1
|
Hsu CY, Chandramoorthy HC, Mohammed JS, Al-Hasnaawei S, Yaqob M, Kundlas M, Samikan K, Sahoo S, Sunori SK, Abbas ZA. Exosomes as key mediators in immune and cancer cell interactions: insights in melanoma progression and therapy. Arch Dermatol Res 2025; 317:729. [PMID: 40252131 DOI: 10.1007/s00403-025-04237-4] [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: 02/27/2025] [Revised: 03/25/2025] [Accepted: 04/02/2025] [Indexed: 04/21/2025]
Abstract
Exosomes (30-150 nm) are small extracellular vesicles that are secreted by cells into the extracellular environment and are known to mediate cell-to-cell communication. Exosomes contain proteins, lipids, and RNA molecules in relative abundance, capable of modifying the activity of target cells. Melanoma-derived exosomes (MEXs) promote the transfer of oncogenic signals and immunosuppressive factors into immune cells, resulting in a bias of the immune response towards tumor-promoting processes. MEXs could suppress the activation and proliferation of T cells and dendritic cells and induce differentiation of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs). They can induce apoptosis of antigen-specific CD8 + T cells and promote the transfer of tumor antigens, resulting in immune evasion. Specifically, MEXs can shuttle cytokines like interleukin-10 (IL-10) and transforming growth factor-β (TGF-β) to immune cells or express programmed death-ligand 1 (PD-L1 or CD274), creating an immune-suppressive microenvironment that promotes tumorigenesis. Since exosomes preferentially accumulate in melanoma tissues, this targeted delivery could enhance the bioavailability of treatments while limiting side effects. Here, we review the molecular composition of melanoma-derived exosomes, their mechanisms of action, and their potential as therapeutic targets or biomarkers in melanoma. The summarizations of these mechanisms to appropriately influence exosome-mediated interactions could yield new tactics to elicit anti-melanoma immunity or augment the therapeutic effects of current therapies.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University, Tempe Campus, Phoenix, AZ, 85004, USA.
| | - Harish C Chandramoorthy
- Department of Microbiology and Clinical Parasitology, College of Medicine and Central Research Laboratories, King Khalid University, Abha, Saudi Arabia
| | | | - Shaker Al-Hasnaawei
- College of Pharmacy, the Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Mohammed Yaqob
- Department of Biology, Mazaya University College, Dhiqar, Iraq
| | - Mayank Kundlas
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Krishnakumar Samikan
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Samir Sahoo
- Department of General Medicine, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - S K Sunori
- Graphic Era Hill University, Bhimtal, Uttarakhand, India
- Graphic Era Deemed to be University, Dehradun, Uttarakhand, 248002, India
| | - Zainab Ahmed Abbas
- College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| |
Collapse
|
2
|
Wang ZL, Qiu SY, Sun YQ, Du XJ, Xu CF, Cao ZY, Lu ZD. An injectable oncolytic hydrogel platform for in situ dendritic cell vaccination to boost antitumor immunity. Biomater Sci 2025. [PMID: 40243662 DOI: 10.1039/d5bm00284b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Dendritic cell (DC) vaccines hold significant promise in cancer therapy due to their ability to induce durable anti-tumor immune responses. However, traditional ex vivo DC vaccines face considerable challenges, including complex preparation, limited DC persistence post-reinfusion, and variable efficacy. To overcome these limitations, we developed an injectable thermosensitive hydrogel (LC-Gel) that incorporates the oncolytic peptide LTX-315 and the chemokine CCL21 to generate in situ DC vaccines aimed at enhancing anti-tumor immunity. Our findings show that LC-Gel facilitates the intratumoral release of LTX-315, triggering the immunogenic cell death (ICD) of tumor cells and exposing tumor antigens. Simultaneously, the sustained release of CCL21 from LC-Gel efficiently recruits DCs to capture these antigens, leading to robust T cell activation. Consequently, intratumoral injection of LC-Gel generates a potent in situ DC vaccine, enhancing anti-tumor T cell immunity and inhibiting the growth of orthotopic breast tumors. Moreover, LC-Gel is shown to trigger long-term immune memory for eliciting a distant anti-tumor effect. In summary, our study introduces an innovative in situ DC vaccination strategy using an injectable oncolytic hydrogel platform for cancer immunotherapy.
