1
|
Khabipov A, Miebach L, Lenz M, Kersting S, Bekeschus S. RAW264.7 Macrophages as a Polarization Model in the Context of Pancreatic Cancer and Chemokine Release. BIOLOGY 2025; 14:320. [PMID: 40282185 PMCID: PMC12024713 DOI: 10.3390/biology14040320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025]
Abstract
The TME is a critical niche for determining the fate of cancer therapy. Tumor cells often polarize nontumor cells, including immune cells, in the TME to favor cancer growth. In pancreatic cancer, macrophages are associated with poor therapy outcomes and unfavorable survival, especially when rendered into M2 macrophages. The latter show features also found in so-called tumor-associated macrophages (TAM), which are described as protecting and propelling tumor growth. In this context, it has been understudied which pancreatic cancer chemokines contribute to macrophage polarization. To this end, we analyzed murine RAW264.7 macrophages and Panc02 and PDA6606 pancreatic cancer cells in mono- and coculture to identify release patterns of 13 chemokines. Artificial macrophage polarization confirmed prominent changes in surface receptor and chemokine secretion profiles. Strikingly, RAW264.7 cocultures with Panc02 or PDA6606 were congruent in showing elevated levels of CCL2, CCL5, CCL17, CCL20, CCL22, CXCL5, and CXCL10. Further underlining the suitability of our in vitro model, both pancreatic cancer cell lines showed similar modulation of the critical macrophage polarization markers arginase, CD206, and iNOS, as well as chemokine receptors CCR2 and CCR4. Collectively, we demonstrated that our model is suitable for testing the roles and functions of chemokines in macrophage polarization by pancreatic cancer cells.
Collapse
Affiliation(s)
- Aydar Khabipov
- Department of General, Visceral, Thoracic, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Lea Miebach
- Department of General, Visceral, Thoracic, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Maik Lenz
- Department of General, Visceral, Thoracic, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Stephan Kersting
- Department of General, Visceral, Thoracic, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Sander Bekeschus
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
- Department of Dermatology and Venerology, Rostock University Medical Center, Strempelstr. 13, 18057 Rostock, Germany
| |
Collapse
|
2
|
Wang S, Kong L, Wang L, Zhuang Y, Guo C, Zhang Y, Cui H, Gu X, Wu J, Jiang C. Viral expression of NE/PPE enhances anti-colorectal cancer efficacy of oncolytic adenovirus by promoting TAM M1 polarization to reverse insufficient effector memory/effector CD8 + T cell infiltration. J Exp Clin Cancer Res 2025; 44:97. [PMID: 40082916 PMCID: PMC11907943 DOI: 10.1186/s13046-025-03358-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/05/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Oncolytic adenoviruses are among the most widely utilized oncolytic viruses due to their notable anti-tumor and gene expression capabilities, and modification of ADVs to create armed adenoviruses remains a popular research direction. Nonetheless, immune suppression triggered by ADV and targeted enhancements based on this limitation have been relatively unexplored. METHODS Flow cytometry was employed to assess immune infiltration in the tumor microenvironment following ADV therapy. Targeted novel recombinant oncolytic viruses, ADVNE and ADVPPE, were designed, and their antitumor efficacy, safety, and ability to reshape immune infiltration were evaluated in both subcutaneous tumor models in mice and in vitro experiments. Immune cell depletion assays confirmed the critical role of macrophages. The impact of HMGB1 on macrophage polarization was investigated using shRNA, qRT-PCR, ELISA, and flow cytometry. Furthermore, the importance of TLR4 and its downstream pathways was validated through immunoprecipitation, Western blotting, homozygous knockout mice, and TLR4 inhibitors. RESULTS We demonstrated that ADV limits the infiltration of effector memory/effector CD8 + T cells (TEM/TE) within the tumor microenvironment. To address this, we leveraged the strong capacity of NE or PPE to recruit TEM/TE by constructing novel recombinant oncolytic adenoviruses, ADVNE or ADVPPE, armed with NE or PPE. These recombinant viruses induce pyroptosis in colorectal cancer cells accompanied by the release of HMGB1. HMGB1 binds to TLR4 on the surface of macrophages, activating the MyD88-NFκB-NLRP3 (ASC) pathway and promoting M1 polarization of TAMs, thereby increasing TEM/TE cell infiltration and enhancing antitumor efficacy. CONCLUSIONS In summary, this study presents the development of the novel oncolytic adenoviruses ADVNE and ADVPPE with enhanced anti-tumor efficacy and provides an in-depth exploration of their specific anti-tumor mechanisms. These findings indicate promising clinical therapeutic prospects and offer new insights for advancing oncolytic adenovirus therapies.
Collapse
Affiliation(s)
- Shuo Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, Nanjing, 210093, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250021, China
| | - Lingkai Kong
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, Nanjing, 210093, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250021, China
| | - Linpei Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, 362000, China
| | - Yan Zhuang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, Nanjing, 210093, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250021, China
| | - Ciliang Guo
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, Nanjing, 210093, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250021, China
| | - Yuxin Zhang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, Nanjing, 210093, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250021, China
| | - Huawei Cui
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, Nanjing, 210093, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250021, China
| | - Xiaosong Gu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250021, China.
| | - Junhua Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, Nanjing, 210093, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250021, China.
| | - Chunping Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, National Institute of Healthcare Data Science at Nanjing University, Nanjing University, Nanjing, 210093, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250021, China.
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, 362000, China.
| |
Collapse
|
3
|
Toghraie FS, Bayat M, Hosseini MS, Ramezani A. Tumor-infiltrating myeloid cells; mechanisms, functional significance, and targeting in cancer therapy. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01051-y. [PMID: 39998754 DOI: 10.1007/s13402-025-01051-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 02/27/2025] Open
Abstract
Tumor-infiltrating myeloid cells (TIMs), which encompass tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), and tumor-associated dendritic cells (TADCs), are of great importance in tumor microenvironment (TME) and are integral to both pro- and anti-tumor immunity. Nevertheless, the phenotypic heterogeneity and functional plasticity of TIMs have posed challenges in fully understanding their complexity roles within the TME. Emerging evidence suggested that the presence of TIMs is frequently linked to prevention of cancer treatment and improvement of patient outcomes and survival. Given their pivotal function in the TME, TIMs have recently been recognized as critical targets for therapeutic approaches aimed at augmenting immunostimulatory myeloid cell populations while depleting or modifying those that are immunosuppressive. This review will explore the important properties of TIMs related to immunity, angiogenesis, and metastasis. We will also document the latest therapeutic strategies targeting TIMs in preclinical and clinical settings. Our objective is to illustrate the potential of TIMs as immunological targets that may improve the outcomes of existing cancer treatments.
Collapse
Affiliation(s)
- Fatemeh Sadat Toghraie
- Institute of Biotechnology, Faculty of the Environment and Natural Sciences, Brandenburg University of Technology, Cottbus, Germany
| | - Maryam Bayat
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Sadat Hosseini
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Amin Ramezani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
4
|
Qi X, Lian Y, Fan Z, Wang H, Jiang H, He M, Li L, Huang J, Wan Y. Electroacupuncture normalized tumor vasculature by downregulating glyoxalase-1 to polarize tumor-associated macrophage to M1 phenotype in triple-negative breast cancer. Int Immunopharmacol 2025; 147:113988. [PMID: 39778275 DOI: 10.1016/j.intimp.2024.113988] [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: 07/04/2024] [Revised: 12/18/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Triple-negative breast cancer is a particularly aggressive type of breast cancer that is closely associated with abnormal vascularization within the tumor. However, traditional anti-VEGF therapies and other treatments have limited efficacy. Tumor-associated macrophages (TAMs) induce and regulate tumor angiogenesis. In recent years, regulating TAMs polarization has become a hot topic for research with objectives to normalize tumor vasculature and improve drug delivery and the tumor microenvironment. Our previous studies have found that peritumoral electroacupuncture (EA) can regulate tumor angiogenesis, but the underlying mechanism remains unclear. METHODS In this study, we examined the phenotype of TAMs and inflammatory factors to observe the effect of peritumoral electroacupuncture on the phenotypic polarization of TAMs. Based on this, we evaluated the structure and function of tumor vasculature. Finally, we conducted a preliminary exploration of the mechanism underlying the regulation of TAMs phenotypic polarization by peritumoral electroacupuncture. RESULTS In this study, we found that peritumoral electroacupuncture could promote the phenotypic polarization of TAMs toward the M1 type, thereby reducing microvascular density in tumor tissue, increasing pericyte coverage, improving the stability of the basement membrane, promoting vascular maturation, and enhancing perfusion while reducing tissue hypoxia. CONCLUSIONS Peritumoral electroacupuncture can promote the phenotypic polarization of TAMs toward the M1 type, leading to normalization of tumor vascular structure and function. The mechanism may be related to the downregulation of glyoxalase-1 and subsequent activation of the MGO-AGEs/RAGE axis.
Collapse
Affiliation(s)
- Xuewei Qi
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Yanyan Lian
- Chaoyang District Hospital of Traditional Chinese Medicine, Beijing, China
| | - Zhenjia Fan
- Beijing University of Chinese Medicine, Beijing, China
| | - Hui Wang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Honglin Jiang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mengyang He
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Liling Li
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jinchang Huang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Yuxiang Wan
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
5
|
Yang Z, Chen Y, Miao Y, Yan H, Chen K, Xu Y, Su L, Zhang L, Yan Y, Chi H, Fu J, Wang L. Elucidating stearoyl metabolism and NCOA4-mediated ferroptosis in gastric cancer liver metastasis through multi-omics single-cell integrative mendelian analysis: advancing personalized immunotherapy strategies. Discov Oncol 2025; 16:46. [PMID: 39812999 PMCID: PMC11735723 DOI: 10.1007/s12672-025-01769-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The metabolism of stearoyl-GPE plays a key role in the liver metastasis of gastric cancer. This investigation delves into the mechanisms underlying the intricate tumor microenvironment (TME) heterogeneity triggered by stearoyl metabolism in gastric cancer with liver metastasis (LMGC), offering novel perspectives for LMGC. OBJECTIVE Utilizing Mendelian randomization, we determined that stearoyl metabolism significantly contributes to the progression of gastric cancer (GC). Following this, bulk transcriptome analyses and single-cell multiomics techniques to investigate the roles of stearoyl-GPE metabolism-related genes, particularly NCOA4, in regulating LMGC TME. RESULTS Our analysis highlights the crucial role of stearoyl metabolism in modulating the complex microenvironment of LMGC, particularly impacting monocyte cells. Through single-cell sequencing and spatial transcriptomics, we have identified key metabolic genes specific to stearoyl metabolism within the monocyte cell population, including NCOA4. Regarding the relationship between ferroptosis, stearoyl metabolism, and LMGC findings, it is plausible that stearoyl metabolism and LMGC pathways intersect with mechanisms involved in ferroptosis. Ferroptosis, characterized by iron-dependent lipid peroxidation, represents a regulated form of cell death. The activity of Stearoyl-CoA desaturase (SCD), a critical enzyme in stearoyl metabolism, has been associated with the modulation of lipid composition and susceptibility to ferroptosis. Furthermore, the LMGC is integral to cellular processes related to oxidative stress and lipid metabolism, both of which are significant factors in the context of ferroptosis. CONCLUSION This study enhances the understanding of the relationship between stearoyl metabolism and ferroptosis in promoting liver metastasis of gastric cancer and its role in the regulation of tumor heterogeneity. In addition, this study contributes to a deeper understanding of the dynamics of gastric cancer tumor microenvironment (TME) and provides a basis for the development of better interventions to combat cancer metastasis.
Collapse
Affiliation(s)
- Zhongqiu Yang
- Department of General Surgery, Dazhou Central Hospital, Dazhou, 635000, China
| | - Yuquan Chen
- School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, 3004, Australia
| | - Yaping Miao
- General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Haisheng Yan
- General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Kexin Chen
- General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yaoqin Xu
- General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Lanqian Su
- School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Lanyue Zhang
- School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yalan Yan
- School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Hao Chi
- School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- Western Institute of Digital-Intelligent Medicine, 401329, Chongqing, China.
| | - Jin Fu
- Department of Laboratory Medicine, Chonggang General Hospital, Chongqing, 400080, China.
| | - Lexin Wang
- Western Institute of Digital-Intelligent Medicine, 401329, Chongqing, China.
| |
Collapse
|
6
|
Wang S, Xu S, Li J, Wang N, Zheng Y, Wang Z. XIAOPI formula inhibits chemoresistance and metastasis of triple-negative breast cancer by suppressing extracellular vesicle/CXCL1-induced TAM/PD-L1 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156039. [PMID: 39303510 DOI: 10.1016/j.phymed.2024.156039] [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: 03/20/2024] [Revised: 07/02/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is challenged by the low chemotherapy response and poor prognosis. Emerging evidence suggests that cytotoxic chemotherapy may lead to the pro-metastatic tumor microenvironment (TME) by eliciting pro-tumor extracellular vesicles (EVs) from cancer cells. However, the precise mechanisms and therapeutic approaches remain inadequately understood. PURPOSE This study aims to determine whether XIAOPI formula (Chinese name XIAOPI San, XPS), a nationally sanctioned medication for mammary hyperplasia, can chemosensitize TNBC by remodeling the TME via modulating EV signaling, and exploring its underlying mechanisms. METHODS Multiple methodologies, such as EV isolation, transmission electron microscope, flow cytometry, dual-luciferase reporter assays, co-immunoprecipitation and in vivo breast cancer xenograft, were employed to elucidate the effect and molecular mechanisms of XPS on paclitaxel-induced EV signaling (EV-dead) of TNBC. RESULTS XPS, at non-toxic concentrations, synergized with PTX to inhibit the invasion and chemoresistance of TNBC cells co-cultured with macrophages. Compared to EV-dead, XPS co-treatment-elicited EVs (EV-deadXPS) exhibited a decreased capacity to promote the invasion, chemoresistance and cancer stem cell subpopulation of the co-cultured TNBC cells. Mechanistically, XPS administration led to a reduction in CXCL1 cargo in EV-dead, and thereby attenuated its activation effect on macrophage polarization into M2 phenotype through the transcriptional downregulation of PD-L1 expression. Furthermore, XPS effectively reduced the number of EV-dead from TNBC cells by inhibiting CXCL1-mediated intraluminal vesicle (ILV) biogenesis in multivesicular bodies (MVBs). Moreover, molecular explorations revealed that XPS impaired ILV biogenesis by disrupting the RAB31/FLOT2 complex via suppressing the CXCL1/Myc signaling. Importantly, XPS significantly chemosensitized paclitaxel to inhibit TNBC growth and metastasis in vivo by suppressing EV-deadCXCL1-induced PD-L1 activation and M2 polarization of macrophages. CONCLUSION This pioneering study not only sheds novel light on EV-deadCXCL1 as a potential therapeutic target to suppress TNBC chemoresistance and metastasis, but also provides XPS as a promising adjuvant formula to chemosensitize TNBC by remodeling EV-deadCXCL1-mediated immunosuppressive TME.
