51
|
Duan W, Zhou Z, Huang Y, Cui Y, Jin X, Liu R, Chen L. Euphorbia helioscopia L. inhibits lung tumorigenesis through alleviating exhausted T cell induced by chronic inflammation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119097. [PMID: 39537116 DOI: 10.1016/j.jep.2024.119097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/06/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Euphorbia helioscopia L. (ZQ) is a very effective traditional Chinese medicine for treating pneumonia and lung cancer. However, the effects and mechanisms by which ZQ prevents lung tumorigenesis in the presence of chronic inflammation remain unexplored. AIM To examine the effects and mechanisms of ZQ in alleviating chronic inflammation-induced T cell exhaustion and inhibiting lung tumorigenesis. METHODS A mice model of lung tumorigenesis under chronic inflammation conditions was established by repeated administration of lipopolysaccharide (LPS) and exposure to the tobacco carcinogen nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). Mice were treated with ZQ (0.9, 1.8, and 3.6 g/kg/day) for 25 weeks. Lung pathology and tumor incidence were assessed, and inflammatory cytokine levels in bronchoalveolar lavage fluid (BALF) and serum were measured. Additionally, the proportions of CD3+ T, CD4+ T, and CD8+ T cells and their inhibitory receptors expression were evaluated. In vitro, T cell exhaustion models were induced using inflammatory-conditioned media, followed by treatment with ZQ (0.5, 2, 8 μg/mL). T cell exhaustion markers and characteristics were analyzed, and molecular mechanisms were explored using RNA sequencing and Immunoblotting analysis. RESULTS In vivo, ZQ significantly reduced inflammatory infiltration and lung damage, tumor incidence, number, size, and lung and spleen indices in mice. It also markedly lowered the levels of pro-inflammatory cytokines and immunosuppressive cytokines in BALF and serum. Additionally, ZQ improved the proportions of CD3+ T, CD4+ T, and CD8+ T cells and significantly decreased the expression of inhibitory receptors on CD4+ T and CD8+ T cells in the lung tissues and spleen. In vitro, ZQ effectively alleviated T cell exhaustion induced by the inflammatory environment, marked by reduced expression of inhibitory receptors, increased cytokine secretion, improved proliferation, and enhanced tumoricidal activity. RNA sequencing revealed that ZQ significantly downregulated the JAK-STAT signaling and upregulated stemness-associated pathways. Immunoblotting results indicated that treatment with ZQ markedly reduced the phosphorylation of Signal transducer and activator of transcription 3 (STAT3) and increased the expression of T cell factor -1/7 (TCF1/7). CONCLUSION ZQ inhibits lung tumorigenesis in LPS/NNK-treated mice through alleviating exhausted T cells induced by chronic inflammation, which is attributed to the suppression of STAT3 activation and the maintenance of stemness characteristics in T cells. These findings provide experimental evidence for the potential use of ZQ in preventing and treating lung tumourigenesis in patients with chronic inflammation and the clinical management of lung cancer patients with concomitant chronic inflammation.
Collapse
Affiliation(s)
- Wenbin Duan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Key Laboratory for Evaluation on Anti-Tumor Effect of Chinese Medicine by Strengthening Body Resistance to Eliminate Pathogenic Factors, Nanchang, 330006, China; Key Laboratory of Effective Material Basis of TCM, Jiangxi Province, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| | - Ziye Zhou
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Yuqing Huang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Yaru Cui
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Key Laboratory of Effective Material Basis of TCM, Jiangxi Province, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| | - Xuhui Jin
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Ronghua Liu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Lanying Chen
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Key Laboratory for Evaluation on Anti-Tumor Effect of Chinese Medicine by Strengthening Body Resistance to Eliminate Pathogenic Factors, Nanchang, 330006, China; Key Laboratory of Effective Material Basis of TCM, Jiangxi Province, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| |
Collapse
|
52
|
Suri C, Pande B, Suhasini Sahithi L, Swarnkar S, Khelkar T, Verma HK. Metabolic crossroads: unravelling immune cell dynamics in gastrointestinal cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:7. [PMID: 40051496 PMCID: PMC11883236 DOI: 10.20517/cdr.2024.164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 03/09/2025]
Abstract
Metabolic reprogramming within the tumor microenvironment (TME) plays a critical role in driving drug resistance in gastrointestinal cancers (GI), particularly through the pathways of fatty acid oxidation and glycolysis. Cancer cells often rewire their metabolism to sustain growth and reshape the TME, creating conditions such as nutrient depletion, hypoxia, and acidity that impair antitumor immune responses. Immune cells within the TME also undergo metabolic alterations, frequently adopting immunosuppressive phenotypes that promote tumor progression and reduce the efficacy of therapies. The competition for essential nutrients, particularly glucose, between cancer and immune cells compromises the antitumor functions of effector immune cells, such as T cells. Additionally, metabolic by-products like lactate and kynurenine further suppress immune activity and promote immunosuppressive populations, including regulatory T cells and M2 macrophages. Targeting metabolic pathways such as fatty acid oxidation and glycolysis presents new opportunities to overcome drug resistance and improve therapeutic outcomes in GI cancers. Modulating these key pathways has the potential to reinvigorate exhausted immune cells, shift immunosuppressive cells toward antitumor phenotypes, and enhance the effectiveness of immunotherapies and other treatments. Future strategies will require continued research into TME metabolism, the development of novel metabolic inhibitors, and clinical trials evaluating combination therapies. Identifying and validating metabolic biomarkers will also be crucial for patient stratification and treatment monitoring. Insights into metabolic reprogramming in GI cancers may have broader implications across multiple cancer types, offering new avenues for improving cancer treatment.
Collapse
Affiliation(s)
- Chahat Suri
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton AB T6G 1Z2, Canada
| | - Babita Pande
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India
| | | | | | - Tuneer Khelkar
- Department of Botany and Biotechnology, Govt. Kaktiya P G College, Jagdalpur 494001, India
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lung Health and Immunity, Comprehensive Pneumology Center, Helmholtz Zentrum, Munich 85764, Germany
| |
Collapse
|
53
|
Wang Y, Dong Q, Yuan M, Hu J, Lin P, Yan Y, Wang Y, Wang Y. Effects of metabolism upon immunity: Targeting myeloid-derived suppressor cells for the treatment of breast cancer is a promising area of study. Int Immunopharmacol 2025; 147:113892. [PMID: 39740506 DOI: 10.1016/j.intimp.2024.113892] [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: 10/24/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025]
Abstract
Breast cancer (BC) ranks among the most prevalent malignancies affecting women, with advanced-stage patients facing an increased mortality risk. Myeloid-derived suppressor cells (MDSCs) contribute significantly to poor prognostic outcomes. Research has concentrated predominantly on the immunological mechanisms underlying MDSC functions, but a comprehensive investigation into the metabolic interactions between BC cells and MDSCs is lacking. In a hypoxic tumor microenvironment (TME), BC cells can enhance aerobic-glycolysis rates, upregulate expression of key lipid metabolism enzymes such as cluster of differentiation (CD) 36 and 5-lipoxygenase (5-LOX), accelerate glutamine (Gln) uptake, and elevate extracellular adenosine (eADO) levels, thereby fostering MDSC proliferation and amplifying immune suppression. Concurrently, alterations in the metabolic state of MDSCs also influence BC progression. To ensure adequate proliferative resources, MDSCs upregulate the pentose phosphate pathway and expedite glycolysis for energy supply while increasing the expression of fatty acid transport proteins (FATPs) such as CD36 and fatty acid transporter 2 (FATP2) to maintain intracellular lipid availability, thereby enhancing their adaptability within the TME. Furthermore, MDSCs undermine T-cell anti-tumor efficacy by depleting essential amino acids (AAs), such as arginine (Arg), tryptophan (Trp), and cysteine (Cys), required for T-cell function. This review elucidates how pharmacological agents such as metformin, liver X receptor (LXR) agonists, and 6-diazo-5-oxo-L-norleucine (DON) can augment anti-cancer treatment efficacy by targeting metabolic pathways in MDSCs. We systematically delineate the mechanisms governing interactions between BC cells and MDSCs from a metabolic standpoint while summarizing therapeutic strategies to modulate metabolism within MDSCs. Our review provides a framework for optimizing MDSC applications in BC immunotherapy.
Collapse
Affiliation(s)
- Yulin Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qiutong Dong
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Menghan Yuan
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingxian Hu
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Peizhe Lin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yijing Yan
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yu Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yanyan Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
54
|
Xu R, Lin P, Zheng J, Lin Y, Mai Z, Lu Y, Chen X, Zhou Z, Cui L, Zhao X. Orchestrating cancer therapy: Recent advances in nanoplatforms harmonize immunotherapy with multifaceted treatments. Mater Today Bio 2025; 30:101386. [PMID: 39742149 PMCID: PMC11683241 DOI: 10.1016/j.mtbio.2024.101386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/17/2024] [Accepted: 12/05/2024] [Indexed: 01/03/2025] Open
Abstract
Advancements in cancer therapy have increasingly focused on leveraging the synergistic effects of combining immunotherapy with other treatment modalities, facilitated by the use of innovative nanoplatforms. These strategies aim to augment the efficacy of standalone treatments while addressing their inherent limitations. Nanoplatforms enable precise delivery and controlled release of therapeutic agents, which enhances treatment specificity and reduces systemic toxicity. This review highlights the critical role of nanomaterials in enhancing immunotherapy when combined with chemotherapy, radiotherapy, photodynamic therapy, photothermal therapy, and sonodynamic therapy. Additionally, it addresses current challenges, including limited in vivo studies, difficulties in standardizing and scaling production, complexities of combination therapies, lack of comparative analyses, and the need for personalized treatments. Future directions involve refining nanoplatform engineering for improved targeting and minimizing adverse effects, alongside large animal studies to establish the long-term efficacy and safety of these combined therapeutic strategies. These efforts aim to translate laboratory successes into clinically viable treatments, significantly improving therapeutic outcomes and advancing the field of oncology.
Collapse
Affiliation(s)
- Rongwei Xu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Pei Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yunfan Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Zizhao Mai
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Ye Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Xu Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Zihao Zhou
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
- School of Dentistry, University of California, Los Angeles, Los Angeles, 90095, CA, USA
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| |
Collapse
|
55
|
Li C, Xue Y, Yinwang E, Ye Z. The Recruitment and Immune Suppression Mechanisms of Myeloid-Derived Suppressor Cells and Their Impact on Bone Metastatic Cancer. Cancer Rep (Hoboken) 2025; 8:e70044. [PMID: 39947253 PMCID: PMC11825175 DOI: 10.1002/cnr2.70044] [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: 05/18/2024] [Revised: 09/16/2024] [Accepted: 10/04/2024] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND MDSCs are immature neutrophils and monocytes with immunosuppressive potentials, involving mononuclear MDSCs (M-MDSCs) and polymorphonuclear MDSCs (PMN-MDSCs). RECENT FINDINGS They are significant components of the tumor microenvironment (TME). Besides, recent studies also verified that MDSCs also facilitated the progression of bone metastasis by regulating the network of cytokines and the function of immune cells. CONCLUSION It is necessary to summarize the mechanisms of MDSC recruitment and immunosuppression, and their impact on bone metastasis.
Collapse
Affiliation(s)
- Chengyuan Li
- Department of Orthopedic Surgery, the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Yucheng Xue
- Department of Orthopedic Surgery, the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Eloy Yinwang
- Department of Orthopedic Surgery, the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Zhaoming Ye
- Department of Orthopedic Surgery, the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Orthopedics Research Institute of Zhejiang UniversityHangzhouChina
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
56
|
Cheng X, Shao P, Wang X, Jiang J, Chen J, Zhu J, Zhu W, Li Y, Zhang J, Chen J, Huang Z. Myeloid-Derived Suppressor Cell Accumulation Drives Intestinal Fibrosis through mCCL6/hCCL15 Chemokine-Mediated Fibroblast Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411711. [PMID: 39739231 PMCID: PMC11848553 DOI: 10.1002/advs.202411711] [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: 09/22/2024] [Indexed: 01/02/2025]
Abstract
Intestinal fibrosis, a severe complication of Crohn's disease (CD), is linked to chronic inflammation, but the precise mechanism by which immune-driven intestinal inflammation leads to fibrosis development is not fully understood. This study investigates the role of myeloid-derived suppressor cells (MDSCs) in intestinal fibrosis in CD patients and a 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced mouse model. Elevated MDSCs are observed in inflamed intestinal tissues prior to fibrosis and their sustained presence in fibrotic tissues of both CD patients and murine models. Depletion of MDSCs significantly reduces fibrosis, highlighting their key role in the fibrotic process. Mechanistically, MDSC-derived mCCL6 activates fibroblasts via the CCR1-MAPK signaling, and interventions targeting this axis, including neutralizing antibodies, a CCR1 antagonist, or fibroblast-specific Ccr1 knockout mice reduce fibrosis. In CD patients with stenosis, human CCL15, analogous to mCCL6, is found to be elevated in MDSCs and activated fibroblasts. Additionally, CXCR2 and CCR2 ligands are identified as key mediators of MDSC recruitment in intestinal fibrosis. Blocking MDSC recruitment with CXCR2 and CCR2 antagonists alleviates intestinal fibrosis. These findings suggest that strategies targeting MDSC recruitment and mCCL6/hCCL15 signaling could offer therapeutic benefits for intestinal fibrosis.
