western blot (WB). By establishing a xenograft mouse model, we studied the impact of P2RX1-blocked neutrophils on the efficacy of ICI treatment in the GC microenvironment. RESULTS In GC, clinical analysis revealed increased infiltration of P2RX1-lowly expressed neutrophil subsets and increased expression of PD-L1. In vitro experiments demonstrated that abnormal expression of P2RX1 affected neutrophil function. Furthermore, the blockage or knockdown of P2RX1 in neutrophils modulated CD8+ T cell function, promoting GC progression. In in vivo experiments, the blockage of P2RX1 in neutrophils inhibited the effectiveness of ICI treatment in the GC microenvironment. CONCLUSION This project validated that the loss of P2RX1 in neutrophils induces CD8+ T cell dysfunction and affects the GC development, indicating that P2RX1 may be an accurate biomarker for predicting ICI response, thus providing a theoretical basis for the clinical application of ICI.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Medical Oncology, Ma'anshan People's Hospital, 519 Hunan East Road, Huashan District, Ma'anshan City, Anhui Province 243000, China.
| | - Fenglin Zhang
- Department of Medical Oncology, Ma'anshan People's Hospital, 519 Hunan East Road, Huashan District, Ma'anshan City, Anhui Province 243000, China
| | - Zhi Liu
- Department of Pathology, Ma'anshan People's Hospital, 519 Hunan East Road, Huashan District, Ma'anshan City, Anhui Province 243000, China
| | - Min Li
- Department of Medical Oncology, Ma'anshan People's Hospital, 519 Hunan East Road, Huashan District, Ma'anshan City, Anhui Province 243000, China
| | - Ge Wu
- Department of Medical Oncology, Ma'anshan People's Hospital, 519 Hunan East Road, Huashan District, Ma'anshan City, Anhui Province 243000, China
| | - Hui Li
- Department of Medical Oncology, Ma'anshan People's Hospital, 519 Hunan East Road, Huashan District, Ma'anshan City, Anhui Province 243000, China
| |
Collapse
|
15
|
Juodžiukynienė N, Lasienė K, Savickienė N, Aniulienė A. Mast Cell Density in Squamous Cell Carcinoma of Skin in Dogs and Cats. Animals (Basel) 2025; 15:316. [PMID: 39943085 PMCID: PMC11816034 DOI: 10.3390/ani15030316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/12/2025] [Accepted: 01/17/2025] [Indexed: 02/16/2025] Open
Abstract
The purpose of the present study was to evaluate mast cell density in squamous cell carcinoma tissues of dogs and cats to assess species differences. Skin squamous cell carcinoma tissues from dogs (n = 15: n = 10 from body sites and n = 5 nail bed specimens) and cats (n = 15, n = 10 from ears and n = 5 nasal planum specimens) were examined. Intratumoral mast cell density (IMCD), peritumoral mast cell density (PMCD) and total mast cells density (TMCD) as a sum of IMCD and PMCD were calculated from Giemsa-stained slides at high magnification in 1 mm2 using an Olympus microscope (Olympus BX41, Tokyo, Japan) equipped with a digital Olympus DP72 image camera and CellSensDimension software V1.16). Both intratumoral and peritumoral tissues of the squa.mous cell carcinoma were divided into two categories: (1) loose, well-vascularized, rich in lymphocytes and plasmocytes, macrophages and neutrophils; and (2) fibrous, with few or no lymphocytes, plasmocytes, macrophages and neutrophils (the presence of neutrophils can be associated with actinic keratosis, mechanical irritation of the tumor in some anatomical areas during scratching with teeth, but, in general, neutrophils are associated with more invasive squamous cell carcinoma). In cats, a markedly higher total number of mast cells was found, and the number was also higher in intratumoral and peritumoral tissues. A similar tendency was found in both dogs and cats-a markedly higher number of mastocytes was found in both peritumoral and intratumoral loose, well-vascularized connective tissue. Conversely, lower numbers of mast cells were found in both intratumoral and peritumoral compact fibrous tissue in both animal species.
Collapse
Affiliation(s)
- Nomeda Juodžiukynienė
- Department of Veterinary Pathobiology, Faculty of Veterinary Medicine, Veterinary Academy, Lithuanian University of Health Sciences, A.Mickevičius Str. 9, LT-44307 Kaunas, Lithuania;
| | - Kristina Lasienė
- Deparment of Histology and Embryology, Faculty of Medicine, Medical Academy, Lithuanian University of Health Sciences, A.Mickevičius Str. 9, LT-44307 Kaunas, Lithuania;
| | - Nijolė Savickienė
- Department of Pharmacognosy, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, A.Mickevičius Str. 9, LT-44307 Kaunas, Lithuania;
| | - Albina Aniulienė
- Department of Veterinary Pathobiology, Faculty of Veterinary Medicine, Veterinary Academy, Lithuanian University of Health Sciences, A.Mickevičius Str. 9, LT-44307 Kaunas, Lithuania;
| |
Collapse
|
16
|
Glaviano A, Lau HSH, Carter LM, Lee EHC, Lam HY, Okina E, Tan DJJ, Tan W, Ang HL, Carbone D, Yee MYH, Shanmugam MK, Huang XZ, Sethi G, Tan TZ, Lim LHK, Huang RYJ, Ungefroren H, Giovannetti E, Tang DG, Bruno TC, Luo P, Andersen MH, Qian BZ, Ishihara J, Radisky DC, Elias S, Yadav S, Kim M, Robert C, Diana P, Schalper KA, Shi T, Merghoub T, Krebs S, Kusumbe AP, Davids MS, Brown JR, Kumar AP. Harnessing the tumor microenvironment: targeted cancer therapies through modulation of epithelial-mesenchymal transition. J Hematol Oncol 2025; 18:6. [PMID: 39806516 PMCID: PMC11733683 DOI: 10.1186/s13045-024-01634-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025] Open
Abstract
The tumor microenvironment (TME) is integral to cancer progression, impacting metastasis and treatment response. It consists of diverse cell types, extracellular matrix components, and signaling molecules that interact to promote tumor growth and therapeutic resistance. Elucidating the intricate interactions between cancer cells and the TME is crucial in understanding cancer progression and therapeutic challenges. A critical process induced by TME signaling is the epithelial-mesenchymal transition (EMT), wherein epithelial cells acquire mesenchymal traits, which enhance their motility and invasiveness and promote metastasis and cancer progression. By targeting various components of the TME, novel investigational strategies aim to disrupt the TME's contribution to the EMT, thereby improving treatment efficacy, addressing therapeutic resistance, and offering a nuanced approach to cancer therapy. This review scrutinizes the key players in the TME and the TME's contribution to the EMT, emphasizing avenues to therapeutically disrupt the interactions between the various TME components. Moreover, the article discusses the TME's implications for resistance mechanisms and highlights the current therapeutic strategies toward TME modulation along with potential caveats.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Hannah Si-Hui Lau
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lukas M Carter
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Donavan Jia Jie Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Wency Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Michelle Yi-Hui Yee
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Xiao Zi Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Lina H K Lim
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Ruby Yun-Ju Huang
- School of Medicine and Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, 23538, Lübeck, Germany
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Experimental Therapeutics (ET) Graduate Program, University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Tullia C Bruno
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Bin-Zhi Qian
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, W12 0BZ, UK
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Salem Elias
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Saurabh Yadav
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Minah Kim
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Caroline Robert
- Department of Cancer Medicine, Inserm U981, Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif, France
- Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, Paris, France
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Tao Shi
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, NY, USA
| | - Simone Krebs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironment Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Matthew S Davids
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jennifer R Brown
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| |
Collapse
|
17
|
Zhang X, Zhang ZH, Liu YM, Zhao SL, Zhao XT, Zhang LZ, Gu CD, Zhao Y. Investigating lung cancer microenvironment from cell segmentation of pathological image and its application in prognostic stratification. Sci Rep 2025; 15:1704. [PMID: 39799232 PMCID: PMC11724888 DOI: 10.1038/s41598-025-85532-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025] Open
Abstract
Lung cancer, particularly adenocarcinoma, ranks high in morbidity and mortality rates worldwide, with a relatively low five-year survival rate. To achieve precise prognostic assessment and clinical intervention for patients, thereby enhancing their survival prospects, there is an urgent need for more accurate stratification schemes. Currently, the TNM staging system is predominantly used in clinical practice for prognostic evaluation, but its accuracy is constrained by the reliance on physician experience. Although biomarker discovery based on molecular pathology offers a new perspective for prognostic assessment, its dependence on expensive gene panel testing limits its widespread clinical application. Pathological images contain abundant diagnostic information, providing a new avenue for prognostic evaluation. In this study, we employed advanced Hover-Net technology to accurately quantify the abundance of epithelial cells, lymphocytes, macrophages, and neutrophils from pathological images, and delved into the clinical and biological significance of these cellular abundances. Our research findings reveal that, in contrast to patients classified as N0 stage, those belonging to the N1 stage demonstrated a marked elevation in the infiltration of epithelial cells, lymphocytes, macrophages, and neutrophils. Notably, the infiltration patterns of lymphocytes and neutrophils exhibited an inverse relationship with the activation status of numerous pivotal gene pathways, including the HALLMARK_HEME_METABOLISM pathway. Furthermore, our analysis distinguished FABP7 as a prognostic biomarker, exhibiting pronounced differential expression between patients with high and low levels of neutrophil infiltration, indicate that cellular abundance analysis based on pathological images can provide a more accurate and cost-effective prognostic evaluation, offering new strategies for the clinical management of lung adenocarcinoma.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116011, China
- Clinical Skills Center of Academic Affairs Office of Dalian Medical University, Dalian Medical University, Dalian, 116044, China
| | - Zi-Han Zhang
- Department of Intelligent Medical Engineering, Medical School of Tianjin University, Tianjin University, Tianjin, 300072, China
| | - Yong-Min Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116011, China
| | - Shi-Lei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116011, China
| | - Xu-Tong Zhao
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116011, China
| | - Li-Zhi Zhang
- Department of Pathology, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116011, China
| | - Chun-Dong Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116011, China.
