1
|
Liu S, Liu C, He Y, Li J. Benign non-immune cells in tumor microenvironment. Front Immunol 2025; 16:1561577. [PMID: 40248695 PMCID: PMC12003390 DOI: 10.3389/fimmu.2025.1561577] [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: 01/20/2025] [Accepted: 02/24/2025] [Indexed: 04/19/2025] Open
Abstract
The tumor microenvironment (TME) is a highly complex and continuous evolving ecosystem, consisting of a diverse array of cellular and non-cellular components. Among these, benign non-immune cells, including cancer-associated fibroblasts (CAFs), adipocytes, endothelial cells (ECs), pericytes (PCs), Schwann cells (SCs) and others, are crucial factors for tumor development. Benign non-immune cells within the TME interact with both tumor cells and immune cells. These interactions contribute to tumor progression through both direct contact and indirect communication. Numerous studies have highlighted the role that benign non-immune cells exert on tumor progression and potential tumor-promoting mechanisms via multiple signaling pathways and factors. However, these benign non-immune cells may play different roles across cancer types. Therefore, it is important to understand the potential roles of benign non-immune cells within the TME based on tumor heterogeneity. A deep understanding allows us to develop novel cancer therapies by targeting these cells. In this review, we will introduce several types of benign non-immune cells that exert on different cancer types according to tumor heterogeneity and their roles in the TME.
Collapse
Affiliation(s)
- Shaowen Liu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chunhui Liu
- The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - Yuan He
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jun Li
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Department of Molecular Pathology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
2
|
Reghukumar SK, Inkielewicz-Stepniak I. Tumour cell-induced platelet aggregation in breast cancer: Scope of metal nanoparticles. Biochim Biophys Acta Rev Cancer 2025; 1880:189276. [PMID: 39921012 DOI: 10.1016/j.bbcan.2025.189276] [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: 09/12/2024] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/10/2025]
Abstract
Breast cancer is a major cause of cancer-related mortality among the female population worldwide. Among the various factors promoting breast cancer metastasis, the role of cancer-cell platelet interactions leading to tumour cell-induced platelet aggregation (TCIPA) has garnered significant attention recently. Our state-of-the-art literature review verifies the implications of metal nanoparticles in breast cancer research and TCIPA-specific breast cancer metastasis. We have evaluated in vitro and in vivo research data as well as clinical investigations within the scope of this topic presented in the last ten years. Nanoparticle-based drug delivery platforms in cancer therapy can combat the growing concerns of multi-drug resistance, the alarming rates of chemotherapy-induced toxicities and cancer progression. Metal nanoparticles conjugated with chemotherapeutics can outperform their free drug counterparts in achieving targeted drug delivery and desired drug concentration inside the tumour tissue with minimal toxic effects. Existing data highlights the potential of metal nanoparticles as a promising tool for targeting the platelet-specific interactions associated with breast cancer metastasis including TCIPA.