Collapse
Affiliation(s)
- Xiaohui Cheng
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Pingwen Shao
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - XinTong Wang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Juan Jiang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Jiahui Chen
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Jie Zhu
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Weiming Zhu
- Department of General SurgeryJinling HospitalSchool of MedicineNanjing UniversityNanjingJiangsu210002China
| | - Yi Li
- Department of General SurgeryJinling HospitalSchool of MedicineNanjing UniversityNanjingJiangsu210002China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Jiangning Chen
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
- State Key Laboratory of Analytical Chemistry for Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Zhen Huang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
- NJU Xishan Institute of Applied BiotechnologyXishan DistrictWuxiJiangsu214101China
| |
Collapse
|
57
|
Yan J, Jiang Z, Zhang S, Yu Q, Lu Y, Miao R, Tang Z, Fan J, Wu L, Duda DG, Zhou J, Yang X. Spatial‒temporal heterogeneities of liver cancer and the discovery of the invasive zone. Clin Transl Med 2025; 15:e70224. [PMID: 39924620 PMCID: PMC11807767 DOI: 10.1002/ctm2.70224] [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: 01/13/2025] [Accepted: 01/19/2025] [Indexed: 02/11/2025] Open
Abstract
Solid tumours are intricate and highly heterogeneous ecosystems, which grow in and invade normal organs. Their progression is mediated by cancer cells' interaction with different cell types, such as immune cells, stromal cells and endothelial cells, and with the extracellular matrix. Owing to its high incidence, aggressive growth and resistance to local and systemic treatments, liver cancer has particularly high mortality rates worldwide. In recent decades, spatial heterogeneity has garnered significant attention as an unfavourable biological characteristic of the tumour microenvironment, prompting extensive research into its role in liver tumour development. Advances in spatial omics have facilitated the detailed spatial analysis of cell types, states and cell‒cell interactions, allowing a thorough understanding of the spatial and temporal heterogeneities of tumour microenvironment and informing the development of novel therapeutic approaches. This review illustrates the latest discovery of the invasive zone, and systematically introduced specific macroscopic spatial heterogeneities, pathological spatial heterogeneities and tumour microenvironment heterogeneities of liver cancer.
Collapse
Affiliation(s)
- Jiayan Yan
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Zhifeng Jiang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Shiyu Zhang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Qichao Yu
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
- BGI‐ShenzhenBeishan Industrial ZoneShenzhenChina
| | - Yijun Lu
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Runze Miao
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Zhaoyou Tang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Jia Fan
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Liang Wu
- BGI‐ShenzhenBeishan Industrial ZoneShenzhenChina
| | - Dan G. Duda
- Steele Laboratories for Tumor BiologyDepartment of Radiation OncologyMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Jian Zhou
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Xinrong Yang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| |
Collapse
|
58
|
Xie Z, Shao J, Shen Z, Ye Z, Okada Y, Okuzaki D, Okada N, Tachibana M. HDAC1-3 inhibition triggers NEDD4-mediated CCR2 downregulation and attenuates immunosuppression in myeloid-derived suppressor cells. Cancer Immunol Immunother 2025; 74:81. [PMID: 39891718 PMCID: PMC11787094 DOI: 10.1007/s00262-024-03931-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/21/2024] [Indexed: 02/03/2025]
Abstract
Myeloid-derived suppressor cells (MDSCs) play a critical role in cancer progression and resistance, thus representing promising targets for immunotherapy. Despite the established role of histone deacetylases (HDACs) in epigenetic regulation of cell fate and function, their specific impact on MDSCs remains elusive. We sought to investigate the effects and underlying mechanisms of HDAC on MDSCs using various HDAC inhibitors. Our results indicate that HDAC1-3 inhibitors reduce CCR2 expression, a chemokine receptor that mediates the migration of monocytic (M-)MDSCs to tumors and attenuated the immunosuppressive activity of MDSCs. In an orthotropic hepatocellular carcinoma (HCC) murine model, HDAC1-3 inhibitors reduced the infiltration of M-MDSCs, increased the number of natural killer cells in tumors, and suppressed tumor growth. Our results also suggest that HDAC1-3 inhibitors potentiate the antitumor effects of anti-programmed cell death protein 1 antibodies. ATAC-seq and RNA-seq analyses revealed 115 genes epigenetically upregulated by HDAC1-3 inhibitors, primarily linked to transcriptional regulation and ubiquitination. We further elucidated that HDAC1-3 inhibitors facilitate CCR2 protein degradation through ubiquitination-mediated by NEDD4 E3 ligase. Our findings reveal a novel mechanism of action of HDAC1-3 inhibitors in MDSCs and suggest a potential synergistic immunotherapy strategy for clinical benefit in HCC.
Collapse
Affiliation(s)
- Zhiqi Xie
- Wuyi First People's Hospital, Affiliated Hospital, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Jinjin Shao
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, 310053, China
| | - Zeren Shen
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Zhichao Ye
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, 310053, China
| | - Yoshiaki Okada
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Daisuke Okuzaki
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Frontier Research Center, Osaka University, Osaka, 565-0871, Japan
| | - Naoki Okada
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Masashi Tachibana
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan.
- Laboratory for Context-Dependent Cell Immunology, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan.
| |
Collapse
|
59
|
Karadima E, Chavakis T, Alexaki VI. Arginine metabolism in myeloid cells in health and disease. Semin Immunopathol 2025; 47:11. [PMID: 39863828 PMCID: PMC11762783 DOI: 10.1007/s00281-025-01038-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Metabolic flexibility is key for the function of myeloid cells. Arginine metabolism is integral to the regulation of myeloid cell responses. Nitric oxide (NO) production from arginine is vital for the antimicrobial and pro-inflammatory responses. Conversely, the arginase 1 (ARG1)-dependent switch between the branch of NO production and polyamine synthesis downregulates inflammation and promotes recovery of tissue homeostasis. Creatine metabolism is key for energy supply and proline metabolism is required for collagen synthesis. Myeloid ARG1 also regulates extracellular arginine availability and T cell responses in parasitic diseases and cancer. Cancer, surgery, sepsis and persistent inflammation in chronic inflammatory diseases, such as neuroinflammatory diseases or arthritis, are associated with dysregulation of arginine metabolism in myeloid cells. Here, we review current knowledge on arginine metabolism in different myeloid cell types, such as macrophages, neutrophils, microglia, osteoclasts, tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs) and myeloid-derived suppressor cells (MDSCs). A deeper understanding of the function of arginine metabolism in myeloid cells will improve our knowledge on the pathology of several diseases and may set the platform for novel therapeutic applications.
Collapse
Affiliation(s)
- Eleftheria Karadima
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.
| |
Collapse
|
60
|
Wang R, Liu Y, Liu M, Zhang M, Li C, Xu S, Tang S, Ma Y, Wu X, Fei W. Combating tumor PARP inhibitor resistance: Combination treatments, nanotechnology, and other potential strategies. Int J Pharm 2025; 669:125028. [PMID: 39638266 DOI: 10.1016/j.ijpharm.2024.125028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/14/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
PARP (poly (ADP-ribose) polymerase) inhibitors (PARPi) have demonstrated significant potential in cancer treatment, particularly in tumors with breast cancer susceptibility gene (BRCA) mutations and other DNA repair deficiencies. However, the development of resistance to PARPi has become a major challenge in their clinical application. The emergence of drug resistance leads to reduced efficacy of the PARPi over time, impacting long-term treatment outcomes and survival rates. PARPi resistance in tumors often arises as cells activate alternative DNA repair pathways or evade the effect of PARPi, diminishing therapeutic effectiveness. Consequently, overcoming resistance is crucial for maintaining treatment efficacy and improving patient prognosis. This paper reviews the strategies to overcome PARPi resistance through combination treatment and nanotechnology therapy. We first review the current combination therapies with PARPi, including anti-angiogenic therapies, radiotherapies, immunotherapies, and chemotherapies, and elucidate their mechanisms for overcoming PARPi resistance. Additionally, this paper focuses on the application of nanotechnology in improving the effectiveness of PARPi and overcoming drug resistance. Subsequently, this paper presents several promising strategies to tackle PARPi resistance, including but not limited to: structural modifications of PARPi, deployment of gene editing systems, implementation of "membrane lipid therapy," and modulation of cellular metabolism in tumors. By integrating these strategies, this research will provide comprehensive approaches to overcome the resistance of PARPi in cancer treatment and offer guidance for future research and clinical practice.
Collapse
Affiliation(s)
- Rong Wang
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yunxi Liu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Mingqi Liu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Meng Zhang
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Chaoqun Li
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Shanshan Xu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Sangsang Tang
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yidan Ma
- YiPeng Subdistrict Community Healthcare Center, Hangzhou 311225, China
| | - Xiaodong Wu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Weidong Fei
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
61
|
Zhao Y, Zhao X, Wang X, Ma Z, Yan J, Li S, Wang N, Jiao J, Cui J, Zhang G. Polyphenol-mediated assembly of toll-like receptor 7/8 agonist nanoparticles for effective tumor immunotherapy. Acta Biomater 2025; 193:417-428. [PMID: 39746528 DOI: 10.1016/j.actbio.2024.12.060] [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: 10/07/2024] [Revised: 12/24/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Toll-like receptor (TLR) 7/8 agonists have shown significant potential in tumor immunotherapy. However, the limited pharmacokinetic properties and systemic toxicity resulting from off-target effects limits their biomedical applications. We here report the polyphenol-mediated assembly of resiquimod (R848, a TLR7/8 agonist) nanoparticles (RTP NPs) to achieve tumor-selective immunotherapy while avoiding systemic adverse effects. Upon intravenous administration, the prepared RTP NPs are effectively accumulated at tumor sites, which increase their bioavailability and reduce systemic inflammation. RTP NPs can trigger a potent antitumor immune response in a mouse tumor model to inhibit tumor growth. Additionally, after subcutaneous injection at the tail base, RTP NPs efficiently migrate to the lymph nodes, where they elicit immune memory to prevent tumorigenesis. This study underscores the potential application of polyphenol-mediated assembly in developing nanomedicines with reduced toxicity for tumor-specific immunotherapy. STATEMENT OF SIGNIFICANCE: Toll-like receptor agonist (R848) nanoparticles for tumor-selective immunotherapy were synthesized through polyphenol-mediated assembly, a method that simplifies preparation process and minimizes potential side effects. Intravenously administered these nanoparticles effectively extended circulation time, enhanced tumor enrichment, and reduced systemic inflammation, thus augmenting the bioavailability and minimizing the side effects of R848. The nanoparticles significantly inhibited tumor growth by triggering a potent antitumor immune response, including dendritic cell maturation, macrophage polarization, T-cell infiltration, and cytokine secretion. Moreover, after subcutaneous injection at the tail base, they can elicit immune memory to prevent tumorigenesis.
Collapse
Affiliation(s)
- Yilei Zhao
- The Second Affiliated Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Xiaonan Zhao
- The Second Affiliated Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Xuechun Wang
- The Second Affiliated Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Zilin Ma
- The Second Affiliated Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Jie Yan
- The Second Affiliated Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Songyan Li
- The Second Affiliated Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Ning Wang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Jianwei Jiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology Chinese Academy of Sciences, Beijing 100101, China.
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China.
| | - Guiqiang Zhang
- The Second Affiliated Hospital, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China.
| |
Collapse
|
62
|
Zhu Y, Chen J, Chen C, Tang R, Xu J, Shi S, Yu X. Deciphering mechanical cues in the microenvironment: from non-malignant settings to tumor progression. Biomark Res 2025; 13:11. [PMID: 39849659 PMCID: PMC11755887 DOI: 10.1186/s40364-025-00727-9] [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: 11/11/2024] [Accepted: 01/05/2025] [Indexed: 01/25/2025] Open
Abstract
The tumor microenvironment functions as a dynamic and intricate ecosystem, comprising a diverse array of cellular and non-cellular components that precisely orchestrate pivotal tumor behaviors, including invasion, metastasis, and drug resistance. While unraveling the intricate interplay between the tumor microenvironment and tumor behaviors represents a tremendous challenge, recent research illuminates a crucial biological phenomenon known as cellular mechanotransduction. Within the microenvironment, mechanical cues like tensile stress, shear stress, and stiffness play a pivotal role by activating mechanosensitive effectors such as PIEZO proteins, integrins, and Yes-associated protein. This activation initiates cascades of intrinsic signaling pathways, effectively linking the physical properties of tissues to their physiological and pathophysiological processes like morphogenesis, regeneration, and immunity. This mechanistic insight offers a novel perspective on how the mechanical cues within the tumor microenvironment impact tumor behaviors. While the intricacies of the mechanical tumor microenvironment are yet to be fully elucidated, it exhibits distinct physical attributes from non-malignant tissues, including elevated solid stresses, interstitial hypertension, augmented matrix stiffness, and enhanced viscoelasticity. These traits exert notable influences on tumor progression and treatment responses, enriching our comprehension of the multifaceted nature of the microenvironment. Through this innovative review, we aim to provide a new lens to decipher the mechanical attributes within the tumor microenvironment from non-malignant contexts, broadening our knowledge on how these factors promote or inhibit tumor behaviors, and thus offering valuable insights to identify potential targets for anti-tumor strategies.