Collapse
Affiliation(s)
- Zi-Lu Wang
- School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China.
| | - Si-Yu Qiu
- School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China.
| | - Yi-Qun Sun
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China
| | - Xiao-Jiao Du
- School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China.
| | - Cong-Fei Xu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China
| | - Zi-Yang Cao
- Department of General Surgery, Guangzhou First People's Hospital, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P. R. China.
| | - Zi-Dong Lu
- School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China.
| |
Collapse
|
3
|
Nour MA, Rajabivahid M, Mehdi MSS, Tahmasebi S, Dashtgol SN, Dehghani-Ghorbi M, Vanan AG, Ghorbaninezhad F. A new era in melanoma immunotherapy: focus on DCs metabolic reprogramming. Cancer Cell Int 2025; 25:149. [PMID: 40234886 PMCID: PMC12001691 DOI: 10.1186/s12935-025-03781-3] [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: 12/11/2024] [Accepted: 04/03/2025] [Indexed: 04/17/2025] Open
Abstract
Melanoma, being one of the most dangerous forms of skin cancer, is characterized by its aggressive and metastatic nature, with the potential to develop resistance to various treatments. This resistance makes the disease challenging to treat, emphasizing the need for new treatment strategies. Within the tumor microenvironment (TME), melanoma cells exploit metabolic shifts, particularly glycolysis, to create an immunosuppressive TME that prevents dendritic cells (DCs) from functioning properly. Essential metabolic alterations such as lactate and lipid accumulation, and lack of tryptophan disrupt DC maturation, antigen presentation, and T cell activation. In recent years, melanoma immunotherapy has increasingly focused on reprogramming the metabolism of DCs. This review paper aims to provide insights into the metabolic suppression of melanoma-associated DCs, allowing the design of therapeutic strategies based on metabolic interventions to promote or restore DC function. This contribution reviews the metabolic reprogramming of DCs as a new approach for melanoma immunotherapy.
Collapse
Affiliation(s)
- Mina Afrashteh Nour
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mansour Rajabivahid
- Department of Internal Medicine, Valiasr Hospital, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Marjan Sadat Seyed Mehdi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Safa Tahmasebi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepideh Nasirzadeh Dashtgol
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Tehran Medical Branch, Islamic Azad University, Tehran, Iran
| | - Mahmoud Dehghani-Ghorbi
- Hematology-Oncology Department, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ahmad Ghorbani Vanan
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Farid Ghorbaninezhad
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Wei J, Li D, Long H, Han M. Immune microenvironment of tumor-draining lymph nodes: insights for immunotherapy. Front Immunol 2025; 16:1562797. [PMID: 40292299 PMCID: PMC12021902 DOI: 10.3389/fimmu.2025.1562797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Tumor-draining lymph nodes (TDLNs) play a crucial role in modulating tumor immune responses and influencing the efficacy of immunotherapy. However, our current understanding of the microenvironment within these lymph nodes remains limited. Tumors not only impair the anti-tumor activity of CD8+ T cells by creating an immunosuppressive microenvironment, but they also facilitate immune evasion and promote metastasis by altering the structure and function of TDLNs. Research has shown that tumor-specific memory CD8+ T cells (TTSM) within TDLNs are essential for the efficacy of immune checkpoint inhibitors, such as PD-1/PD-L1 blockers. Moreover, the abnormal structure of TDLNs, along with the presence of immunosuppressive cells-such as regulatory T cells (Tregs), regulatory B cells (Bregs), and immunosuppressive dendritic cells (DCs)-contributes to tumor-mediated immune evasion. Therefore, gaining a deeper understanding of the immune microenvironment within TDLNs is essential for improving the effectiveness of immunotherapies and developing novel therapeutic strategies. This review explores various TDLN-based therapeutic strategies, addressing the controversies surrounding lymph node dissection, the use of TDLNs as a source of tumor-infiltrating lymphocytes (TILs) for therapy, targeting immunosuppressive cells within TDLNs, and methods to reverse the structural abnormalities of TDLNs. These strategies offer valuable insights and potential directions for advancing tumor immunotherapy.
