1
|
Xie Y, Mi X, Xing Y, Dai Z, Pu Q. Past, present, and future of exosomes research in cancer: A bibliometric and visualization analysis. Hum Vaccin Immunother 2025; 21:2488551. [PMID: 40207548 PMCID: PMC11988232 DOI: 10.1080/21645515.2025.2488551] [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: 01/21/2025] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer seriously threatens the lives and health of people worldwide, and exosomes seem to play an important role in managing cancer effectively, which has attracted extensive attention from researchers in recent years. This study aimed to scientifically visualize exosomes research in cancer (ERC) through bibliometric analysis, reviewing the past, summarizing the present, and predicting the future, with a view to providing valuable insights for scholars and policy makers. Researches search and data collection from Web of Science Core Collection and clinical trial.gov. Calculations and visualizations were performed using Microsoft Excel, VOSviewer, Bibliometrix R-package, and CiteSpace. As of December 1, 2024, and March 8, 2025, we identified 8,001 ERC-related publications and 107 ERC-related clinical trials, with an increasing trend in annual publications. Our findings supported that China, Nanjing Medical University, and International Journal of Molecular Sciences were the most productive countries, institutions, and journals, respectively. Whiteside, Theresa L. had the most publications, while Théry, C was the most co-cited scholar. In addition, Cancer Research was the most co-cited journal. Spatial and temporal distribution of clinical trials was the same as for publications. High-frequency keywords were "extracellular vesicle," "microRNA" and "biomarker." Additional, "surface functionalization," "plant," "machine learning," "nanomaterials," "promotes metastasis," "engineered exosomes," and "macrophage-derived exosomes" were promising research topics. Our study comprehensively and visually summarized the structure, hotspots, and evolutionary trends of ERC. It would inspire subsequent studies from a macroscopic perspective and provide a basis for rational allocation of resources and identification of collaborations among researchers.
Collapse
Affiliation(s)
- Yafei Xie
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Xingqi Mi
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yikai Xing
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Zhangyi Dai
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Qiang Pu
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Wu J, Qian Y, Yang K, Zhang S, Zeng E, Luo D. Innate immune cells in vascular lesions: mechanism and significance of diversified immune regulation. Ann Med 2025; 57:2453826. [PMID: 39847394 PMCID: PMC11758805 DOI: 10.1080/07853890.2025.2453826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/18/2024] [Accepted: 01/06/2025] [Indexed: 01/24/2025] Open
Abstract
Angiogenesis is a complex physiological process. In recent years, the immune regulation of angiogenesis has received increasing attention, and innate immune cells, which are centred on macrophages, are thought to play important roles in vascular neogenesis and development. Various innate immune cells can act on the vasculature through a variety of mechanisms, with commonalities as well as differences and synergistic effects, which are crucial for the progression of vascular lesions. In recent years, monotherapy with antiangiogenic drugs has encountered therapeutic bottlenecks because of the short-term effect of 'vascular normalization'. The combination treatment of antiangiogenic therapy and immunotherapy breaks the traditional treatment pattern. While it has a remarkable curative effect and survival benefits, it also faces many challenges. This review focuses on innate immune cells and mainly introduces the regulatory mechanisms of monocytes, macrophages, natural killer (NK) cells, dendritic cells (DCs) and neutrophils in vascular lesions. The purpose of this paper was to elucidate the underlying mechanisms of angiogenesis and development and the current research status of innate immune cells in regulating vascular lesions in different states. This review provides a theoretical basis for addressing aberrant angiogenesis in disease processes or finding new antiangiogenic immune targets in inflammation and tumor.
Collapse
Affiliation(s)
- Jinjing Wu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yulu Qian
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Kuang Yang
- Queen Mary University of London, Nanchang University, Nanchang, China
| | - Shuhua Zhang
- Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Jiangxi Cardiovascular Research Institute, Nanchang, Jiangxi, China
| | - Erming Zeng
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Daya Luo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
3
|
Yuan Z, Li J, Na Q. Recent advances in biomimetic nanodelivery systems for the treatment of glioblastoma. Colloids Surf B Biointerfaces 2025; 252:114668. [PMID: 40168694 DOI: 10.1016/j.colsurfb.2025.114668] [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: 03/04/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
Glioblastoma remain one of the deadliest malignant tumors in the central nervous system, largely due to their aggressiveness, high degree of heterogeneity, and the protective barrier of the blood-brain barrier (BBB). Conventional therapies including surgery, chemotherapy and radiotherapy often fail to improve patient prognosis due to limited drug penetration and non-specific toxicity. We then present recent advances in biomimetic nanodelivery systems, focusing on cell membrane coatings, nanoenzymes, and exosome-based carriers. By mimicking endogenous biological functions, these systems demonstrate improved immune evasion, enhanced BBB traversal, and selective drug release within the tumor microenvironment. Nevertheless, we acknowledge unresolved bottlenecks related to large-scale production, stability, and the intricacies of regulatory compliance. Looking forward, we propose an interdisciplinary roadmap that combines materials engineering, cellular biology, and clinical expertise. Through this collaborative approach, this work aims to optimize biomimetic nanodelivery for glioma therapy and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Zhenru Yuan
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Jing Li
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Qi Na
- General Hospital of Northern Theater Command, Liaoning 110016, China.
| |
Collapse
|
4
|
Issa H, Singh L, Lai KS, Parusheva-Borsitzky T, Ansari S. Dynamics of inflammatory signals within the tumor microenvironment. World J Exp Med 2025; 15:102285. [DOI: 10.5493/wjem.v15.i2.102285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/31/2024] [Accepted: 01/11/2025] [Indexed: 04/16/2025] Open
Abstract
Tumor stroma, or tumor microenvironment (TME), has been in the spotlight during recent years for its role in tumor development, growth, and metastasis. It consists of a myriad of elements, including tumor-associated macrophages, cancer-associated fibroblasts, a deregulated extracellular matrix, endothelial cells, and vascular vessels. The release of proinflammatory molecules, due to the inflamed microenvironment, such as cytokines and chemokines is found to play a pivotal role in progression of cancer and response to therapy. This review discusses the major key players and important chemical inflammatory signals released in the TME. Furthermore, the latest breakthroughs in cytokine-mediated crosstalk between immune cells and cancer cells have been highlighted. In addition, recent updates on alterations in cytokine signaling between chronic inflammation and malignant TME have also been reviewed.
Collapse
Affiliation(s)
- Hala Issa
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Lokjan Singh
- Department of Microbiology, Karnali Academy of Health Sciences, Jumla 21200, Karnali, Nepal
| | - Kok-Song Lai
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Tina Parusheva-Borsitzky
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Shamshul Ansari
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| |
Collapse
|
5
|
Fu Y, Yang Q, Xu N, Zhang X. MiRNA affects the advancement of breast cancer by modulating the immune system's response. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167759. [PMID: 40037267 DOI: 10.1016/j.bbadis.2025.167759] [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: 10/29/2024] [Revised: 02/05/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Breast cancer (BC), which is the most common tumor in women, has greatly endangered women's lives and health. Currently, patients with BC receive comprehensive treatments, including surgery, chemotherapy, radiotherapy, endocrine therapy, and targeted therapy. According to the latest research, the development of BC is closely related to the inflammatory immune response, and the immunogenicity of BC has steadily been recognized. As such, immunotherapy is one of the promising and anticipated forms of treatment for BC. The potential values of miRNA in the diagnosis and prognosis of BC have been established, and aberrant expression of associated miRNA can either facilitate or inhibit progression of BC. In the tumor immune microenvironment (TME), miRNAs are considered to be an essential molecular mechanism by which tumor cells interact with immunocytes and immunologic factors. Aberrant expression of miRNAs results in reprogramming of tumor cells actively, which may suppress the generation and activation of immunocytes and immunologic factors, avoid tumor cells apoptosis, and ultimately result in uncontrolled proliferation and deterioration. Therefore, through activating and regulating the immunocytes related to tumors and associated immunologic factors, miRNA can contribute to the advancement of BC. In this review, we assessed the function of miRNA and associated immune system components in regulating the advancement of BC, as well as the potential and viability of using miRNA in immunotherapy for BC.
Collapse
Affiliation(s)
- Yeqin Fu
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China; Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China
| | - Qiuhui Yang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 310006, China
| | - Ning Xu
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China; School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, China
| | - Xiping Zhang
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
6
|
Swami R, Popli P, Sal K, Challa RR, Vallamkonda B, Garg M, Dora CP. A review on biomacromolecular ligand-directed nanoparticles: New era in macrophage targeting. Int J Biol Macromol 2025; 306:141740. [PMID: 40058437 DOI: 10.1016/j.ijbiomac.2025.141740] [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: 06/06/2024] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 05/11/2025]
Abstract
Traditional drug delivery strategies often have side effects due to uneven drug distribution leading to the subtherapeutic impacts. Ligand-modified nanoparticles offer a revolutionary approach to precise drug delivery. These modified nanoparticles can potentially target macrophages, which is crucial for defense and disease progression efficiently. Out of many classes of ligands, biomacromolecular ligands emerged as potential ligands for directing these nanoparticles to macrophages due to their consecutive receptors over the macrophage surface, assisting easy internalization and thus supporting elevated efficacy and reduced toxicity. This approach could significantly improve treatment for diseases like cancer, tuberculosis, etc. by directing drugs to macrophages and reducing side effects. By leveraging nanotechnology and biomacromolecular-based ligand-directed targeting, we can achieve more precise and effective treatments, paving the way for advancements in precision medicine.
Collapse
Affiliation(s)
- Rajan Swami
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Pankaj Popli
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Komal Sal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | | | - Madhukar Garg
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | |
Collapse
|
7
|
Sun J, Zhou S, Sun Y, Zeng Y. The clinical significance and potential therapeutic target of tumor-associated macrophage in non-small cell lung cancer. Front Med (Lausanne) 2025; 12:1541104. [PMID: 40370720 PMCID: PMC12076932 DOI: 10.3389/fmed.2025.1541104] [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: 12/07/2024] [Accepted: 03/27/2025] [Indexed: 05/16/2025] Open
Abstract
One of the leading causes of cancer-related mortality globally is non-small cell lung cancer (NSCLC). It has become a significant public health concern due to its rising incidence rate and fatality. Tumor-associated macrophage (TAM) is important in the tumor microenvironment (TME) of NSCLC because they have an impact on the development, metastasis, and incidence of tumors. As a crucial element of the TME, TAM contributes to tumor immune evasion, facilitates tumor proliferation and metastasis, and modulates tumor angiogenesis, immunosuppression, and treatment resistance through the secretion of diverse cytokines, chemokines, and growth factors. Consequently, TAM assumes a multifaceted and intricate function in the onset, progression, and therapeutic response of NSCLC, serving as a crucial focal point for comprehending the tumor microenvironment and formulating novel therapeutic methods. The study aims to review the biological properties and potential processes of TAM in NSCLC, investigate its involvement in the clinical of NSCLC patients, and discuss its potential as a therapeutic target.
Collapse
Affiliation(s)
- Jiazheng Sun
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sirui Zhou
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yalu Sun
- Affiliated Hospital of Jining Medical University, Jining, China
| | - Yulan Zeng
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Wang H, Yang Y, Zhang G, Yang G, Wang Y, Liu L, Du J. Roles of anoikis in hepatocellular carcinoma therapy and the assessment of anoikis-regulatory molecules as therapeutic targets. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04088-w. [PMID: 40183941 DOI: 10.1007/s00210-025-04088-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
As the fourth leading cause of death from cancer and the sixth most common neoplasm in the world, hepatocellular carcinoma (HCC) is responsible for ninety percent of all primary liver cancers. There are four mechanisms that contribute to the spread of cancer: the separation of cells from the primary neoplasm, their survivability during metastasis, extravasation, and the development of secondary tumors at remote locations. In addition to its role in the development of a scaffold for cell adhesion, the extracellular matrix (ECM) also plays a role in the stimulation of signal transduction and the regulation of essential cellular mechanisms, including proliferation, migration, differentiation, and viability. The disruption of cell-ECM interactions and the ensuing separation of cells from the primary ECM trigger anoikis, a form of programmed cell death. One of the most effective factors in suppressing anoikis is ECM receptors from the integrin family. Cell migration, proliferation, and survival are primarily governed by the formation of physical connections with the cytoskeleton and the conveyance of signals between cells and the ECM via integrin receptors.