Collapse
Affiliation(s)
- Shengqi Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Breast Disease Specialist Hospital of Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, PR China; The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, PR China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Shang Xu
- Breast Disease Specialist Hospital of Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, PR China; The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Jing Li
- Breast Disease Specialist Hospital of Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, PR China; The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Neng Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, PR China; The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Yifeng Zheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Breast Disease Specialist Hospital of Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, PR China; The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Zhiyu Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Breast Disease Specialist Hospital of Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, PR China; The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, PR China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, PR China.
| |
Collapse
|
7
|
Kim YJ, Nanda SS, Jiang F, Pyo SY, Han JY, Koh SS, Kang TH. Pancreatic Adenocarcinoma Up-Regulated Factor (PAUF) Transforms Human Monocytes into Alternative M2 Macrophages with Immunosuppressive Action. Int J Mol Sci 2024; 25:11545. [PMID: 39519098 PMCID: PMC11547018 DOI: 10.3390/ijms252111545] [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: 10/02/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Tumor-associated macrophages (TAMs) in the tumor microenvironment (TME) promote immune evasion, cancer cell proliferation, and metastasis. Ongoing research is focused on finding ways to prevent tumor growth by inhibiting TAM polarization, which has shown a correlation with unfavorable prognosis in clinical studies. Pancreatic adenocarcinoma up-regulated factor (PAUF) is a protein secreted from pancreatic cancer (PC) and acts as a TME modulator that affects the TME by acting on not only cancer cells but also stromal cells and immune cells. Tumor cells can evade the immune system by PAUF binding to Toll-like receptor (TLR) in monocytes, as this research shows. In this study, the examination centered around the recruitment of human monocytes by PAUF and the subsequent differentiation into macrophages. In an in vitro chemotaxis assay, PAUF induced chemotactic migration of TLR2-mediated monocytes. In addition, PAUF induced differentiation of monocytes into M2 macrophages, which was verified based on expressing surface markers and cytokines and morphological analysis. The inhibition of T cell proliferation and function was observed in differentiated M2 macrophages. To conclude, these findings indicate that PAUF functions as a promoter of cancer progression by regulating the recruitment and differentiation of macrophages within TMEs, ultimately causing immunosuppression.
Collapse
Affiliation(s)
- Yeon Jeong Kim
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
- Department of Biomedical Sciences, Dong-A University, Busan 49315, Republic of Korea
| | - Sitansu Sekhar Nanda
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| | - Fen Jiang
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| | - Seung Yeon Pyo
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| | - Jin-Yeong Han
- Department of Laboratory Medicine, College of Medicine, Dong-A University, Busan 49201, Republic of Korea;
| | - Sang Seok Koh
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| | - Tae Heung Kang
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| |
Collapse
|
8
|
Fan Q, Fu ZW, Xu M, Lv F, Shi JS, Zeng QQ, Xiong DH. Research progress of tumor-associated macrophages in immune checkpoint inhibitor tolerance in colorectal cancer. World J Gastrointest Oncol 2024; 16:4064-4079. [DOI: 10.4251/wjgo.v16.i10.4064] [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: 06/05/2024] [Revised: 08/03/2024] [Accepted: 08/16/2024] [Indexed: 09/26/2024] Open
Abstract
The relevant mechanism of tumor-associated macrophages (TAMs) in the treatment of colorectal cancer patients with immune checkpoint inhibitors (ICIs) is discussed, and the application prospects of TAMs in reversing the treatment tolerance of ICIs are discussed to provide a reference for related studies. As a class of drugs widely used in clinical tumor immunotherapy, ICIs can act on regulatory molecules on cells that play an inhibitory role-immune checkpoints-and kill tumors in the form of an immune response by activating a variety of immune cells in the immune system. The sensitivity of patients with different types of colorectal cancer to ICI treatment varies greatly. The phenotype and function of TAMs in the colorectal cancer microenvironment are closely related to the efficacy of ICIs. ICIs can regulate the phenotypic function of TAMs, and TAMs can also affect the tolerance of colorectal cancer to ICI therapy. TAMs play an important role in ICI resistance, and making full use of this target as a therapeutic strategy is expected to improve the immunotherapy efficacy and prognosis of patients with colorectal cancer.
Collapse
Affiliation(s)
- Qi Fan
- Intestinal Center, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Zheng-Wei Fu
- Intestinal Center, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Ming Xu
- Intestinal Center, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Feng Lv
- Intestinal Center, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Jia-Song Shi
- Intestinal Center, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Qi-Qi Zeng
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
| | - De-Hai Xiong
- Intestinal Center, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| |
Collapse
|
9
|
Padzińska-Pruszyńska I, Kucharzewska P, Matejuk A, Górczak M, Kubiak M, Taciak B, Król M. Macrophages: Key Players in the Battle against Triple-Negative Breast Cancer. Int J Mol Sci 2024; 25:10781. [PMID: 39409110 PMCID: PMC11476577 DOI: 10.3390/ijms251910781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a challenging subtype of breast cancer characterized by the absence of estrogen and progesterone receptors and HER2 expression, leading to limited treatment options and a poorer prognosis. TNBC is particularly prevalent in premenopausal African-descent women and is associated with aggressive tumor behavior and higher metastatic potential. Tumor-associated macrophages (TAMs) are abundantly present within the TNBC microenvironment and play pivotal roles in promoting tumor growth, progression, and metastasis through various mechanisms, including immune suppression and enhancement of angiogenesis. This review provides an in-depth overview of TNBC, focusing on its epidemiology, its molecular characteristics, and the critical influence of TAMs. It discusses the pathological and molecular aspects that define TNBC's aggressive nature and reviews current and emerging therapeutic strategies aimed at targeting these dynamics. Special attention is given to the role of TAMs, exploring their potential as therapeutic targets due to their significant impact on tumor behavior and patient outcomes. This review aims to highlight the complexities of the TNBC landscape and to present the innovative approaches that are currently being pursued to improve therapeutic efficacy and patient survival.
Collapse
Affiliation(s)
- Irena Padzińska-Pruszyńska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.P.-P.); (P.K.); (M.G.); (M.K.); (B.T.)
| | - Paulina Kucharzewska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.P.-P.); (P.K.); (M.G.); (M.K.); (B.T.)
| | - Agata Matejuk
- Department of Immunology, Collegium Medicum, University of Zielona Góra, 65-417 Zielona Góra, Poland;
| | - Małgorzata Górczak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.P.-P.); (P.K.); (M.G.); (M.K.); (B.T.)
| | - Małgorzata Kubiak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.P.-P.); (P.K.); (M.G.); (M.K.); (B.T.)
| | - Bartłomiej Taciak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.P.-P.); (P.K.); (M.G.); (M.K.); (B.T.)
| | - Magdalena Król
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.P.-P.); (P.K.); (M.G.); (M.K.); (B.T.)
| |
Collapse
|
10
|
Zhang JY, Su YH, Wang X, Yao X, Du JZ. Recent Progress on Nanomedicine-Mediated Repolarization of Tumor-Associated Macrophages for Cancer Immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2001. [PMID: 39425549 DOI: 10.1002/wnan.2001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/07/2024] [Accepted: 09/18/2024] [Indexed: 10/21/2024]
Abstract
Tumor-associated macrophages (TAMs) constitute the largest number of immune cells in the tumor microenvironment (TME). They play an essential role in promoting tumor progression and metastasis, which makes them a potential therapeutic target for cancer treatment. TAMs are usually divided into two categories: pro-tumoral M2-like TAMs and antitumoral M1 phenotypes at either extreme. The reprogramming of M2-like TAMs toward a tumoricidal M1 phenotype is of particular interest for the restoration of antitumor immunity in cancer immunotherapy. Notably, nanomedicines have shown great potential for cancer therapy due to their unique structures and properties. This review will briefly describe the biological features and roles of TAMs in tumor, and then discuss recent advances in nanomedicine-mediated repolarization of TAMs for cancer immunotherapy. Finally, perspectives on nanomedicine-mediated repolarization of TAMs for effective cancer immunotherapy are also presented.
Collapse
Affiliation(s)
- Jing-Yang Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, China
| | - Yun-He Su
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, China
| | - Xu Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, China
| | - Xueqing Yao
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jin-Zhi Du
- School of Medicine, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
11
|
Liang X, Huang R, Ping X, Deng W, Xiang S, Wang Z, Cao J. Upregulation of CKS2 in immunosuppressive cells is associated with metastasis and poor prognosis in prostate cancer: a single-cell RNA-sequencing analysis. Transl Cancer Res 2024; 13:3996-4009. [PMID: 39262475 PMCID: PMC11385535 DOI: 10.21037/tcr-23-2100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 07/11/2024] [Indexed: 09/13/2024]
Abstract
Background Metastasis worsens prostate cancer (PCa) prognosis, with the immunosuppressive microenvironment playing a key role in bone metastasis. This study aimed to investigate how an immunosuppressive environment promotes PCa metastasis and worsens prognosis of patients with PCa. Methods Candidate oncogenes were identified through analysis of the Gene Expression Omnibus (GEO) database. A prognostic model was developed for the purpose of identifying target genes. A single-cell RNA sequencing data from GEO database was used to analyze the localization of target genes in the tumor microenvironment. A pan-cancer analysis was conducted to study the cancer-causing potential of target genes across different types of tumors. Results Fifty-one genes were found to be differentially expressed in bone metastasis compared to non-metastatic PCa, with CKS2 identified as the most significant gene associated with poor prognosis. CKS2 was shown to be linked to an immunosuppressive microenvironment and osteoclastic bone metastases, as shown by its negative correlation with immune cell infiltration and osteoblast-related gene expression. Moreover, CKS2 was found in immunosuppressive cells and was linked to bone metastasis in PCa. It was also overexpressed in different types of tumors, making it as an oncogenic gene. Conclusions This research offers a new perspective on the potential utility of CKS2 as a therapeutic target for the prevention of metastatic PCa.
Collapse
Affiliation(s)
- Xiaoxing Liang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Urology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Renlun Huang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Urology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Xinyue Ping
- The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Deng
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Urology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Songtao Xiang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Urology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Zhichao Wang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Urology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Jiadong Cao
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Urology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
12
|
Gu Q, Qi A, Wang N, Zhou Z, Zhou X. Macrophage dynamics in prostate cancer: Molecular to therapeutic insights. Biomed Pharmacother 2024; 177:117002. [PMID: 38960836 DOI: 10.1016/j.biopha.2024.117002] [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: 04/09/2024] [Revised: 06/08/2024] [Accepted: 06/17/2024] [Indexed: 07/05/2024] Open
Abstract
This review provides an in-depth examination of the role that tumor-associated macrophages (TAMs) play in the progression of prostate cancer (PCa), with a particular focus on the factors influencing the polarization of M1 and M2 macrophages and the implications of targeting these cells for cancer progression. The development and prognosis of PCa are significantly influenced by the behavior of macrophages within the tumor microenvironment. M1 macrophages typically exhibit anti-tumor properties by secreting pro-inflammatory cytokines such as interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α), thereby enhancing the immune response. Conversely, M2 macrophages contribute to tumor cell migration and invasion through the production of factors like arginase-1 (Arg1) and interleukin-10 (IL-10). This review not only explores the diverse factors that affect macrophage polarization but also delves into the potential therapeutic strategies targeting macrophage polarization, including the critical roles of non-coding RNA and exosomes in regulating this process. The polarization state of macrophages is highlighted as a key determinant in PCa progression, offering a novel perspective for clinical treatment. Future research should concentrate on gaining a deeper understanding of the molecular mechanisms underlying macrophage polarization and on developing effective targeted therapeutic strategies. The exploration of the potential of combination therapies to improve treatment efficacy is also emphasized. By emphasizing the importance of macrophages as a therapeutic target in PCa, this review aims to provide valuable insights and research directions for clinicians and researchers.