| | - Yi Zhao
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
18
|
Zakurdaev EI, Bagateliya ZA, Titov KS, Elkhouli E, Chizhikov NP, Kharina DV. The Role of Tumor-Associated Neutrophils in Early Luminal HER2-Negative Breast Cancer Progression. Asian Pac J Cancer Prev 2025; 26:207-213. [PMID: 39874003 PMCID: PMC12082427 DOI: 10.31557/apjcp.2025.26.1.207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 01/11/2025] [Indexed: 01/30/2025] Open
Abstract
OBJECTIVES To study the predictive role of tumor-associated neutrophils in early luminal HER2-negative breast cancer. MATERIALS AND METHODS This is a retrospective study conducted on 60 women cases aged from 31 to 79 years underwent surgery for luminal HER2-negative ductal breast cancer in tertiary care cancer centre. We first estimated basic morphological signs: tumor size, tumor grade (by Nottingham Histologic Score), tumor infiltrating lymphocytes (TILs), Lymphovascular invasion, hormonal receptors status, proliferative index, and regional lymph nodes metastasis. The expression of intratumoral neutrophils was studied by CD15 immunohistochemistry which was performed using tissue microarrays. The total number of intratumoral neutrophils, were counted in 5 high-power fields. RESULTS According to the Nottingham histologic score system, grade I cases were detected in 10 cases (16%), grade II in 34 cases (57%), and grade III in 16 cases (27%). Lymphovascular invasion was determined in 23 cases (38%), and perineural invasion in 14 cases (23%). Number of TILs varied from 0 to 14 (counted in 5 HPF) and averaged 4.2±0.5. Luminal A tumor phenotype was detected in 35 cases (58%), and luminal B HER2-negative in 25 cases (42%). Nineteen (32%) women had metastases in regional lymph nodes (N+). The number of tumor microenvironment neutrophils in luminal HER2-negative breast carcinomas ranged from 1 to 10 (counted in 5 HPF) with an average value of 2.7±0.4. High tumor-associated neutrophils concentration significantly correlated with tumor size (<5mm and >20mm) with p=0.05, high grade (p=0.01), high proliferative index ((r=0.67; p=0.05), TILs (p=0.05), Lymphovascular space invasion (p=0.01)and positive regional lymph nodes metastasis (p=0.001), but not perineural invasion (p=0.1) and also, did not correlate with the expression of estrogen (r=0.18) and progesterone (r=0.14) receptors. CONCLUSION Tumor-associated neutrophils strongly predict a worse prognosis in early luminal HER2-negative breast cancer.
Collapse
MESH Headings
- Humans
- Female
- Neutrophils/pathology
- Neutrophils/metabolism
- Neutrophils/immunology
- Middle Aged
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Breast Neoplasms/surgery
- Retrospective Studies
- Receptor, ErbB-2/metabolism
- Aged
- Adult
- Lymphatic Metastasis
- Prognosis
- Disease Progression
- Follow-Up Studies
- Lymphocytes, Tumor-Infiltrating/pathology
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/secondary
- Carcinoma, Ductal, Breast/surgery
- Biomarkers, Tumor/metabolism
- Neoplasm Invasiveness
- Receptors, Estrogen/metabolism
Collapse
Affiliation(s)
| | | | | | - Ekbal Elkhouli
- Pathology Department, Mansoura University, Mansoura, Egypt.
| | | | | |
Collapse
|
19
|
Song D, Yin X, Che C. Distinct Gene Expression and Immune Features Between Different Neutrophil Extracellular Trap-Related Osteosarcoma Subtypes. Appl Biochem Biotechnol 2025; 197:55-72. [PMID: 39096473 DOI: 10.1007/s12010-024-05021-2] [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: 07/23/2024] [Indexed: 08/05/2024]
Abstract
We sought to determine neutrophil extracellular trap (NET)-related genes' potential value in improving the efficacy of diagnosis and identifying novel therapeutic targets for osteosarcoma. Data were obtained from TARGET, GEO, and CCLE database. Differentially expressed genes were identified between the subtypes based on NET-related genes. PPI network was constructed using STRING, following by ClueGO enrichment analysis. Infiltration of immune cells was calculated by ssGSEA. Risk Score model was built by LASSO Cox regression analysis. Western blot and qRT-PCR were applied to validate the expression of genes used in the model. We identified 19 NET-related genes with prognostic potential in osteosarcoma using univariate Cox regression analysis. Patients from TARGET were clustered into two subtypes with distinct prognosis and immune features. 381 DEGs were identified between the two NET subtypes. Risk Score based on BST1, SELPLG, FPR1 and TNFRSF10C was reliable to predict the prognosis of osteosarcoma patients. The four genes expressed significantly lower in osteosarcoma than normal cells. Low Risk Score individuals only existed in C1 subtype with better prognosis. Osteosarcoma were clustered into two subtypes based on NET-related genes. Risk Score model constructed by four NET-related gene was able to independently predict the prognosis of osteosarcoma.
Collapse
Affiliation(s)
- Delei Song
- Department of West Hospital Orthopaedic Trauma, Zibo Central Hospital, No. 54 Gongqingtuan Road, Zibo, 255036, China
| | - Xuqing Yin
- Department of East Hospital Orthopaedic Trauma, Zibo Central Hospital, Zibo, 255036, China
| | - Chunqing Che
- Department of West Hospital Orthopaedic Trauma, Zibo Central Hospital, No. 54 Gongqingtuan Road, Zibo, 255036, China.
| |
Collapse
|
20
|
Chakra MA, Lassila R, El Beayni N, Mott SL, O'Donnell MA. Prognostic role of the neutrophil/lymphocyte ratio in high-risk BCG-naïve non-muscle-invasive bladder cancer treated with intravesical gemcitabine/docetaxel. BJU Int 2025; 135:125-132. [PMID: 39082304 DOI: 10.1111/bju.16486] [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: 10/17/2024]
Abstract
OBJECTIVES To investigate the role of pretreatment neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) in the prediction of response to sequential intravesical therapy, gemcitabine and docetaxel (Gem/Doce), given to patients with bacille Calmette-Guérin (BCG)- naïve high-risk non-muscle-invasive bladder cancer (NMIBC). PATIENTS AND METHODS A retrospective analysis was conducted on 115 patients who received intravesical Gem/Doce for high-risk NMIBC between January 2011 and December 2021. Data were computed as the median (interquartile range [IQR]) or mean (standard deviation [sd]). Cox regression analysis was performed to determine if neutrophilia, NLR, platelet counts, and PLR before instillation therapy were predictive of recurrence-free survival (RFS) and overall survival (OS). Predictive performance was estimated using Uno's C-statistic. RESULTS The median (IQR) follow-up for the overall cohort was 23 (13-36) months. The mean (sd) values for NLR, PLR and platelet counts were 3.4 (2.3), 142.2 (85.5), and 225.2 (75.1) × 109/L, respectively. NLR was associated with RFS, with a hazard ratio of 1.32 (95% confidence interval CI 1.19-1.46). Concordance analysis showed that NLR had a good ability to predict RFS (C-index: 0.7, P < 0.01). The PLR and platelet count were not associated with RFS and did not predict recurrence. In terms of OS, none of these cellular inflammatory markers showed any prediction value. CONCLUSION Pre-treatment NLR provides some predictive accuracy for RFS in high-risk BCG-naïve patients receiving Gem/Doce. Further prospective trials are needed to validate this finding.
Collapse
Affiliation(s)
- Mohamad Abou Chakra
- Department of Urology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Riitta Lassila
- Unit of Coagulation Disorders, Department of Hematology, Helsinki University Central Hospital, Helsinki, Finland
- Research Program Unit in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Nancy El Beayni
- Unit of Coagulation Disorders, Department of Hematology, Helsinki University Central Hospital, Helsinki, Finland
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Sarah L Mott
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Michael A O'Donnell
- Department of Urology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| |
Collapse
|
21
|
Shindyapin VV, Gubernatorova EO, Gorshkova EA, Chicherina NR, Sysonov FA, Yakovleva AS, Bogdanova DA, Demidov ON, Samsonova MV, Baklaushev VP, Yusubalieva GM, Drutskaya MS. Myeloid Cell Mobilization and Recruitment by Human Mesothelioma in NSG-SGM3 Mice. Cells 2024; 13:2135. [PMID: 39768223 PMCID: PMC11675005 DOI: 10.3390/cells13242135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Malignant pleural mesothelioma is a neoplasm that is often detected late due to nonspecific symptoms. This study utilized NSG-SGM3 mice to examine interactions between a human-derived mesothelioma reporter cell line (MZT-Luc2-mCherry) and the host's myeloid compartment. Tumor growth was assessed using optical tomography, while cytokine/chemokine production was analyzed via multiplex assay. Histological and immunohistochemical analyses validated the epithelioid mesothelioma phenotype. In vitro mesothelioma cells secreted factors associated with myeloid cell chemoattraction and functions supporting the previously reported myeloid-biased secretory phenotype. In line with this, post-engraftment analysis revealed increased neutrophil-like Ly6G+ populations and decreased Ly6C+ inflammatory monocytes in the blood of tumor-bearing mice. Significant Ly6G+ cell infiltration was observed in the tumor, while CD11b+ myeloid cells were localized primarily in the tumor periphery. Tumor lysates showed increased levels of neutrophil chemoattractants and G-CSF, suggesting a previously not reported role of neutrophils in mesothelioma progression. This novel model provides a platform for studying mesothelioma-host interactions, focusing on the myeloid compartment. It may also serve as a tool to facilitate the development of new therapeutic strategies targeting myeloid cell-mediated mechanisms in mesothelioma.
Collapse
Affiliation(s)
- Vadim V. Shindyapin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.O.G.); (E.A.G.); (F.A.S.); (A.S.Y.); (V.P.B.); (G.M.Y.)
- Department of Immunobiology and Biomedicine, Scientific Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, 354340 Krasnodar Krai, Russia; (N.R.C.); (D.A.B.); (O.N.D.)
| | - Ekaterina O. Gubernatorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.O.G.); (E.A.G.); (F.A.S.); (A.S.Y.); (V.P.B.); (G.M.Y.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Ekaterina A. Gorshkova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.O.G.); (E.A.G.); (F.A.S.); (A.S.Y.); (V.P.B.); (G.M.Y.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Nelya R. Chicherina
- Department of Immunobiology and Biomedicine, Scientific Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, 354340 Krasnodar Krai, Russia; (N.R.C.); (D.A.B.); (O.N.D.)
| | - Fedor A. Sysonov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.O.G.); (E.A.G.); (F.A.S.); (A.S.Y.); (V.P.B.); (G.M.Y.)
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Anastasia S. Yakovleva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.O.G.); (E.A.G.); (F.A.S.); (A.S.Y.); (V.P.B.); (G.M.Y.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Daria A. Bogdanova
- Department of Immunobiology and Biomedicine, Scientific Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, 354340 Krasnodar Krai, Russia; (N.R.C.); (D.A.B.); (O.N.D.)
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia
| | - Oleg N. Demidov
- Department of Immunobiology and Biomedicine, Scientific Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, 354340 Krasnodar Krai, Russia; (N.R.C.); (D.A.B.); (O.N.D.)
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia
- INSERM UMR1231, University of Burgundy, 21078 Dijon, France
| | - Mariya V. Samsonova
- Pulmonology Research Institute, Federal Medical-Biological Agency of Russian Federation, 115682 Moscow, Russia;
| | - Vladimir P. Baklaushev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.O.G.); (E.A.G.); (F.A.S.); (A.S.Y.); (V.P.B.); (G.M.Y.)