Collapse
Affiliation(s)
- Yicheng Zhu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jiaoshun Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Chen Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Rong Tang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
63
|
Zhang L, Jiang H, Ma H. Progress in immune microenvironment, immunotherapy and prognostic biomarkers in pediatric osteosarcoma. Front Immunol 2025; 16:1548527. [PMID: 39911380 PMCID: PMC11794274 DOI: 10.3389/fimmu.2025.1548527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/06/2025] [Indexed: 02/07/2025] Open
Abstract
Pediatric osteosarcoma, the most prevalent primary malignant bone tumor in children, is marked by aggressive progression and a generally poor prognosis. Despite advances in treatment, including multi-agent chemotherapy, survival rates remain suboptimal, with metastasis, particularly to the lungs, contributing significantly to mortality. The tumor microenvironment plays a crucial role in osteosarcoma progression, with immune cells such as tumor-associated macrophages and T lymphocytes significantly influencing tumor behavior. The immunosuppressive environment, dominated by M2 macrophages, contributes to immune evasion and poor therapeutic outcomes, though recent findings suggest the potential for reprogramming these cells to enhance immune responses. This review provides a comprehensive overview of the immune landscape in pediatric osteosarcoma, with a focus on the role of immune cells and their interactions within the tumor microenvironment (TME). It examines the impact of immune checkpoints, genetic mutations, and inflammatory pathways on osteosarcoma progression, highlighting their contribution to tumor immune evasion and disease advancement. Additionally, emerging immunotherapeutic strategies, such as immune checkpoint inhibitors, macrophage reprogramming, and antibody-based therapies, are summarized in detail, showcasing their potential to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Orthopedics, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Haoming Jiang
- Department of Orthopedics, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Haichao Ma
- Department of Pediatrics, Shenzhen University General Hospital, Shenzhen, China
| |
Collapse
|
64
|
Zhang X, Sun K, Zhong B, Yan L, Cheng P, Wang Q. PMN-MDSCs are responsible for immune suppression in anti-PD-1 treated TAP1 defective melanoma. Clin Transl Oncol 2025:10.1007/s12094-024-03840-7. [PMID: 39825997 DOI: 10.1007/s12094-024-03840-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/24/2024] [Indexed: 01/20/2025]
Abstract
INTRODUCTION The transporter associated with antigen processing (TAP) is a key component of the classical HLA I antigen presentation pathway. Our previous studies have demonstrated that the downregulation of TAP1 contributes to tumor progression and is associated with an increased presence of myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment. However, it remains unclear whether the elevation of MDSCs leads to immune cell exhaustion in tumors lacking TAP1. In this study, we established mouse models of tumors with TAP1 deficiency, and we employed PMN-MDSC depletion to investigate their impact on the immune microenvironment within the tumors. We found that MDSC depletion significantly altered the immune-suppressive effects of TAP1-deficient tumor when anti-PD-1 treatment was administered. Targeting PMN-MDSC may be a promising therapeutic strategy for the treatment of tumors with TAP1 deficiency during ICB treatment. METHODS Immunohistochemistry (IHC) was conducted to assess TAP1 expression in mouse melanoma tissues. Ly6G, F4/80, and NKp46 markers were detected in B16 parental and TAP1 knockout tissues, respectively. To enhance anti-tumor immunity, hyperthermia-treated B16F10 WT cell suspension was injected prior to tumor cell introduction. Subsequently, we established B16F10 TAP1 knockout and WT melanoma mouse models. Tumors were collected, and the immune microenvironment was monitored accordingly. Anti-Ly6G antibody was administered to deplete polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). Finally, flow cytometry analysis for immune infiltration, quantitative PCR for cytokine levels, and immunofluorescence assays were performed to analyze the immune response. RESULTS The level of Ly6G+ cell infiltration was significantly higher in samples exhibiting low TAP1 expression, while no differences were observed in the infiltration of F4/80+ cells or NKp46+ cells. Furthermore, the immune-suppressive effects associated with PMN-MDSCs were reversed following their elimination; this resulted in an increase in CD8+ T cells and a higher ratio of CD8+ T cells to Tregs, while the infiltration of innate immune cells remained unaffected. Functional markers of these immune cells indicated an active anti-tumoral immune response following the removal of PMN-MDSCs. Quantitative PCR analysis indicated elevated levels of TNF-α and IL-6, accompanied by decreased levels of TGF-β in the tumor microenvironment of TAP1. CONCLUSIONS Our data indicate that myeloid-derived suppressor cells (PMN-MDSCs) play an essential role in creating a tumorigenic immune microenvironment in TAP1 knockout tumors. Therefore, targeting PMN-MDSCs may become a promising therapeutic strategy for the treatment of tumors with TAP1 deficiency during ICB treatment.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510013, Guangdong, China
| | - Kaijun Sun
- Weifang People's Hospital, The First Affiliated Hospital of Shandong Second Medical University, Weifang, 261041, Shandong, China
| | - Bingzheng Zhong
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510013, Guangdong, China
| | - Likun Yan
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Pengrui Cheng
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510013, Guangdong, China
| | - Qiang Wang
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510013, Guangdong, China.
| |
Collapse
|
65
|
Wang H, Zhou F, Qin W, Yang Y, Li X, Liu R. Metabolic regulation of myeloid-derived suppressor cells in tumor immune microenvironment: targets and therapeutic strategies. Theranostics 2025; 15:2159-2184. [PMID: 39990210 PMCID: PMC11840731 DOI: 10.7150/thno.105276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/11/2024] [Indexed: 02/25/2025] Open
Abstract
Cancer remains a major challenge to global public health, with rising incidence and high mortality rates. The tumor microenvironment (TME) is a complex system of immune cells, fibroblasts, extracellular matrix (ECM), and blood vessels that form a space conducive to cancer cell proliferation. Myeloid-derived suppressor cells (MDSCs) are abundant in tumors, and they drive immunosuppression through metabolic reprogramming in the TME. This review describes how metabolic pathways such as glucose metabolism, lipid metabolism, amino acid metabolism, and adenosine metabolism have a significant impact on the function of MDSCs by regulating their immunosuppressive activity and promoting their survival and expansion in tumors. The review also explores key metabolic targets in MDSCs and strategies to modulate MDSC metabolism to improve the tumor immune microenvironment and enhance anti-tumor immune responses. Understanding these pathways can provide insight into potential therapeutic targets for modulating MDSC activity and improving outcomes of cancer immunotherapies.
Collapse
Affiliation(s)
- Hong Wang
- School of Life Sciences, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Fei Zhou
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Wenqing Qin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Yun Yang
- School of Life Sciences, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| |
Collapse
|
66
|
Glaviano A, Lau HSH, Carter LM, Lee EHC, Lam HY, Okina E, Tan DJJ, Tan W, Ang HL, Carbone D, Yee MYH, Shanmugam MK, Huang XZ, Sethi G, Tan TZ, Lim LHK, Huang RYJ, Ungefroren H, Giovannetti E, Tang DG, Bruno TC, Luo P, Andersen MH, Qian BZ, Ishihara J, Radisky DC, Elias S, Yadav S, Kim M, Robert C, Diana P, Schalper KA, Shi T, Merghoub T, Krebs S, Kusumbe AP, Davids MS, Brown JR, Kumar AP. Harnessing the tumor microenvironment: targeted cancer therapies through modulation of epithelial-mesenchymal transition. J Hematol Oncol 2025; 18:6. [PMID: 39806516 PMCID: PMC11733683 DOI: 10.1186/s13045-024-01634-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025] Open
Abstract
The tumor microenvironment (TME) is integral to cancer progression, impacting metastasis and treatment response. It consists of diverse cell types, extracellular matrix components, and signaling molecules that interact to promote tumor growth and therapeutic resistance. Elucidating the intricate interactions between cancer cells and the TME is crucial in understanding cancer progression and therapeutic challenges. A critical process induced by TME signaling is the epithelial-mesenchymal transition (EMT), wherein epithelial cells acquire mesenchymal traits, which enhance their motility and invasiveness and promote metastasis and cancer progression. By targeting various components of the TME, novel investigational strategies aim to disrupt the TME's contribution to the EMT, thereby improving treatment efficacy, addressing therapeutic resistance, and offering a nuanced approach to cancer therapy. This review scrutinizes the key players in the TME and the TME's contribution to the EMT, emphasizing avenues to therapeutically disrupt the interactions between the various TME components. Moreover, the article discusses the TME's implications for resistance mechanisms and highlights the current therapeutic strategies toward TME modulation along with potential caveats.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Hannah Si-Hui Lau
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lukas M Carter
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Donavan Jia Jie Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Wency Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Michelle Yi-Hui Yee
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Xiao Zi Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Lina H K Lim
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Ruby Yun-Ju Huang
- School of Medicine and Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, 23538, Lübeck, Germany
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Experimental Therapeutics (ET) Graduate Program, University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Tullia C Bruno
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Bin-Zhi Qian
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, W12 0BZ, UK
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Salem Elias
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Saurabh Yadav
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Minah Kim
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Caroline Robert
- Department of Cancer Medicine, Inserm U981, Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif, France
- Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, Paris, France
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Tao Shi
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, NY, USA
| | - Simone Krebs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironment Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Matthew S Davids
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jennifer R Brown
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| |
Collapse
|
67
|
Kong Y, Li J, Zhao X, Wu Y, Chen L. CAR-T cell therapy: developments, challenges and expanded applications from cancer to autoimmunity. Front Immunol 2025; 15:1519671. [PMID: 39850899 PMCID: PMC11754230 DOI: 10.3389/fimmu.2024.1519671] [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: 10/30/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025] Open
Abstract
Chimeric Antigen Receptor (CAR)-T cell therapy has rapidly emerged as a groundbreaking approach in cancer treatment, particularly for hematologic malignancies. However, the application of CAR-T cell therapy in solid tumors remains challenging. This review summarized the development of CAR-T technologies, emphasized the challenges and solutions in CAR-T cell therapy for solid tumors. Also, key innovations were discussed including specialized CAR-T, combination therapies and the novel use of CAR-Treg, CAR-NK and CAR-M cells. Besides, CAR-based cell therapy have extended its reach beyond oncology to autoimmune disorders. We reviewed preclinical experiments and clinical trials involving CAR-T, Car-Treg and CAAR-T cell therapies in various autoimmune diseases. By highlighting these cutting-edge developments, this review underscores the transformative potential of CAR technologies in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Yanwei Wu
- School of Medicine, Shanghai University, Shanghai, China
| | - Liang Chen
- School of Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
68
|
He S, Zheng L, Qi C. Myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment and their targeting in cancer therapy. Mol Cancer 2025; 24:5. [PMID: 39780248 PMCID: PMC11707952 DOI: 10.1186/s12943-024-02208-3] [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: 09/26/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
The advent of immunotherapy represents a significant breakthrough in cancer treatment, with immune checkpoint inhibitors (ICIs) targeting PD-1 and CTLA-4 demonstrating remarkable therapeutic efficacy. However, patient responses to immunotherapy vary significantly, with immunosuppression within the tumor microenvironment (TME) being a critical factor influencing this variability. Immunosuppression plays a pivotal role in regulating cancer progression, metastasis, and reducing the success rates of immunotherapy. Myeloid-derived suppressor cells (MDSCs), due to their potent immunosuppressive capabilities, emerged as major negative regulators within the TME, facilitating tumor immune evasion by modulating various immune cells. In addition to their immunosuppressive functions, MDSCs also promote tumor growth and metastasis through non-immunological mechanisms, such as angiogenesis and the formation of pre-metastatic niches. Consequently, MDSCs in the TME are key regulators of cancer immune responses and potential therapeutic targets in cancer treatment. This review describes the origins and phenotypes of MDSCs, their biological roles in tumor progression, and regulatory mechanisms, with a focus on current therapeutic approaches targeting tumor-associated MDSCs. Furthermore, the synergistic effects of targeting MDSCs in combination with immunotherapy are explored, aiming to provide new insights and directions for cancer therapy.