Collapse
Affiliation(s)
- Hongyu Wang
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China
| | - Yawen Yang
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China
| | - Gan Zhang
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China
| | - Guang Yang
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China
| | - Ying Wang
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China
| | - Lu Liu
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China
| | - Juan Du
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China.
| |
Collapse
|
9
|
Ding C, Gao H, Zhang L, Lu Z, Zhang B, Li D, Sun F. The signature based on interleukin family and receptors identified IL19 and IL20RA in promoting nephroblastoma progression through STAT3 pathway. Sci Rep 2025; 15:11639. [PMID: 40185911 PMCID: PMC11971410 DOI: 10.1038/s41598-025-96094-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Wilms tumor (WT) is a common renal malignancy in pediatric patients. Interleukin (receptors) (IL(R)s) play significant roles in tumor biology, however, their specific involvement in WT remains inadequately understood. We employed univariate Cox regression analysis to screen for certain IL(R) genes associated with prognosis and then analyzed their expression patterns. A prognostic model was constructed based on five selected IL(R)s using the LASSO Cox regression algorithm. To further elucidate the relationship between the prognostic model and the immune microenvironment, we conducted immune-related analyses. Additionally, we performed experiments to verify the roles of IL20RA and IL19 in WT. Finally, CNV, methylation and pan-cancer analysis were performed for IL19 and IL20RA. Our analysis ultimately identified five genes associated with prognosis: IL20RA, IL19, IL24, IL11 and IL17RD. The prognostic model incorporating these five genes demonstrated robust predictive power in both training and validation cohorts. Notably, IL19 and IL20RA were found to promote epithelial-mesenchymal transition (EMT) through the STAT3/SNAIL pathway, thereby contributing to tumor progression. Furthermore, significant differences in immune function and checkpoint expression were observed between the two groups. The high-risk group exhibiting a lower TIDE score, which suggests a potentially better response to immunotherapy. This study introduces a novel IL(R)-based prognostic signature for WT, highlighting IL20RA as a potential therapeutic target. These findings offer valuable insights for future studies on WT.
Collapse
Affiliation(s)
- Chen Ding
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, 107 Wenhua West Road, Jinan, 250012, China
| | - Hongjie Gao
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Liting Zhang
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, 107 Wenhua West Road, Jinan, 250012, China
| | - Zhiyi Lu
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, 107 Wenhua West Road, Jinan, 250012, China
| | - Bowen Zhang
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, 107 Wenhua West Road, Jinan, 250012, China
| | - Ding Li
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, 107 Wenhua West Road, Jinan, 250012, China
| | - Fengyin Sun
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, 107 Wenhua West Road, Jinan, 250012, China.
| |
Collapse
|
10
|
Fatima S. Tumor Microenvironment: A Complex Landscape of Cancer Development and Drug Resistance. Cureus 2025; 17:e82090. [PMID: 40351953 PMCID: PMC12066109 DOI: 10.7759/cureus.82090] [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] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
Cancer is responsible for nearly one in six global fatalities, making it a major health issue worldwide. Despite advancements in early detection, surgery, and targeted therapies, effective treatment remains challenging due to the complexity and heterogeneity of the disease. A key factor in cancer progression and resistance to treatment is the tumor microenvironment (TME). It is a complex ecosystem comprising cancer cells, stromal cells, immune cells, extracellular matrix (ECM), and soluble factors like cytokines and chemokines. These components interact dynamically to influence tumor growth, metastasis, immune evasion, and treatment resistance. Cancer cells drive the formation of the TME by releasing signaling molecules, while stromal cells, such as fibroblasts and endothelial cells, support tumor metabolism, angiogenesis, and invasion. Immune cells within the TME can either suppress or promote tumor progression, depending on their activation state. Additionally, the TME can promote the growth of immunosuppressive cells that aid cancer cells in evading immune surveillance, such as regulatory T-cells and myeloid-derived suppressor cells. The TME also impedes drug delivery by creating defective blood vessels, contributing to drug resistance. Recent technological advancements have deepened our understanding of the TME, revealing its role in immune modulation, metabolism, and extracellular matrix remodeling. As a result, targeting the TME has become a promising strategy to overcome treatment resistance and improve cancer therapy outcomes.
Collapse
Affiliation(s)
- Sohaila Fatima
- Pathology, College of Medicine, King Khalid University, Abha, SAU
| |
Collapse
|
11
|
Naldi L, Peri A, Fibbi B. Apelin/APJ: Another Player in the Cancer Biology Network. Int J Mol Sci 2025; 26:2986. [PMID: 40243599 PMCID: PMC11988549 DOI: 10.3390/ijms26072986] [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: 12/30/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
The apelinergic system exerts multiple biological activities in human pathologies, including cancer. Overactivation of apelin/APJ, which has been detected in many malignant tumors, and the strong correlation with progression-free and overall survival, suggested the role of an oncogene for the apelin gene. Emerging evidence sheds new light on the effects of apelin on cellular functions and homeostasis in cancer cells and supports a direct role for this pathway on different hallmarks of cancer: "sustaining proliferative signaling", "resisting cell death", "activating invasion and metastasis", "inducing/accessing vasculature", "reprogramming cellular metabolism", "avoiding immune destruction" and "tumor-promoting inflammation", and "enabling replicative immortality". This article reviews the currently available literature on the intracellular processes regulated by apelin/APJ, focusing on those pathways correlated with tumor development and progression. Furthermore, the association between the activity of the apelinergic axis and the resistance of cancer cells to oncologic treatments (chemotherapy, immunotherapy, radiation) suggests apelin/APJ as a possible target to potentiate traditional therapies, as well as to develop diagnostic and prognostic applications. This issue will be also covered in the review.
Collapse
Affiliation(s)
- Laura Naldi
- “Pituitary Diseases and Sodium Alterations” Unit, AOU Careggi, 50139 Florence, Italy; (L.N.); (B.F.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| | - Alessandro Peri
- “Pituitary Diseases and Sodium Alterations” Unit, AOU Careggi, 50139 Florence, Italy; (L.N.); (B.F.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| | - Benedetta Fibbi
- “Pituitary Diseases and Sodium Alterations” Unit, AOU Careggi, 50139 Florence, Italy; (L.N.); (B.F.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| |
Collapse
|
12
|
Nishida A, Andoh A. The Role of Inflammation in Cancer: Mechanisms of Tumor Initiation, Progression, and Metastasis. Cells 2025; 14:488. [PMID: 40214442 PMCID: PMC11987742 DOI: 10.3390/cells14070488] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/14/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
Inflammation is an essential component of the immune response that protects the host against pathogens and facilitates tissue repair. Chronic inflammation is a critical factor in cancer development and progression. It affects every stage of tumor development, from initiation and promotion to invasion and metastasis. Tumors often create an inflammatory microenvironment that induces angiogenesis, immune suppression, and malignant growth. Immune cells within the tumor microenvironment interact actively with cancer cells, which drives progression through complex molecular mechanisms. Chronic inflammation is triggered by factors such as infections, obesity, and environmental toxins and is strongly linked to increased cancer risk. However, acute inflammatory responses can sometimes boost antitumor immunity; thus, inflammation presents both challenges and opportunities for therapeutic intervention. This review examines how inflammation contributes to tumor biology, emphasizing its dual role as a critical factor in tumorigenesis and as a potential therapeutic target.
Collapse
Affiliation(s)
- Atsushi Nishida
- Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu 520-2192, Shiga, Japan;
| | | |
Collapse
|
13
|
Xiang J, Wang J, Xiao H, Huang C, Wu C, Zhang L, Qian C, Xiang D. Targeting tumor-associated macrophages in colon cancer: mechanisms and therapeutic strategies. Front Immunol 2025; 16:1573917. [PMID: 40191202 PMCID: PMC11968422 DOI: 10.3389/fimmu.2025.1573917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/10/2025] [Indexed: 04/09/2025] Open
Abstract
Colon cancer (CC) remains a primary contributor to cancer-related fatalities worldwide, driven by difficulties in early diagnosis and constrained therapeutic options. Recent studies underscore the importance of the tumor microenvironment (TME), notably tumor-associated macrophages (TAMs), in fostering malignancy progression and therapy resistance. Through their inherent plasticity, TAMs facilitate immunosuppression, angiogenic processes, metastatic spread, and drug tolerance. In contrast to M1 macrophages, which promote inflammatory and tumoricidal responses, M2 macrophages support tumor expansion and dissemination by exerting immunosuppressive and pro-angiogenic influences. Consequently, manipulating TAMs has emerged as a potential avenue to enhance treatment effectiveness. This review outlines the origins, polarization states, and functions of TAMs in CC, highlights their role in driving tumor advancement, and surveys ongoing efforts to target these cells for better patient outcomes. Emerging therapeutic strategies aimed at modulating TAM functions - including depletion strategies, reprogramming approaches that shift M2-polarized TAMs toward an M1 phenotype, and inhibition of key signaling pathways sustaining TAM-mediated immunosuppression-are currently under active investigation. These approaches hold promise in overcoming TAM - induced resistance and improving immunotherapeutic efficacy in CC.
Collapse
Affiliation(s)
- Jianqin Xiang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Jian Wang
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Huihui Xiao
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Chengchen Huang
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Chunrong Wu
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Lin Zhang
- Department of Gastroenterology, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Chenyuan Qian
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Debing Xiang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing, China
| |
Collapse
|
14
|
Zhao WJ, Wang ML, Zhao YF, Zhao WP, Huang QH, Lu ZW, Jia F, Shi JJ, Liu BS, Han WH, Lu HW, Zhang BC, Wang ZX. Pan-cancer analysis reveals SMARCAL1 expression is associated with immune cell infiltration and poor prognosis in various cancers. Sci Rep 2025; 15:6591. [PMID: 39994264 PMCID: PMC11850860 DOI: 10.1038/s41598-025-88955-9] [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: 06/07/2024] [Accepted: 02/03/2025] [Indexed: 02/26/2025] Open
Abstract
Although immune checkpoint inhibition in particular has shown promise in cancer immunotherapy, it is not always efficient. Recent studies suggest that SMARCAL1 may play a role in tumor immune evasion, yet its pan-cancer role is unclear. We conducted a comprehensive analysis of SMARCAL1 using TCGA, GTEx, and CCLE databases, evaluating its expression, genetic alterations, epigenetic modifications, and their clinical correlations across 33 cancer types. Our findings indicate that SMARCAL1 is overexpressed in several cancers, such as Glioma, LUAD, KIRC, and LIHC, impacting prognosis. Elevated SMARCAL1 is linked to poor outcomes in Glioma, LUAD, and LIHC but correlates with better survival in KIRC. We also found significant associations between SMARCAL1 expression and DNA methylation in 13 cancers. Furthermore, SMARCAL1 expression correlates with immune infiltration, suggesting it as a potential therapeutic target in cancer immunotherapy. This study underscores the need for further research on SMARCAL1 to enhance immunotherapeutic strategies.
Collapse
Affiliation(s)
- Wu-Jie Zhao
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Meng-Lei Wang
- Department of Digestive Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Yun-Fang Zhao
- Jitang College of North China University of Science and Technology, Tangshan, 063000, Hebei, China
| | - Wen-Peng Zhao
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Qiong-Hui Huang
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, 350108, Fujian, China
| | - Zhen-Wei Lu
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, 350108, Fujian, China
| | - Fang Jia
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, Guangdong, China
| | - Jin-Jin Shi
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Bo-Sen Liu
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Wan-Hong Han
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Han-Wen Lu
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Bing-Chang Zhang
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China.
| | - Zhan-Xiang Wang
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China.
| |
Collapse
|
15
|
Kuhl GC, Tangney M. Bacterial-Mediated In Situ Engineering of Tumour-Associated Macrophages for Cancer Immunotherapy. Cancers (Basel) 2025; 17:723. [PMID: 40075571 PMCID: PMC11899205 DOI: 10.3390/cancers17050723] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/29/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Tumour-associated macrophages (TAMs) are critical components of the tumour microenvironment (TME), significantly influencing cancer progression and treatment resistance. This review aims to explore the innovative use of engineered bacteria to reprogram TAMs, enhancing their anti-tumour functions and improving therapeutic outcomes. METHODS We conducted a systematic review following a predefined protocol. Multiple databases were searched to identify relevant studies on TAMs, their phenotypic plasticity, and the use of engineered bacteria for reprogramming. Inclusion and exclusion criteria were applied to select studies, and data were extracted using standardised forms. Data synthesis was performed to summarise the findings, focusing on the mechanisms and therapeutic benefits of using non-pathogenic bacteria to modify TAMs. RESULTS The review summarises the findings that engineered bacteria can selectively target TAMs, promoting a shift from the tumour-promoting M2 phenotype to the tumour-fighting M1 phenotype. This reprogramming enhances pro-inflammatory responses and anti-tumour activity within the TME. Evidence from various studies indicates significant tumour regression and improved immune responses following bacterial therapy. CONCLUSIONS Reprogramming TAMs using engineered bacteria presents a promising strategy for cancer therapy. This approach leverages the natural targeting abilities of bacteria to modify TAMs directly within the tumour, potentially improving patient outcomes and offering new insights into immune-based cancer treatments. Further research is needed to optimise these methods and assess their clinical applicability.