- Pulmonology Research Institute, Federal Medical-Biological Agency of Russian Federation, 115682 Moscow, Russia;
- Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies, Federal Medical-Biological Agency of Russian Federation, 123098 Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical-Biological Agency of Russian Federation, 117513 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.O.G.); (E.A.G.); (F.A.S.); (A.S.Y.); (V.P.B.); (G.M.Y.)
- Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies, Federal Medical-Biological Agency of Russian Federation, 123098 Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical-Biological Agency of Russian Federation, 117513 Moscow, Russia
| | - Marina S. Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.O.G.); (E.A.G.); (F.A.S.); (A.S.Y.); (V.P.B.); (G.M.Y.)
- Department of Immunobiology and Biomedicine, Scientific Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, 354340 Krasnodar Krai, Russia; (N.R.C.); (D.A.B.); (O.N.D.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
22
|
Wu Y, Shang J, Zhang X, Li N. Advances in molecular imaging and targeted therapeutics for lymph node metastasis in cancer: a comprehensive review. J Nanobiotechnology 2024; 22:783. [PMID: 39702277 PMCID: PMC11657939 DOI: 10.1186/s12951-024-02940-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 10/19/2024] [Indexed: 12/21/2024] Open
Abstract
Lymph node metastasis is a critical indicator of cancer progression, profoundly affecting diagnosis, staging, and treatment decisions. This review article delves into the recent advancements in molecular imaging techniques for lymph nodes, which are pivotal for the early detection and staging of cancer. It provides detailed insights into how these techniques are used to visualize and quantify metastatic cancer cells, resident immune cells, and other molecular markers within lymph nodes. Furthermore, the review highlights the development of innovative, lymph node-targeted therapeutic strategies, which represent a significant shift towards more precise and effective cancer treatments. By examining cutting-edge research and emerging technologies, this review offers a comprehensive overview of the current and potential impact of lymph node-centric approaches on cancer diagnosis, staging, and therapy. Through its exploration of these topics, the review aims to illuminate the increasingly sophisticated landscape of cancer management strategies focused on lymph node assessment and intervention.
Collapse
Affiliation(s)
- Yunhao Wu
- Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Jin Shang
- Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xinyue Zhang
- The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Nu Li
- The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
23
|
Qian L, Ji Z, Mei L, Zhao J. IGF2BP2 promotes lung adenocarcinoma progression by regulating LOX1 and tumor-associated neutrophils. Immunol Res 2024; 73:16. [PMID: 39688738 DOI: 10.1007/s12026-024-09563-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 11/07/2024] [Indexed: 12/18/2024]
Abstract
Lung adenocarcinoma (LUAD) is the common form of lung cancer and is prone to distant metastasis. IGF2BP2, an m6A modification regulator, is upregulated in lung cancer tissue, but its specific role within the LUAD tumor microenvironment (TME) is unknown. We explored the role of IGF2BP2 in the progression of LUAD. IGF2BP2 expression in LUAD patient specimens and controls was evaluated through bioinformatics, Western blot, and immunohistochemistry. LUAD subcutaneous and orthotopic xenograft tumor models were established, alongside a co-culture system of tumor-associated neutrophils (TANs) and A549 cells. Functional assays assessed IGF2BP2's role under treatment with JX5, an IGF2BP2 inhibitor. Mechanistic assays explored the interaction between IGF2BP2 and LOX1 in 293T cells. IGF2BP2 was significantly upregulated in LUAD tissues, with higher expression levels predicting worse prognosis for patients (p < 0.001). In subcutaneous and orthotopic xenograft models, treatment with JX5, an IGF2BP2 inhibitor, reduced tumor size, volume, and weight (p < 0.001). JX5 also significantly reduced the concentrations of pro-inflammatory cytokines in peripheral blood (p < 0.01 and p < 0.001). Flow cytometry analysis indicated JX5 reduced CD11b+Ly6G+/CD45+ cells (TANs) in the TME (p < 0.001). Mechanistically, JX5 downregulated LOX1 expression in vivo, and co-culture experiments further demonstrated that IGF2BP2 promotes LUAD progression through LOX1-mediated regulation of TAN activity (p < 0.01 and p < 0.001). Overexpression of LOX1 reversed the inhibitory effects of JX5 on TAN infiltration, tumor cell viability, and apoptosis (p < 0.01 and p < 0.001). Additionally, RNA immunoprecipitation revealed that IGF2BP2 binds to LOX1 mRNA at its m6A modification site, stabilizing LOX1 and enhancing its function in the TME. Knockdown of IGF2BP2 accelerated LOX1 mRNA degradation, confirming its role in maintaining LOX1 stability (p < 0.01 and p < 0.001). IGF2BP2 recognizes and stabilizes LOX1 through m6A modification, contributing to TAN-mediated LUAD progression. Overall, these findings offer new insights into LUAD progression and demonstrate that IGF2BP2 is a key regulator that promotes tumor advancement, highlighting the IGF2BP2-LOX1 axis as a potential therapeutic target for LUAD.
Collapse
Affiliation(s)
- Long Qian
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, 215006, Jiangsu, China
- Department of Thoracic Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, 223300, China
| | - Zhuqing Ji
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
- Department of Medical Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, 223300, China
| | - Lingyun Mei
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
24
|
Maggi E, Munari E, Landolina N, Mariotti FR, Azzarone B, Moretta L. T cell landscape in the microenvironment of human solid tumors. Immunol Lett 2024; 270:106942. [PMID: 39486594 DOI: 10.1016/j.imlet.2024.106942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024]
Abstract
T cells are the main effectors involved in anti-tumor immunity, mediating most of the adaptive response towards cancer. After priming in lymph nodes, tumor antigens-specific naïve T lymphocytes proliferate and differentiate into effector CD4+ and CD8+ T cells that migrate from periphery into tumor sites aiming to eliminate cancer cells. Then while most effector T cells die, a small fraction persists and recirculates as long-lived memory T cells which generate enhanced immune responses when re-encountering the same antigen. A number of T (and non-T) cell subsets, stably resides in non-lymphoid peripheral tissues and may provide rapid immune response independently of T cells recruited from blood, against the reemergence of cancer cells. When tumor grows, however, tumor cells have evaded immune surveillance of effector cells (NK and CTL cells) which are exhausted, thus favoring the local expansion of T (and non-T) regulatory cells. In this review, the current knowledge of features of T cells present in the tumor microenvironment (TME) of solid adult and pediatric tumors, the mechanisms upregulating immune-checkpoint molecules and transcriptional and epigenetic landscapes leading to dysfunction and exhaustion of T effector cells are reviewed. The interaction of T cells with cancer- or TME non-neoplastic cells and their secreted molecules shape the T cell profile compromising the intrinsic plasticity of T cells and, therefore, favoring immune evasion. In this phase regulatory T cells contribute to maintain a high immunosuppressive TME thus facilitating tumor cell proliferation and metastatic spread. Despite the advancements of cancer immunotherapy, many tumors are unresponsive to immune checkpoint inhibitors, or therapeutical vaccines or CAR T cell-based adoptive therapy: some novel strategies to improve these T cell-based treatments are lastly proposed.
Collapse
Affiliation(s)
- Enrico Maggi
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS 00146 Rome, Italy
| | - Enrico Munari
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona 37126, Italy
| | - Nadine Landolina
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS 00146 Rome, Italy
| | | | - Bruno Azzarone
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS 00146 Rome, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS 00146 Rome, Italy.
| |
Collapse
|
25
|
Eren E, Das J, Tollefsbol TO. Polyphenols as Immunomodulators and Epigenetic Modulators: An Analysis of Their Role in the Treatment and Prevention of Breast Cancer. Nutrients 2024; 16:4143. [PMID: 39683540 PMCID: PMC11644657 DOI: 10.3390/nu16234143] [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: 11/05/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Breast cancer poses a substantial health challenge for women globally. Recently, there has been a notable increase in scholarly attention regarding polyphenols, primarily attributed to not only the adverse effects associated with conventional treatments but also their immune-preventive impacts. Polyphenols, nature-derived substances present in vegetation, including fruits and vegetables, have received considerable attention in various fields of science due to their probable wellness merits, particularly in the treatment and hindrance of cancer. This review focuses on the immunomodulatory effects of polyphenols in breast cancer, emphasizing their capacity to influence the reaction of adaptive and innate immune cells within the tumor-associated environment. Polyphenols are implicated in the modulation of inflammation, the enhancement of antioxidant defenses, the promotion of epigenetic modifications, and the support of immune functions. Additionally, these compounds have been shown to influence the activity of critical immune cells, including macrophages and T cells. By targeting pathways involved in immune evasion, polyphenols may augment the capacity of the defensive system to detect and eliminate tumors. The findings suggest that incorporating polyphenol-rich foods into the diet could offer a promising, collaborative (integrative) approach to classical breast cancer remedial procedures by regulating how the defense mechanism interacts with the disease.
Collapse
Affiliation(s)
- Esmanur Eren
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (E.E.); (J.D.)
| | - Jyotirmoyee Das
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (E.E.); (J.D.)
| | - Trygve O. Tollefsbol
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (E.E.); (J.D.)
- Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- O’Neal Comprehensive Cancer Research, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
26
|
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
|
27
|
Yang S, Fei C. Prognostic value of systemic immune-inflammation index and systemic inflammation response index for oral cancers: A systematic review and meta-analysis. Med Oral Patol Oral Cir Bucal 2024; 29:e822-e831. [PMID: 39396135 PMCID: PMC11584967 DOI: 10.4317/medoral.26779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/16/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND The systemic immune-inflammation index (SII) and systemic inflammation response index (SIRI) are commonly used prognostic indicators for a variety of cancers. However, their utility in oral cancers is unknown. We systematically examined evidence on the ability of SII and SIRI to predict overall survival (OS) and disease-free survival (DFS) after oral cancers. MATERIAL AND METHODS Embase, PubMed, Web of Science, and Scopus were searched for oral cancer studies reporting OS or DFS based on SII or SIRI. Articles published up to 25th May 2024 were included. RESULTS 17 studies were eligible (14 on SII and 3 on SIRI). Pretreatment high SII scores were found to be significantly linked with poor OS (HR: 1.62 95% 1.26, 2.08 I2=88%) and DFS (HR: 1.62 95% 1.25, 2.27 I2=86%) after oral cancer. Similarly, high SIRI was associated with worse OS in oral cancer patients (HR: 1.60 95% 1.31, 1.94 I2=0%). All results were unchanged on sensitivity analysis. Subgroup analysis based on location, cancer type, sample size, treatment, cut-off, methods of determining cut-off, analysis method, and study quality showed mixed results. CONCLUSIONS Acknowledging the limitations of current evidence, it seems that both SII and SIRI can predict the prognosis of oral cancers. High SII and SIRI are both associated with worse OS while high SII also predicts worse DFS.