Collapse
Affiliation(s)
- Qiannan Gu
- China Pharmaceutical University, School of Basic Medicine and Clinical Pharmacy, Nanjing, Jiangsu 210009, China
| | - Anning Qi
- Medical Laboratory, Liuhe People's Hospital of Jiangsu Province, Nanjing, Jiangsu 211500, China
| | - Ne Wang
- Jiangning Hospital Tiandi New City Branch, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 211198, China
| | - Zhenxian Zhou
- Nanjing Second People's Hospital, Jiangsu Province 211103, China
| | - Xiaohui Zhou
- China Pharmaceutical University, School of Basic Medicine and Clinical Pharmacy, Nanjing, Jiangsu 210009, China; Jiangning Outpatient Department of China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
13
|
Li W, Yuan Q, Li M, He X, Shen C, Luo Y, Tai Y, Li Y, Deng Z, Luo Y. Research advances on signaling pathways regulating the polarization of tumor-associated macrophages in lung cancer microenvironment. Front Immunol 2024; 15:1452078. [PMID: 39144141 PMCID: PMC11321980 DOI: 10.3389/fimmu.2024.1452078] [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: 06/20/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024] Open
Abstract
Lung cancer (LC) is one of the most common cancer worldwide. Tumor-associated macrophages (TAMs) are important component of the tumor microenvironment (TME) and are closely related to the stages of tumor occurrence, development, and metastasis. Macrophages are plastic and can differentiate into different phenotypes and functions under the influence of different signaling pathways in TME. The classically activated (M1-like) and alternatively activated (M2-like) represent the two polarization states of macrophages. M1 macrophages exhibit anti-tumor functions, while M2 macrophages are considered to support tumor cell survival and metastasis. Macrophage polarization involves complex signaling pathways, and blocking or regulating these signaling pathways to enhance macrophages' anti-tumor effects has become a research hotspot in recent years. At the same time, there have been new discoveries regarding the modulation of TAMs towards an anti-tumor phenotype by synthetic and natural drug components. Nanotechnology can better achieve combination therapy and targeted delivery of drugs, maximizing the efficacy of the drugs while minimizing side effects. Up to now, nanomedicines targeting the delivery of various active substances for reprogramming TAMs have made significant progress. In this review, we primarily provided a comprehensive overview of the signaling crosstalk between TAMs and various cells in the LC microenvironment. Additionally, the latest advancements in novel drugs and nano-based drug delivery systems (NDDSs) that target macrophages were also reviewed. Finally, we discussed the prospects of macrophages as therapeutic targets and the barriers to clinical translation.
Collapse
Affiliation(s)
- Wenqiang Li
- Department of Respiratory and Critical Care Medicine, Zigong First People’s Hospital, Zigong, Sichuan, China
| | - Quan Yuan
- Department of Respiratory and Critical Care Medicine, Zigong First People’s Hospital, Zigong, Sichuan, China
| | - Mei Li
- West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoyu He
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Chen Shen
- West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yurui Luo
- West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yunze Tai
- West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Li
- Department of Respiratory and Critical Care Medicine, Zigong First People’s Hospital, Zigong, Sichuan, China
- West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiping Deng
- Department of Respiratory and Critical Care Medicine, Zigong First People’s Hospital, Zigong, Sichuan, China
| | - Yao Luo
- Department of Respiratory and Critical Care Medicine, Zigong First People’s Hospital, Zigong, Sichuan, China
- West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
14
|
Pei X, Zhang SL, Qiu BQ, Zhang PF, Liu TS, Wang Y. Cancer Cell Secreted Legumain Promotes Gastric Cancer Resistance to Anti-PD-1 Immunotherapy by Enhancing Macrophage M2 Polarization. Pharmaceuticals (Basel) 2024; 17:951. [PMID: 39065799 PMCID: PMC11279811 DOI: 10.3390/ph17070951] [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: 05/27/2024] [Revised: 06/19/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
The interaction between cancer cells and immune cells plays critical roles in gastric cancer (GC) progression and immune evasion. Forced legumain (LGMN) is one of the characteristics correlated with poor prognosis in gastric cancer patients. However, the role of gastric-cancer-secreted LGMN (sLGMN) in modulating the tumor immune microenvironment and the biological effect on the immune evasion of gastric cancer remains unclear. In this study, we found that forced expression of sLGMN in gastric cancer serum correlates with increased M2 macrophage infiltration in GC tissues and predicted resistance to anti-PD-1 immunotherapy. Mechanistically, gastric cancer cells secrete LGMN via binding to cell surface Integrin αvβ3, then activate Integrin αvβ3/PI3K (Phosphatidylinositol-4,5-bisphosphate3-kinase)/AKT (serine/threonine kinase)/mTORC2 (mammalian target of rapamycin complex 2) signaling, promote metabolic reprogramming, and polarize macrophages from the M1 to the M2 phenotype. Either blocking LGMN, Integrin αv, or knocking out Integrin αv expression and abolishing the LGMN/Integrin αvβ3 interaction significantly inhibits metabolic reprogramming and polarizes macrophages from the M1 to the M2 phenotype. This study reveals a critical molecular crosstalk between gastric cancer cells and macrophages through the sLGMN/Integrinαvβ3/PI3K/AKT/mTORC2 axis in promoting gastric cancer immune evasion and resistance to anti-PD-1 immunotherapy, indicating that the sLGMN/Integrinαvβ3/PI3K/AKT/mTORC2 axis may act as a promising therapeutic target.
Collapse
Affiliation(s)
- Xu Pei
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (X.P.); (S.-L.Z.); (P.-F.Z.)
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330030, China;
| | - Shi-Long Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (X.P.); (S.-L.Z.); (P.-F.Z.)
| | - Bai-Quan Qiu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330030, China;
| | - Peng-Fei Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (X.P.); (S.-L.Z.); (P.-F.Z.)
- Department of Medical Oncology, Shanghai Geriatric Medical Center, Shanghai 201104, China
| | - Tian-Shu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (X.P.); (S.-L.Z.); (P.-F.Z.)
- Cancer Center, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, China
- Center of Evidence-Based Medicine, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, China
| | - Yan Wang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (X.P.); (S.-L.Z.); (P.-F.Z.)
- Cancer Center, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, China
- Center of Evidence-Based Medicine, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, China
- Shanghai Medical College, Zhongshan Hospital Immunotherapy Translational Research Center, Shanghai 200032, China
| |
Collapse
|
15
|
Salmaninejad A, Layeghi SM, Falakian Z, Golestani S, Kobravi S, Talebi S, Yousefi M. An update to experimental and clinical aspects of tumor-associated macrophages in cancer development: hopes and pitfalls. Clin Exp Med 2024; 24:156. [PMID: 39003350 PMCID: PMC11246281 DOI: 10.1007/s10238-024-01417-w] [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: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
Tumor-associated macrophages (TAMs) represent one of the most abundant tumor-infiltrating stromal cells, and their normal function in tumor microenvironment (TME) is to suppress tumor cells by producing cytokines which trigger both direct cell cytotoxicity and antibody-mediated immune response. However, upon prolonged exposure to TME, the classical function of these so-called M1-type TAMs can be converted to another type, "M2-type," which are recruited by tumor cells so that they promote tumor growth and metastasis. This is the reason why the accumulation of TAMs in TME is correlated with poor prognosis in cancer patients. Both M1- and M2-types have high degree of plasticity, and M2-type cells can be reprogrammed to M1-type for therapeutic purposes. This characteristic introduces TAMs as promising target for developing novel cancer treatments. In addition, inhibition of M2-type cells and blocking their recruitment in TME, as well as their depletion by inducing apoptosis, are other approaches for effective immunotherapy of cancer. In this review, we summarize the potential of TAMs to be targeted for cancer immunotherapy and provide an up-to-date about novel strategies for targeting TAMs.
Collapse
Affiliation(s)
- Arash Salmaninejad
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Pediatric Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| | - Sepideh Mehrpour Layeghi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Falakian
- Department of Laboratory Science, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Shahin Golestani
- Department of Ophthalmology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepehr Kobravi
- Department of Oral and Maxillofacial Surgery, Tehran Azad University, Tehran, Iran
| | - Samaneh Talebi
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Yousefi
- Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
16
|
Pan X, Han T, Zhao Z, Wang X, Fang X. Emerging Nanotechnology in Preclinical Pancreatic Cancer Immunotherapy: Driving Towards Clinical Applications. Int J Nanomedicine 2024; 19:6619-6641. [PMID: 38975321 PMCID: PMC11227336 DOI: 10.2147/ijn.s466459] [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: 02/29/2024] [Accepted: 06/16/2024] [Indexed: 07/09/2024] Open
Abstract
The high malignant degree and poor prognosis of pancreatic cancer (PC) pose severe challenges to the basic research and clinical translation of next-generation therapies. The rise of immunotherapy has improved the treatment of a variety of solid tumors, while the application in PC is highly restricted by the challenge of immunosuppressive tumor microenvironment. The latest progress of nanotechnology as drug delivery platform and immune adjuvant has improved drug delivery in a variety of disease backgrounds and enhanced tumor therapy based on immunotherapy. Based on the immune loop of PC and the status quo of clinical immunotherapy of tumors, this article discussed and critically analyzed the key transformation difficulties of immunotherapy adaptation to the treatment of PC, and then proposed the rational design strategies of new nanocarriers for drug delivery and immune regulation, especially the design of combined immunotherapy. This review also put forward prospective views on future research directions, so as to provide information for the new means of clinical treatment of PC combined with the next generation of nanotechnology and immunotherapy.
Collapse
Affiliation(s)
- Xuan Pan
- Department of Hepato-Biliary-Pancreatic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, People’s Republic of China
| | - Ting Han
- Department of Gastroenterology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, People’s Republic of China
| | - Zixuan Zhao
- The Translational Research Institute for Neurological Disorders of Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, People’s Republic of China
- The Institute of Brain Science, Wannan Medical College, Wuhu, 241000, People’s Republic of China
| | - Xiaoming Wang
- Department of Hepato-Biliary-Pancreatic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, People’s Republic of China
| | - Xiaosan Fang
- Department of Hepato-Biliary-Pancreatic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, People’s Republic of China
| |
Collapse
|
17
|
Wang S, Li J, Xu S, Wang N, Pan B, Yang B, Zheng Y, Zhang J, Peng F, Peng C, Wang Z. Baohuoside I chemosensitises breast cancer to paclitaxel by suppressing extracellular vesicle/CXCL1 signal released from apoptotic cells. J Extracell Vesicles 2024; 13:e12493. [PMID: 39051750 PMCID: PMC11270583 DOI: 10.1002/jev2.12493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 06/29/2024] [Indexed: 07/27/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype and chemotherapy is the cornerstone treatment for TNBC. Regrettably, emerging findings suggest that chemotherapy facilitates pro-metastatic changes in the tumour microenvironment. Extracellular vesicles (EVs) have been highly implicated in cancer drug resistance and metastasis. However, the effects of the EVs released from dying cancer cells on TNBC prognosis and corresponding therapeutic strategies have been poorly investigated. This study demonstrated that paclitaxel chemotherapy elicited CXCL1-enriched EVs from apoptotic TNBC cells (EV-Apo). EV-Apo promoted the chemoresistance and invasion of co-cultured TNBC cells by polarizing M2 macrophages through activating PD-L1 signalling. However, baohuoside I (BHS) remarkably sensitized the co-cultured TNBC cells to paclitaxel chemotherapy via modulating EV-Apo signalling. Mechanistically, BHS remarkably decreased C-X-C motif chemokine ligand 1 (CXCL1) cargo within EV-Apo and therefore attenuated macrophage M2 polarization by suppressing PD-L1 activation. Additionally, BHS decreased EV-Apo release by diminishing the biogenesis of intraluminal vesicles (ILVs) within multivesicular bodies (MVBs) of TNBC cells. Furthermore, BHS bound to the LEU104 residue of flotillin 2 (FLOT2) and interrupted its interaction with RAS oncogene family member 31 (RAB31), leading to the blockage of RAB31-FLOT2 complex-driven ILV biogenesis. Importantly, BHS remarkably chemosensitised paclitaxel to inhibit TNBC metastasis in vivo by suppressing EV-ApoCXCL1-induced PD-L1 activation and M2 polarization of tumour-associated macrophages (TAMs). This pioneering study sheds light on EV-ApoCXCL1 as a novel therapeutic target to chemosensitise TNBC, and presents BHS as a promising chemotherapy adjuvant to improve TNBC chemosensitivity and prognosis by disturbing EV-ApoCXCL1 biogenesis.