Collapse
Affiliation(s)
- Shuyan He
- Department of Tumor Center, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin, Jiangsu, China
| | - Lu Zheng
- Clinical Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Chunjian Qi
- Laboratory of Oncology, Basic Research Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China.
| |
Collapse
|
69
|
Shao X, Zhao X, Wang B, Fan J, Wang J, An H. Tumor microenvironment targeted nano-drug delivery systems for multidrug resistant tumor therapy. Theranostics 2025; 15:1689-1714. [PMID: 39897552 PMCID: PMC11780529 DOI: 10.7150/thno.103636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/10/2024] [Indexed: 02/04/2025] Open
Abstract
In recent years, nano-drug delivery systems (Nano-DDS) that target the tumor microenvironment (TME) to overcome multidrug resistance (MDR) have become a research hotspot in the field of cancer therapy. By precisely targeting the TME and regulating its unique pathological features, such as hypoxia, weakly acidic pH, and abnormally expressed proteins, etc., these Nano-DDS enable effective delivery of therapeutic agents and reversal of MDR. This scientific research community is increasing its investment in the development of diversified systems and exploring their anti-drug resistance potential. Therefore, it is particularly important to conduct a comprehensive review of the research progress of TME-targeted Nano-DDS in recent years. After a brief introduction of TME and tumor MDR, the design principle and structure of liposomes, polymer micelles and inorganic nanocarriers are focused on, and their characteristics as TME-targeted nanocarriers are described. It also demonstrates how these systems break through the cancer MDR treatment through various targeting mechanisms, discusses their synthetic innovation, research results and resistance overcoming mechanisms. The review was concluded with deliberations on the key challenges and future outlooks of targeting TME Nano-DDS in cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | - Jinping Wang
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, 300401, Tianjin, PR China
| | - Hailong An
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, 300401, Tianjin, PR China
| |
Collapse
|
70
|
Yan Q, Mohammadpour H. Platelet-activating factor: a potential therapeutic target to improve cancer immunotherapy. Mol Oncol 2025; 19:11-14. [PMID: 39558859 PMCID: PMC11705722 DOI: 10.1002/1878-0261.13758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/20/2024] Open
Abstract
The tumor microenvironment (TME) fosters cancer progression by supporting the differentiation and proliferation of myeloid-derived suppressor cells (MDSCs), which play a critical role in suppressing immune responses and facilitating tumor growth. Recent findings by Dahal et al. reveal that platelet-activating factor (PAF), a lipid mediator elevated in the TME, contributes to the differentiation of neutrophils into immunosuppressive neutrophils. They showed that inhibiting PAF signaling reduces MDSC-mediated immunosuppression, thereby enhancing cytotoxic T-cell activity. This approach may improve cancer immunotherapy outcomes, particularly when combined with checkpoint blockade therapies, suggesting a promising avenue for therapeutic development.
Collapse
Affiliation(s)
- Qi Yan
- Department of Cell Stress BiologyRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
| | - Hemn Mohammadpour
- Department of Cell Stress BiologyRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
| |
Collapse
|
71
|
Kim J, Dan KB, Kim KU, Min H. 20(S)-ginsenoside Rg3 alleviates DSS-induced colitis by promoting ERK-dependent maturation of MDSCs into M2 macrophages. Biomed Pharmacother 2025; 182:117789. [PMID: 39721327 DOI: 10.1016/j.biopha.2024.117789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are immature myeloid cells with immunosuppressive functions that play various roles in tumors and inflammatory diseases. In colitis, MDSCs accumulate in the inflamed colon, where they mature into M2-polarized macrophages and modulate inflammatory responses. Ginsenosides, active components of ginseng, have been shown to display colitis-alleviating effects in mouse models. However, the detailed mechanisms underlying these effects are incompletely understood. This study explores the impact of ginsenosides on MDSC functions and differentiation, focusing on their potential to mitigate inflammatory symptoms in colitis. Among the 15 ginsenosides tested, Rg3(R) and Rg3(S) were found to promote the maturation of MDSCs into M2 macrophages at non-cytotoxic concentrations. This was confirmed by the increased expression of ARG1, an immunosuppressive marker. These effects were attributed to the activation of the ERK pathway, as confirmed by selective ERK inhibition. In a DSS-induced colitis mouse model, oral administration of Rg3(S) alleviated disease severity and increased MDSC differentiation into M2 macrophages in colon lamina propria, highlighting its therapeutic potential colitis.
Collapse
Affiliation(s)
- Jisu Kim
- College of Pharmacy, Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea.
| | - Kang-Bin Dan
- College of Pharmacy, Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea.
| | - Ki-Uk Kim
- College of Pharmacy, Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea.
| | - Hyeyoung Min
- College of Pharmacy, Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea.
| |
Collapse
|
72
|
Ren J, Ying J, Liu H, Hu S, Li J, Zhou D. Stimulator of Interferon Genes Signal in Lung Cancer Regulates Differentiation of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment Via the Interferon Regulatory Factor 3/NF-κB Pathway. J Interferon Cytokine Res 2025; 45:29-37. [PMID: 39772902 DOI: 10.1089/jir.2024.0150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
Background: This study was designed to explore the action mechanism of stimulator of interferon genes (STING) on the differentiation of myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment of lung cancer. Methods: Bioinformatics analysis yielded a potential pathway for STING to regulate MDSC differentiation, the interferon regulatory factor 3 (IRF3)/NF-κB axis. The transfection efficiency of STING overexpression plasmid and small interfering RNA against IRF3 (siIRF3) was examined by quantitative real-time polymerase chain reaction (qRT-PCR). After transfection, A9 cells were co-cultured with extracted bone marrow cells (BMCs). MDSC differentiation, protein expression of the IRF3/NF-κB pathway, and changes in nuclear translocation of NF-κB were analyzed by flow cytometry, Western blot, and immunofluorescence staining experiments. A transplanted tumor mouse model was used for in vivo experiments. After cyclic diadenyl monophosphate (CDA; STING agonist) treatment, changes in MDSC differentiation and protein expression of the IRF3/NF-κB axis in transplanted tumors were verified by immunohistochemical staining, qRT-PCR, and Western blot. Results: Coculture of A9 cells and BMCs promoted MDSC differentiation, inhibited activation of IRF3/NF-κB signal in A9 cells, and boosted nuclear translocation of NF-κB. However, after the upregulation of STING, IRF3/NF-κB signal was activated, while MDSC differentiation and nuclear translocation of NF-κB were inhibited. SiIRF3 reversed the effects of STING overexpression. In vivo, CDA dampened MDSC differentiation and promoted protein expression of the IRF3/NF-κB axis. Conclusion: STING signal in lung cancer cells inhibits MDSC differentiation through activation of the IRF3/NF-κB pathway.
Collapse
Affiliation(s)
- Jiaojiao Ren
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, China
| | - Jun Ying
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, China
| | - Haijian Liu
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, China
| | - Shanshan Hu
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, China
| | - Jiangdong Li
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, China
| | - Danfei Zhou
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, China
| |
Collapse
|
73
|
Yun H, Dong F, Wei X, Yan X, Zhang R, Zhang X, Wang Y. Role and value of the tumor microenvironment in the progression and treatment resistance of gastric cancer (Review). Oncol Rep 2025; 53:14. [PMID: 39611496 PMCID: PMC11622107 DOI: 10.3892/or.2024.8847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/08/2024] [Indexed: 11/30/2024] Open
Abstract
Gastric cancer (GC) is characterized by a complex and heterogeneous tumor microenvironment (TME) that significantly influences disease progression and treatment outcomes. The tumor stroma, which is composed of a variety of cell types such as cancer‑associated fibroblasts, immune cells and vascular components, displays significant spatial and temporal diversity. These stromal elements engage in dynamic crosstalk with cancer cells, shaping their proliferative, invasive and metastatic potential. Furthermore, the TME is instrumental in facilitating resistance to traditional chemotherapy, specific treatments and immunotherapy strategies. Understanding the underlying mechanisms by which the GC microenvironment evolves and supports tumor growth and therapeutic resistance is critical for developing effective treatment strategies. The present review explores the latest progress in understanding the intricate interactions between cancer cells and their immediate environment in GC, highlighting the implications for disease pathogenesis and therapeutic interventions.
Collapse
Affiliation(s)
- Heng Yun
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Fangde Dong
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Xiaoqin Wei
- Department of Pain, The Second People's Hospital of Baiyin, Baiyin, Gansu 730900, P.R. China
| | - Xinyong Yan
- Department of Proctology, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Ronglong Zhang
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Xiuyu Zhang
- Department of Gastroenterology, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Yulin Wang
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| |
Collapse
|
74
|
Cortellino S, D'Angelo M, Quintiliani M, Giordano A. Cancer knocks you out by fasting: Cachexia as a consequence of metabolic alterations in cancer. J Cell Physiol 2025; 240:e31417. [PMID: 39245862 DOI: 10.1002/jcp.31417] [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: 04/28/2024] [Revised: 07/18/2024] [Accepted: 08/09/2024] [Indexed: 09/10/2024]
Abstract
Neoplastic transformation reprograms tumor and surrounding host cell metabolism, increasing nutrient consumption and depletion in the tumor microenvironment. Tumors uptake nutrients from neighboring normal tissues or the bloodstream to meet energy and anabolic demands. Tumor-induced chronic inflammation, a high-energy process, also consumes nutrients to sustain its dysfunctional activities. These tumor-related metabolic and physiological changes, including chronic inflammation, negatively impact systemic metabolism and physiology. Furthermore, the adverse effects of antitumor therapy and tumor obstruction impair the endocrine, neural, and gastrointestinal systems, thereby confounding the systemic status of patients. These alterations result in decreased appetite, impaired nutrient absorption, inflammation, and shift from anabolic to catabolic metabolism. Consequently, cancer patients often suffer from malnutrition, which worsens prognosis and increases susceptibility to secondary adverse events. This review explores how neoplastic transformation affects tumor and microenvironment metabolism and inflammation, leading to poor prognosis, and discusses potential strategies and clinical interventions to improve patient outcomes.
Collapse
Affiliation(s)
- Salvatore Cortellino
- Laboratory of Molecular Oncology, Responsible Research Hospital, Campobasso, Italy
- Scuola Superiore Meridionale (SSM), School for Advanced Studies, Federico II University, Naples, Italy
- SHRO Italia Foundation ETS, Candiolo, Turin, Italy
| | - Margherita D'Angelo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, USA
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
75
|
Qu F, Wang G, Wen P, Liu X, Zeng X. Knowledge mapping of immunotherapy for breast cancer: A bibliometric analysis from 2013 to 2022. Hum Vaccin Immunother 2024; 20:2335728. [PMID: 38563136 PMCID: PMC10989689 DOI: 10.1080/21645515.2024.2335728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024] Open
Abstract
Breast cancer is the leading cause of cancer-related death among women globally. Immunotherapy has emerged as a major milestone in contemporary oncology. This study aims to conduct a bibliometric analysis in the field of immunotherapy for breast cancer, providing a comprehensive overview of the current research status, identifying trends and hotspots in research topics. We searched and retrieved data from the Web of Science Core Collection, and performed a bibliometric analysis of publications on immunotherapy for breast cancer from 2013 to 2022. Current status and hotspots were evaluated by co-occurrence analysis using VOSviewer. Evolution and bursts of knowledge base were assessed by co-citation analysis using CiteSpace. Thematic evolution by bibliometrix package was used to discover keywords trends. The attribution and collaboration of countries/regions, institutions and authors were also explored. A total of 7,975 publications were included. In co-occurrence analysis of keywords, 6 major clusters were revealed: tumor microenvironment, prognosis biomarker, immune checkpoints, novel drug delivery methods, immune cells and therapeutic approaches. The top three most frequently mentioned keywords were tumor microenvironment, triple-negative breast cancer, and programmed cell death ligand 1. The most productive country, institution and author were the USA (2926 publications), the University of Texas MD Anderson Cancer Center (219 publications), and Sherene Loi (28 publications), respectively. There has been a rapid growth in studies on immunotherapy for breast cancer worldwide. This research area has gained increasing attention from different countries and institutions. With the rising incidence of breast cancer, immunotherapy represents a research field of significant clinical value and potential.
Collapse
Affiliation(s)
- Fanli Qu
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing, China
| | - Guanwen Wang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing, China
| | - Ping Wen
- School of Medicine, Chongqing University, Chongqing, China
| | - Xiaoyu Liu
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaohua Zeng
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
76
|
Nie Y, Schalper KA, Chiang A. Mechanisms of immunotherapy resistance in small cell lung cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:55. [PMID: 39802951 PMCID: PMC11724353 DOI: 10.20517/cdr.2024.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/05/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025]
Abstract
Small-cell lung cancer (SCLC) is an aggressive neuroendocrine tumor with a poor prognosis. Although the addition of immunotherapy to chemotherapy has modestly improved outcomes, most patients rapidly develop resistance. Resistance to immunotherapy can be broadly categorized into primary resistance and acquired resistance, as proposed by the Society for Immunotherapy of Cancer (SITC) consensus definition. Primary resistance occurs in the setting of failure to respond to immune checkpoint inhibitors (ICIs), while acquired resistance develops after initial response. The mechanisms of acquired and primary resistance to ICI are not well understood in SCLC, denoting an area of critical unmet need. Both intrinsic and extrinsic mechanisms play significant roles in immunotherapy resistance. Intrinsic mechanisms include defects in antigen presentation, mutations in key genes, reduced tumor immunogenicity, and epigenetic alterations. Extrinsic mechanisms involve the tumor microenvironment (TME), which is a complex interplay of both tumor- and immunosuppressive immune cells, vasculature, and microbiome. An understanding of these resistance mechanisms is crucial for developing novel therapeutic strategies to advance effective immunotherapy in patients with SCLC, a critical area of unmet need.