Collapse
Affiliation(s)
- Gabriela Christina Kuhl
- Cancer Research @UCC, College of Medicine and Health, University College Cork, T12 K8AF Cork, Ireland;
| | - Mark Tangney
- Cancer Research @UCC, College of Medicine and Health, University College Cork, T12 K8AF Cork, Ireland;
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland
| |
Collapse
|
16
|
Li M, Wang Z, Huang B, Lai Y, Zhang M, Lin C. Integrated analysis of M2 macrophage-related gene prognostic model and single-cell sequence to predict immunotherapy response in lung adenocarcinoma. Front Genet 2025; 16:1519677. [PMID: 39963673 PMCID: PMC11830816 DOI: 10.3389/fgene.2025.1519677] [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: 10/30/2024] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
Background Lung adenocarcinoma (LUAD) patients have high heterogeneity. The significance and clinical value of M2 macrophage-related genes in LUAD require further exploration. We aimed to construct a prognostic signature to predict the immunotherapy efficacy and prognosis in LUAD. Methods GSE26939 and GSE19188 chips were downloaded from the Gene Expression Omnibus (GEO). Weighted gene co-expression network analysis (WGCNA) and least absolute shrinkage and selection operator (LASSO) analysis were used to screen M2 macrophage-related prognostic genes. A signature based on M2 macrophage-related prognostic genes was established and used to predict the prognosis and immunotherapy efficacy in LUAD. Results Twenty-two M2 macrophage-related genes associated with the prognosis of LUAD were confirmed using WGCNA, and then two molecular subtypes were identified with significantly different survival, gene expressions, and clinic characteristics were classified. LASSO analysis identified nine M2 macrophage-related prognostic genes to establish a risk signature, classifying patients into low- and high-risk groups. Data indicated that low-risk patients had better survival. Moreover, the signature was an independent prognostic factor for LUAD and a potential biomarker for patients receiving immunotherapy. Single-cell transcriptome analysis may provide important information on molecular subtypes and heterogeneity. Conclusion Risk signature based on M2 macrophage-related genes is a valuable tool for predicting prognosis and immunotherapy response in patients with LUAD.
Collapse
Affiliation(s)
- Meifang Li
- Department of Medical Oncology, Fujian Cancer Hospital, Clinical Oncology School of Fujian Medical University, Fuzhou, China
| | - Zhiping Wang
- Department of Radiation Oncology, Fujian Cancer Hospital, Clinical Oncology School of Fujian Medical University, Fuzhou, China
| | - Bin Huang
- Department of Radiation Oncology, Fujian Cancer Hospital, Clinical Oncology School of Fujian Medical University, Fuzhou, China
| | - Yanyun Lai
- Department of Medical Oncology, Fujian Cancer Hospital, Clinical Oncology School of Fujian Medical University, Fuzhou, China
| | - Meng Zhang
- Department of Radiation Oncology, Fujian Cancer Hospital, Clinical Oncology School of Fujian Medical University, Fuzhou, China
| | - Cheng Lin
- Department of Radiation Oncology, Fujian Cancer Hospital, Clinical Oncology School of Fujian Medical University, Fuzhou, China
| |
Collapse
|
17
|
Dalpati N, Rai SK, Sharma P, Sarangi PP. Integrins and integrin-driven secretory pathways as multi-dimensional regulators of tumor-associated macrophage recruitment and reprogramming in tumor microenvironment. Matrix Biol 2025; 135:55-69. [PMID: 39645091 DOI: 10.1016/j.matbio.2024.12.003] [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: 06/30/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Integrins, a group of transmembrane receptors, play a crucial role in mediating the interactions between cells and extracellular matrix (ECM) proteins. The intracellular signaling initiated by these cell-matrix interactions in leukocytes mediates many essential cellular processes such as survival, migration, metabolism, and other immunological functions. Macrophages, as phagocytes, participate in both proinflammatory and anti-inflammatory processes, including progression. Numerous reports have shown that the integrin-regulated secretome, comprising cytokines, chemokines, growth factors, proteases, and other bioactive molecules, is a crucial modulator of macrophage functions in tumors, significantly influencing macrophage programming and reprogramming within the tumor microenvironment (TME) in addition to driving their step-by-step entry process into tumor tissue spaces. Importantly, studies have demonstrated a pivotal role for integrin receptor-mediated secretome and associated signaling pathways in functional reprogramming from anti-tumorigenic to pro-tumorigenic phenotype in tumor-associated macrophages (TAMs). In this comprehensive review, we have provided an in-depth analysis of the latest findings of various key pathways, mediators, and signaling cascades associated with integrin-driven polarization of macrophages in tumors. This manuscript will provide an updated understanding of the modulation of inflammatory monocytes/ macrophages and TAMs by integrin-driven secretory pathways in various functions such as migration, differentiation, and their role in tumor progression, angiogenesis, and metastasis.
Collapse
Affiliation(s)
- Nibedita Dalpati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Shubham Kumar Rai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Prerna Sharma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Pranita P Sarangi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India.
| |
Collapse
|
18
|
Pei Y, Ouyang W, Qi P, Yan Z, Li Y, Zhang X, Zhang C, Cui L. The influence of baseline platelet on mortality risk in stroke and cancer patients: a cross-sectional analysis of the NHANES database. BMC Neurol 2025; 25:30. [PMID: 39844121 PMCID: PMC11752736 DOI: 10.1186/s12883-025-04043-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/16/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Platelet count and function may be closely related to survival and prognosis of stroke and cancer. However, little is known on the impact of platelet count on the patients with a history of stroke and cancer. This study aimed to examine the association between baseline platelet level and all-cause mortality in this population using a cross-sectional analysis. METHODS Participants with a history of stroke and cancer were selected from the database of the National Health and Nutrition Examination Survey from 2007 to 2018. A maximum selected rank statistic was conducted to determine platelet cutoff with the most significant association with mortality. The association between platelet and mortality was characterized visually using restricted cubic spline (RCS). Weighted multivariable Cox regression models were performed to evaluate the association between platelet count and mortality. Time-dependent receiver operating characteristic (ROC) analysis was conducted to assess the accuracy of platelet count in predicting mortality. RESULTS Forty-three (43/113, 38.05%) stroke patients with cancer were alive at a median follow-up of 42 months (interquartile range, 23-74 months). The RCS analysis demonstrated a linear relationship between platelet and mortality (nonlinear, p = 0.352). Mortality in higher-platelet group (> 209 × 109/L, n = 57) was decreased than lower-platelet group (≤ 209 × 109/L, n = 56) (Model 1 HR 0.43, 95% CI 0.24-0.77, p = 0.005) (Model 2 HR 0.58, 95% CI 0.35-0.96, p = 0.03). Subgroup analyses showed no significant interaction between platelet and age, sex, BMI, WBC and neutrophil. The areas under time-dependent ROC curve of the 1-, 2-, 3-, 4- and 5-year survival rates were 0.54, 0.55, 0.57, 0.53, 0.59 for mortality of stroke patients with cancer. CONCLUSIONS Lower platelet count may be an independent predictor of all-cause mortality in population with a history of stroke and cancer. This result may provide valuable insights for the long-term management in stroke patients with cancer.
Collapse
Affiliation(s)
- Yuqi Pei
- Department of Neurology, The Second Hospital of Hebei Medical University, NO.309 Zhonghuabei Street, Hebei, 050000, China
| | - Wei Ouyang
- Department of Neurology, The Second Hospital of Hebei Medical University, NO.309 Zhonghuabei Street, Hebei, 050000, China
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Peiyun Qi
- Department of Neurology, The Second Hospital of Hebei Medical University, NO.309 Zhonghuabei Street, Hebei, 050000, China
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Zhongjie Yan
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Yaoru Li
- The Fourth Department of Cerebrovascular Disease, Cangzhou Central Hospital, No. 50 Xinhua West Road, Yunhe District, Cangzhou, 061000, Hebei Province, China
| | - Xiangjian Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, NO.309 Zhonghuabei Street, Hebei, 050000, China
- Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, 050000, Hebei, China
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, 050000, Hebei, China
| | - Cong Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, NO.309 Zhonghuabei Street, Hebei, 050000, China.
- Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, 050000, Hebei, China.
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, 050000, Hebei, China.
| | - Lili Cui
- Department of Neurology, The Second Hospital of Hebei Medical University, NO.309 Zhonghuabei Street, Hebei, 050000, China.
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, 050000, Hebei, China.
- Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
19
|
Zhong L, Li B, Zhang S, Li Q, Xiao G. Computational Identification of Migrating T cells in Spatial Transcriptomics Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.23.619870. [PMID: 39484480 PMCID: PMC11526994 DOI: 10.1101/2024.10.23.619870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
T cells are the central players in antitumor immunity, and effective tumor killing depends on their ability to infiltrate into the tumor microenvironment (TME) while maintaining normal cytotoxicity. However, late-stage tumors develop immunosuppressive mechanisms that impede T cell movement and induce exhaustion. Investigating T cell migration in human tumors in vivo could provide novel insights into tumor immune escape, although it remains a challenging task. In this study, we developed ReMiTT, a computational method that leverages spatial transcriptomics data to track T cell migration patterns within tumor tissue. Applying ReMiTT to multiple tumor samples, we identified potential migration trails. On these trails, chemokines that promote T-cell trafficking display an increasing trend. Additionally, we identified key genes and pathways enriched on these migration trails, including those involved in cytoskeleton rearrangement, leukocyte chemotaxis, cell adhesion, leukocyte migration, and extracellular matrix (ECM) remodeling. Furthermore, we characterized the phenotypes of T cells along these trails, showing that the migrating T cells are highly proliferative. Our findings introduce a novel approach for studying T cell migration and interactions within the tumor microenvironment (TME), offering valuable insights into tumor-immune dynamics.
Collapse
|
20
|
Yin G, Liu X, Yu X, Tan S, Liu F. Analysis of ICIs alone or in combination rechallenged outcomes after progression from first-line ICIs plus chemotherapy in patients with advanced non-small cell lung cancer. Sci Rep 2025; 15:30. [PMID: 39747923 PMCID: PMC11696066 DOI: 10.1038/s41598-024-83947-7] [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: 08/13/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) plus chemotherapy have become the standard of care for first-line treatment of advanced non-small cell lung cancer (NSCLC) with EGFR/ALK negative. However, there is no clear second-line treatment option after first-line treatment failure. To investigate the efficacy and safety of ICIs alone or in combination rechallenge treatment after first-line ICIs plus chemotherapy progression in advanced NSCLC. We retrospectively analyzed the cases of patients who received ICIs alone or in combination rechallenge treatment after first-line ICIs plus chemotherapy progression in advanced NSCLC at Hunan Cancer Hospital between January 2020 and May 2024. We evaluated the effects of continued immunotherapy on patients' objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and adverse events after first-line treatment progression, and analyzed the relationship between outcomes and clinical characteristics. A total of 154 patients were included, with 146 patients developing resistance, 8 patients showing no progression. The ORR was 16.44%, the DCR was 68.49%, and the median PFS was 4.6 months. Patients treated with the new immune drug therapy had longer PFS than those treated with the original immunotherapy (5.0 months vs. 3.7 months, p = 0.0438). The PFS in patients receiving ICIs plus targeted therapy was significantly longer than that in patients who receiving ICIs alone, chemo-ICIs plus targeted therapy and ICIs plus chemotherapy (chemo-ICIs) (5.7 months vs. 3.6 months vs3.2 months vs. 2.9 months, p = 0.0086). Multivariate analysis showed that treatment regimen was a risk factor for immune rechallenge PFS, but there was no statistical correlation between gender, age, smoking history, pathological type, intermittent treatment or first-line drug resistance and immune rechallenge PFS. Our findings suggest that selecting ICIs plus targeted therapy may improve PFS in patients with advanced NSCLC after first-line chemo-ICIs progression. while replacement with new BSAb/PD-1 may be more beneficial to patients. However, there is a lack of large sample randomized controlled studies and evidence-based medical evidence, and more clinical studies are needed to further confirm.
Collapse
Affiliation(s)
- Guisen Yin
- Department of Pharmacy, Yantai Hospital of Traditional Chinese Medicine, Yantai, 264000, Shandong, China
| | - Xin Liu
- Department of Oncology, Yantai Hospital of Traditional Chinese Medicine, Yantai, 264000, Shandong, China
| | - Xiangtao Yu
- Department of Pharmacy, Yantai Hospital of Traditional Chinese Medicine, Yantai, 264000, Shandong, China
| | - Song Tan
- Department of Oncology, Yantai Hospital of Traditional Chinese Medicine, Yantai, 264000, Shandong, China
| | - Fen Liu
- Department of Pharmacy, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
21
|
Tang W, Li X, Liu H, Xu C, Deng S. The role of macrophages in chronic pain. Cytokine 2025; 185:156813. [PMID: 39577336 DOI: 10.1016/j.cyto.2024.156813] [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: 10/08/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024]
Abstract
Chronic pain typically lasts or recurs for more than three months and is an unpleasant sensory and emotional experience, including neuropathic pain, long-term tissue damage, tumors, and viral or bacterial infections.The unpleasantness associated with pain affects the basic life of patients and has become a truly global problem. Macrophages, a powerful immune effector cell whose functional plasticity leads to polarization into different subtypes and opposite effects in different environments, are also indispensable in the development of pain.In recent years, there has been an increasing number of studies on the effects of macrophages on pain, and there are multiple pathways that regulate macrophage polarization, including lipopolysaccharide induction and IL-4/IL-13 stimulation.In addition, pathways involving macrophages and macrophage polarization have been found to have an exacerbating or mitigating role in the progression of chronic pain, with M1 macrophages generally exacerbating pain progression and M2 macrophages mitigating pain progression.Therefore, modulating macrophage polarization holds great promise as an intervention in chronic pain. In this paper, we synthesize multiple macrophage pathways as well as mechanisms affecting their pain processes in the context of different types of chronic pain, providing new avenues for chronic pain relief.