Collapse
Affiliation(s)
- Jiahuan Wei
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Daozhang Li
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Haixia Long
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Mei Han
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| |
Collapse
|
5
|
Veas Rodriguez J, Piñol M, Sorolla MA, Parisi E, Sorolla A, Santacana M, Ruiz M, Parra G, Bernabeu M, Iglesias M, Aracil C, Escartin A, Vilardell F, Matias-Guiu X, Salud A, Montal R. Comprehensive immunophenotyping of gastric adenocarcinoma identifies an inflamed class of tumors amenable to immunotherapies. J Immunother Cancer 2025; 13:e010024. [PMID: 40102027 PMCID: PMC11927434 DOI: 10.1136/jitc-2024-010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Gastric adenocarcinoma (GAC) imposes a considerable global health burden. Molecular profiling of GAC from the tumor microenvironment perspective through a multi-omics approach is eagerly awaited in order to allow a more precise application of novel therapies in the near future. METHODS To better understand the tumor-immune interface of GAC, we identified an internal cohort of 82 patients that allowed an integrative molecular analysis including mutational profiling by whole-exome sequencing, RNA gene expression of 770 genes associated with immune response, and multiplex protein expression at spatial resolution of 34 immuno-oncology targets at different compartments (tumorous cells and immune cells). Molecular findings were validated in 595 GAC from the TCGA and ACRG external cohorts with available multiomics data. Prediction of response to immunotherapies of the discovered immunophenotypes was assessed in 1039 patients with cancer from external cohorts with available transcriptome data. RESULTS Unsupervised clustering by gene expression identified a subgroup of GAC that includes 52% of the tumors, the so-called Inflamed class, characterized by high tumor immunogenicity and cytotoxicity, particularly in the tumor center at protein level, with enrichment of PIK3CA and ARID1A mutations and increased presence of exhausted CD8+ T cells as well as co-inhibitory receptors such as PD1, CTLA4, LAG3, and TIGIT. The remaining 48% of tumors were called non-inflamed based on the observed exclusion of T cell infiltration, with an overexpression of VEGFA and higher presence of TP53 mutations, resulting in a worse clinical outcome. A 10-gene RNA signature was developed for the identification of tumors belonging to these classes, demonstrating in evaluated datasets comparable clinical utility in predicting response to current immunotherapies when tested against other published gene signatures. CONCLUSIONS Comprehensive immunophenotyping of GAC identifies an inflamed class of tumors that complements previously proposed tumor-based molecular clusters. Such findings may provide the rationale for exploring novel immunotherapeutic approaches for biomarker-enriched populations in order to improve GAC patient's survival.
Collapse
Affiliation(s)
- Joel Veas Rodriguez
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Miquel Piñol
- Department of Pathology, Oncological Pathology Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Maria Alba Sorolla
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Eva Parisi
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Anabel Sorolla
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Maria Santacana
- Scientific and Technical Service of Immunohistochemistry, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Maria Ruiz
- Scientific and Technical Service of Biobank, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Genís Parra
- CNAG-Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mario Bernabeu
- CNAG-Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mar Iglesias
- Department of Pathology, Hospital del Mar, University Pompeu Fabra, Hospital del Mar Research Institute, CIBERONC, Barcelona, Spain
| | - Carles Aracil
- Department of Gastroenterology, Clinical and Experimental Research in Digestive and Hematological Pathology Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Alfredo Escartin
- Department of Surgery, Experimental Surgery Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Felip Vilardell
- Department of Pathology, Oncological Pathology Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Xavier Matias-Guiu
- Department of Pathology, Oncological Pathology Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Antonieta Salud
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Robert Montal
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| |
Collapse
|
6
|
Zeng SM, Qu WQ, Sun YL, Chen KW, Zhao K, Yan JH, Zhang C, Liang CX, Chen Y, Pan T, Yu A, Zhang XZ. MnO 2-Assisted Photosynthetic Bacteria Interfering with the Adenosine-A2AR Metabolic Pathway to Enhance Tumor Photothermal Immunotherapy. ACS NANO 2025; 19:7962-7980. [PMID: 39976374 DOI: 10.1021/acsnano.4c15139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Hypoxia-related adenosine (Ado) exerts an immunosuppressive effect in tumors by binding to the metabolic checkpoint Ado A2A receptors (A2AR), thereby hindering the activation of antitumor immunity induced by immunogenic cell death (ICD). In this study, a MnO2-assisted photosynthetic bacteria (PSB) biohybrid (MnO2@PSB) is developed to enhance tumor photothermal immunotherapy by interfering with the Ado-A2AR metabolic pathway. Specifically, manganese dioxide (MnO2) nanoflowers are conjugated onto PSB by the carbodiimide reaction to construct the biohybrid MnO2@PSB. As a photothermal agent, MnO2@PSB generates heat to "burn" tumor cells under 808 nm laser irradiation, inducing tumor cell ICD. Meanwhile, MnO2@PSB catalyzes the decomposition of endogenous hydrogen peroxide into oxygen to alleviate tumor hypoxia, thereby reducing Ado production and downregulating the expression of A2AR, further reversing the tumor immunosuppressive microenvironment and amplifying the ICD effects. In various mouse 4T1 tumor models, MnO2@PSB can enhance antitumor immune responses, prolong mouse survival, and significantly inhibit tumor growth, recurrence, and metastasis under 808 nm laser irradiation. Collectively, this study provides a direction for enhanced antitumor immunotherapy through regulating metabolic pathways.