Collapse
Affiliation(s)
- Yunan Nie
- Department of Medical Oncology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Kurt A. Schalper
- Department of Medical Oncology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Anne Chiang
- Department of Medical Oncology, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
77
|
DU N, Wan H, Guo H, Zhang X, Wu X. [Myeloid-derived suppressor cells as important factors and potential targets for breast cancer progression]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:785-795. [PMID: 39686697 PMCID: PMC11736353 DOI: 10.3724/zdxbyxb-2024-0353] [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: 07/31/2024] [Accepted: 10/28/2024] [Indexed: 12/18/2024]
Abstract
Recurrence and metastasis remain the leading cause of death in breast cancer patients due to the lack of effective treatment. A microenvironment suitable for cancer cell growth, referred to as pre-metastatic niche (PMN), is formed in distant organs before metastasis occurs. Myeloid-derived suppressor cells (MDSCs) are a heterogenous population of immature myeloid cells with immunosuppressive effects. They can expand in large numbers in breast cancer patients and participate in the formation of PMN. MDSCs can remodel the extracellular matrix of pulmonary vascular endothelial cells and recruit cancer stem cells to promote the lung metastasis of breast cancer. Furthermore, MDSCs facilitate immune evasion of breast cancer cells to impact the efficacy of immunotherapy. It is proposed that MDSCs represent a potential therapeutic target for the inhibition of recurrence and metastasis in breast cancer. Therapeutic strategies targeting MDSCs have shown promising efficacy in preclinical studies and clinical trials. This review presents a summary of the principal factors involved in the recruitment and activation of MDSCs during the formation of PMN, and outlines MDSCs functions such as immunosuppression and the current targeted therapies against MDSCs, aiming to provide new ideas for the treatment of distant metastases in breast cancer.
Collapse
Affiliation(s)
- Nannan DU
- Breast Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China.
| | - Hua Wan
- Breast Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Hailing Guo
- Department of Orthopaedics and Traumatology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Xukuan Zhang
- Breast Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Xueqing Wu
- Breast Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China.
| |
Collapse
|
78
|
Masui H, Kawada K, Obama K. Neutrophil and Colorectal Cancer. Int J Mol Sci 2024; 26:6. [PMID: 39795864 PMCID: PMC11720084 DOI: 10.3390/ijms26010006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
Colorectal cancer (CRC) is often associated with metastasis and recurrence and is the leading cause of cancer-related mortality. In the progression of CRC, recent studies have highlighted the critical role of neutrophils, particularly tumor-associated neutrophils (TANs). TANs have both tumor-promoting and tumor-suppressing activities, contributing to metastasis, immunosuppression, angiogenesis, and epithelial-to-mesenchymal transition. Tumor-promoting TANs promote tumor growth by releasing proteases, reactive oxygen species, and cytokines, whereas tumor-suppressing TANs enhance immune responses by activating T cells and natural killer cells. Understanding the mechanisms underlying TAN mobilization, plasticity, and their role in the tumor microenvironment has revealed potential therapeutic targets. This review provides a comprehensive overview of TAN biology in CRC and discusses both the tumor-promoting and tumor-suppressing functions of neutrophils. Novel therapeutic approaches targeting TANs, such as chemokine receptor antagonists, aim to modulate neutrophil reprogramming and offer promising avenues for improving treatment outcomes of CRC.
Collapse
Affiliation(s)
- Hideyuki Masui
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan; (H.M.); (K.O.)
- Department of Surgery, Hirakata Kohsai Hospital, Osaka 573-0153, Japan
| | - Kenji Kawada
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan; (H.M.); (K.O.)
- Department of Surgery, Kurashiki Central Hospital, Okayama 710-8602, Japan
| | - Kazutaka Obama
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan; (H.M.); (K.O.)
| |
Collapse
|
79
|
Pan J, Liang H, Zhou L, Lu W, Huo B, Liu R, Huang P. SQLE-mediated squalene metabolism promotes tumor immune evasion in pancreatic cancer. Front Immunol 2024; 15:1512981. [PMID: 39763673 PMCID: PMC11701373 DOI: 10.3389/fimmu.2024.1512981] [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: 10/17/2024] [Accepted: 12/05/2024] [Indexed: 03/25/2025] Open
Abstract
Background Squalene epoxidase (SQLE) is a key enzyme in cholesterol biosynthesis and has been shown to negatively affect tumor immunity and is associated with poor outcomes of immunotherapy in various cancers. While most research in this area has focused on the impact of cholesterol on immune functions, the influence of SQLE-mediated squalene metabolism within the tumor immune microenvironment (TIME) remains unexplored. Methods We established an immune-competent mouse model (C57BL/6) bearing mouse pancreatic cancer xenografts (KPC cells) with or without stable SQLE-knockdown (SQLE-KD) to evaluate the impact of SQLE-mediated metabolism on pancreatic cancer growth and immune functions. The effect of squalene on tumor growth and immune cells was tested by direct administration of squalene to C57BL/6 mice bearing KPC tumors. Flow cytometry analysis and immunohistochemical (IHC) staining of immune cells from the tumor tissues were performed to evaluate changes in immune function. We also employed RNA-sequencing to analyze the gene expression profiles in pancreatic cancer cells (PANC-1) treated with or without squalene. RT-PCR and Western blot analyses were used to investigate the relevant molecular mechanisms. Results We show that SQLE is significantly overexpressed in pancreatic cancer, and abrogation of SQLE results in a significant increase in squalene accumulation within tumor cells. The elevated squalene inhibits CXCL1 transcription through its impact on the NF-κB pathway via p65, and thus reduces the recruitment of myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) into the tumor microenvironment. Silencing of SQLE also leads to an increased proportion of CD8+ T cells in the tumor tissues and suppresses tumor growth in vivo. Importantly, direct administration of squalene, the metabolic substrate of SQLE, to immune-competent mice bearing KPC pancreatic cancer tumors causes a substantial decrease in CD206+ TAMs and MDSCs, thus releasing immune suppression and inhibiting tumor growth. Conclusion Our study shows that squalene is an important immune-modulating metabolite that inhibits the infiltration of immune-suppressive cells in TIME, and that SQLE exerts its tumor immune evasion effect by metabolic removal of squalene. Thus, SQLE-mediated squalene metabolic pathway could be a potential target to enhance antitumor immunity in pancreatic cancer.
Collapse
Affiliation(s)
- Junchen Pan
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Haixi Liang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Lin Zhou
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wenhua Lu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Bitao Huo
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Metabolic Innovation Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Rui Liu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Peng Huang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Metabolic Innovation Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
80
|
Xiao M, Zhou J, Zhang W, Ding Y, Guo J, Liang X, Zhu J, Jiao X, Zhai Z, Wang H. Association of immunosuppressive CD45 +CD33 +CD14 - CD10 -HLA-DR -/low neutrophils with poor prognosis in patients with lymphoma and their expansion and activation through STAT3/arginase-1 pathway in vitro. Cytojournal 2024; 21:69. [PMID: 39916996 PMCID: PMC11801647 DOI: 10.25259/cytojournal_165_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/13/2024] [Indexed: 02/09/2025] Open
Abstract
Objective This study aimed to explore the clinical significance of CD45+CD33+CD14-CD10-HLA-DR-/low neutrophils (Cluster of Differentiation 10 [CD10-] neutrophils) in B-cell non-Hodgkin's lymphoma (B-NHL). An amplification system of CD10- neutrophils in vitro was constructed using cytokines, and the mechanisms underlying the cytokine-induced expansion and activation of the CD10- neutrophil subpopulation were investigated. Material and Methods We identified a novel suppressive cell population known as CD10- neutrophils in the peripheral blood of patients with B-NHL in different statuses by flow cytometry and found it to be correlated with interleukin-6 levels, T cell counts, and plasma arginase-1 (Arg-1) levels. We then verified the effect of CD10- neutrophil expression on the prognosis of patients with B-NHL. Furthermore, we described a clinically compatible method for generating granulocyte populations rich in CD10- neutrophils using cultures of peripheral blood-isolated neutrophils supplemented with cytokines in vitro. Arg-1 expression was detected in neutrophils before and after induction by cytokines through reverse-transcription polymerase chain reaction, enzyme-linked immunosorbent assay, and flow cytometry. T-cell proliferation and apoptosis were measured by carboxyfluorescein succinimidyl ester assay and Annexin V-Propidium Iodide stains, and induced cells were exposed to Arg-1 inhibitor and ruxolitinib. signal transducer and activator of transcription 3 (STAT3)/Arg-1 signaling was studied mainly by western blot and chromatin immunoprecipitation experiments. Results We established a correlation between high CD10- neutrophil levels and poorer survival outcomes in patients with B-NHL. Moreover, CD10- neutrophils were positively correlated with interleukin (IL)-6, T-reg cells, and plasma Arg-1 levels and negatively correlated with the absolute number of total T cells. Granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte colony-stimulating factor, and IL-6 could all induce the expansion of CD10- neutrophil phenotype cells in vitro, which exhibit typical immature cellular morphology, and the combination of IL-6 and GM-CSF was the most effective. We confirmed that the STAT3/Arg-1 signaling pathway could be a critical mechanism regulating CD10- neutrophil-mediated immunosuppression in vitro. Conclusion CD10- neutrophils exhibited basic characteristics similar to conventional myeloid-derived suppressor cells. Our observations provide a promising STAT3 or Arg-1 targeting strategy for B-NHL and an important method for generating remarkably amounts of inhibitory granulocyte populations rich in CD10- neutrophils for immunotherapy.
Collapse
Affiliation(s)
- Meng Xiao
- Department of Hematology, Hematological Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui., China
- Department of Hematology, Jining NO.1 People’s Hospital, Jining, Shandong, China
| | - Ji Zhou
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- School of Nursing, Anhui Medical University, Hefei, Anhui, China
- Nursing International Collaboration Research Center of Anhui Province, Hefei, Anhui, China
| | - Wanqiu Zhang
- Department of Hematology, Hematological Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui., China
| | - Yangyang Ding
- Department of Hematology, Hematological Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui., China
| | - Jinjing Guo
- Department of Hematology, Hematological Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui., China
- Department of Laboratory, Fuyang People’s Hospital, Fuyang, China
| | - Xue Liang
- Department of Hematology, Hematological Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui., China
| | - Jinli Zhu
- Department of Hematology, Hematological Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui., China
| | - Xunyi Jiao
- Department of Hematology, Hematological Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui., China
| | - Zhimin Zhai
- Department of Hematology, Hematological Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui., China
| | - Huiping Wang
- Department of Hematology, Hematological Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui., China
| |
Collapse
|
81
|
Kim MS, Kang H, Baek JH, Cho MG, Chung EJ, Kim SJ, Chung JY, Chun KH. Disrupting Notch signaling related HES1 in myeloid cells reinvigorates antitumor T cell responses. Exp Hematol Oncol 2024; 13:122. [PMID: 39702544 DOI: 10.1186/s40164-024-00588-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) are immunosuppressive cells within the tumor microenvironment (TME) that hinder anti-tumor immunity. Notch signaling is a pathway crucial for TAM differentiation and function. Here, we investigate the role of HES1, a downstream target of Notch signaling, in TAM-mediated immunosuppression and explore its potential as a target for cancer immunotherapy. METHODS In this work, we constructed conditional Hes1 knockout mice to selectively delete Hes1 in TAMs. We further analyzed the TME composition, T cell infiltration and activation, and anti-tumor effects in these mice, both alone and in combination with PD-1 checkpoint blockade. RESULTS Our study showed that expression levels of Notch target Hes1 were increase in TAMs and mice with conditional knockout of Hes1 gene in TAMs exhibited decreased tumor growth, with increased infiltration and activation of cytotoxic T cells in tumors. Expression of tumor promoting factors was critically altered in Hes1-conditional KO TAMs, leading to the improved tumor microenvironment. Notably, arginase-1 expression was decreased in Hes1-conditional KO mice. Arg1 is known to deplete arginine and deactivate T cells in the TME. Administration of anti-PD-1 monoclonal antibody inhibited tumor growth to a greater extent in Hes1-conditional KO mice than in WT mice. CONCLUSIONS We identified a pivotal role for the Notch signaling pathway in shaping TAM function, suggesting that T-cell dysfunction in the TME is caused when the Notch target, HES1, in TAMs is upregulated by tumor-associated factors (TAFs), which, in turn, increases the expression of arginase-1. Targeting HES1 in TAMs appears to be a promising strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Myung Sup Kim
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyeokgu Kang
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jung-Hwan Baek
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Moon-Gyu Cho
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Eun Joo Chung
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Seok-Jun Kim
- Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju, 61452, Republic of Korea
- Institute of Well-Aging Medicare & Chosun University G-LAMP Project Group, Chosun University, Gwangju, 61452, Republic of Korea
| | - Joon-Yong Chung
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kyung-Hee Chun
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Institute for Bio-medical Convergence Science and Technology, Yonsei University, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Affiliate Faculty, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.
| |
Collapse
|
82
|
Esposito A, Agostini A, Quero G, Piro G, Priori L, Caggiano A, Scaglione G, Battaglia A, Calegari MA, Salvatore L, Bensi M, Maratta MG, Ceccarelli A, Trovato G, Genovese G, Gurreri E, Ascrizzi S, Martini M, Fiorillo C, Fattorossi A, De Sanctis F, Ugel S, Corbo V, Alfieri S, Tortora G, Carbone C. Colorectal cancer patients-derived immunity-organoid platform unveils cancer-specific tissue markers associated with immunotherapy resistance. Cell Death Dis 2024; 15:878. [PMID: 39632825 PMCID: PMC11618451 DOI: 10.1038/s41419-024-07266-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Colorectal cancer (CRC) is a devastating disease, ranking as the second leading cause of cancer-related deaths worldwide. Immune checkpoint inhibitors (ICIs) have emerged as promising treatments; however, their efficacy is largely restricted to a subgroup of microsatellite instable (MSI) CRCs. In contrast, microsatellite stable (MSS) CRCs, which account for the majority of cases, exhibit variable and generally weaker response to ICIs, with only a subset demonstrating exceptional responsiveness. Identifying novel cancer-specific tissue (CST) markers predictive of immunotherapy response is crucial for refining patient selection and overcoming treatment resistance. In this study, we developed clinically relevant CRC organoids and autologous immune system interaction platforms to model ICI response. We conducted a comprehensive molecular characterization of both responder and non-responder models, identifying CST markers that predict ICI response. Validation of these findings was performed using an independent cohort of patient specimens through multiplex immunofluorescence. Furthermore, we demonstrated that knocking out a key gene from the identified predictive signature in resistant organoids restored immune sensitivity and induced T-cell-mediated apoptosis. Overall, our results provide novel insights into the mechanisms underlying immunotherapy resistance and suggest new markers for enhancing patient selection. These findings may pave the way for new therapeutic options in MSS patients, potentially broadening the cohort of individuals eligible for immunotherapy.