Collapse
Affiliation(s)
- S Yang
- Department of Stomatology The second affiliated hospital of Jiaxing university 1518 Huancheng North Road, Jianshe Street Nanhu District, Jiaxing City, Zhejiang Province, China
| | | |
Collapse
|
28
|
Li GP, Zhang D, Li MH, Yuan FF, Hou XJ, He DJ, Wei XD, Fu YW. Association between the neutrophil-to-lymphocyte ratio and cancer in adults from NHANES 2005-2018: a cross-sectional study. Sci Rep 2024; 14:23678. [PMID: 39390050 PMCID: PMC11467198 DOI: 10.1038/s41598-024-75252-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024] Open
Abstract
Inflammation plays a crucial role in cancer development. The neutrophil-to-lymphocyte ratio (NLR), a measure of inflammation, is obtained from a complete blood count. However, little is known about the association between NLR and cancer in the general adult population in the United States. This study aimed to evaluate whether NLR is associated with cancer in American adults. This retrospective cross-sectional study included 28,016 adult participants from the National Health and Nutrition Examination Survey (NHANES) dataset spanning 2005 to 2018. Data on demographics (age, sex, race, marital status, Poverty-Income Ratio, education level), lifestyle factors (smoking, alcohol consumption, body mass index), medical conditions (hypertension, diabetes, cardiovascular disease), and laboratory parameters (hemoglobin, platelet count, alanine aminotransferase, creatinine, albumin, and lactate dehydrogenase), were collected. Logistic regression analysis was used to investigate the research objectives. Of the total 28,016 participants, 2639 had cancer. The mean age was 49.6 ± 17.6 years, and 50% were male. A positive association between NLR and cancer risk was observed after multivariate adjustment (OR = 1.20, 95% confidence interval (CI) = 1.05-1.36, p = 0.006). Similar patterns were observed in subgroup analyses (all p-values for interaction > 0.05). A higher NLR was directly correlated with an increased risk of developing cancer in adults.
Collapse
Affiliation(s)
- Gang-Ping Li
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, China
| | - Di Zhang
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, China
| | - Ming-Hui Li
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, China
| | - Fang-Fang Yuan
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, China
| | - Xin-Jiang Hou
- College of Medicine, Xi'an International University, Xi'an, 710077, China
| | - Dong-Jie He
- Department of Radiation Oncology, Tangdu Hospital, the Second Affiliated Hospital of Air Force Military Medical University, No. 1 Xinsi Road, Xi'an, 710077, China.
| | - Xu-Dong Wei
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, China.
| | - Yue-Wen Fu
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, China.
| |
Collapse
|
29
|
Yang Y, Yang J, Li L, Shao Y, Liu L, Sun B. Neutrophil chemotaxis score and chemotaxis-related genes have the potential for clinical application to prognosticate the survival of patients with tumours. BMC Cancer 2024; 24:1244. [PMID: 39379856 PMCID: PMC11463147 DOI: 10.1186/s12885-024-12993-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/25/2024] [Indexed: 10/10/2024] Open
Abstract
As frontline cells, the precise recruitment of neutrophils is crucial for resolving inflammation and maintaining the homeostasis of the organism. Increasing evidence suggests the pivotal role of neutrophil chemotaxis in cancer progression and metastasis. Here, we collected clinical data and peripheral blood samples from patients with tumours to examine the alterations in the neutrophil quantity and chemotactic function using the Cell Chemotaxis Analysis Platform (CCAP). Transcriptome sequencing data of pan-cancer were obtained from The Cancer Genome Atlas (TCGA). Using the least absolute shrinkage and selection operator (LASSO) Cox regression model, we selected a total of 29 genes from 155 neutrophil- and chemotaxis-related genes to construct the ChemoScore model. Meanwhile, nomogram-based comprehensive model was established for clinical application. Furthermore, immunofluorescence (IF) staining was employed to assess the relationship between the neutrophils infiltrating and the survival outcomes of tumours. In this observational study, the chemotactic function of neutrophils was notably diminished in patients. The establishment and validation of ChemoScore suggested neutrophil chemotaxis to be a risk factor in most tumours, whereby higher scores were associated with poorer survival outcomes and were correlated with various immune cells and malignant biological processes. Moreover, IF staining of tumour tissue substantiated the adverse correlation between neutrophil infiltration and the survival of patients with lung adenocarcinoma (P = 0.0002) and colon adenocarcinoma (P = 0.0472). Taken together, patients with tumours demonstrated a decrease in chemotactic function. ChemoScore potentially prognosticates the survival of patients with tumours. Neutrophil chemotaxis provides novel directions and theoretical foundations for anti-tumour treatment.
Collapse
Affiliation(s)
- Yunxi Yang
- Research Center for Neutrophil Engineering Technology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, 215002, China
| | - Jun Yang
- Department of Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, 215002, China
| | - Linbin Li
- Research Center for Neutrophil Engineering Technology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, 215002, China
| | - Yiming Shao
- Research Center for Neutrophil Engineering Technology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, 215002, China
- Department of Burns and Plastic Surgery, The Affiliated Hospital of Jining Medical University, Jining, Shandong Province, 272000, China
| | - Lu Liu
- Research Center for Neutrophil Engineering Technology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, 215002, China
| | - Bingwei Sun
- Research Center for Neutrophil Engineering Technology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, 215002, China.
| |
Collapse
|
30
|
Lim MS, Crimmins D. The prognostic utility of the neutrophil to lymphocyte ratio in paediatric brain tumours: a retrospective case control study. Br J Neurosurg 2024:1-7. [PMID: 39324393 DOI: 10.1080/02688697.2024.2406804] [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: 04/08/2024] [Revised: 08/20/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION Paediatric brain tumours (PBT) are the most common cause of death among all childhood cancers. The neutrophil to lymphocyte ratio (NLR) has been shown to prognosticate many adult cancers. There is a paucity of literature on the NLR in PBTs. This study aims to study the link between PBTs and the NLR by comparing the preoperative serum NLR in children under 16 with brain tumours with their outcome in terms of grade of brain tumour and overall survival. METHODS This is a retrospective case control study. The NLRs were compared between patients with benign or malignant PBTs and patients who were alive or dead. Receiver-operating characteristic (ROC) curve analyses were performed and Youden indexes were calculated to evaluate the predictive potential of the NLR. A cut-off point of NLR > 4 was selected for the calculation of odds ratios. RESULTS A total of 515 patients were included in this study. 53.8% were male. 66.2% had benign PBTs. 81.0% were alive at the time of the study. Patients with malignant PBTs had a higher NLR compared to patients with benign PBTs (p = 0.0066**). There was no difference in the NLR between patients who were dead compared to those who were alive (p = 0.1682 ns). The NLR had a Youden's index of 0.1567 to predict malignant PBTs and 0.1285 to predict survival. CONCLUSION A high NLR was associated with an increased odds of having a malignant PBT but a reliable cut-off point was not identified and the underlying mechanisms for this remain unknown. The NLR is a poor diagnostic biomarker due to its poor overall sensitivity and specificity. More research is required to further study the role of immunity in PBTs.
Collapse
Affiliation(s)
- Ming-Sheng Lim
- Department of Neurosurgery, Temple Street Children's Hospital, Dublin, Ireland
- Department of Neurosurgery, Beaumont Hospital, Dublin, Ireland
| | - Darach Crimmins
- Department of Neurosurgery, Temple Street Children's Hospital, Dublin, Ireland
- Department of Neurosurgery, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
31
|
Zhong Y, Chen L, Ding F, Ou W, Zhang X, Weng S. Assessing microvascular invasion in HBV-related hepatocellular carcinoma: an online interactive nomogram integrating inflammatory markers, radiomics, and convolutional neural networks. Front Oncol 2024; 14:1401095. [PMID: 39351352 PMCID: PMC11439624 DOI: 10.3389/fonc.2024.1401095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 08/22/2024] [Indexed: 10/04/2024] Open
Abstract
Objective The early recurrence of hepatocellular carcinoma (HCC) correlates with decreased overall survival. Microvascular invasion (MVI) stands out as a prominent hazard influencing post-resection survival status and metastasis in patients with HBV-related HCC. The study focused on developing a web-based nomogram for preoperative prediction of MVI in HBV-HCC. Materials and methods 173 HBV-HCC patients from 2017 to 2022 with complete preoperative clinical data and Gadopentetate dimeglumine-enhanced magnetic resonance images were randomly divided into two groups for the purpose of model training and validation, using a ratio of 7:3. MRI signatures were extracted by pyradiomics and the deep neural network, 3D ResNet. Clinical factors, blood-cell-inflammation markers, and MRI signatures selected by LASSO were incorporated into the predictive nomogram. The evaluation of the predictive accuracy involved assessing the area under the receiver operating characteristic (ROC) curve (AUC), the concordance index (C-index), along with analyses of calibration and decision curves. Results Inflammation marker, neutrophil-to-lymphocyte ratio (NLR), was positively correlated with independent MRI radiomics risk factors for MVI. The performance of prediction model combined serum AFP, AST, NLR, 15 radiomics features and 7 deep features was better than clinical and radiomics models. The combined model achieved C-index values of 0.926 and 0.917, with AUCs of 0.911 and 0.907, respectively. Conclusion NLR showed a positive correlation with MRI radiomics and deep learning features. The nomogram, incorporating NLR and MRI features, accurately predicted individualized MVI risk preoperatively.
Collapse
Affiliation(s)
- Yun Zhong
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Lingfeng Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Fadian Ding
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Wenshi Ou
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiang Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shangeng Weng
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
32
|
Koc DC, Mănescu IB, Mănescu M, Dobreanu M. A Review of the Prognostic Significance of Neutrophil-to-Lymphocyte Ratio in Nonhematologic Malignancies. Diagnostics (Basel) 2024; 14:2057. [PMID: 39335736 PMCID: PMC11431542 DOI: 10.3390/diagnostics14182057] [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: 07/01/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Biomarkers are crucial in cancer diagnostics, prognosis, and surveillance. Extensive research has been dedicated to identifying biomarkers that are broadly applicable across multiple cancer types and can be easily obtained from routine investigations such as blood cell counts. One such biomarker, the neutrophil-to-lymphocyte ratio (NLR), has been established as a prognostic marker in cancer. However, due to the dynamic nature of cancer diagnosis and treatment, periodic updates are necessary to keep abreast of the vast amount of published data. In this review, we searched the PubMed database and analyzed and synthesized recent literature (2018-February 2024) on the role of NLR in predicting clinical outcomes in nonhematologic malignancies. The search was conducted using the PubMed database. We included a total of 88 studies, encompassing 28,050 human subjects, and categorized the findings into four major groups: gastrointestinal cancer, cancers of the urinary tract and reproductive system, lung cancer, and breast cancer. Our analysis confirms that NLR is a reliable prognostic indicator in cancer, and we discuss the specific characteristics, limitations, and exceptions associated with its use. The review concludes with a concise Q&A section, presenting the most relevant take-home messages in response to five key practical questions on this topic.