Collapse
Affiliation(s)
- Si-Min Zeng
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Wen-Qiang Qu
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Yu-Liang Sun
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Ke-Wei Chen
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Kai Zhao
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Jian-Hua Yan
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Cheng Zhang
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Chun-Xiao Liang
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Yu Chen
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Ting Pan
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Aixi Yu
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Xian-Zheng Zhang
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital & Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| |
Collapse
|
7
|
Xiao F, Liu Y, Wang X. The Efficacy of Immune Checkpoint Inhibitors in the EGFR Mutant and Wild-Type Non-Small Cell Lung Cancer Is Positively Associated With the Maturation and Abundance of Dendritic Cells. Thorac Cancer 2025; 16:e70049. [PMID: 40130724 PMCID: PMC11934210 DOI: 10.1111/1759-7714.70049] [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: 01/17/2025] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Dendritic cells (DCs) are known to be crucial in initiating immune responses, but their role in regulating immune checkpoint inhibitor (ICI) efficacy in EGFR mutant NSCLC remains unclear. METHODS Peripheral blood mononuclear cells (PBMCs) were co-cultured with EGFR mutant cells to evaluate immune scores and DC maturation via high-throughput sequencing. TIDE scores were used to predict the efficacy of ICI treatment. Gene set enrichment analysis (GSEA) was carried out on DCs to explore the signaling pathway changes underlying the diverse responses to ICIs. RESULTS A significant decrease in CD8+ T lymphocytes and cytotoxicity scores was found in EGFR mutant LUAD compared to wild-type (p < 0.001). Three datasets (GSE135222, GSE126044, and GSE136961) showed that higher DC gene expression was associated with a more favorable response to ICIs (p = 0.028). The CSE241934 dataset showed that the number of conventional DC 1 (cDC1) was higher in the ICI-sensitive group. The TIDE model suggested that cDC1 was associated with ICIs efficacy. However, GSE32863, GSE75037, and GSE72094 showed no differences in cDC subpopulations between EGFR mutant and wild-type LUAD. EGFR mutant cells exhibited more suppression in the expression of HLA-DR, CD40, CD83, and CD86 than the control group. The TIDE model suggested DC maturity was associated with ICI efficacy. GSE241934-IIT showed that DC maturity was more abundant in the ICI-sensitive group than that in the resistant group. CONCLUSIONS Both the number and maturation capacity of DCs are positively correlated with ICI efficacy. The cause of poor ICI efficacy in EGFR mutant LUAD is more likely to be low DC maturity, not number, compared to EGFR wild-type LUAD.
Collapse
Affiliation(s)
- Fengqi Xiao
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanShandongChina
| | - Yanguo Liu
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanShandongChina
| | - Xiuwen Wang
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanShandongChina
| |
Collapse
|
8
|
Ma K, Wang L, Li W, Tang T, Ma B, Zhang L, Zhang L. Turning cold into hot: emerging strategies to fire up the tumor microenvironment. Trends Cancer 2025; 11:117-134. [PMID: 39730243 DOI: 10.1016/j.trecan.2024.11.011] [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: 08/14/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/29/2024]
Abstract
The tumor microenvironment (TME) is a complex, highly structured, and dynamic ecosystem that plays a pivotal role in the progression of both primary and metastatic tumors. Precise assessment of the dynamic spatiotemporal features of the TME is crucial for understanding cancer evolution and designing effective therapeutic strategies. Cancer is increasingly recognized as a systemic disease, influenced not only by the TME, but also by a multitude of systemic factors, including whole-body metabolism, gut microbiome, endocrine signaling, and circadian rhythm. In this review, we summarize the intrinsic, extrinsic, and systemic factors contributing to the formation of 'cold' tumors within the framework of the cancer-immunity cycle. Correspondingly, we discuss potential strategies for converting 'cold' tumors into 'hot' ones to enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Kaili Ma
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China
| | - Lin Wang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China
| | - Wenhui Li
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China
| | - Tingting Tang
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Bo Ma
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Liyuan Zhang
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, China; PRAG Therapy Center, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, China.