Collapse
Affiliation(s)
- Weikang Tang
- School of Medicine, Tarim University, Alaer, 843300 Xinjiang, China
| | - Xuan Li
- School of Medicine, Tarim University, Alaer, 843300 Xinjiang, China
| | - Huixia Liu
- School of Medicine, Tarim University, Alaer, 843300 Xinjiang, China
| | - Chunyan Xu
- School of Medicine, Tarim University, Alaer, 843300 Xinjiang, China
| | - Siyao Deng
- School of Medicine, Tarim University, Alaer, 843300 Xinjiang, China..
| |
Collapse
|
22
|
Yuan S, Zhu L, Luo Y, Chen X, Jing H, Wang J, Su X, Liang M, Zhuang Z. Igniting tumour microenvironment in triple-negative breast cancer using a mannose/hyaluronic acid dual-coated Ganoderma polysaccharide-superparamagnetic iron oxide nanocomplex for combinational therapies. J Drug Target 2025; 33:111-126. [PMID: 39470031 DOI: 10.1080/1061186x.2024.2408721] [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: 07/16/2024] [Revised: 08/31/2024] [Accepted: 09/21/2024] [Indexed: 10/30/2024]
Abstract
Eliciting tumour microenvironment (TME) activation in triple-negative breast cancer (TNBC) is crucial for effective anti-tumour therapies. The aim of this study is to employ pharmaceutical approaches to precisely deliver Ganoderma polysaccharide (GPS) to tumour sites, thereby enhancing TME activation. We first established a direct link between the accumulation of GPS within tumours and its efficacy in the TME activation. Building upon this insight, we then engineered a mannose/hyaluronic acid dual-coated GPS-loaded superparamagnetic iron oxide nanocomplex (Man/HA/GPS-SPIONs) with a particle size of 33.8 ± 1.6 nm and a zeta potential of -22.4 ± 3.5 mV, capable of precise tumour accumulation through magnet-assisted targeting and internalisation by tumour-associated macrophages (TAMs) and tumour cells, facilitated by dual ligand modification. In vitro, Man/HA/GPS-SPIONs effectively induced M1 polarisation of macrophages (CD86+ cells: 38.6 ± 2.8%), curbed 4T1 cell proliferation (viability: 47.3 ± 2.9%) and heightened Th1 cytokine release. Significantly, in vivo, Man/HA/GPS-SPIONs notably suppressed tumour growth (tumour index: 0.048 ± 0.005), fostered M1 polarisation of TAMs (CD45+F4/80+CD86+ cells: 26.1 ± 7.2%), consequently bolstering intratumoural T cytotoxic cells. This enhancement was intricately tied to the efficient co-delivery of GPS and iron ions to the tumours, made possible by the Man/HA/GPS-SPIONs delivery system. The synergistic effects with paclitaxel (PTX, inhibition rate: 61.2 ± 4.3%) and PD-1 inhibitors (inhibition rate: 69.8 ± 7.6%) underscored the translational potential of this approach. By harnessing a well-conceived iron-based drug delivery strategy, this study amplifies the tumour immune modulatory potential of natural polysaccharides, offering insightful guidance for interventions in the TME and synergistic therapies.
Collapse
Affiliation(s)
- Shaofei Yuan
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, P.R. China
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Linjia Zhu
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Yi Luo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Xiaoqiang Chen
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Haibo Jing
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Jiaqi Wang
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Xiangyu Su
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Meizhen Liang
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Zhixiang Zhuang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, P.R. China
| |
Collapse
|
23
|
Chan HW, Kuo DY, Shueng PW, Chuang HY. Visualizing the Tumor Microenvironment: Molecular Imaging Probes Target Extracellular Matrix, Vascular Networks, and Immunosuppressive Cells. Pharmaceuticals (Basel) 2024; 17:1663. [PMID: 39770505 PMCID: PMC11676442 DOI: 10.3390/ph17121663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
The tumor microenvironment (TME) is a critical factor in cancer progression, driving tumor growth, immune evasion, therapeutic resistance, and metastasis. Understanding the dynamic interactions within the TME is essential for advancing cancer management. Molecular imaging provides a non-invasive, real-time, and longitudinal approach to studying the TME, with techniques such as positron emission tomography (PET), magnetic resonance imaging (MRI), and fluorescence imaging offering complementary strengths, including high sensitivity, spatial resolution, and intraoperative precision. Recent advances in imaging probe development have enhanced the ability to target and monitor specific components of the TME, facilitating early cancer diagnosis, therapeutic monitoring, and deeper insights into tumor biology. By integrating these innovations, molecular imaging offers transformative potential for precision oncology, improving diagnostic accuracy and treatment outcomes through a comprehensive assessment of TME dynamics.
Collapse
Affiliation(s)
- Hui-Wen Chan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Beitou Dist., Taipei City 112, Taiwan;
| | - Deng-Yu Kuo
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Pei-Wei Shueng
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei City 112, Taiwan
| | - Hui-Yen Chuang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Beitou Dist., Taipei City 112, Taiwan;
| |
Collapse
|
24
|
Kuźnicki J, Janicka N, Białynicka-Birula B, Kuźnicki W, Chorążyczewska H, Deszcz I, Kulbacka J. How to Use Macrophages Against Cancer. Cells 2024; 13:1948. [PMID: 39682696 PMCID: PMC11639767 DOI: 10.3390/cells13231948] [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: 10/06/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Numerous studies have demonstrated the significant influence of immune cells on cancer development and treatment. This study specifically examines tumor-associated macrophages (TAMs), detailing their characteristics and roles in tumorigenesis and analyzing the impact of the ratio of TAM subtypes on patient survival and prognosis. It is established that TAMs interact with immunotherapy, radiotherapy, and chemotherapy, thereby influencing the efficacy of these treatments. Emerging therapies are explored, such as the use of nanoparticles (NPs) for drug delivery to target TAMs and modify the tumor microenvironment (TME). Additionally, novel anticancer strategies like the use of chimeric antigen receptor macrophages (CAR-Ms) show promising results. Investigations into the training of macrophages using magnetic fields, plasma stimulation, and electroporation are also discussed. Finally, this study presents prospects for the combination of TAM-based therapies for enhanced cancer treatment outcomes.
Collapse
Affiliation(s)
- Jacek Kuźnicki
- Students Scientific Group No.148, Faculty of Medicine, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (J.K.); (B.B.-B.); (H.C.)
| | - Natalia Janicka
- Students Scientific Group No.148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Barbara Białynicka-Birula
- Students Scientific Group No.148, Faculty of Medicine, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (J.K.); (B.B.-B.); (H.C.)
| | - Wojciech Kuźnicki
- Department of External Beam Radiotherapy, Nicolaus Copernicus Multidisciplinary Centre for Oncology and Traumatology, Pabianicka 62, 93-513 Łódź, Poland;
| | - Hanna Chorążyczewska
- Students Scientific Group No.148, Faculty of Medicine, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (J.K.); (B.B.-B.); (H.C.)
| | - Iwona Deszcz
- Department of Immunopathology and Molecular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine Santariškių g. 5, LT-08406 Vilnius, Lithuania
| |
Collapse
|
25
|
Dalpati N, Rai SK, Dash SP, Kumar P, Singh D, Sarangi PP. Integrins α5β1 and αvβ3 Differentially Participate in the Recruitment and Reprogramming of Tumor-associated Macrophages in the In Vitro and In Vivo Models of Breast Tumor. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1553-1568. [PMID: 39330703 DOI: 10.4049/jimmunol.2400180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024]
Abstract
Tumor-associated macrophages (TAMs) drive the protumorigenic responses and facilitate tumor progression via matrix remodeling, angiogenesis, and immunosuppression by interacting with extracellular matrix proteins via integrins. However, the expression dynamics of integrin and its correlation with TAM functional programming in the tumors remain unexplored. In this study, we examined surface integrins' role in TAM recruitment and phenotypic programming in a 4T1-induced murine breast tumor model. Our findings show that integrin α5β1 is upregulated in CD11b+Ly6Chi monocytes in the bone marrow and blood by day 10 after tumor induction. Subsequent analysis revealed elevated integrin α5β1 expression on tumor-infiltrating monocytes (Ly6ChiMHC class II [MHCII]low) and M1 TAMs (F4/80+Ly6ClowMHCIIhi), whereas integrin αvβ3 was predominantly expressed on M2 TAMs (F4/80+Ly6ClowMHCIIlow), correlating with higher CD206 and MERTK expression. Gene profiling of cells sorted from murine tumors showed that CD11b+Ly6G-F4/80+α5+ TAMs had elevated inflammatory genes (IL-6, TNF-α, and STAT1/2), whereas CD11b+Ly6G-F4/80+αv+ TAMs exhibited a protumorigenic phenotype (IL-10, Arg1, TGF-β, and STAT3/6). In vitro studies demonstrated that blocking integrin α5 and αv during macrophage differentiation from human peripheral blood monocytes reduced cell spreading and expression of CD206 and CD163 in the presence of specific matrix proteins, fibronectin, and vitronectin. Furthermore, RNA sequencing data analysis (GEO dataset: GSE195857) from bone marrow-derived monocytes and TAMs in 4T1 mammary tumors revealed differential integrin α5 and αv expression and their association with FAK and SRC kinase. In line with this, FAK inhibition during TAM polarization reduced SRC, STAT1, and STAT6 phosphorylation. In conclusion, these findings underscore the crucial role of integrins in TAM recruitment, polarization, and reprogramming in tumors.
Collapse
Affiliation(s)
- Nibedita Dalpati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Shubham Kumar Rai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Shiba Prasad Dash
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Puneet Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Divya Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Pranita P Sarangi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| |
Collapse
|
26
|
Costa S, Rodrigues J, Vieira C, Dias S, Viegas J, Castro F, Sarmento B, Leite Pereira C. Advancing osteosarcoma 3D modeling in vitro for novel tumor microenvironment-targeted therapies development. J Control Release 2024; 376:1068-1085. [PMID: 39505219 DOI: 10.1016/j.jconrel.2024.10.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/08/2024]
Abstract
Osteosarcoma (OS) represents one of the most common primary bone cancers affecting children and young adults. The available treatments have remained unimproved for the past decades, hampered by the poor knowledge of OS etiology/pathophysiology and the lack of innovative, predictive and biologically relevant in vitro models, that can recapitulate the 3D OS tumor microenvironment (TME). Here, we report the development and characterization of an innovative 3D model of OS, composed of OS tumor cells, immune cells (macrophages) and mesenchymal stem cells (MSCs), that formed a multicellular tissue spheroid (MCTS). This fully humanized 3D model was shown to accurately mimic the native histological features of OS, while innately leading to the polarization of macrophages towards an M2-like phenotype, highly aggressive and pro-tumor profile. Upon the exposure to immunomodulatory molecules, the MCTS were shown to be responsive by shifting macrophages polarization, and dramatically altering the TME secretome. In agreement, when treated with immunomodulatory/stimulatory nanoparticles (NPSs), we were able to revert the TME secretome towards an anti-inflammatory profile. This study establishes an advanced 3D OS model capable of shedding light on macrophages and MSCs contributions to disease progression, paving the way for the development of innovative therapeutic approaches targeting the OS TME, while providing a biologically relevant in vitro tool for the efficacy screening of novel OS therapeutic approaches.
Collapse
Affiliation(s)
- Sofia Costa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - João Rodrigues
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Carolina Vieira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; FMUP - Faculdade de Medicina, Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Sofia Dias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Juliana Viegas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Flávia Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS-CESPU - Instituto Universitário de Ciências da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Catarina Leite Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.
| |
Collapse
|
27
|
Ge Y, Jiang L, Yang C, Dong Q, Tang C, Xu Y, Zhong X. Interactions between tumor-associated macrophages and regulated cell death: therapeutic implications in immuno-oncology. Front Oncol 2024; 14:1449696. [PMID: 39575419 PMCID: PMC11578871 DOI: 10.3389/fonc.2024.1449696] [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: 06/15/2024] [Accepted: 10/21/2024] [Indexed: 11/24/2024] Open
Abstract
Tumor-associated macrophages (TAMs) play a pivotal role in sculpting the tumor microenvironment and influencing cancer progression, particularly through their interactions with various forms of regulated cell death (RCD), including apoptosis, pyroptosis, ferroptosis, and necroptosis. This review examines the interplay between TAMs and these RCD pathways, exploring the mechanisms through which they interact to promote tumor growth and advancement. We examine the underlying mechanisms of these intricate interactions, emphasizing their importance in cancer progression and treatment. Moreover, we present potential therapeutic strategies for targeting TAMs and manipulating RCD to enhance anti-tumor responses. These strategies encompass reprogramming TAMs, inhibiting their recruitment, and selectively eliminating them to enhance anti-tumor functions, alongside modulating RCD pathways to amplify immune responses. These insights offer a novel perspective on tumor biology and provide a foundation for the development of more efficacious cancer therapies.