Collapse
Affiliation(s)
- Shengqi Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
- State Key Laboratory of Southwestern Chinese Medicine ResourcesChengduUniversity of Traditional Chinese MedicineChengduSichuanChina
- Breast Disease Specialist Hospital of Guangdong Provincial Hospital ofChinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer Medicine, Disciplineof Integrated Chinese and Western MedicineThe Second Clinical College ofGuangzhou University of Chinese MedicineGuangzhouChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical SciencesGuangdong Provincial Hospital of Chinese MedicineGuangzhouChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouChina
| | - Jing Li
- Breast Disease Specialist Hospital of Guangdong Provincial Hospital ofChinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer Medicine, Disciplineof Integrated Chinese and Western MedicineThe Second Clinical College ofGuangzhou University of Chinese MedicineGuangzhouChina
| | - Shang Xu
- Breast Disease Specialist Hospital of Guangdong Provincial Hospital ofChinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer Medicine, Disciplineof Integrated Chinese and Western MedicineThe Second Clinical College ofGuangzhou University of Chinese MedicineGuangzhouChina
| | - Neng Wang
- The Research Center of Integrative Cancer Medicine, Disciplineof Integrated Chinese and Western MedicineThe Second Clinical College ofGuangzhou University of Chinese MedicineGuangzhouChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouChina
- The Research Center for Integrative Medicine, School of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouChina
| | - Bo Pan
- Breast Disease Specialist Hospital of Guangdong Provincial Hospital ofChinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer Medicine, Disciplineof Integrated Chinese and Western MedicineThe Second Clinical College ofGuangzhou University of Chinese MedicineGuangzhouChina
| | - Bowen Yang
- Breast Disease Specialist Hospital of Guangdong Provincial Hospital ofChinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer Medicine, Disciplineof Integrated Chinese and Western MedicineThe Second Clinical College ofGuangzhou University of Chinese MedicineGuangzhouChina
| | - Yifeng Zheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
- Breast Disease Specialist Hospital of Guangdong Provincial Hospital ofChinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer Medicine, Disciplineof Integrated Chinese and Western MedicineThe Second Clinical College ofGuangzhou University of Chinese MedicineGuangzhouChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouChina
| | - Juping Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
- Breast Disease Specialist Hospital of Guangdong Provincial Hospital ofChinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer Medicine, Disciplineof Integrated Chinese and Western MedicineThe Second Clinical College ofGuangzhou University of Chinese MedicineGuangzhouChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouChina
| | - Fu Peng
- Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education Ministry and Sichuan ProvinceWest China School of Pharmacy, Sichuan UniversityChengduChina
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine ResourcesChengduUniversity of Traditional Chinese MedicineChengduSichuanChina
| | - Zhiyu Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
- Breast Disease Specialist Hospital of Guangdong Provincial Hospital ofChinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer Medicine, Disciplineof Integrated Chinese and Western MedicineThe Second Clinical College ofGuangzhou University of Chinese MedicineGuangzhouChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical SciencesGuangdong Provincial Hospital of Chinese MedicineGuangzhouChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouChina
| |
Collapse
|
18
|
Huang S, He L, Zhao Y, Wei Y, Wang Q, Gao Y, Jiang X. TREM1 + tumor-associated macrophages secrete CCL7 to promote hepatocellular carcinoma metastasis. J Cancer Res Clin Oncol 2024; 150:320. [PMID: 38914803 PMCID: PMC11196310 DOI: 10.1007/s00432-024-05831-1] [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: 02/18/2024] [Accepted: 06/03/2024] [Indexed: 06/26/2024]
Abstract
PURPOSE Tumor-associated macrophages (TAMs) play a critical role in hepatocellular carcinoma (HCC) progression and metastasis. Systematic investigation of the cross-talk between TAMs and HCC may help in searching for the critical target to guard against HCC metastasis. METHODS AND RESULTS Herein, we found that TREM1 highly expressed in HCC tissue by analyzing the data obtain from GEO database. Interestingly, the results indicated that TREM1 was primarily expressed by monocytes. Immune infiltration studies further validated that TREM1 expression was positively related with increased infiltration of macrophages in HCC tissues. In vitro, we observed that TREM1 knockdown significantly abrogated the effect of TAMs in promoting the metastasis and epithelial-mesenchymal transition (EMT) of HCC cells. Additionally, cytokine array detection identified CCL7 as the main responsive cytokine following with TREM1 knockdown in TAMs. CONCLUSION Taken together, our findings strongly suggested that high expression of TREM1 was positively associated with metastasis and poor prognosis of HCC. Furthermore, TAMs expressing TREM1 contribute to EMT-based metastasis through secreting CCL7. These results provide a novel insight into the potential development of targeting the TREM1/CCL7 pathway for preventing metastatic HCC.
Collapse
Affiliation(s)
- Simin Huang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
- Liver Cancer Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Longguang He
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangdong Guangzhou, 510282, China
- Department of Hepatobiliary Surgery, Gaozhou People's Hospital, Guangdong Gaozhou, 525000, China
| | - Yufei Zhao
- Department of Gastrointestinal Surgery, Lab of Surgery, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Yuxuan Wei
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
- Liver Cancer Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Qiwen Wang
- Department of Gastrointestinal Surgery, Lab of Surgery, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Yi Gao
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangdong Guangzhou, 510282, China.
| | - Xiaofeng Jiang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.
- Liver Cancer Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.
| |
Collapse
|
19
|
Wang C, Gao Q, Wu J, Lu M, Wang J, Ma T. The Biological Role of Macrophage in Lung and Its Implications in Lung Cancer Immunotherapy. Adv Biol (Weinh) 2024; 8:e2400119. [PMID: 38684453 DOI: 10.1002/adbi.202400119] [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: 02/29/2024] [Revised: 04/07/2024] [Indexed: 05/02/2024]
Abstract
The lungs are the largest surface of the body and the most important organ in the respiratory system, which are constantly exposed to the external environment. Tissue Resident Macrophages in lung constitutes the important defense against external pathogens. Macrophages connects the innate and adaptive immune system, and also plays important roles in carcinogenesis and cancer immunotherapy. Lung cancer is the leading cause of cancer-related death worldwide, with an overall five-year survival rate of only 21%. Macrophages that infiltrate or aggregate in lung tumor microenvironment are defined as tumor-associated macrophages (TAMs). TAMs are the main components of immune cells in the lung tumor microenvironment. The differentiation and maturation process of TAMs can be roughly divided into two different types: classical activation pathway produces M1 tumor-associated macrophages, and bypass activation pathway produces M2 tumor-associated macrophages. Studies have found that TAMs are related to tumor invasion, metastasis, and treatment resistance, and show potential as a new target for tumor immunotherapy. Therefore, the biological function of macrophages in lung and the role of TAMs in the occurrence, development, and treatment of lung cancer are discussed in this paper.
Collapse
Affiliation(s)
- Chenyang Wang
- Cancer Research Center, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Qing Gao
- Cancer Research Center, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Jinghong Wu
- Cancer Research Center, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Mingjun Lu
- Cancer Research Center, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Jinghui Wang
- Cancer Research Center, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Teng Ma
- Cancer Research Center, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| |
Collapse
|
20
|
Su P, Li O, Ke K, Jiang Z, Wu J, Wang Y, Mou Y, Jin W. Targeting tumor‑associated macrophages: Critical players in tumor progression and therapeutic strategies (Review). Int J Oncol 2024; 64:60. [PMID: 38695252 PMCID: PMC11087038 DOI: 10.3892/ijo.2024.5648] [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: 03/01/2024] [Accepted: 04/19/2024] [Indexed: 05/12/2024] Open
Abstract
Tumor‑associated macrophages (TAMs) are essential components of the tumor microenvironment (TME) and display phenotypic heterogeneity and plasticity associated with the stimulation of bioactive molecules within the TME. TAMs predominantly exhibit tumor‑promoting phenotypes involved in tumor progression, such as tumor angiogenesis, metastasis, immunosuppression and resistance to therapies. In addition, TAMs have the potential to regulate the cytotoxic elimination and phagocytosis of cancer cells and interact with other immune cells to engage in the innate and adaptive immune systems. In this context, targeting TAMs has been a popular area of research in cancer therapy, and a comprehensive understanding of the complex role of TAMs in tumor progression and exploration of macrophage‑based therapeutic approaches are essential for future therapeutics against cancers. The present review provided a comprehensive and updated overview of the function of TAMs in tumor progression, summarized recent advances in TAM‑targeting therapeutic strategies and discussed the obstacles and perspectives of TAM‑targeting therapies for cancers.
Collapse
Affiliation(s)
- Pengfei Su
- Department of General Surgery, Cancer Center, Division of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Ou Li
- Department of General Surgery, Cancer Center, Division of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Kun Ke
- Department of General Surgery, Cancer Center, Division of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Zhichen Jiang
- Department of General Surgery, Cancer Center, Division of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Jianzhang Wu
- Department of General Surgery, Cancer Center, Division of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Yuanyu Wang
- Department of General Surgery, Cancer Center, Division of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Yiping Mou
- Department of General Surgery, Cancer Center, Division of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Weiwei Jin
- Department of General Surgery, Cancer Center, Division of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| |
Collapse
|
21
|
Chen C, Huang R, Wang N, Zheng Y, Zhou J, Yang B, Wang X, Zhang J, Pan B, Chen Z, Wang S, Wang Z, Xiang S. Fu-Zheng-Yi-Liu Formula inhibits the stem cells and metastasis of prostate cancer via tumor-associated macrophages/C-C motif chemokine ligand 5 pathway in tumor microenvironment. Chin J Nat Med 2024; 22:501-514. [PMID: 38906598 DOI: 10.1016/s1875-5364(24)60653-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Indexed: 06/23/2024]
Abstract
Prostate cancer (PCa) is the second most common malignancy among men globally. The Fu-Zheng-Yi-Liu (FZYL) Formula has been widely utilized in the treatment of PCa. This study investigates whether the FZYL Formula can inhibit PCa by targeting the TAMs/CCL5 pathway. We conducted in vitro co-cultures and in vivo co-injections of PCa cells and TAMs to mimic their interaction. Results showed that the FZYL Formula significantly reduced the proliferation, colony formation, subpopulations of PCSCs, and sphere-formation efficacy of PCa cells, even in the presence of TAM co-culture. Additionally, the Formula markedly decreased the migration, invasion, and epithelial-mesenchymal transition (EMT) of PCa cells induced by TAMs. The FZYL Formula also reversed M2 phenotype polarization in TAMs and dose-dependently reduced their CCL5 expression and secretion, with minimal cytotoxicity observed. Mechanistic studies confirmed that the TAMs/CCL5 axis is a critical target of the FZYL Formula, as the addition of exogenous CCL5 partially reversed the formula's inhibitory effects on PCSCs self-renewal in the co-culture system. Importantly, the Formula also significantly inhibited the growth of PCa xenografts, bone metastasis, and PCSCs activity in vivo by targeting the TAMs/CCL5 pathway. Overall, this study not only elucidates the immunomodulatory mechanism of the FZYL Formula in PCa therapy but also highlights the TAMs/CCL5 axis as a promising therapeutic target.
Collapse
Affiliation(s)
- Chiwei Chen
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China
| | - Renlun Huang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China
| | - Neng Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou 510000, China; The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Yifeng Zheng
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Jianfu Zhou
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China
| | - Bowen Yang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China
| | - Xuan Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China
| | - Juping Zhang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China
| | - Bo Pan
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China
| | - Zhiqiang Chen
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China
| | - Shengqi Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou 510000, China.
| | - Zhiyu Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou 510000, China; The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510000, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510000, China.
| | - Songtao Xiang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China.
| |
Collapse
|
22
|
Elkhamisy FAA, Aboelkomsan EA, Sallam MK, Eesa AN. Cytoplasmic PPARγ Significantly Correlates With P53 Immunohistochemical Expression and Tumor Size in Localized Tenosynovial Giant Cell Tumor. Cureus 2024; 16:e60377. [PMID: 38882990 PMCID: PMC11178509 DOI: 10.7759/cureus.60377] [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] [Accepted: 05/15/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Tenosynovial giant cell tumor (TGCT) is a monoarticular fibrohistiocytic benign or locally aggressive soft tissue tumor that originates from the synovium of joints, bursae, and tendon sheaths. It has an inflammatory neoplastic nature, with a clinical presentation ranging from pain, swelling, stiffness, and limited range of movement to joint instability and blockage. Its uncommon incidence leads to a poorly understood pathogenesis. Localized forms of TGCT (LTGCT) can cause significant morbidity, interfere with daily patient activities, and decrease the patient's quality of life in challenging cases. This study aimed to investigate the immunohistochemical expression of PPARγ (peroxisome proliferator-activated receptor gamma) and P53 in LTGCT to understand the disease better and offer potential therapeutic targets. METHODS The study is cross-sectional, in which 27 LTGCT cases were collected from the Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt. Solitary and multiple LTGCT cases retrieved between January 2018 and December 2022 were included, and immunohistochemically stained with anti-PPARγ and P53 antibodies. The TGCT samples were excluded if they were insufficient for sectioning, processing, and interpretation, over-fixed, had process artifacts, or were of the diffuse TGCT type. Scoring of stain expression was performed by ImageJ (National Institutes of Health, Bethesda, MD) analysis using the threshold method and was expressed in percent area/high power field. Clinicopathological correlations were analyzed. RESULTS All the 27 collected LTGCT cases were located in the small joints of patients' hands. Cases with solitary LGTCTs constituted 55.6% (n = 15), while 44.4% (n = 12) had multiple LTGCTs related to one affected site/case (e.g., multiple tumors in one finger). PPARγ was expressed in the cytoplasm of mononuclear and multinucleated tumor cells and foamy histiocytes, while P53 expression was mainly in mononuclear cells' nuclei. PPARγ significantly correlated with P53 expression (r = 0.9 and P = 0.000). PPARγ (r = 0.4 and P = 0.02) and P53 (r = 0.5 and P = 0.01) were positively correlated with tumor size. Only P53 expression was positively correlated with tumor multiplicity (r = 0.4 and P = 0.03). Using the receiver operating characteristic curve test, the P53 cutoff score detecting the multiplicity of TGCTs was ≥20.5%, with a 75% sensitivity and 80% specificity. CONCLUSION PPARγ and P53 have a significant role in LTGCT growth, while P53 plays a role in tumor multiplicity. They can be possible targets in LTGCTs unfit for excision.
Collapse
Affiliation(s)
| | | | - Marwa K Sallam
- Medical Microbiology and Immunology Department, Faculty of Medicine, Cairo University, Giza, EGY
| | - Ahmed N Eesa
- Pathology Department, Faculty of Medicine, Cairo University, Giza, EGY
| |
Collapse
|
23
|
Calmon MS, Lemos FFB, Silva Luz M, Rocha Pinheiro SL, de Oliveira Silva LG, Correa Santos GL, Rocha GR, Freire de Melo F. Immune pathway through endometriosis to ovarian cancer. World J Clin Oncol 2024; 15:496-522. [PMID: 38689629 PMCID: PMC11056862 DOI: 10.5306/wjco.v15.i4.496] [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: 12/15/2023] [Revised: 01/29/2024] [Accepted: 03/18/2024] [Indexed: 04/22/2024] Open
Abstract
Endometriosis is an estrogen-dependent inflammatory disease, defined by the presence of functional endometrial tissue outside of the uterine cavity. This disease is one of the main gynecological diseases, affecting around 10%-15% women and girls of reproductive age, being a common gynecologic disorder. Although endometriosis is a benign disease, it shares several characteristics with invasive cancer. Studies support that it has been linked with an increased chance of developing endometrial ovarian cancer, representing an earlier stage of neoplastic processes. This is particularly true for women with clear cell carcinoma, low-grade serous carcinoma and endometrioid. However, the carcinogenic pathways between both pathologies remain poorly understood. Current studies suggest a connection between endometriosis and endometriosis-associated ovarian cancers (EAOCs) via pathways associated with oxidative stress, inflammation, and hyperestrogenism. This article aims to review current data on the molecular events linked to the development of EAOCs from endometriosis, specifically focusing on the complex relationship between the immune response to endometriosis and cancer, including the molecular mechanisms and their ramifications. Examining recent developments in immunotherapy and their potential to boost the effectiveness of future treatments.