| | - Lianjun Zhang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
9
|
Jiang Y, Liu Y, Huang H, Zhao T, Zhao Z, Gao Y. Effect of RAS mutations and related immune characteristics on the prognosis of patients with MSI-H/dMMR colorectal cancer. Cancer Immunol Immunother 2025; 74:78. [PMID: 39891700 PMCID: PMC11787098 DOI: 10.1007/s00262-024-03926-9] [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: 10/15/2024] [Accepted: 12/16/2024] [Indexed: 02/03/2025]
Abstract
PURPOSE Microsatellite high instability/deficient mismatch repair (MSI-H/dMMR) colorectal cancer (CRC) has an active tumor microenvironment, rendering it more sensitive to immune checkpoint inhibitors. Given that studies involving patients with MSI-H colorectal cancer with RAS mutations are scarce, we explored the effect of RAS mutations on the TME in patients with MSI-H/dMMR cancer and identified potential prognostic factors. METHODS Seventy-five patients diagnosed with MSI-H/dMMR colorectal cancer were retrospectively enrolled and divided into RAS-mutant and -wild-type groups. The expression levels of CD11c+ dendritic cells, CD4+ T cells, CD8+ T cells, and regulatory T cell (Treg) markers were detected, and prognostic factors were analyzed. RESULTS RAS-mutant MSI-H colorectal patients were more likely to have: (1) higher platelet values; (2) shorter disease-free survival (DFS); (3) lower infiltrated numbers of CD11c+ dendritic cells, CD4+ T lymphocytes, and CD8+ T lymphocytes, and higher infiltrated numbers of Foxp3+ Treg cells. In MSI-H/dMMR CRC patients: (1) the high CD11c + , CD4 +, and CD8 + cells infiltration group had longer DFS than the low-infiltration group, and Foxp3 + cells infiltration was not significantly correlated with DFS; (2) the RAS mutation status, number of CD11c+ cells infiltrated, and carbohydrate antigen 19-9 (CA19-9) level were the potential prognostic factors. CONCLUSION RAS mutations in patients with MSI-H/dMMR CRC may reduce the infiltration of CD11c+ dendritic cells, CD4+ T cells, and CD8+ T cells, and increase the infiltration of Foxp3+ Treg cells to affect the tumor microenvironment of patients. RAS gene status, CD11c + cells infiltration, and CA19-9 level were potential prognostic factors for MSI-H/dMMR CRC.
Collapse
Affiliation(s)
- Yupeng Jiang
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yuyao Liu
- Department of Oncology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, 410004, China
| | - Hong Huang
- Guilin Medical University, Guilin, 541000, China
| | - Tiantian Zhao
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Zengyi Zhao
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yawen Gao
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
10
|
Ascic E, Pereira CF. Transcription factor-mediated reprogramming to antigen-presenting cells. Curr Opin Genet Dev 2025; 90:102300. [PMID: 39721321 DOI: 10.1016/j.gde.2024.102300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024]
Abstract
Antigen-presenting cells (APCs) are a heterogenous group of immune cells composed by dendritic cells (DCs) and macrophages (Mϕ), which are critical for orchestrating immunity against cancer or infections. Several strategies have been explored to generate APC subsets, including enrichment from peripheral blood and differentiation from pluripotent or multipotent cells. During development, the generation of APC subsets is instructed by transcription factors (TFs). Direct cell reprogramming, also known as transdifferentiation, offers an approach to harness combinations of TFs to generate APCs from unrelated somatic cells, including cancer cells. In this review, we summarize the transcriptional specification of DC subsets, highlight transcriptional networks for their generation, and discuss future applications of DC reprogramming in cancer immunotherapy.