Collapse
Affiliation(s)
- Defne Cigdem Koc
- Medical Campus Hamburg, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 11-15 Albert-Einstein-Ring, 22761 Hamburg, Germany; (D.C.K.); (I.B.M.)
| | - Ion Bogdan Mănescu
- Medical Campus Hamburg, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 11-15 Albert-Einstein-Ring, 22761 Hamburg, Germany; (D.C.K.); (I.B.M.)
- Department of Laboratory Medicine, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 38 Gheorghe Marinescu, 540142 Targu Mures, Romania
| | - Măriuca Mănescu
- Department of Pediatrics, Emergency County Clinical Hospital of Targu Mures, 50 Gheorghe Marinescu, 540136 Targu Mures, Romania
| | - Minodora Dobreanu
- Department of Laboratory Medicine, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 38 Gheorghe Marinescu, 540142 Targu Mures, Romania
- Clinical Laboratory, Emergency County Clinical Hospital of Targu Mures, 50 Gheorghe Marinescu, 540136 Targu Mures, Romania
| |
Collapse
|
33
|
Bharadwaj D, Mandal M. Tumor microenvironment: A playground for cells from multiple diverse origins. Biochim Biophys Acta Rev Cancer 2024; 1879:189158. [PMID: 39032537 DOI: 10.1016/j.bbcan.2024.189158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Tumor microenvironment is formed by various cellular and non-cellular components which interact with one another and form a complex network of interactions. Some of these cellular components also attain a secretory phenotype and release growth factors, cytokines, chemokines etc. in the surroundings which are capable of inducing even greater number of signalling networks. All these interactions play a decisive role in determining the course of tumorigenesis. The treatment strategies against cancer also exert their impact on the local microenvironment. Such interactions and anticancer therapies have been found to induce more deleterious outcomes like immunosuppression and chemoresistance in the process of tumor progression. Hence, understanding the tumor microenvironment is crucial for dealing with cancer and chemoresistance. This review is an attempt to develop some understanding about the tumor microenvironment and different factors which modulate it, thereby contributing to tumorigenesis. Along with summarising the major components of tumor microenvironment and various interactions taking place between them, it also throws some light on how the existing and potential therapies exert their impact on these dynamics.
Collapse
Affiliation(s)
- Deblina Bharadwaj
- Department of Biotechnology, KIT-Kalaignarkarunanidhi Institute of Technology, Coimbatore, Tamil Nadu, India.
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, India.
| |
Collapse
|
34
|
Menyhart O, Fekete JT, Győrffy B. Inflammation and Colorectal Cancer: A Meta-Analysis of the Prognostic Significance of the Systemic Immune-Inflammation Index (SII) and the Systemic Inflammation Response Index (SIRI). Int J Mol Sci 2024; 25:8441. [PMID: 39126008 PMCID: PMC11312822 DOI: 10.3390/ijms25158441] [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: 06/06/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 08/12/2024] Open
Abstract
The overall prognosis for colorectal cancer (CRC) remains challenging as the survival time varies widely, even in patients with the same stage of disease. Recent studies suggest prognostic relevance of the novel markers of systemic inflammation, the systemic immune-inflammation index (SII), and the systemic inflammation response index (SIRI). We conducted a comprehensive meta-analysis to assess the prognostic significance of the SII and the SIRI in CRC. We searched the relevant literature for observational studies, and random effects models were employed to conduct a statistical analysis using the metaanalysisonline.com platform. Pooled effect sizes were reported with hazard ratios (HRs) and corresponding 95% confidence intervals (CI). Data from 29 studies published between 2016 and 2024, comprising 10,091 participants, were included in our meta-analysis on SII. CRC patients with high SII levels had worse disease outcomes, which were associated with poor OS (HR: 1.75; 95% CI: 1.4-2.19) and poor PFS/DFS/RFS (HR: 1.25; 95% CI: 1.18-1.33). This increased risk of worse OS was present irrespective of the treatment strategy, sample size (<220 and ≥220), and cutoff used to define high and low SII (<550 and ≥550) groups. Based on data from five studies comprising 2362 participants, we found a strong association between the high SIRI and worse OS (HR: 2.65; 95% CI: 1.6-4.38) and DFS/RFS (HR: 2.04; 95% CI: 1.42-2.93). According to our results, both the SII and SIRI hold great promise as prognostic markers in CRC. Further validations are needed for their age- and stage-specific utility in the clinical routine.
Collapse
Affiliation(s)
- Otilia Menyhart
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, Hungarian Research Network, 1117 Budapest, Hungary; (O.M.); (J.T.F.)
- Department of Bioinformatics, Semmelweis University, 1094 Budapest, Hungary
| | - János Tibor Fekete
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, Hungarian Research Network, 1117 Budapest, Hungary; (O.M.); (J.T.F.)
- Department of Bioinformatics, Semmelweis University, 1094 Budapest, Hungary
| | - Balázs Győrffy
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, Hungarian Research Network, 1117 Budapest, Hungary; (O.M.); (J.T.F.)
- Department of Bioinformatics, Semmelweis University, 1094 Budapest, Hungary
- Department of Biophysics, Medical School, University of Pecs, 7624 Pecs, Hungary
| |
Collapse
|
35
|
Zhang Y, Lee C, Geng S, Wang J, Bohara U, Hou J, Yi Z, Li L. Immune-enhancing neutrophils reprogrammed by subclinical low-dose endotoxin in cancer treatment. EMBO Mol Med 2024; 16:1886-1900. [PMID: 39009886 PMCID: PMC11319772 DOI: 10.1038/s44321-024-00100-7] [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: 10/24/2023] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 07/17/2024] Open
Abstract
Despite the re-emergence of the pioneering "Coley's toxin" concept in anti-cancer immune therapies highlighted by check-point inhibitors and CAR-T approaches, fundamental mechanisms responsible for the immune-enhancing efficacy of low-dose "Coley's toxin" remain poorly understood. This study examines the novel reprogramming of immune-enhancing neutrophils by super-low dose endotoxin conducive for anti-cancer therapies. Through integrated analyses including scRNAseq and functional characterizations, we examined the efficacy of reprogrammed neutrophils in treating experimental cancer. We observed that neutrophils trained by super-low dose endotoxin adopt a potent immune-enhancing phenotype characterized by CD177loCD11bloCD80hiCD40hiDectin2hi. Both murine and human neutrophils trained by super-low dose endotoxin exhibit relieved suppression of adaptive T cells as compared to un-trained neutrophils. Functionally, neutrophils trained by super-low dose endotoxin can potently reduce tumor burden when transfused into recipient tumor-bearing mice. Mechanistically, Super-low dose endotoxin enables the generation of immune-enhancing neutrophils through activating STAT5 and reducing innate suppressor IRAK-M. Together, our data clarify the long-held mystery of "Coley's toxin" in rejuvenating anti-tumor immune defense, and provide a proof-of-concept in developing innate neutrophil-based anti-tumor therapeutics.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA
| | - Christina Lee
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA
| | - Shuo Geng
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA
| | - Jing Wang
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA
| | - Udipta Bohara
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA
| | - Jacqueline Hou
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA
| | - Ziyue Yi
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA.
| |
Collapse
|
36
|
Kang Q, He L, Zhang Y, Zhong Z, Tan W. Immune-inflammatory modulation by natural products derived from edible and medicinal herbs used in Chinese classical prescriptions. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155684. [PMID: 38788391 DOI: 10.1016/j.phymed.2024.155684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/29/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Edible and medicinal herbs1 (EMHs) refer to a class of substances with dual attribution of food and medicine. These substances are traditionally used as food and also listed in many international pharmacopoeias, including the European Pharmacopoeia, the United States Pharmacopoeia, and the Chinese Pharmacopoeia. Some classical formulas that are widely used in traditional Chinese medicine include a series of EMHs, which have been shown to be effective with obvious characteristics and advantages. Notably, these EMHs and Chinese classical prescriptions2 (CCPs) have also attracted attention in international herbal medicine research because of their low toxicity and high efficiency as well as the rich body of experience for their long-term clinical use. PURPOSE Our purpose is to explore the potential therapeutic effect of EMHs with immune-inflammatory modulation for the study of modern cancer drugs. STUDY DESIGN In the present study, we present a detailed account of some EMHs used in CCPs that have shown considerable research potential in studies exploring modern drugs with immune-inflammatory modulation. METHODS Approximately 500 publications in the past 30 years were collected from PubMed, Web of Science and ScienceDirect using the keywords, such as natural products, edible and medicinal herbs, Chinese medicine, classical prescription, immune-inflammatory, tumor microenvironment and some related synonyms. The active ingredients instead of herbal extracts or botanical mixtures were focused on and the research conducted over the past decade were discussed emphatically and analyzed comprehensively. RESULTS More than ten natural products derived from EMHs used in CCPs are discussed and their immune-inflammatory modulation activities, including enhancing antitumor immunity, regulating inflammatory signaling pathways, lowering the proportion of immunosuppressive cells, inhibiting the secretion of proinflammatory cytokines, immunosuppressive factors, and inflammatory mediators, are summarized. CONCLUSION Our findings demonstrate the immune-inflammatory modulating role of those EMHs used in CCPs and provide new ideas for cancer treatment in clinical settings.
Collapse
Affiliation(s)
- Qianming Kang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Luying He
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yang Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China.
| | - Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
37
|
Boersma B, Poinot H, Pommier A. Stimulating the Antitumor Immune Response Using Immunocytokines: A Preclinical and Clinical Overview. Pharmaceutics 2024; 16:974. [PMID: 39204319 PMCID: PMC11357675 DOI: 10.3390/pharmaceutics16080974] [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: 06/03/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Cytokines are immune modulators which can enhance the immune response and have been proven to be an effective class of immunotherapy. Nevertheless, the clinical use of cytokines in cancer treatment has faced several challenges associated with poor pharmacokinetic properties and the occurrence of adverse effects. Immunocytokines (ICKs) have emerged as a promising approach to overcome the pharmacological limitations observed with cytokines. ICKs are fusion proteins designed to deliver cytokines in the tumor microenvironment by taking advantage of the stability and specificity of immunoglobulin-based scaffolds. Several technological approaches have been developed. This review focuses on ICKs designed with the most impactful cytokines in the cancer field: IL-2, TNFα, IL-10, IL-12, IL-15, IL-21, IFNγ, GM-CSF, and IFNα. An overview of the pharmacological effects of the naked cytokines and ICKs tested for cancer therapy is detailed. A particular emphasis is given on the immunomodulatory effects of ICKs associated with their technological design. In conclusion, this review highlights active ways of development of ICKs. Their already promising results observed in clinical trials are likely to be improved with the advances in targeting technologies such as cytokine/linker engineering and the design of multispecific antibodies with tumor targeting and immunostimulatory functional properties.