Collapse
Affiliation(s)
- Mariana Santos Calmon
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabian Fellipe Bueno Lemos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Marcel Silva Luz
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Samuel Luca Rocha Pinheiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | - Gabriel Lima Correa Santos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Gabriel Reis Rocha
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| |
Collapse
|
24
|
Wang S, Li J, Hong S, Wang N, Xu S, Yang B, Zheng Y, Zhang J, Pan B, Hu Y, Wang Z. Chemotherapy-elicited extracellular vesicle CXCL1 from dying cells promotes triple-negative breast cancer metastasis by activating TAM/PD-L1 signaling. J Exp Clin Cancer Res 2024; 43:121. [PMID: 38654356 PMCID: PMC11036662 DOI: 10.1186/s13046-024-03050-7] [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: 01/16/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, and chemotherapy still serves as the cornerstone treatment functioning by inducing cytotoxic cell death. Notably, emerging evidence suggests that dying cell-released signals may induce cancer progression and metastasis by modulating the surrounding microenvironment. However, the underlying molecular mechanisms and targeting strategies are yet to be explored. METHODS Apoptotic TNBC cells induced by paclitaxel or adriamycin treatment were sorted and their released extracellular vesicles (EV-dead) were isolated from the cell supernatants. Chemokine array analysis was conducted to identify the crucial molecules in EV-dead. Zebrafish and mouse xenograft models were used to investigate the effect of EV-dead on TNBC progression in vivo. RESULTS It was demonstrated that EV-dead were phagocytized by macrophages and induced TNBC metastasis by promoting the infiltration of immunosuppressive PD-L1+ TAMs. Chemokine array identified CXCL1 as a crucial component in EV-dead to activate TAM/PD-L1 signaling. CXCL1 knockdown in EV-dead or macrophage depletion significantly inhibited EV-dead-induced TNBC growth and metastasis. Mechanistic investigations revealed that CXCL1EV-dead enhanced TAM/PD-L1 signaling by transcriptionally activating EED-mediated PD-L1 promoter activity. More importantly, TPCA-1 (2-[(aminocarbonyl) amino]-5-(4-fluorophenyl)-3-thiophenecarboxamide) was screened as a promising inhibitor targeting CXCL1 signals in EVs to enhance paclitaxel chemosensitivity and limit TNBC metastasis without noticeable toxicities. CONCLUSIONS Our results highlight CXCL1EV-dead as a novel dying cell-released signal and provide TPCA-1 as a targeting candidate to improve TNBC prognosis.
Collapse
Affiliation(s)
- Shengqi Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jing Li
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shicui Hong
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Neng Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shang Xu
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bowen Yang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yifeng Zheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Juping Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bo Pan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yudie Hu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhiyu Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China.
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China.
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
25
|
Gao Y, Wu R, Pei Z, Ke C, Zeng D, Li X, Zhang Y. Cell cycle associated protein 1 associates with immune infiltration and ferroptosis in gastrointestinal cancer. Heliyon 2024; 10:e28794. [PMID: 38586390 PMCID: PMC10998105 DOI: 10.1016/j.heliyon.2024.e28794] [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: 07/19/2023] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024] Open
Abstract
Background Cell Cycle-Associated Protein 1 (CAPRIN1) play an important role in cell proliferation, oxidative stress, and inflammatory response. Nonetheless, its role in tumor immunity and ferroptosis is largely unknown in gastrointestinal cancer patients. Methods Through comprehensive bioinformatics, we investigate CAPRIN1 expression patterns and its role in diagnosis, functional signaling pathways, tumor immune infiltration and ferroptosis of different gastrointestinal cancer subtypes. Besides, immunohistochemistry (IHC) and immune blot were used to validate our esophagus cancer clinical data. The ferroptotic features of CAPRIN1 in vitro were assessed through knockdown assays in esophagus cancer cells. Results CAPRIN1 expression was significantly upregulated, correlated with poor prognosis, and served as an independent risk factor for most gastrointestinal cancer. Moreover, CAPRIN1 overexpression positively correlated with gene markers of most infiltrating immune cells, and immune checkpoints. CAPRIN1 knockdown significantly decreased the protein level of major histocompatibility complex class I molecules. We also identified a link between CAPRIN1 and ferroptosis-related genes in gastrointestinal cancer. Knockdown of CAPRIN1 significantly increased the production of lipid reactive oxygen species and malondialdehyde. Inhibition of CAPRIN1 expression promoted ferroptotic cell death induced by RAS-selective lethal 3 and erastin in human esophagus cancer cells. Conclusion Collectively, our results demonstrate that CAPRIN1 is aberrantly expressed in gastrointestinal cancer, is associated with poor prognosis, and could potentially influence immune infiltration and ferroptosis.
Collapse
Affiliation(s)
- Yan Gao
- Department of Pharmacy, Taihe Hospital, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P.R. China
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Ruimin Wu
- Department of Pharmacy, Taihe Hospital, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhijun Pei
- Department of Pharmacy, Taihe Hospital, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Changbin Ke
- Department of Pharmacy, Taihe Hospital, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Daobing Zeng
- Department of Pharmacy, Taihe Hospital, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiaohui Li
- Department of Pharmacy, Taihe Hospital, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P.R. China
- State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
26
|
Lu Q, Kou D, Lou S, Ashrafizadeh M, Aref AR, Canadas I, Tian Y, Niu X, Wang Y, Torabian P, Wang L, Sethi G, Tergaonkar V, Tay F, Yuan Z, Han P. Nanoparticles in tumor microenvironment remodeling and cancer immunotherapy. J Hematol Oncol 2024; 17:16. [PMID: 38566199 PMCID: PMC10986145 DOI: 10.1186/s13045-024-01535-8] [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: 12/30/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer immunotherapy and vaccine development have significantly improved the fight against cancers. Despite these advancements, challenges remain, particularly in the clinical delivery of immunomodulatory compounds. The tumor microenvironment (TME), comprising macrophages, fibroblasts, and immune cells, plays a crucial role in immune response modulation. Nanoparticles, engineered to reshape the TME, have shown promising results in enhancing immunotherapy by facilitating targeted delivery and immune modulation. These nanoparticles can suppress fibroblast activation, promote M1 macrophage polarization, aid dendritic cell maturation, and encourage T cell infiltration. Biomimetic nanoparticles further enhance immunotherapy by increasing the internalization of immunomodulatory agents in immune cells such as dendritic cells. Moreover, exosomes, whether naturally secreted by cells in the body or bioengineered, have been explored to regulate the TME and immune-related cells to affect cancer immunotherapy. Stimuli-responsive nanocarriers, activated by pH, redox, and light conditions, exhibit the potential to accelerate immunotherapy. The co-application of nanoparticles with immune checkpoint inhibitors is an emerging strategy to boost anti-tumor immunity. With their ability to induce long-term immunity, nanoarchitectures are promising structures in vaccine development. This review underscores the critical role of nanoparticles in overcoming current challenges and driving the advancement of cancer immunotherapy and TME modification.
Collapse
Affiliation(s)
- Qiang Lu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, China
| | - Dongquan Kou
- Department of Rehabilitation Medicine, Chongqing Public Health Medical Center, Chongqing, China
| | - Shenghan Lou
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Milad Ashrafizadeh
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China
| | - Amir Reza Aref
- Xsphera Biosciences, Translational Medicine Group, 6 Tide Street, Boston, MA, 02210, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Israel Canadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, USA
| | - Xiaojia Niu
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Yuzhuo Wang
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Pedram Torabian
- Cumming School of Medicine, Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Department of Medical Sciences, University of Calgary, Calgary, AB, T2N 4Z6, Canada
| | - Lingzhi Wang
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore
| | - Gautam Sethi
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore.
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore, Republic of Singapore
| | - Franklin Tay
- The Graduate School, Augusta University, 30912, Augusta, GA, USA
| | - Zhennan Yuan
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Peng Han
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
| |
Collapse
|
27
|
Huang Z, Xiao Z, Yu L, Liu J, Yang Y, Ouyang W. Tumor-associated macrophages in non-small-cell lung cancer: From treatment resistance mechanisms to therapeutic targets. Crit Rev Oncol Hematol 2024; 196:104284. [PMID: 38311012 DOI: 10.1016/j.critrevonc.2024.104284] [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: 10/28/2023] [Revised: 01/20/2024] [Accepted: 01/31/2024] [Indexed: 02/06/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) remains one of the leading causes of cancer-related deaths worldwide. Different treatment approaches are typically employed based on the stage of NSCLC. Common clinical treatment methods include surgical resection, drug therapy, and radiation therapy. However, with the introduction and utilization of immune checkpoint inhibitors, cancer treatment has entered a new era, completely revolutionizing the treatment landscape for various cancers and significantly improving overall patient survival. Concurrently, treatment resistance often poses a critical challenge, with many patients experiencing disease progression following an initial response due to treatment resistance. Increasing evidence suggests that the tumor microenvironment (TME) plays a pivotal role in treatment resistance. Tumor-associated macrophages (TAMs) within the TME can promote treatment resistance in NSCLC by secreting various cytokines activating signaling pathways, and interacting with other immune cells. Therefore, this article will focus on elucidating the key mechanisms of TAMs in treatment resistance and analyze how targeting TAMs can reduce the levels of treatment resistance in NSCLC, providing a comprehensive understanding of the principles and approaches to overcome treatment resistance in NSCLC.
Collapse
Affiliation(s)
- Zhenjun Huang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Ziqi Xiao
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Liqing Yu
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Jiayu Liu
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Yihan Yang
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang 330006, Jiangxi Province, China.
| | - Wenhao Ouyang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| |
Collapse
|
28
|
Yuan F, Hu Y, Xu F, Feng X. A review of obstructive sleep apnea and lung cancer: epidemiology, pathogenesis, and therapeutic options. Front Immunol 2024; 15:1374236. [PMID: 38605948 PMCID: PMC11007033 DOI: 10.3389/fimmu.2024.1374236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 03/12/2024] [Indexed: 04/13/2024] Open
Abstract
Despite undeniable advances in modern medicine, lung cancer still has high morbidity and mortality rates. Lung cancer is preventable and treatable, and it is important to identify new risk factors for lung cancer, especially those that can be treated or reversed. Obstructive sleep apnea (OSA) is a very common sleep-breathing disorder that is grossly underestimated in clinical practice. It can cause, exacerbate, and worsen adverse outcomes, including death and various diseases, but its relationship with lung cancer is unclear. A possible causal relationship between OSA and the onset and progression of lung cancer has been established biologically. The pathophysiological processes associated with OSA, such as sleep fragmentation, intermittent hypoxia, and increased sympathetic nervous excitation, may affect normal neuroendocrine regulation, impair immune function (especially innate and cellular immunity), and ultimately contribute to the occurrence of lung cancer, accelerate progression, and induce treatment resistance. OSA may be a contributor to but a preventable cause of the progression of lung cancer. However, whether this effect exists independently of other risk factors is unclear. Therefore, by reviewing the literature on the epidemiology, pathogenesis, and treatment of lung cancer and OSA, we hope to understand the relationships between the two and promote the interdisciplinary exchange of ideas between basic medicine, clinical medicine, respiratory medicine, sleep medicine, and oncology.
Collapse
Affiliation(s)
- Fang Yuan
- Department of Respiratory, The First Hospital of Jiujiang City, Jiujiang, China
| | - Yanxia Hu
- Department of Respiratory, The First Hospital of Jiujiang City, Jiujiang, China
| | - Fei Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xujun Feng
- Department of Respiratory, The First Hospital of Jiujiang City, Jiujiang, China
- Department of Respiratory and Critical Care Medicine, Sleep Medicine Center, Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
29
|
Erreni M, Fumagalli MR, Zanini D, Candiello E, Tiberi G, Parente R, D’Anna R, Magrini E, Marchesi F, Cappello P, Doni A. Multiplexed Imaging Mass Cytometry Analysis in Preclinical Models of Pancreatic Cancer. Int J Mol Sci 2024; 25:1389. [PMID: 38338669 PMCID: PMC10855072 DOI: 10.3390/ijms25031389] [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: 12/28/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 02/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers. PDAC is characterized by a complex tumor microenvironment (TME), that plays a pivotal role in disease progression and resistance to therapy. Investigating the spatial distribution and interaction of TME cells with the tumor is the basis for understanding the mechanisms underlying disease progression and represents a current challenge in PDAC research. Imaging mass cytometry (IMC) is the major multiplex imaging technology for the spatial analysis of tumor heterogeneity. However, there is a dearth of reports of multiplexed IMC panels for different preclinical mouse models, including pancreatic cancer. We addressed this gap by utilizing two preclinical models of PDAC: the genetically engineered, bearing KRAS-TP53 mutations in pancreatic cells, and the orthotopic, and developed a 28-marker panel for single-cell IMC analysis to assess the abundance, distribution and phenotypes of cells involved in PDAC progression and their reciprocal functional interactions. Herein, we provide an unprecedented definition of the distribution of TME cells in PDAC and compare the diversity between transplanted and genetic disease models. The results obtained represent an important and customizable tool for unraveling the complexities of PDAC and deciphering the mechanisms behind therapy resistance.