Collapse
Affiliation(s)
- A Esposito
- Medical Oncology, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - A Agostini
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - G Quero
- Pancreatic Surgery Unit, Gemelli Pancreatic Advanced Research Center (CRMPG), Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Digestive Surgery Unit, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - G Piro
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.
| | - L Priori
- Medical Oncology, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - A Caggiano
- Medical Oncology, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - G Scaglione
- Department of Anatomic Pathology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - A Battaglia
- Department of Life Science and Public Health, Catholic University of the Sacred Heart, Rome, Italy
| | - M A Calegari
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - L Salvatore
- Medical Oncology, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - M Bensi
- Medical Oncology, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - M G Maratta
- Medical Oncology, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - A Ceccarelli
- Medical Oncology, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - G Trovato
- Medical Oncology, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - G Genovese
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - E Gurreri
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - S Ascrizzi
- Medical Oncology, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - M Martini
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, Messina, Italy
| | - C Fiorillo
- Pancreatic Surgery Unit, Gemelli Pancreatic Advanced Research Center (CRMPG), Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Digestive Surgery Unit, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - A Fattorossi
- Department of Life Science and Public Health, Catholic University of the Sacred Heart, Rome, Italy
| | - F De Sanctis
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - S Ugel
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - V Corbo
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - S Alfieri
- Pancreatic Surgery Unit, Gemelli Pancreatic Advanced Research Center (CRMPG), Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Digestive Surgery Unit, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - G Tortora
- Medical Oncology, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - C Carbone
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.
| |
Collapse
|
83
|
Falcone M, Salhia B, Halbert CH, Torres ETR, Stewart D, Stern MC, Lerman C. Impact of Structural Racism and Social Determinants of Health on Disparities in Breast Cancer Mortality. Cancer Res 2024; 84:3924-3935. [PMID: 39356624 PMCID: PMC11611670 DOI: 10.1158/0008-5472.can-24-1359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/24/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
The striking ethnic and racial disparities in breast cancer mortality are not explained fully by pathologic or clinical features. Structural racism contributes to adverse conditions that promote cancer inequities, but the pathways by which this occurs are not fully understood. Social determinants of health, such as economic status and access to care, account for a portion of this variability, yet interventions designed to mitigate these barriers have not consistently led to improved outcomes. Based on the current evidence from multiple disciplines, we describe a conceptual model in which structural racism and racial discrimination contribute to increased mortality risk in diverse groups of patients by promoting adverse social determinants of health that elevate exposure to environmental hazards and stress; these exposures in turn contribute to epigenetic and immune dysregulation, thereby altering breast cancer outcomes. Based on this model, opportunities and challenges arise for interventions to reduce racial and ethnic disparities in breast cancer mortality.
Collapse
Affiliation(s)
- Mary Falcone
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Bodour Salhia
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Chanita Hughes Halbert
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Evanthia T. Roussos Torres
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Medicine, Division of Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Daphne Stewart
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Medicine, Division of Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mariana C. Stern
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Caryn Lerman
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
84
|
Wang X, Tan B, Liu J, Wang J, Chen M, Yang Q, Zhang X, Li F, Wei Y, Wu K, Ren G, Li H. Echinacoside inhibits tumor immune evasion by downregulating inducible PD-L1 and reshaping tumor immune landscape in breast and colorectal cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156188. [PMID: 39488102 DOI: 10.1016/j.phymed.2024.156188] [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: 06/12/2024] [Revised: 10/01/2024] [Accepted: 10/24/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Targeting PD-L1 has become a crucial approach in tumor immunotherapy. Echinacoside (ECH) is a natural compound known for its extensive biological activities, its impact on antitumor immunity remains uncertain. PURPOSE This work was designed to assess the effects of ECH on the PD-L1/PD-1-mediated tumor immune evasion and its underlying mechanisms. METHODS Flow cytometry and RT-qPCR were utilized to explore the influence of ECH on PD-L1 expression. Western blot was employed to examine the mechanism by which ECH might modulate PD-L1 expression. Flow cytometry was conducted to evaluate the influence of ECH therapy, or the synergistic effects of ECH combined with immune checkpoint blockade (ICB) on tumor immune microenvironment (TIME) in tumor-burden mice. Blood biochemistry tests were used to evaluate the safety of ECH treatment. RESULTS ECH downregulated both the protein and mRNA expression levels of IFN-γ-induced PD-L1 through JAK/STAT1/IRF1 signaling pathway. ECH treatment upregulated the infiltration of IFN-γ+CD8+ T cells and Ki-67+CD8+ T cells, lowered the frequency of TIM-3+PD-1+ T cells, promoted the infiltration of effector CD4+ T cells and total CD8+ T cells while suppressed the percentage of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSC). Moreover, the combination of ECH and anti-PD-1 or anti-CTLA-4 therapy exhibited synergistic anti-tumor effects, reshaping TIME. Blood biochemistry tests unveiled that ECH did not show additional toxicity. CONCLUSION ECH upregulates the expression of inducible PD-L1 through the JAK/STAT1/IRF1 signaling pathway, enhances T cell function, and reshapes the tumor immune landscape into an anti-tumor phenotype. Importantly, ECH markedly enhances the efficacy of ICB treatment, indicating its potential application in anti-tumor therapy.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Binxin Tan
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Oncology, The Affiliated Banan's Hospital of Chongqing Medical University, Chongqing 401320, China
| | - Jiazhou Liu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jing Wang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Head, Neck and Breast Surgery, The First Affiliated Hospital of the University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei 230000, China
| | - Mingjing Chen
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qian Yang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiang Zhang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fan Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yuxian Wei
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ke Wu
- Department of Pharmacy, The Affiliated Dazu's Hospital of Chongqing Medical University, Chongqing 402360, China.
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Hongzhong Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
85
|
Liu QQ, Li HZ, Li SX, Bao Y, Wang TC, Hu C, Xiao YD. CD36-mediated accumulation of MDSCs exerts abscopal immunosuppressive responses in hepatocellular carcinoma after insufficient microwave ablation. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167493. [PMID: 39233261 DOI: 10.1016/j.bbadis.2024.167493] [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: 03/21/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
The immune landscape of distant unablated tumors following insufficient microwave ablation (iMWA) in hepatocellular carcinoma (HCC) remains to be clarified. The objective of this study is to define the abscopal immune landscape in distant unablated tumor before and after iMWA for HCC. Two treatment-naive patients were recruited for tumor tissue sampling, of each with two HCC lesions. Tumor samples were obtained at before and after microwave ablation in distant unablated sites for single-cell RNA sequencing (scRNA-seq). Mouse model with bilateral hepatoma tumors were developed, and distant unablated tumors were analyzed using multicolor immunofluorescence, RNA sequencing and flow cytometry. The scRNA-seq revealed that a reduced proportion of CD8+ T cells and an increased proportion of myeloid-derived suppressor cells (MDSCs) were observed in the distant unablated tumor microenvironment (TME). A notable disruption was observed in the lipid metabolism of tumor-associated immune cells, accompanied by an upregulated expression of CD36 in tumor-infiltrating immune cells in distant unablated tumor. The administration of a CD36 inhibitor has been demonstrated to ameliorate the adverse effects induced by iMWA, primarily by reinstating the anti-tumor responses of T cells in distant unablated tumor. These findings explain the recurrence and progression of tumors after iMWA and provide a new target of immunotherapy for HCC.
Collapse
Affiliation(s)
- Qing-Qing Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Hui-Zhou Li
- Department of Radiology, the Second Xiangya Hospital, Central South University, Changsha 410011, China; Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China.
| | - Shu-Xian Li
- Department of Radiology, the Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | - Yan Bao
- Department of Radiology, the Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | - Tian-Cheng Wang
- Department of Radiology, the Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | - Chao Hu
- Department of Radiology, the Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | - Yu-Dong Xiao
- Department of Radiology, the Second Xiangya Hospital, Central South University, Changsha 410011, China.
| |
Collapse
|
86
|
Green KA, Ma C, Hoffmann FW, Hoffmann PR, Green WR. Depletion of monocytic myeloid-derived suppressor cells in LP-BM5 murine retroviral infection has a positive impact on virus-induced host immunodeficiency. Virology 2024; 600:110247. [PMID: 39307098 PMCID: PMC11560480 DOI: 10.1016/j.virol.2024.110247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/29/2024] [Accepted: 09/16/2024] [Indexed: 11/12/2024]
Abstract
We have shown the induction of CD11b+Ly6C+ monocytic myeloid-derived suppressor cells (M-MDSCs) during infection of B6 mice by LP-BM5 immunodeficiency-inducing retrovirus. We published that the molecular mechanisms of these M-MDSCs vary, and depend on the cell type targeted by the suppression -defined by use of biochemical inhibitors, mouse M-MDSCs knock-out strains and blocking antibodies. These M-MDSCs suppressed proliferation and function of T cells, via nitric oxide synthase/nitric oxide; and that of B cells, ∼50% via INOS/NO along with the negative checkpoint regulator VISTA, reactive nitrogen and oxygen species, and other soluble mediators. Here, LP-BM5 infected mice were treated weekly with 5-Fluorouracil (5-FU), resulting in depletion of peripheral blood and splenic M-MDSCs, reduced MDSC activity, and significantly decreased standard disease parameters of: splenomegaly, impaired B-and T-cell ex vivo polyclonal responses, and viral load. In addition, 5-FU treatment significantly increased percentages of CD4+ and CD8+ T cells.
Collapse
Affiliation(s)
- Kathy A Green
- Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, USA.
| | - Chi Ma
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96813, USA
| | - Fukun W Hoffmann
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96813, USA
| | - Peter R Hoffmann
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96813, USA
| | - William R Green
- Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, USA
| |
Collapse
|
87
|
Lv Y, Zhu J, Ge S, Jiang T, Xu Y, Yao W, Jiang C. The AXL-mediated modulation of myeloid-derived suppressor cells (MDSC) in nasopharyngeal carcinoma. Med Oncol 2024; 42:17. [PMID: 39592496 DOI: 10.1007/s12032-024-02561-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024]
Abstract
AXL has ubiquitous expression in multiple cancers, and is strongly linked to both tumor progression, metastasis, and poor prognosis, as well as anti-tumor immune response suppression and induction of tumor resistance to immunotherapy. Therefore, it is a strong target for cancer intervention. Despite the wide application of AXL inhibitors in clinical trials, the role of AXL in the tumor immune microenvironment (TIME) remains undetermined. Herein, we established cell lines with stable AXL knockdown or overexpression using lentiviral infection. Subsequently, we co-cultured the cells with healthy human blood-derived CD33 + PBMCs. After two days of culture, we evaluated the differentiation of PBMCs into MDSCs. Additionally, the culture supernatants were collected from both the co-culture system and the individual cultures of each cell group to measure the concentrations of IL-6 and GM-CSF. Additionally, we subcutaneously administered nasopharyngeal carcinoma (NPC) cells into mice, and evaluated the association between AXL content and MDSC recruitment in the resulting tumors. We demonstrated that AXL is a critical modulator of MDSC differentiation and accumulation in NPC. It modulates IL-6, GM-CSF, and Toll-like receptor contents to achieve the aforementioned actions. Herein, we revealed a strong and direct link between AXL, cytokines in TIME, and MDSC differentiation and accumulation. Our work highlights novel approaches to optimizing existing immunotherapeutic interventions.