Collapse
Affiliation(s)
- Bart Boersma
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland;
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Hélène Poinot
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
- Translational Research Centre in Oncohaematology, University of Geneva, 1211 Geneva, Switzerland
| | - Aurélien Pommier
- UMR1240 Imagerie Moléculaire et Stratégies Théranostiques INSERM, Université Clermont Auvergne, BP 184, F-63005 Clermont-Ferrand, France
| |
Collapse
|
38
|
Yu C, Zhou G, Shi Z, Yu L, Zhou X. TREM1 facilitates the development of gastric cancer through regulating neutrophil extracellular traps-mediated macrophage polarization. Dig Liver Dis 2024; 56:1237-1247. [PMID: 38151453 DOI: 10.1016/j.dld.2023.12.002] [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: 05/19/2023] [Revised: 12/04/2023] [Accepted: 12/07/2023] [Indexed: 12/29/2023]
Abstract
Triggering receptor expressed on myeloid cell 1 (TREM1) elevation is associated with the unfavorable prognosis of gastric cancer (GC) patients. This work uncovered the effects and mechanism of TREM1 in GC. IHC staining examined TREM1 expression in GC tissues. TREM1-knockout and TREM1 knock-in mice were generated prior to the construction of N-methyl-N'-nitro-N-nitrosoguanidine (MNNG)-induced GC mice model. H&E staining detected the pathological alternations of gastric tissues. IHC staining tested Ki67 expression. Wright-Giemsa staining performed neutrophil counting and flow cytometry analysis measured neutrophil infiltration. ELISA analyzed serum and tissue myeloperoxidase (MPO) levels and serum MPO-DNA levels. Immunofluorescence, Western blotting and related kits detected NETs formation. Immunofluorescence and IHC staining evaluated macrophage polarization. In MNNG-treated GES-1 cells and phorbal myristate acetate (PMA)-treated neutrophils, TREM1 expression was also examined. CCK-8 method and Western blotting assayed cell proliferation. Western blotting and immunofluorescence detected NETs formation. Flow cytometry analysis detected the changes of macrophage typing. TREM1 was overexpressed in tumor tissues, MNNG-treated GES-1 cells and PMA-treated neutrophils. TREM1 deficiency hindered tumor growth, reduced neutrophil infiltration, NETs formation and stimulated M1 macrophage polarization in MNNG-induced GC models. Neutrophil extracellular traps (NETs) degrader DNase-1 countervailed the impacts of TREM1 on MNNG-induced GC models in vivo. Collectively, TREM1 knockdown obstructed NETs-mediated M2 macrophage polarization to hamper GC progression.
Collapse
Affiliation(s)
- Cheng Yu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of General Surgery, Affiliated Changshu Hospital of Nantong University, Changshu, 215500, China
| | - Guoqiang Zhou
- Department of General Surgery, Affiliated Changshu Hospital of Nantong University, Changshu, 215500, China
| | - Zhiliang Shi
- Department of General Surgery, Affiliated Changshu Hospital of Nantong University, Changshu, 215500, China
| | - Liang Yu
- Department of General Surgery, Affiliated Changshu Hospital of Nantong University, Changshu, 215500, China
| | - Xiaojun Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| |
Collapse
|
39
|
Sahin TK, Akyildiz A, Dogan OT, Kavgaci G, Guven DC, Aksoy S. Prognostic Significance of Pan-Immune-Inflammation Value in Patients with HER2-Positive Metastatic Breast Cancer Treated with Trastuzumab Emtansine. Pharmaceuticals (Basel) 2024; 17:824. [PMID: 39065675 PMCID: PMC11279561 DOI: 10.3390/ph17070824] [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: 05/10/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Trastuzumab emtansine (T-DM1) is a mainstay therapy for HER2-positive metastatic breast cancer (mBC). However, identifying patients who will benefit most remains a challenge due to the lack of reliable biomarkers. The recently developed pan-immune-inflammation value (PIV), a novel immune-inflammation marker, could aid in this regard, considering the immunomodulatory effects of T-DM1. Therefore, we aimed to evaluate the association between the PIV and the efficacy of T-DM1 in patients with HER2-positive mBC. A total of 122 HER2-positive mBC patients treated with T-DM1 were included. Receiver operating characteristic (ROC) curve analyses were conducted to determine the optimal PIV threshold value for survival prediction. Kaplan-Meier survival curves and Cox regression analyses were used for univariable and multivariable survival analyses, respectively. The median age was 51 years, and 95.1% of the patients had ECOG PS 0-1. The optimal PIV cutoff value was identified as 338 in ROC analyses (AUC: 0.667, 95% CI: 0.569-0.765, p = 0.002). The multivariate analysis revealed that patients in the high-PIV group had significantly shorter OS (HR: 2.332; 95% CI: 1.408-3.861; p = 0.001) and PFS (HR: 2.423; 95% CI: 1.585-3.702; p < 0.001) than patients in the low-PIV group. Additionally, both ORR and DCR were significantly lower in the high-PIV group (36.6% vs. 61.3%, p = 0.011; 56.1% vs. 76.0%, p = 0.027). Our findings suggest that pre-treatment PIV may be a novel prognostic biomarker for HER2-positive mBC patients receiving T-DM1. A low PIV level is associated with more favorable outcomes. Future prospective studies are warranted to validate these findings and explore the potential utility of PIV in aiding treatment decisions.
Collapse
Affiliation(s)
- Taha Koray Sahin
- Department of Medical Oncology, Hacettepe University Cancer Institute, 06100 Ankara, Turkey; (A.A.); (G.K.); (D.C.G.); (S.A.)
| | - Arif Akyildiz
- Department of Medical Oncology, Hacettepe University Cancer Institute, 06100 Ankara, Turkey; (A.A.); (G.K.); (D.C.G.); (S.A.)
| | - Osman Talha Dogan
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey;
| | - Gozde Kavgaci
- Department of Medical Oncology, Hacettepe University Cancer Institute, 06100 Ankara, Turkey; (A.A.); (G.K.); (D.C.G.); (S.A.)
| | - Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, 06100 Ankara, Turkey; (A.A.); (G.K.); (D.C.G.); (S.A.)
- Medical Oncology Clinic, Health Sciences University, Elazig City Hospital, 23200 Elazig, Turkey
| | - Sercan Aksoy
- Department of Medical Oncology, Hacettepe University Cancer Institute, 06100 Ankara, Turkey; (A.A.); (G.K.); (D.C.G.); (S.A.)
| |
Collapse
|
40
|
Wang KN, Zhou K, Zhong NN, Cao LM, Li ZZ, Xiao Y, Wang GR, Huo FY, Zhou JJ, Liu B, Bu LL. Enhancing cancer therapy: The role of drug delivery systems in STAT3 inhibitor efficacy and safety. Life Sci 2024; 346:122635. [PMID: 38615745 DOI: 10.1016/j.lfs.2024.122635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/14/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024]
Abstract
The signal transducer and activator of transcription 3 (STAT3), a member of the STAT family, resides in the nucleus to regulate genes essential for vital cellular functions, including survival, proliferation, self-renewal, angiogenesis, and immune response. However, continuous STAT3 activation in tumor cells promotes their initiation, progression, and metastasis, rendering STAT3 pathway inhibitors a promising avenue for cancer therapy. Nonetheless, these inhibitors frequently encounter challenges such as cytotoxicity and suboptimal biocompatibility in clinical trials. A viable strategy to mitigate these issues involves delivering STAT3 inhibitors via drug delivery systems (DDSs). This review delineates the regulatory mechanisms of the STAT3 signaling pathway and its association with cancer. It offers a comprehensive overview of the current application of DDSs for anti-STAT3 inhibitors and investigates the role of DDSs in cancer treatment. The conclusion posits that DDSs for anti-STAT3 inhibitors exhibit enhanced efficacy and reduced adverse effects in tumor therapy compared to anti-STAT3 inhibitors alone. This paper aims to provide an outline of the ongoing research and future prospects of DDSs for STAT3 inhibitors. Additionally, it presents our insights on the merits and future outlook of DDSs in cancer treatment.
Collapse
Affiliation(s)
- Kang-Ning Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Kan Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Fang-Yi Huo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jun-Jie Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial, Anyang Sixth People's Hospital, Anyang 45500, China.
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
41
|
Zuo Q, Wu Y, Hu Y, Shao C, Liang Y, Chen L, Guo Q, Huang P, Chen Q. Targeting lipid reprogramming in the tumor microenvironment by traditional Chinese medicines as a potential cancer treatment. Heliyon 2024; 10:e30807. [PMID: 38765144 PMCID: PMC11101863 DOI: 10.1016/j.heliyon.2024.e30807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/21/2024] Open
Abstract
In the last ten years, there has been a notable rise in the study of metabolic abnormalities in cancer cells. However, compared to glucose or glutamine metabolism, less attention has been paid to the importance of lipid metabolism in tumorigenesis. Recent developments in lipidomics technologies have allowed for detailed analysis of lipid profiles within cancer cells and other cellular players present within the tumor microenvironment (TME). Traditional Chinese medicine (TCM) and its bioactive components have a long history of use in cancer treatments and are also being studied for their potential role in regulating metabolic reprogramming within TME. This review focuses on four core abnormalities altered by lipid reprogramming in cancer cells: de novo synthesis and exogenous uptake of fatty acids (FAs), upregulated fatty acid oxidation (FAO), cholesterol accumulation, which offer benefits for tumor growth and metastasis. The review also discusses how altered lipid metabolism impacts infiltrating immune cell function and phenotype as these interactions between cancer-stromal become more pronounced during tumor progression. Finally, recent literature is highlighted regarding how cancer cells can be metabolically reprogrammed by specific Chinese herbal components with potential therapeutic benefits related to lipid metabolic and signaling pathways.