Collapse
Affiliation(s)
- Marco Erreni
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
| | - Maria Rita Fumagalli
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Damiano Zanini
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Ermes Candiello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44b, 10126 Torino, Italy
| | - Giorgia Tiberi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44b, 10126 Torino, Italy
| | - Raffaella Parente
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Raffaella D’Anna
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Elena Magrini
- IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Federica Marchesi
- IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133 Milan, Italy
| | - Paola Cappello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44b, 10126 Torino, Italy
| | - Andrea Doni
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| |
Collapse
|
30
|
Xiong J, Xiao R, Zhao J, Zhao Q, Luo M, Li F, Zhang W, Wu M. Matrix stiffness affects tumor-associated macrophage functional polarization and its potential in tumor therapy. J Transl Med 2024; 22:85. [PMID: 38246995 PMCID: PMC10800063 DOI: 10.1186/s12967-023-04810-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/17/2023] [Indexed: 01/23/2024] Open
Abstract
The extracellular matrix (ECM) plays critical roles in cytoskeletal support, biomechanical transduction and biochemical signal transformation. Tumor-associated macrophage (TAM) function is regulated by matrix stiffness in solid tumors and is often associated with poor prognosis. ECM stiffness-induced mechanical cues can activate cell membrane mechanoreceptors and corresponding mechanotransducers in the cytoplasm, modulating the phenotype of TAMs. Currently, tuning TAM polarization through matrix stiffness-induced mechanical stimulation has received increasing attention, whereas its effect on TAM fate has rarely been summarized. A better understanding of the relationship between matrix stiffness and macrophage function will contribute to the development of new strategies for cancer therapy. In this review, we first introduced the overall relationship between macrophage polarization and matrix stiffness, analyzed the changes in mechanoreceptors and mechanotransducers mediated by matrix stiffness on macrophage function and tumor progression, and finally summarized the effects of targeting ECM stiffness on tumor prognosis to provide insight into this new field.
Collapse
Affiliation(s)
- Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Rourou Xiao
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jiahui Zhao
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Qiuyan Zhao
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Manwen Luo
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Feng Li
- Department of Medical Genetics, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Allergy and Immunology, Wuhan, 430071, China.
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430032, China.
| |
Collapse
|
31
|
Puengel T, Tacke F. Role of Kupffer cells and other immune cells. SINUSOIDAL CELLS IN LIVER DISEASES 2024:483-511. [DOI: 10.1016/b978-0-323-95262-0.00024-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
32
|
Bui I, Bonavida B. Polarization of M2 Tumor-Associated Macrophages (TAMs) in Cancer Immunotherapy. Crit Rev Oncog 2024; 29:75-95. [PMID: 38989739 DOI: 10.1615/critrevoncog.2024053830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
We have witnessed in the last decade new milestones in the treatment of various resistant cancers with new immunotherapeutic modalities. These advances have resulted in significant objective durable clinical responses in a subset of cancer patients. These findings strongly suggested that immunotherapy should be considered for the treatment of all subsets of cancer patients. Accordingly, the mechanisms underlying resistance to immunotherapy must be explored and develop new means to target these resistant factors. One of the pivotal resistance mechanisms in the tumor microenvironment (TME) is the high infiltration of tumor-associated macrophages (TAMs) that are highly immunosuppressive and responsible, in large part, of cancer immune evasion. Thus, various approaches have been investigated to target the TAMs to restore the anti-tumor immune response. One approach is to polarize the M2 TAMS to the M1 phenotype that participates in the activation of the anti-tumor response. In this review, we discuss the various and differential properties of the M1 and M2 phenotypes, the molecular signaling pathways that participate in the polarization, and various approaches used to target the polarization of the M2 TAMs into the M1 anti-tumor phenotype. These approaches include inhibitors of histone deacetylases, PI3K inhibitors, STAT3 inhibitors, TLR agonists, and metabolic reprogramming. Clearly, due to the distinct features of various cancers and their heterogeneities, a single approach outlined above might only be effective against some cancers and not others. In addition, targeting by itself may not be efficacious unless used in combination with other therapeutic modalities.
Collapse
Affiliation(s)
- Indy Bui
- University of California Los Angeles
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine at UCLA, Johnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90025-1747, USA
| |
Collapse
|
33
|
Pu Q, Gao H. The Role of the Tumor Microenvironment in Triple-Positive Breast Cancer Progression and Therapeutic Resistance. Cancers (Basel) 2023; 15:5493. [PMID: 38001753 PMCID: PMC10670777 DOI: 10.3390/cancers15225493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/26/2023] [Accepted: 11/18/2023] [Indexed: 11/26/2023] Open
Abstract
Breast cancer (BRCA) is a highly heterogeneous systemic disease. It is ranked first globally in the incidence of new cancer cases and has emerged as the primary cause of cancer-related death among females. Among the distinct subtypes of BRCA, triple-positive breast cancer (TPBC) has been associated with increased metastasis and invasiveness, exhibiting greater resistance to endocrine therapy involving trastuzumab. It is now understood that invasion, metastasis, and treatment resistance associated with BRCA progression are not exclusively due to breast tumor cells but are from the intricate interplay between BRCA and its tumor microenvironment (TME). Accordingly, understanding the pathogenesis and evolution of the TPBC microenvironment demands a comprehensive approach. Moreover, addressing BRCA treatment necessitates a holistic consideration of the TME, bearing significant implications for identifying novel targets for anticancer interventions. This review expounds on the relationship between critical cellular components and factors in the TPBC microenvironment and the inception, advancement, and therapeutic resistance of breast cancer to provide perspectives on the latest research on TPBC.
Collapse
Affiliation(s)
- Qian Pu
- Department of Breast Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China;
- Oncology Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
| | - Haidong Gao
- Department of Breast Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China;
- Oncology Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
| |
Collapse
|
34
|
Chen C, Wang S, Wang N, Zheng Y, Zhou J, Hong M, Chen Z, Wang S, Wang Z, Xiang S. Icariin inhibits prostate cancer bone metastasis and destruction via suppressing TAM/CCL5-mediated osteoclastogenesis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155076. [PMID: 37716031 DOI: 10.1016/j.phymed.2023.155076] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 09/04/2023] [Accepted: 09/09/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND Bone metastasis occurs in nearly 70% of patients with metastatic prostate cancer (PCa), and represents the leading cause of death in patients with PCa. Emerging evidence has demonstrated the potential activities of icariin in modulating bone metabolism and remodelling the tumor microenvironment (TME). However, whether icariin could inhibit PCa bone metastasis and destruction by modulating the TME as well as the underlying mechanisms remains unclear. PURPOSE This study investigated whether icariin could inhibit PCa bone metastasis and destruction by modulating the bone TME as well as the underlying mechanisms. METHODS Osteoclasts were induced from mouse bone marrow-derived macrophages (BMMs) or Raw264.7 cells. PCa cells were cultured in the conditional medium (CM) of macrophages in vitro or co-injected with macrophages in vivo to simulate their coexistence in the TME. Multiple molecular biology experiments and the mouse RM1-Luc PCa bone metastasis model were used to explore the inhibitory activity and mechanism of icariin on PCa metastasis and bone destruction. RESULTS Icariin treatment significantly suppressed PCa growth, bone metastasis and destruction as well as osteoclastogenesis in vivo. Furthermore, icariin remarkably inhibited osteoclast differentiation, even in the presence of the CM of tumor-associated macrophages (TAMs), while exhibiting no obvious effect on osteoblasts. Moreover, icariin suppressed the M2 phenotype polarization of Raw264.7-derived TAMs and transcriptionally attenuated their CC motif chemokine ligand 5 (CCL5) expression and secretion via inhibiting SPI1. Additionally, CCL5 induced the differentiation and chemotaxis of osteoclast precursor cells by binding with its receptor CCR5. The clinicopathological analysis further verified the positive correlation between the TAM/CCL5/CCR5 axis and osteoclastogenesis within the TME of PCa patients. More importantly, icariin remarkably suppressed PCa metastasis-induced bone destruction in vivo by inhibiting osteoclastogenesis via downregulating the TAM/CCL5 pathway. CONCLUSION Altogether, these results not only implicate icariin as a promising candidate immunomodulator for PCa bone metastasis and destruction but also shed novel insight into targeting TAM/CCL5-mediated osteoclastogenesis as a potential treatment strategy for osteolytic bone metastasis. This study helps to advance the understanding of the crosstalk between bone TME and bone homeostasis.
Collapse
Affiliation(s)
- Chiwei Chen
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shengqi Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Neng Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yifeng Zheng
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianfu Zhou
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Min Hong
- Department of Pathology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhiqiang Chen
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shusheng Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhiyu Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Songtao Xiang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
35
|
Yang D, Kuang T, Zhou Y, Su Y, Shen J, Yu B, Zhao K, Ding Y. Tumor-associated endothelial cell prognostic risk model and tumor immune environment modulation in liver cancer based on single-cell and bulk RNA sequencing: Experimental verification. Int Immunopharmacol 2023; 124:110870. [PMID: 37690233 DOI: 10.1016/j.intimp.2023.110870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 08/08/2023] [Accepted: 08/27/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND To build a prognostic and immunotherapeutic response prediction model for liver cancer based on marker genes of tumor-associated endothelial cell (TEC). METHOD Single cell sequencing data from Gene Expression Omnibus (GEO) liver cancer patients were utilized to identify TEC subpopulations. Models were built from transcriptomic and clinical data of TCGA liver cancer patients. The GSE76427 and ICGC databases were used as independent validation sets. Time-dependent receiver operating characteristic (ROC) curves and Kaplan-Meier curves were used to verify the ability of the model to predict survival. XCELL, TIMER, QUANTISEQ, CIBERSORT, CIBERSORT-ABS, and ssGSEA were applied to evaluate tumor immune cell infiltration. The TIDE score was used to predict the effect of immunotherapy. Immune blockade checkpoint gene, tumor mutational load and GSVA enrichment analyses were further explored. The expression levels of candidate genes were measured and validated by real-time PCR between liver cancer tissues and adjacent nontumor liver tissues. RESULTS Eighty-seven genes were identified as marker genes for TECs. IGFBP3, RHOC, S100A16, FSCN1, and CLEC3B were included in the constructed prognostic model. Time-dependent ROC curve values were higher than 0.700 in both the model and validation groups. The low risk group exhibited high immune cell infiltration and function than the higher risk group. The TIDE score indicated that the low-risk group benefited more from immunotherapy than the high-risk group. The risk score and multiple immune blockade checkpoint genes and immune-related pathways were strongly correlated. CONCLUSION Novel signatures of TEC marker genes showed a powerful ability to predict prognosis and immunotherapy response in patients with liver cancer.
Collapse
Affiliation(s)
- Dashuai Yang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, No. 99 Zhangzhidong Road, Wuchang District, Wuhan 430060, China.
| | - Tianrui Kuang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, No. 99 Zhangzhidong Road, Wuchang District, Wuhan 430060, China.
| | - Yu Zhou
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, No. 99 Zhangzhidong Road, Wuchang District, Wuhan 430060, China.
| | - Yang Su
- Department of Gastrointestinal Surgery, Tongji Hospital, Tongji Medical College in Huazhong University of Science and Technology, Wuhan 430060, Hubei, China.
| | - Jie Shen
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, No. 99 Zhangzhidong Road, Wuchang District, Wuhan 430060, China.
| | - Bin Yu
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, No. 99 Zhangzhidong Road, Wuchang District, Wuhan 430060, China.
| | - Kailiang Zhao
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, No. 99 Zhangzhidong Road, Wuchang District, Wuhan 430060, China.
| | - Youming Ding
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, No. 99 Zhangzhidong Road, Wuchang District, Wuhan 430060, China.
| |
Collapse
|
36
|
Shang L, Zhong Y, Yao Y, Liu C, Wang L, Zhang W, Liu J, Wang X, Sun C. Subverted macrophages in the triple-negative breast cancer ecosystem. Biomed Pharmacother 2023; 166:115414. [PMID: 37660651 DOI: 10.1016/j.biopha.2023.115414] [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: 06/16/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023] Open
Abstract
Tumor-associated macrophages (TAMs) are the most critical effector cells of innate immunity and the most abundant tumor-infiltrating immune cells. They play a key role in the clearance of apoptotic bodies, regulation of inflammation, and tissue repair to maintain homeostasis in vivo. With the progression of triple-negative breast cancer(TNBC), TAMs are "subverted" from tumor-promoting immune cells to tumor-promoting immune suppressor cells, which play a significant role in tumor development and are considered potential targets for cancer therapy. Here, we explored how macrophages, as the most important part of the TNBC ecosystem, are "subverted" to drive cancer evolution and the uniqueness of TAMs in TNBC progression and metastasis. Similarly, we discuss the rationale and available evidence for TAMs as potential targets for TNBC therapy.
Collapse
Affiliation(s)
- Linxiao Shang
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai 264000, China
| | - Yuting Zhong
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250022, China
| | - Yan Yao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250022, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China
| | - Lu Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250022, China
| | - Wenfeng Zhang
- School of Traditional Chinese Medicine, Macau University of Science and Technology, Macao Special Administrative Region, Macau 999078, China
| | - Jingyang Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250022, China
| | - Xue Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250022, China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, China.
| |
Collapse
|
37
|
Sun Y, Jiang G, Wu Q, Ye L, Li B. The role of tumor-associated macrophages in the progression, prognosis and treatment of endometrial cancer. Front Oncol 2023; 13:1213347. [PMID: 37810971 PMCID: PMC10556650 DOI: 10.3389/fonc.2023.1213347] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/18/2023] [Indexed: 10/10/2023] Open
Abstract
Tumor-associated macrophages (TAMs) are the main immune cells in the tumor microenvironment (TME) of endometrial cancer (EC). TAMs recruitment and polarization in EC is regulated by the TME of EC, culminating in a predominantly M2-like macrophage infiltration. TAMs promote lymphatic angiogenesis through cytokine secretion, aid immune escape of EC cells by synergizing with other immune cells, and contribute to the development of EC through secretion of exosomes so as to promoting EC development. EC is a hormone- and metabolism-dependent cancer, and TAMs promote EC through interactions on estrogen receptor (ER) and metabolic factors such as the metabolism of glucose, lipids, and amino acids. In addition, we have explored the predictive significance of some TAM-related indicators for EC prognosis, and TAMs show remarkable promise as a target for EC immunotherapy.