Collapse
|
3
|
Feng K, Cai X, Qiao G. Platelet-derived growth factor subunit B overexpression promotes lung cancer tumor growth and metastasis: The role of glucose metabolism. Cytojournal 2025; 22:33. [PMID: 40260069 PMCID: PMC12010884 DOI: 10.25259/cytojournal_190_2024] [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: 09/14/2024] [Accepted: 01/09/2025] [Indexed: 04/23/2025] Open
Abstract
Objective Lung cancer represents a formidable global health challenge due to its substantial prevalence and mortality rates. Metabolic reprogramming, especially the transition to aerobic glycolysis, plays a pivotal role in the progression of lung cancer by sustaining the energy demands for rapid tumor proliferation. The prominent involvement of platelet-derived growth factor subunit B (PDGFB) in promoting the growth and metastasis of lung cancer through specific signaling cascades is well established in. Nonetheless, further research is imperative to elucidate the intricate regulatory mechanisms of PDGFB in glucose metabolism and its implications for the advancement of lung cancer. Our study is dedicated to exploring the effect of PDGFB on lung cancer by modulating glucose metabolism. Material and Methods First, we determined the expression patterns of PDGFB in various lung cancer cell lines (A549, H460, HCC827, and H1975) using quantitative real-time polymerase chain reaction and Western blot analyses. We measured the expression levels of PDGFB and Ki-67 in tumor tissues from lung cancer patients through immunohistochemistry. We then transfected lung cancer cells with a PDGFB overexpression (PDGFB OE) plasmid. The effects of PDGFB OE and galactose + PDGFB OE co-treatment on cell migration and invasion characteristics were assessed using wound healing and Transwell assays. The impact of PDGFB OE and galactose + PDGFB OE co-treatment on the proliferation capacity of lung cancer cells was evaluated through colony formation and 5-ethynyl-2'-deoxyuridine staining assays. We also measured the effects of PDGFB OE on mitochondrial function and glycolytic capacity in lung cancer cells using extracellular acidification rate assay (ECAR) measurement methods. Results Elevated levels of PDGFB expression were markedly detected in various lung cancer cell lines, notably A549 and H460 (P < 0.001). This observation was validated by the analysis of tumor samples from patients with lung cancer who exhibited heightened PDGFB expression in tumor tissues (P < 0.001). Moreover, an association was found between increased levels of Ki67 expression and elevated PDGFB expression (P < 0.001). The upregulation of PDGFB was linked to heightened migratory (P < 0.001), invasive (P < 0.001), and proliferative (P < 0.001) capacities of the cells. Furthermore, an elevation in lactate levels and ECAR (P < 0.001) was noted in the PDGFB OE group, along with increased levels of glycolysis-related regulatory proteins. The inhibition of aerobic glycolysis with galactose effectively mitigated the PDGFB-induced enhancement of lung cancer cell proliferation and migration. Conclusion By affecting glucose metabolism, PDGFB drives the growth and metastasis of lung cancer, underscoring its potential as a promising therapeutic target for the management of this complex disease.
Collapse
Affiliation(s)
- Kai Feng
- Department of Thoracic Surgery II, Binzhou People’s Hospital, Binzhou, China
| | - Xiaoping Cai
- Department of Oncology II, Binzhou People’s Hospital, Binzhou, China
| | - Gaofeng Qiao
- Department of Thoracic Surgery, Shandong Public Health Clinical Center, Shandong University, Jinan City, Shandong, China
| |
Collapse
|
4
|
Wan M, Pan S, Shan B, Diao H, Jin H, Wang Z, Wang W, Han S, Liu W, He J, Zheng Z, Pan Y, Han X, Zhang J. Lipid metabolic reprograming: the unsung hero in breast cancer progression and tumor microenvironment. Mol Cancer 2025; 24:61. [PMID: 40025508 PMCID: PMC11874147 DOI: 10.1186/s12943-025-02258-1] [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: 12/03/2024] [Accepted: 02/02/2025] [Indexed: 03/04/2025] Open
Abstract
Aberrant lipid metabolism is a well-recognized hallmark of cancer. Notably, breast cancer (BC) arises from a lipid-rich microenvironment and depends significantly on lipid metabolic reprogramming to fulfill its developmental requirements. In this review, we revisit the pivotal role of lipid metabolism in BC, underscoring its impact on the progression and tumor microenvironment. Firstly, we delineate the overall landscape of lipid metabolism in BC, highlighting its roles in tumor progression and patient prognosis. Given that lipids can also act as signaling molecules, we next describe the lipid signaling exchanges between BC cells and other cellular components in the tumor microenvironment. Additionally, we summarize the therapeutic potential of targeting lipid metabolism from the aspects of lipid metabolism processes, lipid-related transcription factors and immunotherapy in BC. Finally, we discuss the possibilities and problems associated with clinical applications of lipid‑targeted therapy in BC, and propose new research directions with advances in spatiotemporal multi-omics.