Collapse
Affiliation(s)
- Yifei Ge
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lixue Jiang
- Department of Breast Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Chengru Yang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qingfu Dong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Chengwu Tang
- Department of Hepatopancreatobiliary Surgery, Huzhou Key Laboratory of Translational Medicine, First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Department of Hepatopancreatobiliary Surgery, Huzhou Key Laboratory of Translational Medicine, First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xiangyu Zhong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
28
|
Ulas A, Temel B, Kos FT. Comparison of Prognostic Values of Seven Immune Indexes in Advanced Non-Small-Cell Lung Cancer Treated with Nivolumab: How Effective Can They Be Regarding Our Treatment Decisions? MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1792. [PMID: 39596977 PMCID: PMC11596302 DOI: 10.3390/medicina60111792] [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: 09/26/2024] [Revised: 10/27/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024]
Abstract
Background and Objectives: In this study, we evaluated the impact of seven immune indexes on treatment response and survival outcomes in advanced non-small-cell lung cancer (NSCLC) patients receiving second-line and subsequent nivolumab treatment under real-life conditions. Materials and Methods: The pan-immune inflammation value (PIV), systemic immune inflammation value (SII), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), monocyte-to-lymphocyte ratio (MLR), derived neutrophil-to-lymphocyte ratio (d-NLR), and prognostic nutritional index (PNI) were calculated. All immune indexes were classified as low and high based on cut-off values. Kaplan-Meier and Cox hazard models were used for survival analysis. Results: The median follow-up was 22.0 months (6.0-96.0). The median overall survival (OS) was 30.0 months and the median progression-free survival (PFS) was 7.0 months. In the univariate analysis, comorbidity (p = 0.03) and nivolumab use for more than eight cycles (p < 0.0001) were associated with an increase in PFS, while smoking history (p < 0.005) and d-NLR (p < 0.05) were more effective regarding OS. Patients who received more than eight cycles of nivolumab had longer median PFS (4 vs. 19 months, p < 0.001) and OS (23 vs. 43 months, p < 0.001). We found longer median OS in the PLR (45.7 vs. 75.4 months; p = 0.05), PIV (53.0 vs. 66.4 months; p = 0.19), SII (50.0 vs. 71.9 vs. months, p = 0.19), and NLR (49.9 vs. 74.55 months, p = 0.10) indexes in nivolumab long-term users (high vs. low groups, respectively). In short-term users of nivolumab, only d-NLR median OS (high vs. low, 19 vs. 75.2 months, p = 0.07) was different. Complete and partial response rates to nivolumab treatment were higher in the PNI-high group (p = 0.04). Conclusions: In these real-life data, we determined that the PLR, PIV, SII, and NLR indexes were effective in the prognosis of patients who received PD1 inhibitor nivolumab for a long time, and the d-NLR index was effective in those who developed progression in a short time. We found that the PNI was effective in patients who responded well to ICI treatment.
Collapse
Affiliation(s)
- Arife Ulas
- Department of Medical Oncology, University of Health Sciences, Bursa City Education and Research Hospital, 16059 Bursa, Turkey
| | - Beyza Temel
- Department of Internal Medicine, University of Health Sciences, Bursa City Education and Research Hospital, 16059 Bursa, Turkey;
| | - Fahriye Tugba Kos
- Department of Medical Oncology, University of Health Sciences, Ankara City Education and Research Hospital, 06290 Ankara, Turkey;
| |
Collapse
|
29
|
Ji Y, Wang Y, Zhang N, Yang J, Li J, Zheng H, Wang L, Wang W, Li J. Mechanism of LMNB1 activating GPR84 through JAK-STAT pathway to mediate M2 macrophage polarization in lung cancer. Hum Immunol 2024; 85:111150. [PMID: 39357468 DOI: 10.1016/j.humimm.2024.111150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND It is reported that G protein-coupled receptor 84 (GPR84) can participate in inflammation and immune regulation to repress anti-tumor responses. However, the function of GPR84 in lung cancer (LC) and its potential molecular mechanisms are still largely unknown. METHODS Bioinformatics and molecular experiments were employed to assess the expression of GPR84 in LC. The pathways enriched by GPR84 were analyzed by the Kyoto Encyclopedia of Genes and Genomes. Bioinformatics prediction identified the potential upstream regulatory factors of GPR84, which were verified through dual luciferase and chromatin immunoprecipitation experiments. Cell viability was measured by methyl thiazolyl tetrazolium assay. The expression levels of key proteins related to the janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway such as JAK2, p-JAK2, p-STAT3, and STAT3 were detected by western blot. Macrophages were co-cultured with LC cells. Flow cytometry was employed to examine the proportion of mannose receptor-positive cells. The expression levels of M2 polarization marker genes chitinase-like protein 3, arginase-1, and found in inflammatory zone 1 were measured by quantitative reverse transcription polymerase chain reaction. We applied an enzyme-linked immunosorbent assay to determine levels of cytokines (interleukin-10 and transforming growth factor beta) to evaluate the M2 macrophage polarization. RESULTS GPR84 was highly expressed in LC and substantially enriched in the JAK-STAT pathway. GPR84 facilitated the M2 polarization of macrophages in LC. Adding the JAK-STAT pathway inhibitor weakened the promoting effect of GPR84 overexpression on M2 macrophage polarization. Furthermore, GPR84 also had an upstream regulatory factor lamin B1 (LMNB1). Knocking down LMNB1 blocked the JAK-STAT signaling pathway to repress M2 macrophage polarization in LC, while overexpression of GPR84 reversed the impact of LMNB1 knockdown on macrophage polarization. CONCLUSION The project suggested that the LMNB1/GPR84 axis can facilitate M2 polarization of macrophages in LC by triggering the JAK-STAT pathway. Targeting LMNB1/GPR84 or blocking the JAK-STAT pathway may be a novel approach for LC diagnosis and treatment.
Collapse
Affiliation(s)
- Yuanyuan Ji
- Department of Oncology, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Yuekun Wang
- Department of Oncology, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Ning Zhang
- Department of Oncology, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Junhong Yang
- Department of Oncology, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Jing Li
- Department of Oncology, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Hui Zheng
- Department of Oncology, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Lihua Wang
- Department of Oncology, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Weijie Wang
- Department of Surgical Oncology, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China.
| | - Junkuo Li
- Department of Pathology, Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China.
| |
Collapse
|
30
|
Shahgoli VK, Noorolyai S, Ahmadpour Youshanlui M, Saeidi H, Nasiri H, Mansoori B, Holmskov U, Baradaran B. Inflammatory bowel disease, colitis, and cancer: unmasking the chronic inflammation link. Int J Colorectal Dis 2024; 39:173. [PMID: 39465427 PMCID: PMC11513726 DOI: 10.1007/s00384-024-04748-y] [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] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND Chronic inflammation is a significant driver in the development of various diseases, including cancer. Colitis-associated colorectal cancer (CA-CRC) refers to the increased risk of colorectal cancer in individuals with chronic inflammatory bowel diseases (IBD) such as ulcerative colitis and Crohn's disease. METHODS This narrative review examines the link between chronic inflammation and CA-CRC. A comprehensive literature search was conducted using PubMed, Scopus, and Web of Science, focusing on studies published between 2000 and 2024. Studies were selected based on relevance to the role of inflammation in CA-CRC, specifically targeting molecular pathways and clinical implications. Both clinical and mechanistic studies were reviewed. CONCLUSION Sustained inflammation in the colon fosters a pro-tumorigenic environment, leading to the initiation and progression of CA-CRC. Prevention strategies must focus on controlling chronic inflammation, optimizing IBD management, and implementing regular screenings. Emerging therapies targeting key inflammatory pathways and immune responses, along with microbiome modulation, hold promise for reducing CA-CRC risk. Understanding these molecular mechanisms provides a path toward personalized treatment and better outcomes for patients with IBD at risk of colorectal cancer.
Collapse
Affiliation(s)
- Vahid Khaze Shahgoli
- Faculty of Medicine, Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Saeed Noorolyai
- Faculty of Medicine, Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hossein Saeidi
- Faculty of Medicine, Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Nasiri
- Faculty of Medicine, Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, USA
| | - Uffe Holmskov
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Behzad Baradaran
- Faculty of Medicine, Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
31
|
Budek M, Nuszkiewicz J, Czuczejko J, Maruszak-Parda M, Wróblewska J, Wojtasik J, Hołyńska-Iwan I, Pawłowska M, Woźniak A, Szewczyk-Golec K. Searching for New Biomarkers of Neuroendocrine Tumors: A Comparative Analysis of Chromogranin A and Inflammatory Cytokines in Patients with Neuroendocrine Tumors. Curr Oncol 2024; 31:6110-6132. [PMID: 39451760 PMCID: PMC11506232 DOI: 10.3390/curroncol31100456] [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: 09/02/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
Neuroendocrine neoplasms (NENs) present a diagnostic challenge due to their heterogeneous nature and non-specific clinical manifestations. This study aimed to explore novel biomarkers for NENs. Serum chromogranin A (CgA) levels and a panel of 48 inflammatory cytokines were analyzed in a cohort of 84 NEN patients and 40 healthy controls using enzyme-linked immunosorbent assay (ELISA) and multiplex ELISA. Significant alterations in cytokine levels were observed in the NEN patients compared to the controls, including elevated levels of pro-inflammatory cytokines, such as interleukin (IL)-6, IL-8, and tumor necrosis factor alpha (TNF-α), and reduced levels of angiogenic factors like platelet-derived growth factor-BB (PDGF-BB) and tumor necrosis factor beta (TNF-β). Notably, cytokines such as growth-regulated alpha protein (GRO-α) and TNF-β demonstrated strong potential as diagnostic markers, with receiver operating characteristic (ROC) curve analyses showing high sensitivity and specificity. Additionally, a positive correlation was found between CgA levels and several inflammatory cytokines, suggesting their synergistic role in tumor progression. These findings highlight the limited reliability of CgA alone as a diagnostic marker and underscore the importance of a multi-marker approach in diagnosing and monitoring NENs. Further research on a larger cohort is necessary to validate these biomarkers and their potential clinical applications.
Collapse
Affiliation(s)
- Marlena Budek
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland; (M.B.); (J.N.); (J.W.); (M.P.); (A.W.)
| | - Jarosław Nuszkiewicz
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland; (M.B.); (J.N.); (J.W.); (M.P.); (A.W.)
| | - Jolanta Czuczejko
- Department of Psychiatry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland;
- Department of Nuclear Medicine, Oncology Centre Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland;
| | - Marta Maruszak-Parda
- Department of Nuclear Medicine, Oncology Centre Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland;
| | - Joanna Wróblewska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland; (M.B.); (J.N.); (J.W.); (M.P.); (A.W.)
| | - Jakub Wojtasik
- Centre for Statistical Analysis, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland;
| | - Iga Hołyńska-Iwan
- Department of Pathobiochemistry and Clinical Chemistry, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 87-100 Toruń, Poland;
| | - Marta Pawłowska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland; (M.B.); (J.N.); (J.W.); (M.P.); (A.W.)
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland; (M.B.); (J.N.); (J.W.); (M.P.); (A.W.)
| | - Karolina Szewczyk-Golec
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland; (M.B.); (J.N.); (J.W.); (M.P.); (A.W.)
| |
Collapse
|
32
|
Muteeb G, Khafaga DS, El-Morsy MT, Farhan M, Aatif M, Hosney M. Targeting tumor-associated macrophages with nanocarrier-based treatment for breast cancer: A step toward developing innovative anti-cancer therapeutics. Heliyon 2024; 10:e37217. [PMID: 39309874 PMCID: PMC11415663 DOI: 10.1016/j.heliyon.2024.e37217] [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: 01/18/2024] [Revised: 08/06/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Tumor-associated macrophages (TAMs) promote tumor advancement in many ways, such as inducing angiogenesis and the formation of new blood vessels that provide tumors with nourishment and oxygen. TAMs also facilitate tumor invasion and metastasis by secreting enzymes that degrade the extracellular matrix and generating pro-inflammatory cytokines that enhance the migration of tumor cells. TAMs also have a role in inhibiting the immune response against malignancies. To accomplish this, they release immunosuppressive cytokines such as IL-10, and TAMs can hinder the function of T cells and natural killer cells, which play crucial roles in the immune system's ability to combat cancer. The role of TAMs in breast cancer advancement is a complex and dynamic field of research. Therefore, TAMs are a highly favorable focus for innovative breast cancer treatments. This review presents an extensive overview of the correlation between TAMs and breast cancer development as well as its role in the tumor microenvironment (TME) shedding light on their impact on tumor advancement and immune evasion mechanisms. Notably, our study provides an innovative approach to employing nanomedicine approaches for targeted TAM therapy in breast cancer, providing an in-depth overview of recent advances in this emerging field.