Collapse
Affiliation(s)
- Yu Lv
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Jiahui Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Sichen Ge
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Tao Jiang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Yajia Xu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Weige Yao
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Chengyi Jiang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China.
| |
Collapse
|
88
|
Shen Y, Zhong B, Zheng W, Wang D, Chen L, Song H, Pan X, Mo S, Jin B, Cui H, Zhan H, Luo F, Liu J. Rg3-lipo biomimetic delivery of paclitaxel enhances targeting of tumors and myeloid-derived suppressor cells. J Clin Invest 2024; 134:e178617. [PMID: 39545407 PMCID: PMC11563678 DOI: 10.1172/jci178617] [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: 12/20/2023] [Accepted: 09/25/2024] [Indexed: 11/17/2024] Open
Abstract
Liposomal drug delivery systems have revolutionized traditional cytotoxic drugs. However, the relative instability and toxicity of the existing liposomal drug delivery systems compromised their efficacy. Herein, we present Rg3-lipo, an innovative drug delivery system using a glycosyl moiety-enriched ginsenoside (Rg3). This system is distinguished by its glycosyl moieties exposed on the liposomal surface. These moieties imitate human cell membranes to stabilize and evade phagocytic clearance. The Rg3-lipo system loaded with paclitaxel (PTX-Rg3-lipo) demonstrated favorable bioavailability and safety in Sprague-Dawley rats, beagle dogs, and cynomolgus monkeys. With its glycosyl moieties recognizing tumor cells via the glucose transporter Glut1, PTX-Rg3-lipo inhibited gastric, breast, and esophageal cancers in human cancer cell lines, tumor-bearing mice, and patient-derived xenograft models. These glycosyl moieties selectively targeted myeloid-derived suppressor cells (MDSCs) through the glucose transporter Glut3 to attenuate their immunosuppressive effect. The mechanism study revealed that Rg3-lipo suppressed glycolysis and downregulated the transcription factors c-Maf and Mafb overcoming the MDSC-mediated immunosuppressive microenvironment and enhancing PTX-Rg3-lipo's antitumor effect. Taken together, we supply substantial evidence for its advantageous bioavailability and safety in multiple animal models, including nonhuman primates, and Rg3-lipo's dual targeting of cancer cells and MDSCs. Further investigation regarding Rg3-lipo's druggability will be conducted in clinical trials.
Collapse
Affiliation(s)
- Yuru Shen
- Department of Digestive Diseases and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Bin Zhong
- Department of Digestive Diseases and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Biotherapy Research Center, Fudan University, Shanghai, China
| | - Wanwei Zheng
- Department of Digestive Diseases and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Dan Wang
- Xiamen Ginposome Pharmaceutical Co. Ltd., Xiamen, China
| | - Lin Chen
- Department of Digestive Diseases and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Huan Song
- Department of Digestive Diseases and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xuanxuan Pan
- Department of Digestive Diseases and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Shaocong Mo
- Department of Digestive Diseases and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Bryan Jin
- Department of Digestive Diseases and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Haoshu Cui
- Department of Digestive Diseases and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Huaxing Zhan
- Xiamen Ginposome Pharmaceutical Co. Ltd., Xiamen, China
| | - Feifei Luo
- Department of Digestive Diseases and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jie Liu
- Department of Digestive Diseases and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
89
|
Qiu Z, Fan J, He J, Huang X, Yang Z, Sheng Q, Jin L. Causal relationship between cancer and immune cell traits: A two-sample mendelian randomization study. Heliyon 2024; 10:e39732. [PMID: 39583800 PMCID: PMC11582454 DOI: 10.1016/j.heliyon.2024.e39732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/26/2024] Open
Abstract
Background Observational studies provide evidence of correlations between cancer and the immune system. Previous research has established associations between immune traits and the propensity for developing certain cancers. However, a systematic exploration of these connections remains largely uncharted. Therefore, further investigation is needed to examine the causal association between cancer and immune cell traits using Mendelian randomization (MR) approach. Methods We identified genetic instruments for breast cancer (BC), lung cancer (LC), endometrial cancer (EC), ovarian cancer (OC), prostate cancer (PC), and their subtype cancers to investigate their potential causal impact on immune traits. Data on cancer and immune cell traits were obtained from the IEU Open GWAS project. To assess whether these five cancer types and subtype cancers have a causal association with immune cell traits, we conducted two-sample MR analyses. Additionally, we conducted bidirectional MR analyses to examine the direction of causal relationships and adjusted for potentially related pleiotropy through multivariable MR analysis. Results We have identified several causal relationships between different types of cancer and immune traits. We found that breast cancer may influence 49 immune cell traits, endometrial cancer may influence 38, lung cancer may influence 25, ovarian cancer may influence 19, and prostate cancer may influence 28. Among these, breast cancer and lung cancer were associated with four common immune traits: CD25 on IgD- CD38dim, CD25 on sw mem, CD24 on IgD- CD38-, and CD25 on IgD- CD38-. Lung cancer and prostate cancer shared four immune traits: CD25 on IgD+ CD24+, CD25 on IgD+ CD38-, CD66b on CD66b++ myeloid cell, DN (CD4-CD8-) AC. Endometrial cancer and ovarian cancer shared two immune traits: TD DN (CD4-CD8-) %DN, EM DN (CD4-CD8-) %DN. Breast cancer and endometrial cancer shared one immune trait: CD20 on IgD- CD38dim. Endometrial cancer and prostate cancer shared one immune trait: CCR2 on myeloid DC. Lastly, breast cancer, lung cancer, and prostate cancer shared one immune trait: CD25 on CD24+ CD27+. Additionally, we identified specific immune traits that may serve as protective or risk factors for cancers. We found 14 immune traits may influence breast cancer, 9 immune traits may influence endometrial cancer, 22 immune traits may influence lung cancer, 9 immune traits may influence ovarian cancer, and 14 immune traits may influence prostate cancer. Among these, breast cancer and prostate cancer shared three immune traits: HLA DR++ monocyte %monocyte, HLA DR on plasmacytoid DC, and HLA DR on DC. Lung cancer and ovarian cancer shared one immune trait: CD62L- monocyte %monocyte. Prostate cancer and endometrial cancer shared one immune trait: HLA DR on CD33dim HLA DR + CD11b+. Lastly, ovarian cancer and prostate cancer shared one immune trait: CD3 on resting Treg. Conclusions Our MR study suggests a potential relationship between immune traits and cancers, particularly highlighting 14 immune traits that are simultaneously influenced by two or three of five cancer types, while also indicating that 6 immune traits may simultaneously contribute to the development of two of the cancers. This elucidation enables us to reveal a significant involvement of immune traits in cancer progression, providing critical insights into how immune traits affect cancer susceptibility.
Collapse
Affiliation(s)
- Zejing Qiu
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Jingjing Fan
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Jun He
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Xingxing Huang
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Zuyi Yang
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Qinsong Sheng
- Department of Colorectal Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lijun Jin
- Department of Traditional Chinese Medicine, Hangzhou Shangcheng District People's Hospital, Hangzhou, China
| |
Collapse
|
90
|
Zhu C, Liao JY, Liu YY, Chen ZY, Chang RZ, Chen XP, Zhang BX, Liang JN. Immune dynamics shaping pre-metastatic and metastatic niches in liver metastases: from molecular mechanisms to therapeutic strategies. Mol Cancer 2024; 23:254. [PMID: 39543660 PMCID: PMC11562679 DOI: 10.1186/s12943-024-02171-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
Liver metastases are commonly detected in the advanced stages of various malignant tumors, representing a significant clinical challenge. Throughout the process of liver metastases formation, immune cells play a pivotal role, particularly in the pre-metastatic and metastatic niches within the liver. Immune cells establish extensive and intricate interactions with tumor cells and other components in the liver, collectively promoting and sustaining the growth of liver metastases. Despite the limited efficacy of existing therapeutic modalities against some advanced liver metastases, novel immune-based treatment approaches are continuously being explored and validated. Building on the systematic elucidation of the immunosuppressive characteristics of liver metastases, we explored the potential of novel immunotherapies applicable to patients with liver metastases from multiple dimensions.
Collapse
Affiliation(s)
- Chang Zhu
- Hepatic Surgery Center, and Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Jing-Yu Liao
- Hepatic Surgery Center, and Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Yi-Yang Liu
- Hepatic Surgery Center, and Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Ze-Yu Chen
- Hepatic Surgery Center, and Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Rui-Zhi Chang
- Hepatic Surgery Center, and Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Xiao-Ping Chen
- Hepatic Surgery Center, and Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China
| | - Bi-Xiang Zhang
- Hepatic Surgery Center, and Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China.
| | - Jun-Nan Liang
- Hepatic Surgery Center, and Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, P. R. China.
| |
Collapse
|
91
|
Donaubauer AJ, Scheer I, Fietkau R, Gaipl US, Frey B. Flow cytometry-based monitoring of myeloid-derived suppressor cells in the peripheral blood of patients with solid tumors. Methods Cell Biol 2024; 191:135-150. [PMID: 39824553 DOI: 10.1016/bs.mcb.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2025]
Abstract
Myeloid-derived suppressor cells (MDSCs) ameliorate inflammation by inhibiting T cell responses. In pathological conditions, such as autoimmunity, chronic infections or cancer they accumulate in the periphery. In cancer, MDSCs can also be part of the tumor microenvironment and are associated with a worse prognosis and limited response to immunotherapy. Nowadays attempts are made to specifically target MDSCs in cancer therapy. Still, the role of MDSCs in standard cancer treatment modalities, such as radiotherapy remains mostly elusive. Here, we describe a flow cytometry-based method to determine and monitor monocytic and granulocytic-derived MDSCs directly from whole blood in an easy, fast and reliable assay. As specific surface markers for MDSCs are lacking, the assay follows a gating strategy that excludes successively the main immune cells types and analyzes the remaining events for a set of molecules that are expressed on MDSCs. This assay is especially appropriate for longitudinal analyses and clinical trials and is suitable for being integrated into more complex immunophenotyping panels to generate a comprehensive immune status.
Collapse
Affiliation(s)
- Anna-Jasmina Donaubauer
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Ilka Scheer
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany; FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, Erlangen, Germany
| | - Udo S Gaipl
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany; FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, Erlangen, Germany.
| | - Benjamin Frey
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany; FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, Erlangen, Germany
| |
Collapse
|
92
|
Santerre JP, Yang Y, Du Z, Wang W, Zhang X. Biomaterials' enhancement of immunotherapy for breast cancer by targeting functional cells in the tumor micro-environment. Front Immunol 2024; 15:1492323. [PMID: 39600709 PMCID: PMC11588700 DOI: 10.3389/fimmu.2024.1492323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Immunotherapy for breast cancer is now being considered clinically, and more recently, the number of investigations aimed specifically at nano-biomaterials-assisted immunotherapy for breast cancer treatment is growing. Alterations of the breast cancer micro-environment can play a critical role in anti-tumor immunity and cancer development, progression and metastasis. The improvement and rearrangement of tumor micro-environment (TME) may enhance the permeability of anti-tumor drugs. Therefore, targeting the TME is also an ideal and promising option during the selection of effective nano-biomaterial-based immuno-therapeutic strategies excepted for targeting intrinsic resistant mechanisms of the breast tumor. Although nano-biomaterials designed to specifically release loaded anti-tumor drugs in response to tumor hypoxia and low pH conditions have shown promises and the diversity of the TME components also supports a broad targeting potential for anti-tumor drug designs, yet the applications of nano-biomaterials for targeting immunosuppressive cells/immune cells in the TME for improving the breast cancer treating outcomes, have scarcely been addressed in a scientific review. This review provides a thorough discussion for the application of the different forms of nano-biomaterials, as carrier vehicles for breast cancer immunotherapy, targeting specific types of immune cells in the breast tumor microenvironment. In parallel, the paper provides a critical analysis of current advances/challenges with leading nano-biomaterial-mediated breast cancer immunotherapeutic strategies. The current review is timely and important to the cancer research field and will provide a critical tool for nano-biomaterial design and research groups pushing the clinical translation of new nano-biomaterial-based immuno-strategies targeting breast cancer TME, to further open new avenues for the understanding, prevention, diagnosis and treatment of breast cancer, as well as other cancer types.
Collapse
Affiliation(s)
- J. Paul Santerre
- The School of Basic Medicine, Binzhou Medical University, Yantai, Shandong, China
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Yangyang Yang
- The School of Basic Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Ziwei Du
- The School of Basic Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Wenshuang Wang
- Department of Gynecology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Xiaoqing Zhang
- The School of Basic Medicine, Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
93
|
Jangid AK, Kim K. Phenylboronic acid-functionalized biomaterials for improved cancer immunotherapy via sialic acid targeting. Adv Colloid Interface Sci 2024; 333:103301. [PMID: 39260104 DOI: 10.1016/j.cis.2024.103301] [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: 03/16/2024] [Revised: 06/16/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
Phenylboronic acid (PBA) is recognized as one of the most promising cancer cell binding modules attributed to its potential to form reversible and dynamic boronic ester covalent bonds. Exploring the advanced chemical versatility of PBA is crucial for developing new anticancer therapeutics. The presence of a specific Lewis acidic boron atom-based functional group and a Π-ring-connected ring has garnered increasing interest in the field of cancer immunotherapy. PBA-derivatized functional biomaterials can form reversible bonds with diols containing cell surface markers and proteins. This review primarily focuses on the following topics: (1) the importance and versatility of PBA, (2) different PBA derivatives with pKa values, (3) specific key features of PBA-mediated biomaterials, and (4) cell surface activity for cancer immunotherapy applications. Specific key features of PBA-mediated materials, including sensing, bioadhesion, and gelation, along with important synthesis strategies, are highlighted. The utilization of PBA-mediated biomaterials for cancer immunotherapy, especially the role of PBA-based nanoparticles and PBA-mediated cell-based therapeutics, is also discussed. Finally, a perspective on future research based on PBA-biomaterials for immunotherapy applications is presented.