Collapse
Affiliation(s)
- Yihan Sun
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Genyi Jiang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qianhua Wu
- School of Medicine, Tongji University, Shanghai, China
| | - Lei Ye
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bilan Li
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
38
|
Zhang J, Wang S, Zhang D, He X, Wang X, Han H, Qin Y. Nanoparticle-based drug delivery systems to enhance cancer immunotherapy in solid tumors. Front Immunol 2023; 14:1230893. [PMID: 37600822 PMCID: PMC10435760 DOI: 10.3389/fimmu.2023.1230893] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
Immunotherapy has developed rapidly in solid tumors, especially in the areas of blocking inhibitory immune checkpoints and adoptive T-cell transfer for immune regulation. Many patients benefit from immunotherapy. However, the response rate of immunotherapy in the overall population are relatively low, which depends on the characteristics of the tumor and individualized patient differences. Moreover, the occurrence of drug resistance and adverse reactions largely limit the development of immunotherapy. Recently, the emergence of nanodrug delivery systems (NDDS) seems to improve the efficacy of immunotherapy by encapsulating drug carriers in nanoparticles to precisely reach the tumor site with high stability and biocompatibility, prolonging the drug cycle of action and greatly reducing the occurrence of toxic side effects. In this paper, we mainly review the advantages of NDDS and the mechanisms that enhance conventional immunotherapy in solid tumors, and summarize the recent advances in NDDS-based therapeutic strategies, which will provide valuable ideas for the development of novel tumor immunotherapy regimen.
Collapse
Affiliation(s)
- Jiaxin Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Siyuan Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Daidi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin He
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xue Wang
- Academy of Medical Science, School of Basic Medical Science, Zhengzhou University, Zhengzhou, China
| | - Huiqiong Han
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
39
|
Zhang L, Cui S, Ding N, Zhang J, Cui E, Xiang Q, Zhou Z, Sun B, Wang Y, Hong H, Ma Y, Yang D. Tumor-Associated Macrophages Regulating a Polymer Nanoplatform for Synergistic Treatment of Breast Tumors. ACS APPLIED MATERIALS & INTERFACES 2023; 15:34527-34539. [PMID: 37462215 DOI: 10.1021/acsami.3c05497] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Tumor-associated macrophages (TAMs) play a critical role in tumor progression and metastasis. Modulation of TAM polarization is one of the most effective strategies to change the immunosuppressive tumor microenvironment (TME). In this study, organic polymer nanoparticles (CPHT) were prepared using hyaluronic acid (HA)-conjugated disulfide-bonded polyethylene imide (PEIS) as a carrier through a self-assembly strategy. These nanoparticles were modified by transferrin (Tf) and loaded with chlorin e6 (Ce6). The results showed that CPHT had good dispersion with a particle size of about 30 nm. CPHT gradually disintegrated under the exposure with a high concentration of glutathione (GSH) in tumor cells, proving the possibility for the controlled release of Ce6 and photodynamic therapy. An in vitro test showed that the uptake of CPHT in tumor cells was mediated by both HA and Tf, indicating the active tumor-targeting capacity of CPHT. CPHT significantly downregulated the ratio of CD206/CD86 and triggered the upregulation of immune factors such as TNF-α and iNOS, suggesting the repolarization of TAMs. We also found that CPHT effectively induced ferroptosis in tumor cells through lipid peroxide accumulation, GSH depletion, and downregulation of lipid peroxidase (GPX4) expression. Animal experiments confirmed that CPHT not only effectively inhibited the growth of tumors in situ but also significantly decelerated the growth of the distal tumor. Elevated levels of CD86 and IFN-γ and decreased expression of CD206 were observed at the tumor sites post CPHT treatment. These results confirmed the value of CPHT as a multifunctional nanoplatform that can tune the TME and provide new hope for tumor treatment.
Collapse
Affiliation(s)
- Li Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Shuai Cui
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Ning Ding
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Jing Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Enna Cui
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Qian Xiang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Zhenghao Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Bo Sun
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Yinan Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Hao Hong
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu 210093, PR China
| | - Yunsu Ma
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
- Jiangsu Yuanlong Hospital Management Co. LTD, Xuzhou, Jiangsu 221000, PR China
| | - Dongzhi Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| |
Collapse
|
40
|
Zhao Z, Sun C, Hou J, Yu P, Wei Y, Bai R, Yang P. Identification of STEAP3-based molecular subtype and risk model in ovarian cancer. J Ovarian Res 2023; 16:126. [PMID: 37386521 DOI: 10.1186/s13048-023-01218-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/20/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND Ovarian cancer (OC) is one of the most common malignancies in women. It has a poor prognosis owing to its recurrence and metastasis. Unfortunately, reliable markers for early diagnosis and prognosis of OC are lacking. Our research aimed to investigate the value of the six-transmembrane epithelial antigen of prostate family member 3 (STEAP3) as a prognostic predictor and therapeutic target in OC using bioinformatics analysis. METHODS STEAP3 expression and clinical data were acquired from The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), and Gene Expression Omnibus (GEO). Unsupervised clustering was used to identify molecular subtypes. Prognosis, tumor immune microenvironment (TIME), stemness indexes, and functional enrichment analysis were compared between two definite clusters. Through the least absolute shrinkage and selection operator (LASSO) regression analysis, a STEAP3-based risk model was developed, and the predictive effectiveness of this signature was confirmed using GEO datasets. A nomogram was used to predict the survival possibility of patients. Additionally, TIME, tumor immune dysfunction and exclusion (TIDE), stemness indexes, somatic mutations, and drug sensitivity were evaluated in different risk groups with OC. STEAP3 protein expression was detected using immunohistochemistry (IHC). RESULTS STEAP3 displayed marked overexpression in OC. STEAP3 is an independent risk factor for OC. Based on the mRNA levels of STEAP3-related genes (SRGs), two distinct clusters were identified. Patients in the cluster 2 (C2) subgroup had a considerably worse prognosis, higher immune cell infiltration, and lower stemness scores. Pathways involved in tumorigenesis and immunity were highly enriched in the C2 subgroup. A prognostic model based on 13 SRGs was further developed. Kaplan-Meier analysis indicated that the overall survival (OS) of high-risk patients was poor. The risk score was significantly associated with TIME, TIDE, stemness indexes, tumor mutation burden (TMB), immunotherapy response, and drug sensitivity. Finally, IHC revealed that STEAP3 protein expression was noticeably elevated in OC, and overexpression of STEAP3 predicted poor OS and relapse-free survival (RFS) of patients. CONCLUSION In summary, this study revealed that STEAP3 reliably predicts patient prognosis and provides novel ideas for OC immunotherapy.
Collapse
Affiliation(s)
- Zouyu Zhao
- First Affiliated Hospital, Shihezi University, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Chongfeng Sun
- First Affiliated Hospital, Shihezi University, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Jishuai Hou
- First Affiliated Hospital, Shihezi University, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Panpan Yu
- First Affiliated Hospital, Shihezi University, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Yan Wei
- First Affiliated Hospital, Shihezi University, Shihezi, China
| | - Rui Bai
- First Affiliated Hospital, Shihezi University, Shihezi, China
| | - Ping Yang
- First Affiliated Hospital, Shihezi University, Shihezi, China.
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China.
| |
Collapse
|
41
|
Sun J, Yu L, Qu X, Huang T. The role of peroxisome proliferator-activated receptors in the tumor microenvironment, tumor cell metabolism, and anticancer therapy. Front Pharmacol 2023; 14:1184794. [PMID: 37251321 PMCID: PMC10213337 DOI: 10.3389/fphar.2023.1184794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/05/2023] [Indexed: 05/31/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) have been extensively studied for over 3 decades and consist of three isotypes, including PPARα, γ, and β/δ, that were originally considered key metabolic regulators controlling energy homeostasis in the body. Cancer has become a leading cause of human mortality worldwide, and the role of peroxisome proliferator-activated receptors in cancer is increasingly being investigated, especially the deep molecular mechanisms and effective cancer therapies. Peroxisome proliferator-activated receptors are an important class of lipid sensors and are involved in the regulation of multiple metabolic pathways and cell fate. They can regulate cancer progression in different tissues by activating endogenous or synthetic compounds. This review emphasizes the significance and knowledge of peroxisome proliferator-activated receptors in the tumor microenvironment, tumor cell metabolism, and anti-cancer treatment by summarizing recent research on peroxisome proliferator-activated receptors. In general, peroxisome proliferator-activated receptors either promote or suppress cancer in different types of tumor microenvironments. The emergence of this difference depends on various factors, including peroxisome proliferator-activated receptor type, cancer type, and tumor stage. Simultaneously, the effect of anti-cancer therapy based on drug-targeted PPARs differs or even opposes among the three peroxisome proliferator-activated receptor homotypes and different cancer types. Therefore, the current status and challenges of the use of peroxisome proliferator-activated receptors agonists and antagonists in cancer treatment are further explored in this review.
Collapse
Affiliation(s)
- Jiaao Sun
- Department of Urology, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Liyan Yu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Xueling Qu
- Dalian Women and Children’s Medical Center(Group), Dalian, Liaoning, China
| | - Tao Huang
- Department of Urology, First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
42
|
Khan SU, Khan MU, Azhar Ud Din M, Khan IM, Khan MI, Bungau S, Hassan SSU. Reprogramming tumor-associated macrophages as a unique approach to target tumor immunotherapy. Front Immunol 2023; 14:1166487. [PMID: 37138860 PMCID: PMC10149956 DOI: 10.3389/fimmu.2023.1166487] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/04/2023] [Indexed: 05/05/2023] Open
Abstract
In the last ten years, it has become increasingly clear that tumor-infiltrating myeloid cells drive not just carcinogenesis via cancer-related inflammatory processes, but also tumor development, invasion, and metastasis. Tumor-associated macrophages (TAMs) in particular are the most common kind of leucocyte in many malignancies and play a crucial role in establishing a favorable microenvironment for tumor cells. Tumor-associated macrophage (TAM) is vital as the primary immune cell subset in the tumor microenvironment (TME).In order to proliferate and spread to new locations, tumors need to be able to hide from the immune system by creating an immune-suppressive environment. Because of the existence of pro-tumoral TAMs, conventional therapies like chemotherapy and radiotherapy often fail to restrain cancer growth. These cells are also to blame for the failure of innovative immunotherapies premised on immune-checkpoint suppression. Understanding the series of metabolic changes and functional plasticity experienced by TAMs in the complex TME will help to use TAMs as a target for tumor immunotherapy and develop more effective tumor treatment strategies. This review summarizes the latest research on the TAMs functional status, metabolic changes and focuses on the targeted therapy in solid tumors.
Collapse
Affiliation(s)
- Safir Ullah Khan
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Munir Ullah Khan
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Muhammad Azhar Ud Din
- Faculty of Pharmacy, Gomal University Dera Ismail Khan KPK, Dera Ismail Khan, Pakistan
| | - Ibrar Muhammad Khan
- Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering Fuyang Normal University, Fuyang, China
| | - Muhammad Imran Khan
- School of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Syed Shams ul Hassan
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
- Department of Natural Product Chemistry, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
43
|
Sadhukhan P, Seiwert TY. The role of macrophages in the tumor microenvironment and tumor metabolism. Semin Immunopathol 2023; 45:187-201. [PMID: 37002376 DOI: 10.1007/s00281-023-00988-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/08/2023] [Indexed: 04/03/2023]
Abstract
The complexity and plasticity of the tumor microenvironment (TME) make it difficult to fully understand the intratumoral regulation of different cell types and their activities. Macrophages play a crucial role in the signaling dynamics of the TME. Among the different subtypes of macrophages, tumor-associated macrophages (TAMs) are often associated with poor prognosis, although some subtypes of TAMs can at the same time improve treatment responsiveness and lead to favorable clinical outcomes. TAMs are key regulators of cancer cell proliferation, metastasis, angiogenesis, extracellular matrix remodeling, tumor metabolism, and importantly immunosuppression in the TME by modulating various chemokines, cytokines, and growth factors. TAMs have been identified as a key contributor to resistance to chemotherapy and cancer immunotherapy. In this review article, we aim to discuss the mechanisms by which TAMs regulate innate and adaptive immune signaling in the TME and summarize recent preclinical research on the development of therapeutics targeting TAMs and tumor metabolism.
Collapse
Affiliation(s)
- Pritam Sadhukhan
- Johns Hopkins University, Skip Viragh Outpatient Cancer Building, Baltimore, MD, 21287, USA
| | - Tanguy Y Seiwert
- Johns Hopkins University, Skip Viragh Outpatient Cancer Building, Baltimore, MD, 21287, USA.