Collapse
Affiliation(s)
- Mengting Wan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuaikang Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, Anhui, China
| | - Benjie Shan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Haizhou Diao
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongwei Jin
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Anhui Medical University, Hefei, China
| | - Ziqi Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, Anhui, China
| | - Shuya Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Wan Liu
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiaying He
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Graduate School of Bengbu Medical University, Bengbu, Anhui Province, China
| | - Zihan Zheng
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Anhui Medical University, Hefei, China
| | - Yueyin Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Xinghua Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Jinguo Zhang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
5
|
Qian C, Huang Y, Zhang S, Yang C, Zheng W, Tang W, Wan G, Wang A, Lu Y, Zhao Y. Integrated identification and mechanism exploration of bioactive ingredients from Salvia miltiorrhiza to induce vascular normalization. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156427. [PMID: 39892310 DOI: 10.1016/j.phymed.2025.156427] [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: 10/22/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND The clinical management of ischemic disease and cancer is complex, with disruptions in local vascular function and tumor angiogenesis contributing to blood stasis, which complicates treatment strategies. Salvia miltiorrhiza, a natural product, is known to restore vascular structure and function. However, its specific roles in concurrently addressing ischemic disease and cancer within the same organism remain poorly understood. PURPOSE This study aimed to explore the material basis, pharmacological effects, and underlying mechanisms of Salvia miltiorrhiza extract (SME) in promoting blood flow recovery in ischemic hindlimbs and inducing tumor vascular normalization. METHODS The pharmacological effects of SME were evaluated in a mouse model combining ischemic hindlimbs and tumors. Mice were administered low (SME-L) or high (SME-H) doses of SME daily, and the gastrocnemius muscle mass and tumor vascular structure were assessed. Laser Doppler perfusion imaging (LDPI) was used to monitor hindlimb blood flow recovery and tumor vascular perfusion. The pharmacokinetics of the key bioactive constituents in SME were characterized by liquid chromatography-mass spectrometry (LC-MS). Interactions between SME's active compounds and predicted targets were investigated using molecular docking, microscale thermophoresis (MST), and luciferase reporter assays. The synergistic effects of the primary components, Tanshinone I (Tan I) and Salvianolic acid A (Sal A), were analyzed through tube formation assays, enzyme-linked immunosorbent assays (ELISA), immunofluorescence staining, and western blot. RESULTS Phytochemical profiling revealed that SME contains several active compounds, including Danshensu, Sal A, Sal B, Tan IIA, and Tan I. SME treatment reduced the frequency of necrotic toes, increased muscle mass, and alleviated hypoxia in the gastrocnemius muscle. SME significantly improved tumor vascular perfusion and notably enhanced pericyte coverage and basement membrane integrity. Pharmacokinetic analysis identified Tan I and Sal A as the key bioactive components that promote vascular normalization. Tan I inhibited FoxO1, preventing endothelial cell activation induced by angiopoietin 2 (Ang2), while Sal A bound to Ang2, facilitating Tie2 activation mediated by Ang1. Both in vitro and in vivo results demonstrated that the combination of Tan I and Sal A exerted a synergistic therapeutic effect on correcting abnormal blood vessels in ischemic hindlimbs and tumors. CONCLUSION Our study innovatively revealed a reliable mouse model wherein the Ang2/Tie2 signaling cascade disrupted the endothelial homeostasis to aggravate the progression of hindlimb ischemia and tumor angiogenesis. This balance can be rescued by the combination therapy of Tan I and Sal A that were both from SME, leading to the occurrence of vascular normalization.