Collapse
Affiliation(s)
- Ghazala Muteeb
- Department of Nursing, College of Applied Medical Sciences, King Faisal University, Al-Ahsa, 31982, Saudi Arabia
| | - Doaa S.R. Khafaga
- Health Sector, Faculty of Science, Galala University, New Galala City, 43511, Suez, Egypt
| | - Manar T. El-Morsy
- Biotechnology Department, Faculty of Science, Cairo University, 12613, Giza, Egypt
| | - Mohd Farhan
- Department of Chemistry, College of Science, King Faisal University, Al Ahsa, 31982, Saudi Arabia
- Department of Basic Sciences, Preparatory Year Deanship, King Faisal University, Al Ahsa, 31982, Saudi Arabia
| | - Mohammad Aatif
- Department of Public Health, College of Applied Medical Sciences, King Faisal University, Al-Ahsa, 31982, Saudi Arabia
| | - Mohamed Hosney
- Zoology Department, Faculty of Science, Cairo University, 12613, Giza, Egypt
| |
Collapse
|
33
|
Maleki R, Ghith A, Heydarlou H, Grzeskowiak LE, Ingman WV. The Role of Breastmilk in Macrophage-Tumour Cell Interactions in Postpartum Breast Cancer. FRONT BIOSCI-LANDMRK 2024; 29:328. [PMID: 39344339 DOI: 10.31083/j.fbl2909328] [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: 06/28/2024] [Revised: 08/22/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Lactation is associated with long-term reduced risk of breast cancer. However, there is a transient increased risk of breast cancer in the 5 to 10 years postpartum and this is associated with a high incidence of metastasis and mortality. Breastmilk is a physiological fluid secreted by the mammary glands intimately connected with breast cells and the microenvironment that may affect postpartum breast cancer development and progression. This study aims to investigate the effect of breastmilk on interactions between breast cancer cells and macrophages in vitro. METHODS Human breastmilk from healthy donors (n = 7) was pooled and incubated with breast cancer (MCF-7 and MDA-MB-231) and macrophage (RAW264.7) cell lines to assess cell proliferation, viability, migration, and expression of key genes associated with epithelial-mesenchymal transition (EMT) and macrophage phenotype. Indirect co-culture studies assessed the effect of breastmilk on interactions between breast cancer cells and macrophages. RESULTS Breastmilk increased the proliferation and viability of breast cancer cells, reduced EMT markers, and reduced cell migration in MDA-MB-231 cells. Breastmilk decreased mRNA expression of interleukin 1B (IL1B) and interleukin 10 (IL10) in macrophages. Reduced EMT marker expression was observed in breast cancer cells co-cultured with macrophages pre-treated with breastmilk. Macrophages co-cultured with breast cancer cells pre-treated with breastmilk exhibited increased expression of a pro-inflammatory cytokine tumor necrosis factor A (TNFA) and pro-inflammatory nitric oxide synthase 2 (NOS2), and reduced expression of cytokines IL10 and transforming growth factor B1 (TGFB1) which are associated with the alternatively-activated macrophage phenotype. CONCLUSIONS Breastmilk has the potential to promote breast cancer proliferation, however, it can also reduce breast cancer progression through inhibition of breast cancer cell migration and regulation of macrophage polarisation. These findings suggest that breastmilk has potential to shape the tumour microenvironment in postpartum breast cancer.
Collapse
Affiliation(s)
- Reza Maleki
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| | - Amna Ghith
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| | - Hanieh Heydarlou
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| | - Luke E Grzeskowiak
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Women's and Children's Hospital, North Adelaide, SA 5006, Australia
| | - Wendy V Ingman
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| |
Collapse
|
34
|
Chakraborty R, Zaw T, Khodlan P, Darido C, Palmisano G, Chien A, Tay A, Ranganathan S, Liu F. Pseudonormal Morphology of Salivary Gland Adenoid Cystic Carcinoma Cells Subverts the Antitumor Reactivity of Immune Cells: A Tumour-Cell-Based Initiation of Immune Evasion. Cancer Rep (Hoboken) 2024; 7:e70019. [PMID: 39324702 PMCID: PMC11425664 DOI: 10.1002/cnr2.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 07/18/2024] [Accepted: 09/09/2024] [Indexed: 09/27/2024] Open
Abstract
INTRODUCTION Salivary gland adenoid cystic carcinoma (ACC), mucoepidermoid carcinoma (MEC) and oral squamous cell carcinoma (OSCC) occurs within the head and neck region. So far immune check point inhibitors failed in ACC. Gipie (CCDC88B) is a microtubule linker protein that activates immune cells. Gipie expressions found in head and neck cancer cells. We hypothesised that the presence of Gipie diminishes anti-tumour reactivity of immune cells towards head and neck cancer. METHOD To determine the effect of Gipie in oral and salivary gland cancer cells, Gipie was silenced in cancer cells in cancer-immune cells co-culture models and we performed 3D Z series confocal imaging, annexin V and immune activation flow cytometry, proteome profiler and discovery phase proteomics. RESULTS ACC cells morphed into pseudonormal morphology in immune co-culture models. Silencing Gipie in ACC cells showed significant increase of apoptotic cells and activated natural killer cells, and lowering of regulatory T cells. Other salivary and oral cancer cells showed negligible effect of Gipie. Proteome profiler and proteomics assay confirmed Gipie affecting proliferation mechanism and immune activated proteins in ACC immune co-culture models. CONCLUSION Overall, we conclude that the presence of Gipie has a confounding role during the ACC-immune cell interaction.
Collapse
Affiliation(s)
- Rajdeep Chakraborty
- Applied Biosciences, Faculty of Science and EngineeringMacquarie UniversitySydneyNew South WalesAustralia
- School of Natural Sciences, Faculty of Science and EngineeringMacquarie UniversitySydneyNew South WalesAustralia
| | - Thiri Zaw
- Australian Proteome Analysis Facility, Faculty of Science and EngineeringMacquarie UniversitySydneyNew South WalesAustralia
| | - Pallavi Khodlan
- Applied Biosciences, Faculty of Science and EngineeringMacquarie UniversitySydneyNew South WalesAustralia
| | - Charbel Darido
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVictoriaAustralia
| | - Giuseppe Palmisano
- School of Natural Sciences, Faculty of Science and EngineeringMacquarie UniversitySydneyNew South WalesAustralia
- GlycoProteomics Laboratory, Department of ParasitologyICB, University of Sao PauloSão PauloSão PauloBrazil
| | - Arthur Chien
- School of Natural Sciences, Faculty of Science and EngineeringMacquarie UniversitySydneyNew South WalesAustralia
| | - Aidan Tay
- Applied Biosciences, Faculty of Science and EngineeringMacquarie UniversitySydneyNew South WalesAustralia
- Australian e‐Health Research Centre, Transformational Bioinformatics GroupCSIRONew South WalesAustralia
| | - Shoba Ranganathan
- Applied Biosciences, Faculty of Science and EngineeringMacquarie UniversitySydneyNew South WalesAustralia
| | - Fei Liu
- School of Natural Sciences, Faculty of Science and EngineeringMacquarie UniversitySydneyNew South WalesAustralia
| |
Collapse
|
35
|
Yin M, Liu Z, Zhou Y, Li W, Yan J, Cao D, Yin L. Two-pronged anti-cancer nanovaccines enpowered by exogenous/endogenous tumor-associated antigens. J Control Release 2024; 373:358-369. [PMID: 39009083 DOI: 10.1016/j.jconrel.2024.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/01/2024] [Accepted: 07/09/2024] [Indexed: 07/17/2024]
Abstract
Cancer vaccines based on single-source (exogenous or endogenous) tumor-associated antigens (TAAs) are often challenged by the insufficient T cell response and the immunosuppressive tumor microenvironment (TME). Herein, a dual TAAs-boosted nanovaccine based on cancer cell (4T1) membrane-cloaked, CO-immobilized Prussian blue nanoparticles (4T1-PB-CO NPs) is developed and coupled with anti-interleukin (IL)-10 therapy to maximize the efficacy of antitumor immunotherapy. 4T1 cell membrane not only endows NPs with tumor targeting ability, but also serves as exogenous TAAs to trigger CD4+ T cell response and M1-phenotype polarization of tumor-associated macrophages. Under near-infrared light irradiation, 4T1-PB-CO NPs release CO to induce immunogenic cell death (ICD) of tumor cells, thus generating endogenous TAAs to activate CD8+ T cell response. Meanwhile, ICD triggers release of damage-associated molecular patterns, which can promote DC maturation to amplify the antitumor T cell response. When combined with anti-IL-10 that reverses the immunosuppressive TME, 4T1-PB-CO NPs efficiently suppress the primary tumors and produce an abscopal effect to inhibit distant tumors in a breast tumor-bearing mouse model. Such a two-pronged cancer vaccine represents a promising paradigm for robust antitumor immunotherapy.
Collapse
Affiliation(s)
- Mengyuan Yin
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China
| | - Zhongmin Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China
| | - Yang Zhou
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| | - Wei Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China
| | - Jing Yan
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China
| | - Desheng Cao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China
| | - Lichen Yin
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China.
| |
Collapse
|
36
|
Moon TJ, Ta HM, Bhalotia A, Paulsen KE, Hutchinson DW, Arkema GM, Choi AS, Haynie MG, Ogunnaike L, Dever M, Wang LL, Karathanasis E. Nanoparticles targeting immune checkpoint protein VISTA induce potent antitumor immunity. J Immunother Cancer 2024; 12:e008977. [PMID: 39209454 PMCID: PMC11367342 DOI: 10.1136/jitc-2024-008977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Immune checkpoint protein V-domain immunoglobulin suppressor of T cell activation (VISTA) controls antitumor immunity and is a valuable target for cancer immunotherapy. Previous mechanistic studies have indicated that VISTA impairs the toll-like receptor (TLR)-mediated activation of myeloid antigen-presenting cells, promoting the expansion of myeloid-derived suppressor cells, and suppressing tumor-reactive cytotoxic T cell function. METHODS The aim of this study was to develop a dual-action lipid nanoparticle (dual-LNP) coloaded with VISTA-specific siRNA and TLR9 agonist CpG oligonucleotide. We used three murine preclinical tumor models, melanoma YUMM1.7, melanoma B16F10, and colon carcinoma MC38 to assess the functional synergy of the two cargoes of the dual LNP and therapeutic efficacy. RESULTS The dual-LNP synergistically augmented antitumor immune responses and rejected large established tumors whereas LNPs containing VISTA siRNA or CpG alone were ineffective. In comparison with therapies using the soluble CpG and a VISTA-specific monoclonal antibody, the dual-LNP demonstrated superior therapeutic efficacy yet with reduced systemic inflammatory cytokine production. In three murine models, the dual-LNP treatment achieved a high cure rate. Tumor rejection was associated with influx of immune cells to tumor tissues, augmented dendritic cell activation, production of proinflammatory cytokines, and improved function of cytotoxic T cells. CONCLUSIONS Our studies show the dual-LNP ensured codelivery of its synergistic cargoes to tumor-infiltrating myeloid cells, leading to simultaneous silencing of VISTA and stimulation of TLR9. As a result, the dual-LNP drove a highly potent antitumor immune response that rejected large aggressive tumors, thus may be a promising therapeutic platform for treating immune-cold tumors.
Collapse
Affiliation(s)
- Taylor J Moon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Hieu Minh Ta
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, Ohio, USA
| | - Anubhuti Bhalotia
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Kai E Paulsen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Diarmuid W Hutchinson
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Gabrielle M Arkema
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Andrew S Choi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Michiko G Haynie
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Laolu Ogunnaike
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Margee Dever
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Li Lily Wang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Efstathios Karathanasis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| |
Collapse
|
37
|
Liu H, Lv Z, Zhang G, Yan Z, Bai S, Dong D, Wang K. Molecular understanding and clinical aspects of tumor-associated macrophages in the immunotherapy of renal cell carcinoma. J Exp Clin Cancer Res 2024; 43:242. [PMID: 39169402 PMCID: PMC11340075 DOI: 10.1186/s13046-024-03164-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: 07/06/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024] Open
Abstract
Renal cell carcinoma (RCC) is one of the most common tumors that afflicts the urinary system, accounting for 90-95% of kidney cancer cases. Although its incidence has increased over the past decades, its pathogenesis is still unclear. Tumor-associated macrophages (TAMs) are the most prominent immune cells in the tumor microenvironment (TME), comprising more than 50% of the tumor volume. By interacting with cancer cells, TAMs can be polarized into two distinct phenotypes, M1-type and M2-type TAMs. In the TME, M2-type TAMs, which are known to promote tumorigenesis, are more abundant than M1-type TAMs, which are known to suppress tumor growth. This ratio of M1 to M2 TAMs can create an immunosuppressive environment that contributes to tumor cell progression and survival. This review focused on the role of TAMs in RCC, including their polarization, impacts on tumor proliferation, angiogenesis, invasion, migration, drug resistance, and immunosuppression. In addition, we discussed the potential of targeting TAMs for clinical therapy in RCC. A deeper understanding of the molecular biology of TAMs is essential for exploring innovative therapeutic strategies for the treatment of RCC.