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| |
Collapse
|
44
|
Zhan C, Jin Y, Xu X, Shao J, Jin C. Antitumor therapy for breast cancer: Focus on tumor-associated macrophages and nanosized drug delivery systems. Cancer Med 2023. [PMID: 36794651 DOI: 10.1002/cam4.5489] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/15/2022] [Accepted: 11/17/2022] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND In breast cancer (BC), tumor-associated macrophages (TAMs) are an important component of the tumor microenvironment and are closely related to poor prognosis. A growing number of studies have focused on the role of TAMs in BC progression and therapeutic strategies targeting TAMs. As an emerging treatment, the application of nanosized drug delivery systems (NDDSs) in the treatment of BC by targeting TAMs has attracted much attention. AIMS This review is to summarize the characteristics and treatment strategies targeting TAMs in BC and to clarify the applications of NDDSs targeting TAMs in the treatment of BC by targeting TAMs. MATERIALS & METHODS The existing results related to characteristics of TAMs in BC, BC treatment strategies by targeting TAMs, and the applications of NDDSs in these strategies are described. Through analyzing these results, the advantages and disadvantages of the treatment strategies using NDDSs are discussed, which could provide advices on designing NDDSs for BC treatment. RESULTS TAMs are one of the most prominent noncancer cell types in BC. TAMs not only promote angiogenesis, tumor growth and metastasis but also lead to therapeutic resistance and immunosuppression. Mainly four strategies have been used to target TAMs for BC therapy, which include depleting macrophages, blocking recruitment, reprogramming to attain an anti-tumor phenotype, and increasing phagocytosis. Since NDDSs can efficiently deliver drugs to TAMs with low toxicity, they are promising approaches for targeting TAMs in tumor therapy. NDDSs with various structures can deliver immunotherapeutic agents and nucleic acid therapeutics to TAMs. In addition, NDDSs can realize combination therapies. DISCUSSION TAMs play a critical role in the progression of BC. An increasing number of strategies have been proposed to regulate TAMs. Compared with free drugs, NDDSs targeting TAMs improve drug concentration, reduce toxicity and realize combination therapies. However, in order to achieve better therapeutic efficacy, there are still some disadvantages that need to be considered in the design of NDDSs. CONCLUSION TAMs play an important role in the progression of BC, and targeting TAMs is a promising strategy for BC therapy. In particular, NDDSs targeting TAMs have unique advantages and are potential treatments for BC.
Collapse
Affiliation(s)
- Cuiping Zhan
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ying Jin
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, China
| | - Xinzhi Xu
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, China.,Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing, China
| | - Jiangbo Shao
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chunxiang Jin
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
45
|
Zhu YH, Zheng JH, Jia QY, Duan ZH, Yao HF, Yang J, Sun YW, Jiang SH, Liu DJ, Huo YM. Immunosuppression, immune escape, and immunotherapy in pancreatic cancer: focused on the tumor microenvironment. Cell Oncol (Dordr) 2023; 46:17-48. [PMID: 36367669 DOI: 10.1007/s13402-022-00741-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic cancer, is characterized by poor treatment response and low survival time. The current clinical treatment for advanced PDAC is still not effective. In recent years, the research and application of immunotherapy have developed rapidly and achieved substantial results in many malignant tumors. However, the translational application in PDAC is still far from satisfactory and needs to be developed urgently. To carry out the study of immunotherapy, it is necessary to fully decipher the immune characteristics of PDAC. This review summarizes the recent progress of the tumor microenvironment (TME) of PDAC and highlights its link with immunotherapy. We describe the molecular cues and corresponding intervention methods, collate several promising targets and progress worthy of further study, and put forward the importance of integrated immunotherapy to provide ideas for future research of TME and immunotherapy of PDAC.
Collapse
Affiliation(s)
- Yu-Heng Zhu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Jia-Hao Zheng
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Qin-Yuan Jia
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Zong-Hao Duan
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Hong-Fei Yao
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Jian Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Yong-Wei Sun
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China.
| | - Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 800 Dongchuan Road, 200240, People's Republic of China.
| | - De-Jun Liu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China.
| | - Yan-Miao Huo
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China.
| |
Collapse
|
46
|
Zwager MC, Bense R, Waaijer S, Qiu SQ, Timmer-Bosscha H, de Vries EGE, Schröder CP, van der Vegt B. Assessing the role of tumour-associated macrophage subsets in breast cancer subtypes using digital image analysis. Breast Cancer Res Treat 2023; 198:11-22. [PMID: 36622544 PMCID: PMC9883348 DOI: 10.1007/s10549-022-06859-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/29/2022] [Indexed: 01/10/2023]
Abstract
PURPOSE The number of M1-like and M2-like tumour-associated macrophages (TAMs) and their ratio can play a role in breast cancer development and progression. Early clinical trials using macrophage targeting compounds are currently ongoing. However, the most optimal detection method of M1-like and M2-like macrophage subsets and their clinical relevance in breast cancer is still unclear. We aimed to optimize the assessment of TAM subsets in different breast cancer subtypes, and therefore related TAM subset numbers and ratio to clinicopathological characteristics and clinical outcome. METHODS Tissue microarrays of 347 consecutive primary Luminal-A, Luminal-B, HER2-positive and triple-negative tumours of patients with early-stage breast cancer were serially sectioned and immunohistochemically stained for the pan-macrophage marker CD68 and the M2-like macrophage markers CD163, CSF-1R and CD206. TAM numbers were quantified using a digital image analysis algorithm. M1-like macrophage numbers were calculated by subtracting M2-like TAM numbers from the total TAM number. RESULTS M2-like markers CD163 and CSF-1R showed a moderate positive association with each other and with CD68 (r ≥ 0.47), but only weakly with CD206 (r ≤ 0.06). CD68 + , CD163 + and CSF-1R + macrophages correlated with tumour grade in Luminal-B tumours (P < 0.001). Total or subset TAM numbers did not correlate with disease outcome in any breast cancer subtype. CONCLUSION In conclusion, macrophages and their subsets can be detected by means of a panel of TAM markers and are related to unfavourable clinicopathological characteristics in Luminal-B breast cancer. However, their impact on outcome remains unclear. Preferably, this should be determined in prospective series.
Collapse
Affiliation(s)
- Mieke C. Zwager
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rico Bense
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Stijn Waaijer
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Si-Qi Qiu
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Diagnosis and Treatment Center of Breast Diseases, Clinical Research Center, Shantou Central Hospital, Shantou, China
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Shantou University Medical College, Shantou, China
| | - Hetty Timmer-Bosscha
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Elisabeth G. E. de Vries
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Carolien P. Schröder
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Medical Oncology, Dutch Cancer Institute, Amsterdam, Netherlands
| | - Bert van der Vegt
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB Groningen, The Netherlands
| |
Collapse
|
47
|
Rao DY, Huang DF, Si MY, Lu H, Tang ZX, Zhang ZX. Role of exosomes in non-small cell lung cancer and EGFR-mutated lung cancer. Front Immunol 2023; 14:1142539. [PMID: 37122754 PMCID: PMC10130367 DOI: 10.3389/fimmu.2023.1142539] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/22/2023] [Indexed: 05/02/2023] Open
Abstract
As an important mediator of information transfer between cells, exosomes play a unique role in regulating tumor growth, supporting vascular proliferation, tumor invasion, and metastasis. Exosomes are widely present in various body fluids, and therefore they can be used as a potential tool for non-invasive liquid biopsy. The present study reviews the role of exosomes in liquid biopsy, tumor microenvironment formation, and epithelial-mesenchymal transition in non-small cell lung cancer (NSCLC). By targeting epidermal growth factor receptor (EGFR) therapy as a first-line treatment for patients with NSCLC, this study also briefly describes the occurrence of EGRF+ exosomes and the role of exosomes and their contents in non-invasive detection and potential therapeutic targets in EGFR-mutated lung cancer.
Collapse
Affiliation(s)
- Ding-Yu Rao
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - De-Fa Huang
- Laboratory Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Mao-Yan Si
- The First Clinical College, Gannan Medical University, Ganzhou, China
| | - Hua Lu
- The First Clinical College, Southern Medical University, Guangzhou, China
| | - Zhi-Xian Tang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- *Correspondence: Zhi-Xian Tang, ; Zu-Xiong Zhang,
| | - Zu-Xiong Zhang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
48
|
Tu J, Wang D, Zheng X, Liu B. Single-cell RNA datasets and bulk RNA datasets analysis demonstrated C1Q+ tumor-associated macrophage as a major and antitumor immune cell population in osteosarcoma. Front Immunol 2023; 14:911368. [PMID: 36814925 PMCID: PMC9939514 DOI: 10.3389/fimmu.2023.911368] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 01/23/2023] [Indexed: 02/09/2023] Open
Abstract
Background Osteosarcoma is the most frequent primary bone tumor with a poor prognosis. Immune infiltration proved to have a strong impact on prognosis. We analyzed single-cell datasets and bulk datasets to confirm the main immune cell populations and their properties in osteosarcoma. Methods The examples in bulk datasets GSE21257 and GSE32981 from the Gene Expression Omnibus database were divided into two immune infiltration level groups, and 34 differentially expressed genes were spotted. Then, we located these genes among nine major cell clusters and their subclusters identified from 99,668 individual cells in single-cell dataset GSE152048 including 11 osteosarcoma patients. Especially, the markers of all kinds of myeloid cells identified in single-cell dataset GSE152048 were set to gene ontology enrichment. We clustered the osteosarcoma samples in the TARGET-OS from the Therapeutically Applicable Research to Generate Effective Treatments dataset into two groups by complete component 1q positive macrophage markers and compared their survival. Results Compared with the low-immune infiltrated group, the high-immune infiltrated group showed a better prognosis. Almost all the 34 differentially expressed genes expressed higher or exclusively among myeloid cells. A group of complete component 1q-positive macrophages was identified from the myeloid cells. In the bulk dataset TARGET-OS, these markers and the infiltration of complete component 1q-positive macrophages related to longer survival. Conclusions Complete component 1q-positive tumor-associated macrophages were the major immune cell population in osteosarcoma, which contributed to a better prognosis.
Collapse
Affiliation(s)
- Jihao Tu
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Duo Wang
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - XiaoTian Zheng
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Bin Liu
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
49
|
Li Y, Li Q, Liu J, Huang Y, Mao J, Zhang G. HSF1 expression in tumor-associated macrophages promotes tumor cell proliferation and indicates poor prognosis in esophageal squamous cell carcinoma. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2022; 25:1682-1689. [PMID: 36586067 DOI: 10.1007/s12094-022-03063-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/24/2022] [Indexed: 01/01/2023]
Abstract
PURPOSE Tumor-associated macrophages (TAMs), are crucial for the survival and development of tumor cells. Heat shock factor 1 (HSF1) is a potent, complex carcinogenesis modulator, and esophageal cancer (EC) patients have a bad prognosis when HSF1 is highly expressed. HSF1's clinical importance and biological role in TAMs are still unknown. METHODS The HSF1 expression profile and patient survival information were analyzed from the TCGA database. The infiltration of different types of immune cells in EC was evaluated based on HSF1 gene expression by Sangerbox 3.0. Immunochemistry was employed to assess HSF1 protein expression in 134 individuals with esophageal squamous cell carcinoma (ESCC), proceeded by association with clinicopathological variables. The role of macrophage-driven HSF1 were observed using HSF1-knockdown THP1 cells. RESULTS High level of HSF1 have a poorer prognosis in individuals with EC. The expressing level of HSF1 was positively related to infiltration of M2 macrophages (P < 0.05). The expression of HSF1 in macrophages was an independent factor for DFS (P = 0.002) and OS (P = 0.002) in ESCC cases. HSF1 was up-regulated in IL-4 stimulation THP1 cells in a time-dependent manner. Under the heat stimulation condition, THP1-derived macrophages were more sensitive than tumor cells. Compared to IL-4 induced-THP1 cells control, the HSF1 knockdown in THP1 cell inhibited the growth and proliferation of ESCC cells. CONCLUSIONS The up-regulation of HSF1 was more rapid and could affect the proliferation of tumor cells in IL4-induced macrophages. The expression of HSF1 in TAMs can also serve as a marker for ESCC prognosis.
Collapse
Affiliation(s)
- Yiqiu Li
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, No. 132 Waihuandong Road, University Town, Guangzhou, 510006, China
| | - Qifan Li
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, No. 132 Waihuandong Road, University Town, Guangzhou, 510006, China
| | - Jiasheng Liu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, No. 132 Waihuandong Road, University Town, Guangzhou, 510006, China
| | - Yuying Huang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, No. 132 Waihuandong Road, University Town, Guangzhou, 510006, China
| | - Jinzhu Mao
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, No. 132 Waihuandong Road, University Town, Guangzhou, 510006, China
| | - Ge Zhang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, No. 132 Waihuandong Road, University Town, Guangzhou, 510006, China.
| |
Collapse
|
50
|
Molecular Features, Prognostic Value, and Cancer Immune Interactions of Angiogenesis-Related Genes in Ovarian Cancer. Reprod Sci 2022; 30:1637-1650. [PMID: 36471217 DOI: 10.1007/s43032-022-01123-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 11/04/2022] [Indexed: 12/12/2022]
Abstract
Angiogenesis is crucial to tumor growth and metastasis; it plays a key role in various cancers development and progression. However, the potential effects of angiogenesis-related genes (ARGs) in ovarian cancer (OC) remain to be further investigated. We discussed the characteristics changes of ARGs in 784 OC samples from genomic and transcriptional levels, as well as their expression patterns based on four distinct datasets. First, 784 OC patients were divided into three molecular subtypes, and the findings indicated that ARG changes were correlated with clinicopathological parameters, prognosis, and immune cell-infiltrating tumor microenvironment (TME). OC patients were subsequently divided into two gene subtypes depending on differentially expressed genes (DEGs) of the abovementioned molecular subtypes. We also established an ARGs-related score (ARGs score) model for evaluating overall survival (OS) and determining the immunological landscape of OC patients, therefore predicting patients' prognosis and therapeutic responses. A lower ARGs' score accompanied by a high mutation frequency implies a higher probability of survival. Furthermore, the ARG score was correlated with the cancer stem cell (CSC) index and chemotherapeutic sensitivity. The significant involvement of ARGs in the tumor-immune-stromal microenvironment, clinicopathological characteristics, and prognosis were established in our systematic investigation of ARGs for OC patients. These discoveries might help us to better understand the role of ARGs in OC, as well as give new insight for predicting the prognosis and providing promising immunotherapy.
Collapse
|