Collapse
Affiliation(s)
- Qian Zuo
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yingchao Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuyu Hu
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Academy of Traditional Chinese Medicine, Guangzhou, China
| | - Cui Shao
- The First Affiliated Traditional Chinese Medicine Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuqi Liang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liushan Chen
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Academy of Traditional Chinese Medicine, Guangzhou, China
| | - Qianqian Guo
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Ping Huang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Qianjun Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Academy of Traditional Chinese Medicine, Guangzhou, China
| |
Collapse
|
42
|
Wendlinger S, Wohlfarth J, Siedel C, Kreft S, Kilian T, Junker S, Schmid L, Sinnberg T, Dischinger U, Heppt MV, Wistuba-Hamprecht K, Meier F, Erpenbeck L, Neubert E, Goebeler M, Gesierich A, Schrama D, Kosnopfel C, Schilling B. Susceptibility of Melanoma Cells to Targeted Therapy Correlates with Protection by Blood Neutrophils. Cancers (Basel) 2024; 16:1767. [PMID: 38730718 PMCID: PMC11083732 DOI: 10.3390/cancers16091767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/25/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
Elevated levels of peripheral blood and tumor tissue neutrophils are associated with poorer clinical response and therapy resistance in melanoma. The underlying mechanism and the role of neutrophils in targeted therapy is still not fully understood. Serum samples of patients with advanced melanoma were collected and neutrophil-associated serum markers were measured and correlated with response to targeted therapy. Blood neutrophils from healthy donors and patients with advanced melanoma were isolated, and their phenotypes, as well as their in vitro functions, were compared. In vitro functional tests were conducted through nonadherent cocultures with melanoma cells. Protection of melanoma cell lines by neutrophils was assessed under MAPK inhibition. Blood neutrophils from advanced melanoma patients exhibited lower CD16 expression compared to healthy donors. In vitro, both healthy-donor- and patient-derived neutrophils prevented melanoma cell apoptosis upon dual MAPK inhibition. The effect depended on cell-cell contact and melanoma cell susceptibility to treatment. Interference with protease activity of neutrophils prevented melanoma cell protection during treatment in cocultures. The negative correlation between neutrophils and melanoma outcomes seems to be linked to a protumoral function of neutrophils. In vitro, neutrophils exert a direct protective effect on melanoma cells during dual MAPK inhibition. This study further hints at a crucial role of neutrophil-related protease activity in protection.
Collapse
Affiliation(s)
- Simone Wendlinger
- Department of Dermatology, University Hospital Würzburg, 97080 Würzburg, Germany
- Mildred Scheel Early Career Center Wuerzburg, University Hospital Wuerzburg, 97080 Würzburg, Germany
| | - Jonas Wohlfarth
- Department of Dermatology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Claudia Siedel
- Department of Dermatology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Sophia Kreft
- Department of Dermatology, University Hospital Würzburg, 97080 Würzburg, Germany
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Teresa Kilian
- Department of Dermatology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Sarah Junker
- Department of Dermatology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Luisa Schmid
- Department of Dermatology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Tobias Sinnberg
- Division of Dermatooncology, Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Ulrich Dischinger
- Department of Endocrinology and Diabetology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Markus V. Heppt
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Kilian Wistuba-Hamprecht
- Skin Cancer Unit, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, 68167 Mannheim, Germany
| | - Friedegund Meier
- Department of Dermatology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Skin Cancer Center at the University Cancer Centre Dresden and National Center for Tumor Diseases, 01307 Dresden, Germany
| | - Luise Erpenbeck
- Department of Dermatology, University of Münster, 48149 Münster, Germany
| | - Elsa Neubert
- Leiden Academic Centre for Drug Research, Leiden University, 2333 Leiden, The Netherlands
- Department of Dermatology, Venereology and Allergology, University Medical Center, Göttingen University, 37075 Göttingen, Germany
| | - Matthias Goebeler
- Department of Dermatology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Anja Gesierich
- Department of Dermatology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - David Schrama
- Department of Dermatology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Corinna Kosnopfel
- Department of Dermatology, University Hospital Würzburg, 97080 Würzburg, Germany
- Mildred Scheel Early Career Center Wuerzburg, University Hospital Wuerzburg, 97080 Würzburg, Germany
- Department of Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Münster, Germany
| | - Bastian Schilling
- Department of Dermatology, University Hospital Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
43
|
Larson AC, Doty KR, Solheim JC. The double life of a chemotherapy drug: Immunomodulatory functions of gemcitabine in cancer. Cancer Med 2024; 13:e7287. [PMID: 38770637 PMCID: PMC11106691 DOI: 10.1002/cam4.7287] [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: 11/19/2023] [Revised: 04/19/2024] [Accepted: 04/28/2024] [Indexed: 05/22/2024] Open
Abstract
Although the development of immunotherapies has been revolutionary in the treatment of several cancers, many cancer types remain unresponsive to immune-based treatment and are largely managed by chemotherapy drugs. However, chemotherapeutics are not infallible and are frequently rendered ineffective as resistance develops from prolonged exposure. Recent investigations have indicated that some chemotherapy drugs have additional functions beyond their normative cytotoxic capacity and are in fact immune-modifying agents. Of the pharmaceuticals with identified immune-editing properties, gemcitabine is well-studied and of interest to clinicians and scientists alike. Gemcitabine is a chemotherapy drug approved for the treatment of multiple cancers, including breast, lung, pancreatic, and ovarian. Because of its broad applications, relatively low toxicity profile, and history as a favorable combinatory partner, there is promise in the recharacterization of gemcitabine in the context of the immune system. Such efforts may allow the identification of suitable immunotherapeutic combinations, wherein gemcitabine can be used as a priming agent to improve immunotherapy efficacy in traditionally insensitive cancers. This review looks to highlight documented immunomodulatory abilities of one of the most well-known chemotherapy agents, gemcitabine, relating to its influence on cells and proteins of the immune system.
Collapse
Affiliation(s)
- Alaina C. Larson
- Eppley Institute for Research in Cancer & Allied DiseasesUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Fred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Kenadie R. Doty
- Eppley Institute for Research in Cancer & Allied DiseasesUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Fred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Joyce C. Solheim
- Eppley Institute for Research in Cancer & Allied DiseasesUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Fred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Department of Biochemistry & Molecular BiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Department of Pathology, Microbiology, & ImmunologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|
44
|
Zhang Y, Nie Y, Liu X, Wan X, Shi Y, Zhang K, Wu P, He J. Tumor metabolic crosstalk and immunotherapy. Clin Transl Oncol 2024; 26:797-807. [PMID: 37740892 DOI: 10.1007/s12094-023-03304-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/08/2023] [Indexed: 09/25/2023]
Abstract
Tumor cells must resist the host's immune system while maintaining growth under harsh conditions of acidity and hypoxia, which indicates that tumors are more robust than normal tissue. Immunotherapeutic agents have little effect on solid tumors, mostly because of the tumor density and the difficulty of penetrating deeply into the tissue to achieve the theoretical therapeutic effect. Various therapeutic strategies targeting the tumor microenvironment (TME) have been developed. Immunometabolic disorders play a dominant role in treatment resistance at both the TME and host levels. Understanding immunometabolic factors and their treatment potential may be a way forward for tumor immunotherapy. Here, we summarize the metabolism of substances that affect tumor progression, the crosstalk between the TME and immunosuppression, and some potential tumor-site targets. We also summarize the progress and challenges of tumor immunotherapy.
Collapse
Affiliation(s)
- Yiwen Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yueli Nie
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- School of Pharmacy, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xitian Wan
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yuanyuan Shi
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Keyong Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Pan Wu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- School of Pharmacy, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jian He
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- School of Pharmacy, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
45
|
Liu H, Gan XM, Sun JM, Yang Q, Zhang DZ, Zuo YQ, Liu FL, Li B, Tan QL, Zhang J. Transcatheter arterial chemoembolisation combined with lenvatinib and cabozantinib in the treatment of advanced hepatocellular carcinoma. Int Immunopharmacol 2024; 130:111510. [PMID: 38422766 DOI: 10.1016/j.intimp.2024.111510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/01/2024] [Accepted: 01/04/2024] [Indexed: 03/02/2024]
Abstract
OBJECTIVE The objective of this study was to evaluate the effect and prognosis of transcatheter arterial chemoembolisation (TACE) combined with lenvatinib and cabozantinib in the treatment of advanced unresectable hepatocellular carcinoma (uHCC) and identify the predictors of prognosis related to cellular inflammation and body mass index (BMI). To the best of our knowledge, this is the first study to report the efficacy and prognosis of TACE combined with lenvatinib and cabozantinib in patients with uHCC and propose the neutrophil-to-lymphocyte ratio (NLR) and the platelet-to-lymphocyte ratio (PLR) as predictors of response and survival outcomes in this context. METHODS The clinicopathologic data of 217 patients with advanced uHCC who underwent TACE combined with systemic therapy (lenvatinib mesylate + cabozantinib) in the Department of Hepatobiliary Surgery, Dazhou Central Hospital between October 2017 and February 2020 were collected retrospectively, and the relevant parameters were analysed and compared. RESULTS Univariate and multivariate logistic regression analyses showed that BMI, NLR, PLR and prothrombin time were independent factors for the objective response rate (ORR) of transformed therapy for uHCC (OR = 0.812 vs 1,290.68 vs 1.067 vs 0.626, 95 % CI: 0.719-0.897 vs 108.081-11,541.137 vs 1.037-1.099 vs 0.414-0.946, respectively, p < 0.05). The results showed that BMI, NLR and PLR had certain predictive values for the ORR in patients with liver cancer undergoing translational therapy (p < 0.05); the combined predictive effect of the three was the best, and the area under the curve (AUC) of BMI + NLR + PLR for predicting the ORR in patients with liver cancer undergoing translational therapy was 0.951 (95 % CI: 0.921, 0.964). A total of 181 patients experienced adverse reactions at different grades, including 104 cases at grade 1, 50 cases at grade 2, 22 cases at grade 3 and 5 cases at grade 4. There was a significant difference in overall survival (OS) between low- and high-NLR groups, low- and high-PLR groups and low- and high-BMI groups (χ2 = 9.644, 8.313 and 10.314, respectively, p < 0.05). There was a significant difference in progression-free survival (PFS) between the low- and high-NLR groups, the low- and high-PLR groups and the low- and high-BMI groups (χ2 = 8.965, 9.783 and 6.343, respectively, p < 0.05). CONCLUSION Transcatheter arterial chemoembolisation combined with lenvatinib and cabozantinib is safe and effective in the treatment of advanced uHCC, with controllable adverse reactions. High NLR and PLR and low BMI values before treatment were independent risk factors for the ORR. Body mass index, NLR and PLR predicted responses to triple switch therapy and survival outcomes in uHCC. Patients with pretreatment NLR ≥ 2.96 and PLR ≥ 184.41 had worse OS and PFS rates. Patients with pretreatment BMI ≥ 23 kg/m2 had improved OS and a reduced risk of death.