Collapse
Affiliation(s)
- Han Liu
- Department of Urology, Shengjing Hospital of China Medical University, #36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Zongwei Lv
- Department of Urology, Shengjing Hospital of China Medical University, #36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Gong Zhang
- Department of Urology, Shengjing Hospital of China Medical University, #36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Zhenhong Yan
- Department of Urology, Shengjing Hospital of China Medical University, #36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Song Bai
- Department of Urology, Shengjing Hospital of China Medical University, #36 Sanhao Street, Shenyang, Liaoning, 110004, China.
| | - Dan Dong
- College of Basic Medical Science, China Medical University, #77 Puhe Road, Shenyang, Liaoning, 110122, China.
| | - Kefeng Wang
- Department of Urology, Shengjing Hospital of China Medical University, #36 Sanhao Street, Shenyang, Liaoning, 110004, China.
| |
Collapse
|
38
|
Sun M, Bai J, Wang H, Li M, Zhou L, Li S. Unraveling the relationship between anoikis-related genes and cancer-associated fibroblasts in liver hepatocellular carcinoma. Heliyon 2024; 10:e35306. [PMID: 39165997 PMCID: PMC11334810 DOI: 10.1016/j.heliyon.2024.e35306] [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: 11/28/2023] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
This study intended to determine the molecular subtypes of liver hepatocellular carcinoma (LIHC) on the strength of anoikis-related genes (ARGs) and to assess their prognostic value and prospective relationship with immune cell infiltration and cancer-associated fibroblasts (CAFs). Univariate Cox regression analysis yielded 66 prognosis-related ARGs and classified LIHC into two distinct subtypes, with subtype A demonstrating overexpression of most prognosis-related ARGs and a significant survival disadvantage. Furthermore, a reliable prediction model was developed using ARGs to evaluate the risk of LIHC patients. This model served as an independent prognostic indicator and a quantitative tool for clinical prognostic prediction. Additionally, subtype-specific differences in immune cell infiltration were observed, and the risk score was potentially linked to immune-related characteristics. Moreover, the study identified a significant association between CAF score and LIHC prognosis, with a low CAF score indicating a favorable patient prognosis. In conclusion, this study reveals the molecular mechanisms underlying the development and progression of LIHC and identifies potential therapeutic targets for the disease.
Collapse
Affiliation(s)
- Meng Sun
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Jiangtao Bai
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Haisong Wang
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Mei Li
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Long Zhou
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Shanfeng Li
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
39
|
Gryziak M, Kraj L, Stec R. The role of tumor-associated macrophages in hepatocellular carcinoma-from bench to bedside: A review. J Gastroenterol Hepatol 2024; 39:1489-1499. [PMID: 38651642 DOI: 10.1111/jgh.16564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 02/19/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024]
Abstract
Hepatocellular carcinoma is one of the most common cancers worldwide. Despite progress in treatment, recurrence after radical treatment is common, and the prognosis remains poor for patients with advanced disease. Therefore, there is a need to identify prognostic and predictive factors for the response to therapy or more intensive surveillance or treatment. Because the tumor microenvironment plays a crucial role in the development of cancer and metastasis, it is a crucial need to understand processes that are involved in carcinogenesis. Within the microenvironment, several immune cells with different roles are present. One of the most important of these is tumor-associated macrophages. These cells may exert either antitumor or protumor roles. Several studies have suggested that tumor-associated macrophages can be used as prognostic markers. Furthermore, they may be involved in resistance to immunotherapy or systemic treatment. As they play an important role in cancer development, tumor-associated macrophages are also a good target for therapy. In this review, we briefly summarize recent progress on knowledge regarding the basic molecular characteristics, impact on prognosis and potential clinical implications of tumor-associated macrophages in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Maciej Gryziak
- Department of Oncology, Medical University of Warsaw, Warsaw, Poland
| | - Leszek Kraj
- Department of Oncology, Medical University of Warsaw, Warsaw, Poland
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology Polish Academy of Sciences, Jastrzebiec, Poland
| | - Rafał Stec
- Department of Oncology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
40
|
Liu T, Yao W, Sun W, Yuan Y, Liu C, Liu X, Wang X, Jiang H. Components, Formulations, Deliveries, and Combinations of Tumor Vaccines. ACS NANO 2024; 18:18801-18833. [PMID: 38979917 DOI: 10.1021/acsnano.4c05065] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Tumor vaccines, an important part of immunotherapy, prevent cancer or kill existing tumor cells by activating or restoring the body's own immune system. Currently, various formulations of tumor vaccines have been developed, including cell vaccines, tumor cell membrane vaccines, tumor DNA vaccines, tumor mRNA vaccines, tumor polypeptide vaccines, virus-vectored tumor vaccines, and tumor-in-situ vaccines. There are also multiple delivery systems for tumor vaccines, such as liposomes, cell membrane vesicles, viruses, exosomes, and emulsions. In addition, to decrease the risk of tumor immune escape and immune tolerance that may exist with a single tumor vaccine, combination therapy of tumor vaccines with radiotherapy, chemotherapy, immune checkpoint inhibitors, cytokines, CAR-T therapy, or photoimmunotherapy is an effective strategy. Given the critical role of tumor vaccines in immunotherapy, here, we look back to the history of tumor vaccines, and we discuss the antigens, adjuvants, formulations, delivery systems, mechanisms, combination therapy, and future directions of tumor vaccines.
Collapse
Affiliation(s)
- Tengfei Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Wenyan Yao
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Wenyu Sun
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Yihan Yuan
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Chen Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xiaohui Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xuemei Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Hui Jiang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| |
Collapse
|
41
|
Mateiou C, Lokhande L, Diep LH, Knulst M, Carlsson E, Ek S, Sundfeldt K, Gerdtsson A. Spatial tumor immune microenvironment phenotypes in ovarian cancer. NPJ Precis Oncol 2024; 8:148. [PMID: 39026018 PMCID: PMC11258306 DOI: 10.1038/s41698-024-00640-8] [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: 12/04/2023] [Accepted: 07/09/2024] [Indexed: 07/20/2024] Open
Abstract
Immunotherapy has largely failed in ovarian carcinoma (OC), likely due to that the vast tumor heterogeneity and variation in immune response have hampered clinical trial outcomes. Tumor-immune microenvironment (TIME) profiling may aid in stratification of OC tumors for guiding treatment selection. Here, we used Digital Spatial Profiling combined with image analysis to characterize regions of spatially distinct TIME phenotypes in OC to assess whether immune infiltration pattern can predict presence of immuno-oncology targets. Tumors with diffuse immune infiltration and increased tumor-immune spatial interactions had higher presence of IDO1, PD-L1, PD-1 and Tim-3, while focal immune niches had more CD163 macrophages and a preliminary worse outcome. Immune exclusion was associated with presence of Tregs and Fibronectin. High-grade serous OC showed an overall stronger immune response and presence of multiple targetable checkpoints. Low-grade serous OC was associated with diffuse infiltration and a high expression of STING, while endometrioid OC had higher presence of CTLA-4. Mucinous and clear cell OC were dominated by focal immune clusters and immune-excluded regions, with mucinous tumors displaying T-cell rich immune niches.
Collapse
Affiliation(s)
- Claudia Mateiou
- Department of Pathology and Cytology, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | | | - Lan Hoa Diep
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Mattis Knulst
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Elias Carlsson
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Sara Ek
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Karin Sundfeldt
- Department of Obstetrics and Gynecology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Anna Gerdtsson
- Department of Immunotechnology, Lund University, Lund, Sweden.
| |
Collapse
|
42
|
Zhang K, Li G, Wang Q, Liu X, Chen H, Li F, Li S, Song X, Li Y. A disulfidptosis-related glucose metabolism and immune response prognostic model revealing the immune microenvironment in lung adenocarcinoma. Front Immunol 2024; 15:1398802. [PMID: 39091494 PMCID: PMC11291233 DOI: 10.3389/fimmu.2024.1398802] [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: 03/10/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024] Open
Abstract
Background Lung adenocarcinoma accounts for the majority of lung cancer cases and impact survival rate of patients severely. Immunotherapy is an effective treatment for lung adenocarcinoma but is restricted by many factors including immune checkpoint expression and the inhibitory immune microenvironment. This study aimed to explore the immune microenvironment in lung adenocarcinoma via disulfidptosis. Methods Public datasets of lung adenocarcinoma from the TCGA and GEO was adopted as the training and validation cohort. Based on the differences in the expression of disulfidptosis -related genes, a glucose metabolism and immune response prognostic model was constructed. The prognostic value and clinical relationship of the model were further explored. Immune-related analyses were performed according to CIBERSORT, ssGSEA, TIDE, IPS. Results We verified that the model could accurately predict the survival expectancy of lung adenocarcinoma patients. Patients with lung adenocarcinoma and a low-risk score had better survival outcomes according to the model. Moreover, the high-risk group tended to have an immunosuppressive effect, as reflected by the immune cell components, phenotypes and functions. We also found that the clinically relevant immune checkpoint CTLA-4 was significantly higher in low-risk group (P<0.05), indicating that the high-risk group may suffer worse tumor immunotherapy efficacy. Finally, we found that this model has accurate predictive value for the efficacy of immune checkpoint blockade in non-small cell lung cancer (P<0.05). Conclusion The prognostic model demonstrated the feasibility of predicting survival and immunotherapy efficacy via disulfidptosis-related genes and will facilitate the development of personalized anticancer therapy.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Gang Li
- Graduate School, Kunming Medical University, Kunming, China
| | - Qin Wang
- Graduate School, Kunming Medical University, Kunming, China
| | - Xin Liu
- Department of Thoracic Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hong Chen
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Fuqiang Li
- Department of Traditional Chinese Medicine, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Shuangyan Li
- Graduate School, Kunming Medical University, Kunming, China
| | - Xinmao Song
- Department of Radiation Oncology, Ear, Nose & Throat Hospital of Fudan University, Shanghai, China
| | - Yi Li
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Kunming, China
| |
Collapse
|
43
|
Du Q, Dickinson A, Nakuleswaran P, Maghami S, Alagoda S, Hook AL, Ghaemmaghami AM. Targeting Macrophage Polarization for Reinstating Homeostasis following Tissue Damage. Int J Mol Sci 2024; 25:7278. [PMID: 39000385 PMCID: PMC11242417 DOI: 10.3390/ijms25137278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Tissue regeneration and remodeling involve many complex stages. Macrophages are critical in maintaining micro-environmental homeostasis by regulating inflammation and orchestrating wound healing. They display high plasticity in response to various stimuli, showing a spectrum of functional phenotypes that vary from M1 (pro-inflammatory) to M2 (anti-inflammatory) macrophages. While transient inflammation is an essential trigger for tissue healing following an injury, sustained inflammation (e.g., in foreign body response to implants, diabetes or inflammatory diseases) can hinder tissue healing and cause tissue damage. Modulating macrophage polarization has emerged as an effective strategy for enhancing immune-mediated tissue regeneration and promoting better integration of implantable materials in the host. This article provides an overview of macrophages' functional properties followed by discussing different strategies for modulating macrophage polarization. Advances in the use of synthetic and natural biomaterials to fabricate immune-modulatory materials are highlighted. This reveals that the development and clinical application of more effective immunomodulatory systems targeting macrophage polarization under pathological conditions will be driven by a detailed understanding of the factors that regulate macrophage polarization and biological function in order to optimize existing methods and generate novel strategies to control cell phenotype.
Collapse
Affiliation(s)
- Qiran Du
- Immuno-Bioengineering Group, School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Anna Dickinson
- Medical School, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (A.D.); (P.N.); (S.A.)
| | - Pruthvi Nakuleswaran
- Medical School, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (A.D.); (P.N.); (S.A.)
| | - Susan Maghami
- Hull York Medical School, University of York, York YO10 5DD, UK;
| | - Savindu Alagoda
- Medical School, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (A.D.); (P.N.); (S.A.)
| | - Andrew L. Hook
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Amir M. Ghaemmaghami
- Immuno-Bioengineering Group, School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
| |
Collapse
|
44
|
Zhu T, Alves SM, Adamo A, Wen X, Corn KC, Shostak A, Johnson S, Shaub ND, Martello SE, Hacker BC, D'Amore A, Bardhan R, Rafat M. Mammary tissue-derived extracellular matrix hydrogels reveal the role of irradiation in driving a pro-tumor and immunosuppressive microenvironment. Biomaterials 2024; 308:122531. [PMID: 38531198 PMCID: PMC11065579 DOI: 10.1016/j.biomaterials.2024.122531] [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: 12/18/2023] [Revised: 03/03/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024]
Abstract
Radiation therapy (RT) is essential for triple negative breast cancer (TNBC) treatment. However, patients with TNBC continue to experience recurrence after RT. The role of the extracellular matrix (ECM) of irradiated breast tissue in tumor recurrence is still unknown. In this study, we evaluated the structure, molecular composition, and mechanical properties of irradiated murine mammary fat pads (MFPs) and developed ECM hydrogels from decellularized tissues (dECM) to assess the effects of RT-induced ECM changes on breast cancer cell behavior. Irradiated MFPs were characterized by increased ECM deposition and fiber density compared to unirradiated controls, which may provide a platform for cell invasion and proliferation. ECM component changes in collagens I, IV, and VI, and fibronectin were observed following irradiation in both MFPs and dECM hydrogels. Encapsulated TNBC cell proliferation and invasive capacity was enhanced in irradiated dECM hydrogels. In addition, TNBC cells co-cultured with macrophages in irradiated dECM hydrogels induced M2 macrophage polarization and exhibited further increases in proliferation. Our study establishes that the ECM in radiation-damaged sites promotes TNBC invasion and proliferation as well as an immunosuppressive microenvironment. This work represents an important step toward elucidating how changes in the ECM after RT contribute to breast cancer recurrence.