Collapse
Affiliation(s)
- Cheng Qian
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Ying Huang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Shan Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Chunmei Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Weiwei Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Weiwei Tang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China
| | - Guiping Wan
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Yang Zhao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| |
Collapse
|
6
|
Tian L, Huang Y, Liu Y, Liu J, Liu Y. Parecoxib inhibits tumorigenesis and angiogenesis in hepatocellular carcinoma through ERK-VEGF/MMPs signaling pathway. IUBMB Life 2024; 76:972-986. [PMID: 38873890 DOI: 10.1002/iub.2861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 05/05/2024] [Indexed: 06/15/2024]
Abstract
Parecoxib, a well-recognized nonsteroidal anti-inflammatory drug, has been reported to possess anticancer properties in various tumor types. In this work, we aimed to investigate the potential anticancer effects of parecoxib on hepatocellular carcinoma (HCC) cells. To assess the impact of parecoxib on HCC cell proliferation, we employed Cell Counting Kit-8, colony formation, and 5-ethynyl-2'-deoxyuridine assays. Hoechst/propidium iodide (PI) double staining and flow cytometry were performed to evaluate apoptosis and cell cycle analysis. Wound healing and transwell assays were utilized to assess cell migration and invasion. Tube formation assay was employed to analyze angiogenesis. Protein levels were determined using western blotting, and mRNA expression levels were assessed using quantitative real-time polymerase chain reaction (PCR). A xenograft mouse model was used to confirm the antitumor effects of parecoxib on HCC tumors in vivo. Our data demonstrated that parecoxib effectively inhibited the proliferation of HCC cells in a dose- and time-dependent manner. In addition, parecoxib induced cell cycle arrest in the G2 phase and promoted apoptosis. Moreover, parecoxib hindered tumor migration and invasion by impeding the epithelial-mesenchymal transition process. Further investigation showed that parecoxib could significantly suppress angiogenesis through the inhibition of extracellular signal-regulated kinase (ERK)-vascular endothelial growth factor (VEGF) axis. Notably, treatment with the ERK activator phorbol myristate acetate upregulated the expression of matrix metalloproteinase (MMP)-2, MMP-9, and VEGF and reversed the function of parecoxib in HCC cells. Besides, parecoxib displayed its antitumor efficacy in vivo. Collectively, our results suggest that parecoxib ameliorates HCC progression by regulating proliferation, cell cycle, apoptosis, migration, invasion, and angiogenesis through the ERK-VEGF/MMPs signaling pathway.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/drug therapy
- Liver Neoplasms/metabolism
- Liver Neoplasms/genetics
- Animals
- Isoxazoles/pharmacology
- Mice
- Cell Proliferation/drug effects
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/metabolism
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor A/genetics
- Apoptosis/drug effects
- Cell Movement/drug effects
- Xenograft Model Antitumor Assays
- Mice, Nude
- Signal Transduction/drug effects
- Mice, Inbred BALB C
- Gene Expression Regulation, Neoplastic/drug effects
- Carcinogenesis/drug effects
- MAP Kinase Signaling System/drug effects
- Matrix Metalloproteinase 9/metabolism
- Matrix Metalloproteinase 9/genetics
- Male
- Cell Line, Tumor
- Angiogenesis
Collapse
Affiliation(s)
- Li Tian
- Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - YuQi Huang
- Department of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
| | - Yan Liu
- Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - JiangWei Liu
- Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Yan Liu
- Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| |
Collapse
|
7
|
Xie Q, Zhou J, He C, Xu Y, Tao F, Hu M. Unlocking the intricacies: Exploring the complex interplay between platelets and ovarian cancer. Crit Rev Oncol Hematol 2024; 202:104465. [PMID: 39097249 DOI: 10.1016/j.critrevonc.2024.104465] [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: 06/07/2024] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024] Open
Abstract
Ovarian cancer, an aggressive malignancy of the female reproductive tract, is frequently linked to an elevated risk of thrombotic events. This association is manifested by a pronounced rise in platelet counts and activation levels. Current research firmly supports the pivotal role of platelets in the oncogenic processes of ovarian cancer, influencing tumor cell proliferation and metastasis. Platelets influence these processes through direct interactions with tumor cells or by secreting cytokines and growth factors that enhance tumor growth, angiogenesis, and metastasis. This review aims to thoroughly dissect the interactions between platelets and ovarian cancer cells, emphasizing their combined role in tumor progression and associated thrombotic events. Additionally, it summarizes therapeutic strategies targeting platelet-cancer interface which show significant promise. Such approaches could not only be effective in managing the primary ovarian tumor but also play a pivotal role in preventing metastasis and attenuating thrombotic complications associated with ovarian cancer.