Collapse
Affiliation(s)
- Hong Liu
- Department of Hepatobiliary Surgery, Dazhou Central Hospital, Dazhou 635000, China; Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Luzhou 646000, China
| | - Xue-Mei Gan
- Department of Hepatology, Dazhou Central Hospital, Dazhou 635000, China
| | - Jian-Ming Sun
- Department of Hepatobiliary Surgery, Dazhou Central Hospital, Dazhou 635000, China
| | - Qin Yang
- Department of Hepatology, Dazhou Central Hospital, Dazhou 635000, China
| | - Dai-Zhong Zhang
- Department of Hepatobiliary Surgery, Dazhou Central Hospital, Dazhou 635000, China
| | - Yong-Qing Zuo
- Department of Hepatology, Dazhou Central Hospital, Dazhou 635000, China
| | - Feng-Ling Liu
- Department of Hepatobiliary Surgery, Dazhou Central Hospital, Dazhou 635000, China
| | - Bo Li
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Luzhou 646000, China
| | - Qi-Liang Tan
- Department of Hepatology, Dazhou Central Hospital, Dazhou 635000, China
| | - Jun Zhang
- Department of Hepatobiliary Surgery, Dazhou Central Hospital, Dazhou 635000, China.
| |
Collapse
|
46
|
Lightsey S, Sharma B. Natural Killer Cell Mechanosensing in Solid Tumors. Bioengineering (Basel) 2024; 11:328. [PMID: 38671750 PMCID: PMC11048000 DOI: 10.3390/bioengineering11040328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Natural killer (NK) cells, which are an exciting alternative cell source for cancer immunotherapies, must sense and respond to their physical environment to traffic to and eliminate cancer cells. Herein, we review the mechanisms by which NK cells receive mechanical signals and explore recent key findings regarding the impact of the physical characteristics of solid tumors on NK cell functions. Data suggest that different mechanical stresses present in solid tumors facilitate NK cell functions, especially infiltration and degranulation. Moreover, we review recent engineering advances that can be used to systemically study the role of mechanical forces on NK cell activity. Understanding the mechanisms by which NK cells interpret their environment presents potential targets to enhance NK cell immunotherapies for the treatment of solid tumors.
Collapse
Affiliation(s)
| | - Blanka Sharma
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 23610, USA;
| |
Collapse
|
47
|
Sharma A, Verwilst P, Li M, Ma D, Singh N, Yoo J, Kim Y, Yang Y, Zhu JH, Huang H, Hu XL, He XP, Zeng L, James TD, Peng X, Sessler JL, Kim JS. Theranostic Fluorescent Probes. Chem Rev 2024; 124:2699-2804. [PMID: 38422393 PMCID: PMC11132561 DOI: 10.1021/acs.chemrev.3c00778] [Citation(s) in RCA: 95] [Impact Index Per Article: 95.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
The ability to gain spatiotemporal information, and in some cases achieve spatiotemporal control, in the context of drug delivery makes theranostic fluorescent probes an attractive and intensely investigated research topic. This interest is reflected in the steep rise in publications on the topic that have appeared over the past decade. Theranostic fluorescent probes, in their various incarnations, generally comprise a fluorophore linked to a masked drug, in which the drug is released as the result of certain stimuli, with both intrinsic and extrinsic stimuli being reported. This release is then signaled by the emergence of a fluorescent signal. Importantly, the use of appropriate fluorophores has enabled not only this emerging fluorescence as a spatiotemporal marker for drug delivery but also has provided modalities useful in photodynamic, photothermal, and sonodynamic therapeutic applications. In this review we highlight recent work on theranostic fluorescent probes with a particular focus on probes that are activated in tumor microenvironments. We also summarize efforts to develop probes for other applications, such as neurodegenerative diseases and antibacterials. This review celebrates the diversity of designs reported to date, from discrete small-molecule systems to nanomaterials. Our aim is to provide insights into the potential clinical impact of this still-emerging research direction.
Collapse
Affiliation(s)
- Amit Sharma
- Amity
School of Chemical Sciences, Amity University
Punjab, Sector 82A, Mohali 140 306, India
| | - Peter Verwilst
- Rega
Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49, Box 1041, 3000 Leuven, Belgium
| | - Mingle Li
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
| | - Dandan Ma
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Nem Singh
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Jiyoung Yoo
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Yujin Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Ying Yang
- School of
Light Industry and Food Engineering, Guangxi
University, Nanning, Guangxi 530004, China
| | - Jing-Hui Zhu
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Haiqiao Huang
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xi-Le Hu
- Key
Laboratory for Advanced Materials and Joint International Research
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, School of Chemistry and
Molecular Engineering, East China University
of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Xiao-Peng He
- Key
Laboratory for Advanced Materials and Joint International Research
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, School of Chemistry and
Molecular Engineering, East China University
of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- National
Center for Liver Cancer, the International Cooperation Laboratory
on Signal Transduction, Eastern Hepatobiliary
Surgery Hospital, Shanghai 200438, China
| | - Lintao Zeng
- School of
Light Industry and Food Engineering, Guangxi
University, Nanning, Guangxi 530004, China
| | - Tony D. James
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
- School
of Chemistry and Chemical Engineering, Henan
Normal University, Xinxiang 453007, China
| | - Xiaojun Peng
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- State
Key Laboratory of Fine Chemicals, Dalian
University of Technology, Dalian 116024, China
| | - Jonathan L. Sessler
- Department
of Chemistry, The University of Texas at
Austin, Texas 78712-1224, United
States
| | - Jong Seung Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
- TheranoChem Incorporation, Seongbuk-gu, Seoul 02841, Korea
| |
Collapse
|
48
|
Li Y, Zheng Y, Tan X, Du Y, Wei Y, Liu S. Extracellular vesicle-mediated pre-metastatic niche formation via altering host microenvironments. Front Immunol 2024; 15:1367373. [PMID: 38495881 PMCID: PMC10940351 DOI: 10.3389/fimmu.2024.1367373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/23/2024] [Indexed: 03/19/2024] Open
Abstract
The disordered growth, invasion and metastasis of cancer are mainly attributed to bidirectional cell-cell interactions. Extracellular vesicles (EVs) secreted by cancer cells are involved in orchestrating the formation of pre-metastatic niches (PMNs). Tumor-derived EVs mediate bidirectional communication between tumor and stromal cells in local and distant microenvironments. EVs carrying mRNAs, small RNAs, microRNAs, DNA fragments, proteins and metabolites determine metastatic organotropism, enhance angiogenesis, modulate stroma cell phenotypes, restructure the extracellular matrix, induce immunosuppression and modify the metabolic environment of organs. Evidence indicates that EVs educate stromal cells in secondary sites to establish metastasis-supportive microenvironments for seeding tumor cells. In this review, we provide a comprehensive overview of PMN formation and the underlying mechanisms mediated by EVs. Potential approaches to inhibit cancer metastasis by inhibiting the formation of PMNs are also presented.
Collapse
Affiliation(s)
- Ying Li
- Department of Blood Transfusion, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan Zheng
- Department of Operating Room, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaojie Tan
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yongxing Du
- Department of Pancreatic and Gastric Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingxin Wei
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Shanglong Liu
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
49
|
Zhou J, Zhou P, Wang J, Song J. Roles of endothelial cell specific molecule‑1 in tumor angiogenesis (Review). Oncol Lett 2024; 27:137. [PMID: 38357478 PMCID: PMC10865172 DOI: 10.3892/ol.2024.14270] [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: 10/03/2023] [Accepted: 01/23/2024] [Indexed: 02/16/2024] Open
Abstract
Angiogenesis plays a crucial role in tumor growth and metastasis, and is heavily influenced by the tumor microenvironment (TME). Endothelial cell dysfunction is a key factor in tumor angiogenesis and is characterized by the aberrant expression of pro-angiogenic factors. Endothelial cell specific molecule-1 (ESM1), also known as endocan, is a marker of endothelial cell dysfunction. Although ESM1 is primarily expressed in normal endothelial cells, dysregulated ESM1 expression has been observed in human tumors and animal tumor models, and implicated in tumor growth, metastasis and angiogenesis. The precise role of ESM1 in tumor angiogenesis and its potential regulatory mechanisms are not yet conclusively defined. However, the aim of the present review was to explore the involvement of ESM1 in the process of tumor angiogenesis in the TME and the characteristics of neovascularization. In addition, the present review discusses the interaction between ESM1 and angiogenic factors, as well as the mechanisms through which ESM1 contributes to tumor angiogenesis. Furthermore, the reciprocal regulation between ESM1 and the TME is explored. Finally, the potential of targeting ESM1 as a therapeutic strategy for tumor angiogenesis is presented.
Collapse
Affiliation(s)
- Jie Zhou
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Ping Zhou
- College of Chinese Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Jinfang Wang
- College of Nursing, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Jie Song
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| |
Collapse
|
50
|
Chen X, Cai Q, Wong K, Shen X, Guan Z. Bioinformatic analysis reveals prognostic value and immunotherapy potential of Siglec-15 in laryngeal squamous cell carcinoma. Heliyon 2024; 10:e25266. [PMID: 38352733 PMCID: PMC10861961 DOI: 10.1016/j.heliyon.2024.e25266] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/16/2024] Open
Abstract
Background Laryngeal squamous cell carcinoma (LSCC) is the ultimate common malignant head and neck cancer with dismal prognosis. The expression pattern and clinical significance of Siglec-15 (Sialic acid-binding immunoglobulin-like lectin 15) in LSCC are poorly understood. In order to lay the groundwork for future immune-related research on Siglec-15 in LSCC, we set out to study its expression and prognostic importance in the disease, as well as to use bioinformatics to investigate the immune features modulated by Siglec-15 in LSCC. Methods ① In order to get the gene expression profile and clinical data for TCGA head and neck cancer (TCGA-HNSC), you may access the relevant data from UCSC xena and use 110 cases of laryngeal cancer as a training set. Two datasets, GSE27020 and GSE25727, were obtained from the GEO databank and utilized as validation sets. These datasets include expression profiles and clinical information. The Siglec-15 gene and immune characteristics were analyzed by bioinformatics methods. ② Retrospectively collected routine paraffin specimens from patients with pathological diagnosis of squamous cell carcinoma from December 2012 to November 2015 in Sun Yat-sen Memorial Hospital and fresh frozen tissue of patients from June 2021 to March 2022. Immunohistochemistry method, immunofluorescence technique and real-time quantitative PCR was used to examine the difference of Siglec-15 appearance in LSCC tissue and adjacent tissue, and its correlation of prognosis, clinic pathological characteristics and CD8+T lymphocyte infiltration. Using human laryngeal cancer cell line (LCC), we studied the influence of Siglec-15 in cell proliferation and invasion. Results We identified Siglec-15 was upregulated in LSCC. The patients in Siglec-15 high expression group had a poor overall survival (OS) based on the clinical information from TGCA and 111 LSCC patients that hospitalized in Sun Yat-sen Memorial Hospital. The COX regression analysis indicated Siglec-15 as an independent predictor for poor prognosis of LSCC. Bioinformatic analysis suggested that the high expression of Siglec-15 shape an immune suppressive tumor microenvironment (TEM), leading to poor response to immunotherapy in LSCC. Siglec-15 enhanced cell invasion and proliferation, as we showed in vitro. Conclusion Our study support Siglec-15 as a potential predictor for LSCC prognosis and an attractive target for LSCC immunotherapy.
Collapse
Affiliation(s)
- Xiaoting Chen
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Heyou Hospital, No. 1 of Heren Road, Junlan Community, Beijiao Town, Shunde District, Foshan City, Guangdong Province, China
| | - Qian Cai
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kaiyi Wong
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ximing Shen
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong Guan
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|