Collapse
Affiliation(s)
- Ervin Ascic
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, BMC A12, 221 84 Lund, Sweden; Wallenberg Center for Molecular Medicine at Lund University, BMC A12, 221 84 Lund, Sweden.
| | - Carlos-Filipe Pereira
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, BMC A12, 221 84 Lund, Sweden; Wallenberg Center for Molecular Medicine at Lund University, BMC A12, 221 84 Lund, Sweden; Asgard Therapeutics AB, Medicon Village, 223 81 Lund, Sweden; CNC - Centre for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês do Pombal, 3004-517 Coimbra, Portugal.
| |
Collapse
|
11
|
Chiang MR, Hsu CW, Pan WC, Tran NT, Lee YS, Chiang WH, Liu YC, Chen YW, Chiou SH, Hu SH. Reprogramming Dysfunctional Dendritic Cells by a Versatile Catalytic Dual Oxide Antigen-Captured Nanosponge for Remotely Enhancing Lung Metastasis Immunotherapy. ACS NANO 2025; 19:2117-2135. [PMID: 39739571 PMCID: PMC11760334 DOI: 10.1021/acsnano.4c09525] [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: 07/16/2024] [Revised: 12/06/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025]
Abstract
Dendritic cells (DCs) play a crucial role in initiating antitumor immune responses. However, in the tumor environment, dendritic cells often exhibit impaired antigen presentation and adopt an immunosuppressive phenotype, which hinders their function and reduces their ability to efficiently present antigens. Here, a dual catalytic oxide nanosponge (DON) doubling as a remotely boosted catalyst and an inducer of programming DCs to program immune therapy is reported. Intravenous delivery of DON enhances tumor accumulation via the marginated target. At the tumor site, DON incorporates cerium oxide nanozyme (CeO2)-coated iron oxide nanocubes as a peroxide mimicry in cancer cells, promoting sustained ROS generation and depleting intracellular glutathione, i.e., chemodynamic therapy (CDT). Upon high-frequency magnetic field (HFMF) irradiation, CDT accelerates the decomposition of H2O2 and the subsequent production of more reactive oxygen species, known as Kelvin's force laws, which promote the cycle between Fe3+/Fe2+ and Ce3+/Ce4+ in a sustainable active surface. HFMF-boosted catalytic DON promotes tumors to release tumor-associated antigens, including neoantigens and damage-associated molecular patterns. Then, the porous DON acts as an antigen transporter to deliver autologous tumor-associated antigens to program DCs, resulting in sustained immune stimulation. Catalytic DON combined with the immune checkpoint inhibitor (anti-PD1) in lung metastases suppresses tumors and improves survival over 40 days.
Collapse
Affiliation(s)
- Min-Ren Chiang
- Department
of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Chin-Wei Hsu
- Department
of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Wan-Chi Pan
- Department
of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Ngoc-Tri Tran
- Department
of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Yu-Sheng Lee
- Department
of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Wen-Hsuan Chiang
- Department
of Chemical Engineering, National Chung
Hsing University, Taichung 402, Taiwan
| | - Yu-Chen Liu
- Laboratory for Human Immunology (Single Cell Genomics), WPI Immunology
Frontier Research Center, Center for Infectious Disease Education and Research
(CiDER)Osaka University, Osaka 565-0871, Japan
| | - Ya-Wen Chen
- National
Institute of Cancer Research, National Health
Research Institutes, Miaoli County 35053, Taiwan
| | - Shih-Hwa Chiou
- Institute
of Pharmacology, College of Medicine, National
Yang Ming Chiao Tung University, Hsinchu, Taipei 112304, Taiwan
- Department
of Medical Research, Veterans General Hospital, Taipei, Taipei 112304, Taiwan
| | - Shang-Hsiu Hu
- Department
of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| |
Collapse
|
12
|
Sun Z, Hu M, Huang X, Song M, Chen X, Bei J, Lin Y, Chen S. Predictive value of dendritic cell-related genes for prognosis and immunotherapy response in lung adenocarcinoma. Cancer Cell Int 2025; 25:13. [PMID: 39810206 PMCID: PMC11730157 DOI: 10.1186/s12935-025-03642-z] [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: 07/25/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Patients with lung adenocarcinoma (LUAD) receiving drug treatment often have an unpredictive response and there is a lack of effective methods to predict treatment outcome for patients. Dendritic cells (DCs) play a significant role in the tumor microenvironment and the DCs-related gene signature may be used to predict treatment outcome. Here, we screened for DC-related genes to construct a prognostic signature to predict prognosis and response to immunotherapy in LUAD patients. METHODS DC-related biological functions and genes were identified using