Collapse
Affiliation(s)
- Qianxin Xie
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie Zhou
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chaonan He
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ye Xu
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangfang Tao
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Mengjiao Hu
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
8
|
Mahasa KJ, Ouifki R, de Pillis L, Eladdadi A. A Role of Effector CD 8 + T Cells Against Circulating Tumor Cells Cloaked with Platelets: Insights from a Mathematical Model. Bull Math Biol 2024; 86:89. [PMID: 38884815 DOI: 10.1007/s11538-024-01323-y] [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: 01/18/2024] [Accepted: 05/31/2024] [Indexed: 06/18/2024]
Abstract
Cancer metastasis accounts for a majority of cancer-related deaths worldwide. Metastasis occurs when the primary tumor sheds cells into the blood and lymphatic circulation, thereby becoming circulating tumor cells (CTCs) that transverse through the circulatory system, extravasate the circulation and establish a secondary distant tumor. Accumulating evidence suggests that circulating effector CD 8 + T cells are able to recognize and attack arrested or extravasating CTCs, but this important antitumoral effect remains largely undefined. Recent studies highlighted the supporting role of activated platelets in CTCs's extravasation from the bloodstream, contributing to metastatic progression. In this work, a simple mathematical model describes how the primary tumor, CTCs, activated platelets and effector CD 8 + T cells participate in metastasis. The stability analysis reveals that for early dissemination of CTCs, effector CD 8 + T cells can present or keep secondary metastatic tumor burden at low equilibrium state. In contrast, for late dissemination of CTCs, effector CD 8 + T cells are unlikely to inhibit secondary tumor growth. Moreover, global sensitivity analysis demonstrates that the rate of the primary tumor growth, intravascular CTC proliferation, as well as the CD 8 + T cell proliferation, strongly affects the number of the secondary tumor cells. Additionally, model simulations indicate that an increase in CTC proliferation greatly contributes to tumor metastasis. Our simulations further illustrate that the higher the number of activated platelets on CTCs, the higher the probability of secondary tumor establishment. Intriguingly, from a mathematical immunology perspective, our simulations indicate that if the rate of effector CD 8 + T cell proliferation is high, then the secondary tumor formation can be considerably delayed, providing a window for adjuvant tumor control strategies. Collectively, our results suggest that the earlier the effector CD 8 + T cell response is enhanced the higher is the probability of preventing or delaying secondary tumor metastases.
Collapse
Affiliation(s)
- Khaphetsi Joseph Mahasa
- Department of Mathematics and Computer Science, National University of Lesotho, Roma, Maseru, Lesotho.
| | - Rachid Ouifki
- Department of Mathematics and Applied Mathematics, Mafikeng Campus, North-West University, Private Bag X2046, Mmabatho, 2735, South Africa
| | | | - Amina Eladdadi
- Division of Mathematical Sciences, The National Science Foundation, Alexandria, VA, USA
| |
Collapse
|
9
|
Ma J, Pang Y, Shang Y, Xie C, Xu X, Chan L, Zhang Z, Wang W. CyTOF analysis revealed platelet heterogeneity in breast cancer patients received T-DM1 treatment. Clin Immunol 2024; 263:110227. [PMID: 38643891 DOI: 10.1016/j.clim.2024.110227] [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: 01/14/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
T-DM1 (Trastuzumab Emtansine) belongs to class of Antibody-Drug Conjugates (ADC), where cytotoxic drugs are conjugated with the antibody Trastuzumab to specifically target HER2-positive cancer cells. Platelets, as vital components of the blood system, intricately influence the immune response to tumors through complex mechanisms. In our study, we examined platelet surface proteins in the plasma of patients before and after T-DM1 treatment, categorizing them based on treatment response. We identified a subgroup of platelets with elevated expression of CD63 and CD9 exclusively in patients with favorable treatment responses, while this subgroup was absent in patients with poor responses. Another noteworthy discovery was the elevated expression of CD36 in the platelet subgroups of patients exhibiting inadequate responses to treatment. These findings suggest that the expression of these platelet surface proteins may be correlated with the prognosis of T-DM1 treatment. These indicators offer valuable insights for predicting the therapeutic response to T-DM1 and may become important references in future clinical practice, contributing to a better understanding of the impact of ADC therapies and optimizing personalized cancer treatment strategies.