Collapse
Affiliation(s)
- Ashok Kumar Jangid
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul, South Korea
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul, South Korea.
| |
Collapse
|
94
|
Vijayan Y, James S, Viswanathan A, Aparna JS, Bindu A, Namitha NN, Anantharaman D, Babu Lankadasari M, Harikumar KB. Targeting acid ceramidase enhances antitumor immune response in colorectal cancer. J Adv Res 2024; 65:73-87. [PMID: 38142035 PMCID: PMC11518951 DOI: 10.1016/j.jare.2023.12.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 12/25/2023] Open
Abstract
INTRODUCTION Acid ceramidase (hereafter referred as ASAH1) is an enzyme in sphingolipid metabolism that converts pro-survival ceramide into sphingosine. ASAH1 has been shown to be overexpressed in certain cancers. However, the role of ASAH1 in colorectal cancer still remain elusive. OBJECTIVE The present study is aimed to understand how ASAH1 regulates colorectal cancer (CRC) progression and resistance to checkpoint inhibitor therapy. METHODS Both pharmacological and genetic silencing of ASAH1 was used in the study. In vitro experiments were done on human and mouse CRC cell lines. The in vivo studies were conducted in NOD-SCID and BALB/c mice models. The combination of ASAH1 inhibitor and checkpoint inhibitor was tested using a syngeneic tumor model of CRC. Transcriptomic and metabolomic analyses were done to understand the effect of ASAH1 silencing. RESULTS ASAH1 is overexpressed in human CRC cases, and silencing the expression resulted in the induction of immunological cell death (ICD) and mitochondrial stress. The ASAH1 inhibitor (LCL-521), either as monotherapy or in combination with an anti-PD-1 antibody, resulted in reduction of tumors and, through induction of type I and II interferon response, activation of M1 macrophages and T cells, leading to enhanced infiltration of cytotoxic T cells. Our findings supported that the combination of LCL-521 and ICIs, which enhances the antitumor responses, and ASAH1 can be a druggable target in CRC.
Collapse
Affiliation(s)
- Yadu Vijayan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India; Manipal Academy of Higher Education (MAHE), Manipal, 576104, India
| | - Shirley James
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Arun Viswanathan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India; Manipal Academy of Higher Education (MAHE), Manipal, 576104, India
| | - Jayasekharan S Aparna
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Anu Bindu
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Narayanan N Namitha
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Devasena Anantharaman
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Manendra Babu Lankadasari
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Kuzhuvelil B Harikumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India.
| |
Collapse
|
95
|
Wang S, Zhang Z, Wang J, Lou Y, Zhu Y, You J, Liu P, Xu LX. Neutrophils promote the activation of monocytes via ROS to boost systemic antitumor immunity after cryo-thermal therapy. Front Immunol 2024; 15:1445513. [PMID: 39555061 PMCID: PMC11563809 DOI: 10.3389/fimmu.2024.1445513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 10/14/2024] [Indexed: 11/19/2024] Open
Abstract
Background The characteristics of the tumor immunosuppressive microenvironment represent a major challenge that limits the efficacy of immunotherapy. Our previous results suggested that cryo-thermal therapy, a tumor ablation system developed in our laboratory, promotes macrophage M1-type polarization and the complete maturation of DCs to remodel the immunosuppressive environment. However, the cells that respond promptly to CTT have not yet been identified. CTT can cause extensive cell death and the release of danger-associated molecular patterns and antigens. Neutrophils are the first white blood cells recruited to sites of damage and acute inflammation. Therefore, we hypothesized that neutrophils are the initial cells that respond to CTT and are involved in the subsequent establishment of antitumor immunity. Methods In this study, we examined the kinetics of neutrophil recruitment after CTT via flow cytometry and immunofluorescence staining and explored the effect of neutrophils on the establishment of systemic antitumor immunity by in vivo neutrophil depletion and in vitro co-culture assays. Results We found that CTT led to a rapid and strong proinflammatory neutrophil response, which was essential for the long-term survival of mice. CTT-induced neutrophils promoted the activation of monocytes via reactive oxygen species and further upregulated the expression of IFN-γ and cytotoxic molecules in T and NK cells. Adoptive neutrophil transfer further enhanced the antitumor efficacy of CTT in tumor models of spontaneous and experimental metastasis. Conclusion These results reveal the important role of neutrophil‒monocyte interactions in the development of anti-tumor immunity and highlight that CTT could be used as an immunotherapy for targeting neutrophils and monocytes to enhance antitumor immunity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ping Liu
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Lisa X. Xu
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
96
|
Matsuoka T, Yashiro M. Molecular Mechanism for Malignant Progression of Gastric Cancer Within the Tumor Microenvironment. Int J Mol Sci 2024; 25:11735. [PMID: 39519285 PMCID: PMC11546171 DOI: 10.3390/ijms252111735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/23/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Gastric cancer (GC) is one of the most common cancers worldwide. Most patients are diagnosed at the progressive stage of GC, and progress in the development of effective anti-GC drugs has been insufficient. The tumor microenvironment (TME) regulates various functions of tumor cells, and interactions between the cellular and molecular components of the TME-e.g., inflammatory cells, fibroblasts, vasculature cells, and innate and adaptive immune cells-promote the aggressiveness of cancer cells and dissemination to distant organs. This review summarizes the roles of various TME cells and molecules in regulating the malignant progression and metastasis of GC. We also address the important roles of signaling pathways in mediating the interaction between cancer cells and the different components of the GC TME. Finally, we discuss the implications of these molecular mechanisms for developing novel and effective therapies targeting molecular and cellular components of the GC TME to control the malignant progression of GC.
Collapse
Affiliation(s)
- Tasuku Matsuoka
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan;
- Institute of Medical Genetics, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan
| | - Masakazu Yashiro
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan;
- Institute of Medical Genetics, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan
| |
Collapse
|
97
|
Xiao J, Guo X, Lin Y, Wang Z. The causal relationship between immune cell-mediated gut microbiota and ulcerative colitis: a bidirectional two-sample, mediation Mendelian randomization analysis. Front Nutr 2024; 11:1433545. [PMID: 39525506 PMCID: PMC11545678 DOI: 10.3389/fnut.2024.1433545] [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: 05/16/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Background Numerous studies have highlighted the close association between gut microbiota and the development of ulcerative colitis (UC), yet research on whether immune cells mediate this process remains scarce. This study utilizes various Mendelian randomization (MR) methods to investigate the causal relationship between gut microbiota and UC, further exploring the mediating role of immune cells in this process. Methods Genome-wide association study (GWAS) summary statistics for 473 gut microbiota, 731 immune cell phenotypes, and UC were obtained from the GWAS catalog database. Single nucleotide polymorphisms (SNP) were used as instrumental variables (IV) to validate the causal relationship between gut microbiota and UC through two-sample MR and Bayesian weighted MR (BWMR), and reverse MR was employed to explore the presence of reverse causal effects. Two-step MR was applied to identify immune cell mediators and evaluate their mediation effects. Results The study revealed a causal relationship between 20 gut microbiota and UC, with 14 microbiota acting as protective factors for UC and 6 as risk factors. Mediation MR identified 26 immune cell mediators, among which the association between CD11b on Mo MDSC and Bifidobacterium bifidum (B. bifidum) was most significant (p = 0.0017, OR = 1.4540, 95% CI: 1.1504-1.8378). Mediation MR analysis indicated that the mediation effect of CD11b on Mo MDSC between B. bifidum and UC was -0.0385, with a mediation effect ratio of 16.67%. Conclusion There is a clear causal relationship between certain gut microbiota and UC, and CD11b on Mo MDSC is a significant mediator between B. bifidum and UC, providing new insights for the clinical treatment of UC.
Collapse
Affiliation(s)
- Jinyin Xiao
- Department of Anorectal, The Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, China
- Graduate School, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Xiajun Guo
- Department of Geriatric, The First People’s Hospital of Xiangtan City, Xiangtan, China
| | - Youwei Lin
- Graduate School, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Zhenquan Wang
- Department of Anorectal, The Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, China
| |
Collapse
|
98
|
Cao Y, Yi Y, Han C, Shi B. NF-κB signaling pathway in tumor microenvironment. Front Immunol 2024; 15:1476030. [PMID: 39493763 PMCID: PMC11530992 DOI: 10.3389/fimmu.2024.1476030] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
The genesis and progression of tumors are multifaceted processes influenced by genetic mutations within the tumor cells and the dynamic interplay with their surrounding milieu, which incessantly impacts the course of cancer. The tumor microenvironment (TME) is a complex and dynamic entity that encompasses not only the tumor cells but also an array of non-cancerous cells, signaling molecules, and the extracellular matrix. This intricate network is crucial in tumor progression, metastasis, and response to treatments. The TME is populated by diverse cell types, including immune cells, fibroblasts, endothelial cells, alongside cytokines and growth factors, all of which play roles in either suppressing or fostering tumor growth. Grasping the nuances of the interactions within the TME is vital for the advancement of targeted cancer therapies. Consequently, a thorough understanding of the alterations of TME and the identification of upstream regulatory targets have emerged as a research priority. NF-κB transcription factors, central to inflammation and innate immunity, are increasingly recognized for their significant role in cancer onset and progression. This review emphasizes the crucial influence of the NF-κB signaling pathway within the TME, underscoring its roles in the development and advancement of cancer. By examining the interactions between NF-κB and various components of the TME, targeting the NF-κB pathway appears as a promising cancer treatment approach.
Collapse
Affiliation(s)
- Yaning Cao
- Department of Blood Transfusion, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu, China
| | - Yanan Yi
- Department of Laboratory Medicine, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Chongxu Han
- Department of Laboratory Medicine, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Bingwei Shi
- Department of Blood Transfusion, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu, China
| |
Collapse
|
99
|
Cai W, Sun T, Qiu C, Sheng H, Chen R, Xie C, Kou L, Yao Q. Stable triangle: nanomedicine-based synergistic application of phototherapy and immunotherapy for tumor treatment. J Nanobiotechnology 2024; 22:635. [PMID: 39420366 PMCID: PMC11488210 DOI: 10.1186/s12951-024-02925-3] [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: 10/03/2023] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
In recent decades, cancer has posed a challenging obstacle that humans strive to overcome. While phototherapy and immunotherapy are two emerging therapies compared to traditional methods, they each have their advantages and limitations. These limitations include easy metastasis and recurrence, low response rates, and strong side effects. To address these issues, researchers have increasingly focused on combining these two therapies by utilizing a nano-drug delivery system due to its superior targeting effect and high drug loading rate, yielding remarkable results. The combination therapy demonstrates enhanced response efficiency and effectiveness, leading to a preparation that is highly targeted, responsive, and with low recurrence rates. This paper reviews several main mechanisms of anti-tumor effects observed in combination therapy based on the nano-drug delivery system over the last five years. Furthermore, the challenges and future prospects of this combination therapy are also discussed.
Collapse
Affiliation(s)
- Wenjing Cai
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325027, China
| | - Tuyue Sun
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Chenyu Qiu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325027, China
| | - Huixiang Sheng
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325027, China
| | - Congying Xie
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, Wenzhou, 325000, China.
- Zhejiang-Hong Kong Precision Theranostics of Thoracic Tumors Joint Laboratory, Wenzhou, 325000, China.
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325027, China.
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, Wenzhou, 325000, China.
- Zhejiang-Hong Kong Precision Theranostics of Thoracic Tumors Joint Laboratory, Wenzhou, 325000, China.
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
100
|
Wang L, Zhu Y, Zhang N, Xian Y, Tang Y, Ye J, Reza F, He G, Wen X, Jiang X. The multiple roles of interferon regulatory factor family in health and disease. Signal Transduct Target Ther 2024; 9:282. [PMID: 39384770 PMCID: PMC11486635 DOI: 10.1038/s41392-024-01980-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/12/2024] [Accepted: 09/10/2024] [Indexed: 10/11/2024] Open
Abstract
Interferon Regulatory Factors (IRFs), a family of transcription factors, profoundly influence the immune system, impacting both physiological and pathological processes. This review explores the diverse functions of nine mammalian IRF members, each featuring conserved domains essential for interactions with other transcription factors and cofactors. These interactions allow IRFs to modulate a broad spectrum of physiological processes, encompassing host defense, immune response, and cell development. Conversely, their pivotal role in immune regulation implicates them in the pathophysiology of various diseases, such as infectious diseases, autoimmune disorders, metabolic diseases, and cancers. In this context, IRFs display a dichotomous nature, functioning as both tumor suppressors and promoters, contingent upon the specific disease milieu. Post-translational modifications of IRFs, including phosphorylation and ubiquitination, play a crucial role in modulating their function, stability, and activation. As prospective biomarkers and therapeutic targets, IRFs present promising opportunities for disease intervention. Further research is needed to elucidate the precise mechanisms governing IRF regulation, potentially pioneering innovative therapeutic strategies, particularly in cancer treatment, where the equilibrium of IRF activities is of paramount importance.
Collapse
Affiliation(s)
- Lian Wang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanghui Zhu
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yali Xian
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yu Tang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Ye
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fekrazad Reza
- Radiation Sciences Research Center, Laser Research Center in Medical Sciences, AJA University of Medical Sciences, Tehran, Iran
- International Network for Photo Medicine and Photo Dynamic Therapy (INPMPDT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Gu He
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiang Wen
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Xian Jiang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|