Collapse
Affiliation(s)
- Tian Zhu
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Steven M Alves
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Arianna Adamo
- Ri.MED Foundation, Palermo, Italy; McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaona Wen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kevin C Corn
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Anastasia Shostak
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | | | - Nicholas D Shaub
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Shannon E Martello
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Benjamin C Hacker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Antonio D'Amore
- Ri.MED Foundation, Palermo, Italy; McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rizia Bardhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA; Nanovaccine Institute, Iowa State University, Ames, IA, USA
| | - Marjan Rafat
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
45
|
Guo M, Hu P, Xie J, Tang K, Hu S, Sun J, He Y, Li J, Lu W, Liu H, Liu M, Yi Z, Peng S. Remodeling the immune microenvironment for gastric cancer therapy through antagonism of prostaglandin E2 receptor 4. Genes Dis 2024; 11:101164. [PMID: 38560505 PMCID: PMC10980949 DOI: 10.1016/j.gendis.2023.101164] [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: 06/08/2023] [Revised: 10/17/2023] [Accepted: 10/31/2023] [Indexed: 04/04/2024] Open
Abstract
Gastric cancer is highly prevalent among digestive tract tumors. Due to the intricate nature of the gastric cancer immune microenvironment, there is currently no effective treatment available for advanced gastric cancer. However, there is promising potential for immunotherapy targeting the prostaglandin E2 receptor subtype 4 (EP4) in gastric cancer. In our previous study, we identified a novel small molecule EP4 receptor antagonist called YY001. Treatment with YY001 alone demonstrated a significant reduction in gastric cancer growth and inhibited tumor metastasis to the lungs in a mouse model. Furthermore, administration of YY001 stimulated a robust immune response within the tumor microenvironment, characterized by increased infiltration of antigen-presenting cells, T cells, and M1 macrophages. Additionally, our research revealed that YY001 exhibited remarkable synergistic effects when combined with the PD-1 antibody and the clinically targeted drug apatinib, rather than fluorouracil. These findings suggest that YY001 holds great promise as a potential therapeutic strategy for gastric cancer, whether used as a standalone treatment or in combination with other drugs.
Collapse
Affiliation(s)
- Mengmeng Guo
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Pan Hu
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jiayi Xie
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Kefu Tang
- Prenatal Diagnosis Center, Department of Clinical Laboratory, Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai 200051, China
| | - Shixiu Hu
- Key Laboratory of Acupuncture and Immunological Effects, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jialiang Sun
- Fengxian Hospital Affiliated to Southern Medical University, Shanghai 201400, China
| | - Yundong He
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jing Li
- Key Laboratory of Acupuncture and Immunological Effects, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Huirong Liu
- Key Laboratory of Acupuncture and Immunological Effects, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Zhengfang Yi
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Shihong Peng
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
- Shanghai Yuyao Biotech Co., Ltd., Shanghai 200241, China
| |
Collapse
|
46
|
Chen Y, Hou Y, Li S, Qin W, Zhang J. The N6-methyladenosine methylation landscape stratifies breast cancer into two subtypes with distinct immunological characteristics. Clin Exp Pharmacol Physiol 2024; 51:e13875. [PMID: 38797522 DOI: 10.1111/1440-1681.13875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 04/15/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024]
Abstract
N6-methyladenosine (m6A) methylation modification affects the tumorigenesis and metastasis of breast cancer (BC). This study investigated the association between m6A regulator-mediated methylation modification patterns and characterization of the tumour microenvironment in BC, as well as their prognostic importance. Public gene expression data and clinical annotations were collected from The Cancer Genome Atlas (TCGA) database, the Gene Expression Omnibus website and the METABRIC program. We analysed the genetic expression, gene-gene interactions, gene mutations and copy number variations using R software. The data were screened for risk genes using the Cox risk regression model, and we developed an algorithm for risk score and its predictive value. Compared to adjacent normal tissue, we identified 16 differentially expressed m6A regulators in BC, including six writers and 10 readers. Under unsupervised clustering, two distinguished modification patterns were identified, cluster C1 and C2. Compared to m6A cluster C2, cluster C1 was found to be more involved in immune-related pathways, with a relatively higher immune score and stromal score (P < 0.05). Patients were divided into two groups based on their risk scores for survival analysis. The patients in the high-risk score group had significantly worse overall survival than patients in the low-risk score group, (P < 0.0001). The TCGA database validation revealed the same prognostic tendency. In summary, our study showed distinct m6A regulator modification patterns contribute to the immunological heterogeneity and diversity of BC. The development of m6A gene signatures and the m6A score aid in the prognostic prediction of patients with BC.
Collapse
Affiliation(s)
- Yang Chen
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yijiang Hou
- School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Shuguang Li
- School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Wenxing Qin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
47
|
Mohamed AH, Ahmed AT, Al Abdulmonem W, Bokov DO, Shafie A, Al-Hetty HRAK, Hsu CY, Alissa M, Nazir S, Jamali MC, Mudhafar M. Interleukin-6 serves as a critical factor in various cancer progression and therapy. Med Oncol 2024; 41:182. [PMID: 38900329 DOI: 10.1007/s12032-024-02422-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
Interleukin-6 (IL-6), a pro-inflammatory cytokine, plays a crucial role in host immune defense and acute stress responses. Moreover, it modulates various cellular processes, including proliferation, apoptosis, angiogenesis, and differentiation. These effects are facilitated by various signaling pathways, particularly the signal transducer and activator of transcription 3 (STAT3) and Janus kinase 2 (JAK2). However, excessive IL-6 production and dysregulated signaling are associated with various cancers, promoting tumorigenesis by influencing all cancer hallmarks, such as apoptosis, survival, proliferation, angiogenesis, invasiveness, metastasis, and notably, metabolism. Emerging evidence indicates that selective inhibition of the IL-6 signaling pathway yields therapeutic benefits across diverse malignancies, such as multiple myeloma, prostate, colorectal, renal, ovarian, and lung cancers. Targeting key components of IL-6 signaling, such as IL-6Rs, gp130, STAT3, and JAK via monoclonal antibodies (mAbs) or small molecules, is a heavily researched approach in preclinical cancer studies. The purpose of this study is to offer an overview of the role of IL-6 and its signaling pathway in various cancer types. Furthermore, we discussed current preclinical and clinical studies focusing on targeting IL-6 signaling as a therapeutic strategy for various types of cancer.
Collapse
Affiliation(s)
- Asma'a H Mohamed
- Biomedical Engineering Department, College of Engineering and Technologies, Al-Mustaqbal University, Babil, Hilla, 51001, Iraq
| | - Abdulrahman T Ahmed
- Department of Nursing, Al-Maarif University College, Ramadi, AL-Anbar Governorate, Iraq.
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraidah, Kingdom of Saudi Arabia
| | - Dmitry Olegovich Bokov
- Institute of Pharmacy named after A.P. Nelyubin, Sechenov First Moscow State Medical University, 8 Trubetskaya St., bldg. 2, Moscow, Russian Federation, 119991
- Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, 2/14 Ustyinsky pr., Moscow, Russian Federation, 109240
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | | | - Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, AZ, 85004, USA
| | - Mohammed Alissa
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Shahid Nazir
- School of Science and Technology, University of New England, Armidale, NSW, Australia
| | - Mohammad Chand Jamali
- Faculty of Medical and Health Sciences, Liwa College, Al Ain, Abu Dhabi, United Arab Emirates
| | - Mustafa Mudhafar
- Department of Medical Physics, College of Applied Medical Sciences, University of Kerbala, Karbala, 56001, Iraq
- Department of Anesthesia Techniques and Intensive Care, Al-Taff University College, Kerbala, 56001, Iraq
| |
Collapse
|
48
|
Lin C, Teng W, Tian Y, Li S, Xia N, Huang C. Immune landscape and response to oncolytic virus-based immunotherapy. Front Med 2024; 18:411-429. [PMID: 38453818 DOI: 10.1007/s11684-023-1048-0] [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: 07/19/2023] [Accepted: 11/15/2023] [Indexed: 03/09/2024]
Abstract
Oncolytic virus (OV)-based immunotherapy has emerged as a promising strategy for cancer treatment, offering a unique potential to selectively target malignant cells while sparing normal tissues. However, the immunosuppressive nature of tumor microenvironment (TME) poses a substantial hurdle to the development of OVs as effective immunotherapeutic agents, as it restricts the activation and recruitment of immune cells. This review elucidates the potential of OV-based immunotherapy in modulating the immune landscape within the TME to overcome immune resistance and enhance antitumor immune responses. We examine the role of OVs in targeting specific immune cell populations, including dendritic cells, T cells, natural killer cells, and macrophages, and their ability to alter the TME by inhibiting angiogenesis and reducing tumor fibrosis. Additionally, we explore strategies to optimize OV-based drug delivery and improve the efficiency of OV-mediated immunotherapy. In conclusion, this review offers a concise and comprehensive synopsis of the current status and future prospects of OV-based immunotherapy, underscoring its remarkable potential as an effective immunotherapeutic agent for cancer treatment.
Collapse
Affiliation(s)
- Chaolong Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China
| | - Wenzhong Teng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China
| | - Yang Tian
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China
| | - Shaopeng Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China.
| | - Chenghao Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
49
|
Hoffmann E, Masthoff M, Kunz WG, Seidensticker M, Bobe S, Gerwing M, Berdel WE, Schliemann C, Faber C, Wildgruber M. Multiparametric MRI for characterization of the tumour microenvironment. Nat Rev Clin Oncol 2024; 21:428-448. [PMID: 38641651 DOI: 10.1038/s41571-024-00891-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 04/21/2024]
Abstract
Our understanding of tumour biology has evolved over the past decades and cancer is now viewed as a complex ecosystem with interactions between various cellular and non-cellular components within the tumour microenvironment (TME) at multiple scales. However, morphological imaging remains the mainstay of tumour staging and assessment of response to therapy, and the characterization of the TME with non-invasive imaging has not yet entered routine clinical practice. By combining multiple MRI sequences, each providing different but complementary information about the TME, multiparametric MRI (mpMRI) enables non-invasive assessment of molecular and cellular features within the TME, including their spatial and temporal heterogeneity. With an increasing number of advanced MRI techniques bridging the gap between preclinical and clinical applications, mpMRI could ultimately guide the selection of treatment approaches, precisely tailored to each individual patient, tumour and therapeutic modality. In this Review, we describe the evolving role of mpMRI in the non-invasive characterization of the TME, outline its applications for cancer detection, staging and assessment of response to therapy, and discuss considerations and challenges for its use in future medical applications, including personalized integrated diagnostics.
Collapse
Affiliation(s)
- Emily Hoffmann
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Max Masthoff
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Wolfgang G Kunz
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Max Seidensticker
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Stefanie Bobe
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Mirjam Gerwing
- Clinic of Radiology, University of Münster, Münster, Germany
| | | | | | - Cornelius Faber
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Moritz Wildgruber
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
50
|
Lee JH, Hallis SP, Kwak MK. Continuous TNF-α exposure in mammary epithelial cells promotes cancer phenotype acquisition via EGFR/TNFR2 activation. Arch Pharm Res 2024; 47:465-480. [PMID: 38734854 DOI: 10.1007/s12272-024-01497-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: 10/11/2023] [Accepted: 04/23/2024] [Indexed: 05/13/2024]
Abstract
Tumor necrosis factor alpha (TNF-α), an abundant inflammatory cytokine in the tumor microenvironment (TME), is linked to breast cancer growth and metastasis. In this study, we established MCF10A cell lines incubated with TNF-α to investigate the effects of continuous TNF-α exposure on the phenotypic change of normal mammary epithelial cells. The established MCF10A-LE cell line, through long-term exposure to TNF-α, displayed cancer-like features, including increased proliferation, migration, and sustained survival signaling even in the absence of TNF-α stimulation. Unlike the short-term exposed cell line MCF10A-SE, MCF10A-LE exhibited elevated levels of epidermal growth factor receptor (EGFR) and subsequent TNF receptor 2 (TNFR2), and silencing of EGFR or TNFR2 suppressed the cancer-like phenotype of MCF10A-LE. Notably, we demonstrated that the elevated levels of NAD(P)H oxidase 4 (NOX4) and the resulting increase in reactive oxygen species (ROS) were associated with EGFR/TNFR2 elevation in MCF10A-LE. Furthermore, mammosphere-forming capacity and the expression of cancer stem cell (CSC) markers increased in MCF10A-LE. Silencing of EGFR reversed these effects, indicating the acquisition of CSC-like properties via EGFR signaling. In conclusion, our results reveal that continuous TNF-α exposure activates the EGFR/TNFR2 signaling pathway via the NOX4/ROS axis, promoting neoplastic changes in mammary epithelial cells within the inflammatory TME.
Collapse
Affiliation(s)
- Jin-Hee Lee
- Integrated Research Institute for Pharmaceutical Sciences, The Catholic University of Korea, Bucheon, Gyeonggi‑do, 14662, Republic of Korea
| | - Steffanus Pranoto Hallis
- Department of Pharmacy and BK21FOUR Advanced Program for SmartPharma Leaders, Graduate School of The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| | - Mi-Kyoung Kwak
- Integrated Research Institute for Pharmaceutical Sciences, The Catholic University of Korea, Bucheon, Gyeonggi‑do, 14662, Republic of Korea.
- Department of Pharmacy and BK21FOUR Advanced Program for SmartPharma Leaders, Graduate School of The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea.
- College of Pharmacy, The Catholic University of Korea, 43 Jibong-ro, Bucheon, Gyeonggi-do, 14662, Republic of Korea.
| |
Collapse
|