Collapse
Affiliation(s)
- Jianli Ma
- Department of Radiation Oncology, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Yuheng Pang
- Department of Breast Surgery, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Yuefeng Shang
- Department of Breast Surgery, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Chufei Xie
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, PR China
| | - Xiaoxue Xu
- Capital Medical University, Beijing, PR China
| | - Liujia Chan
- Capital Medical University, Beijing, PR China
| | - Zhiren Zhang
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin 150001, China.
| | - Wenjing Wang
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, PR China.
| |
Collapse
|
10
|
Qian C, Zhou Y, Zhang T, Dong G, Song M, Tang Y, Wei Z, Yu S, Shen Q, Chen W, Choi JP, Yan J, Zhong C, Wan L, Li J, Wang A, Lu Y, Zhao Y. Targeting PKM2 signaling cascade with salvianic acid A normalizes tumor blood vessels to facilitate chemotherapeutic drug delivery. Acta Pharm Sin B 2024; 14:2077-2096. [PMID: 38799619 PMCID: PMC11121179 DOI: 10.1016/j.apsb.2024.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/22/2024] [Accepted: 02/02/2024] [Indexed: 05/29/2024] Open
Abstract
Aberrant tumor blood vessels are prone to propel the malignant progression of tumors, and targeting abnormal metabolism of tumor endothelial cells emerges as a promising option to achieve vascular normalization and antagonize tumor progression. Herein, we demonstrated that salvianic acid A (SAA) played a pivotal role in contributing to vascular normalization in the tumor-bearing mice, thereby improving delivery and effectiveness of the chemotherapeutic agent. SAA was capable of inhibiting glycolysis and strengthening endothelial junctions in the human umbilical vein endothelial cells (HUVECs) exposed to hypoxia. Mechanistically, SAA was inclined to directly bind to the glycolytic enzyme PKM2, leading to a dramatic decrease in endothelial glycolysis. More importantly, SAA improved the endothelial integrity via activating the β-Catenin/Claudin-5 signaling axis in a PKM2-dependent manner. Our findings suggest that SAA may serve as a potent agent for inducing tumor vascular normalization.
Collapse
Affiliation(s)
- Cheng Qian
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yueke Zhou
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Teng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Guanglu Dong
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mengyao Song
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu Tang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhonghong Wei
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Suyun Yu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qiuhong Shen
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wenxing Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jaesung P. Choi
- Centre for Inflammation, Faculty of Science, Centenary Institute, School of Life Sciences, University of Technology Sydney, Sydney NSW 2050, Australia
| | - Juming Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, China
| | - Chongjin Zhong
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Li Wan
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Jia Li
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney NSW 2109, Australia
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yang Zhao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
11
|
Jiang M, Karsenberg R, Bianchi F, van den Bogaart G. CD36 as a double-edged sword in cancer. Immunol Lett 2024; 265:7-15. [PMID: 38122906 DOI: 10.1016/j.imlet.2023.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
The membrane protein CD36 is a lipid transporter, scavenger receptor, and receptor for the antiangiogenic protein thrombospondin 1 (TSP1). CD36 is expressed by cancer cells and by many associated cells including various cancer-infiltrating immune cell types. Thereby, CD36 plays critical roles in cancer, and it has been reported to affect cancer growth, metastasis, angiogenesis, and drug resistance. However, these roles are partly contradictory, as CD36 has been both reported to promote and inhibit cancer progression. Moreover, the mechanisms are also partly contradictory, because CD36 has been shown to exert opposite cellular effects such as cell division, senescence and cell death. This review provides an overview of the diverse effects of CD36 on tumor progression, aiming to shed light on its diverse pro- and anti-cancer roles, and the implications for therapeutic targeting.
Collapse
Affiliation(s)
- Muwei Jiang
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747AG, Nijenborgh 7, Groningen, the Netherlands
| | - Renske Karsenberg
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747AG, Nijenborgh 7, Groningen, the Netherlands
| | - Frans Bianchi
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747AG, Nijenborgh 7, Groningen, the Netherlands
| | - Geert van den Bogaart
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747AG, Nijenborgh 7, Groningen, the Netherlands.
| |
Collapse
|