single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing. DCs-related gene signature (DCRGS) was constructed using integrated machine learning algorithms. Expression of key genes in clinical samples was examined by real-time q-PCR. Performance of the prognostic model, DCRGS, for the prognostic evaluation, was assessed using a multiple time-dependent receiver operating characteristic (ROC) curve, the R package, "timeROC", and validated using GEO datasets. RESULTS Analysis of scRNA-seq data showed that there is a significant upregulation of LGALS9 expression in DCs isolated from malignant pleural effusion samples. Leveraging the Coxboost and random survival forest combination algorithm, we filtered out six DC-related genes on which a prognostic prediction model, DCRGS, was established. A high predictive capability nomogram was constructed by combining DCRGS with clinical features. We found that patients with a high-DCRGS score had immunosuppression, activated tumor-associated pathways, and elevated somatic mutational load and copy number variant load. In contrast, patients in the low-DCRGS subgroup were resistant to chemotherapy but sensitive to the CTLA-4 immune checkpoint inhibitor and targeted therapy. CONCLUSION We have innovatively established a deep learning-based prediction model, DCRGS, for the prediction of the prognosis of patients with LUAD. The model possesses a strong prognostic prediction performance with high accuracy and sensitivity and could be clinically useful to guide the management of LUAD. Furthermore, the findings of this study could provide an important reference for individualized clinical treatment and prognostic prediction of patients with LUAD.
Collapse
Affiliation(s)
- Zihao Sun
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Mengfei Hu
- Department of Internal Medicine, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230000, China
| | - Xiaoning Huang
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Minghan Song
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Xiujing Chen
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Jiaxin Bei
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
| | - Yiguang Lin
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
- Research & Development Division, Guangzhou Anjie Biomedical Technology Co., Ltd., Guangzhou, 510535, China.
| | - Size Chen
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangzhou, 510080, China.
- Key Laboratory of Monitoring Adverse Reactions Associated with CAR-T Cell Therapy, Guangzhou, 510080, China.
| |
Collapse
|
13
|
Broksø AD, Bendixen L, Fammé S, Mikkelsen K, Jensen TI, Bak RO. Orthogonal transcriptional modulation and gene editing using multiple CRISPR-Cas systems. Mol Ther 2025; 33:71-89. [PMID: 39563029 PMCID: PMC11764084 DOI: 10.1016/j.ymthe.2024.11.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/13/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024] Open
Abstract
CRISPR-Cas-based transcriptional activation (CRISPRa) and interference (CRISPRi) enable transient programmable gene regulation by recruitment or fusion of transcriptional regulators to nuclease-deficient Cas (dCas). Here, we expand on the emerging area of transcriptional engineering and RNA delivery by benchmarking combinations of RNA-delivered dCas and transcriptional modulators. We utilize dCas9 from Staphylococcus aureus and Streptococcus pyogenes for orthogonal transcriptional modulation to upregulate one set of genes while downregulating another. We also establish trimodal genetic engineering by combining orthogonal transcriptional regulation with gene knockout by Cas12a (Acidaminococcus; AsCas12a) ribonucleoprotein delivery. To simplify trimodal engineering, we explore optimal parameters for implementing truncated single guide RNAs (sgRNAs) to make use of SpCas9 for knockout and CRISPRa. We find the Cas9 protein/sgRNA ratio to be crucial for avoiding sgRNA cross-complexation and for balancing knockout and activation efficiencies. We demonstrate high efficiencies of trimodal genetic engineering in primary human T cells while preserving basic T cell health and functionality. This study highlights the versatility and potential of complex genetic engineering using multiple CRISPR-Cas systems in a simple one-step process yielding transient transcriptome modulation and permanent DNA changes. We believe such elaborate engineering can be implemented in regenerative medicine and therapies to facilitate more sophisticated treatments.
Collapse
Affiliation(s)
| | - Louise Bendixen
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Simon Fammé
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | | | | | - Rasmus O Bak
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
| |
Collapse
|
14
|
Yao F, Yuan Q, Yan Y, Liang G, Zhou L, Xu H, Gao S, Zou T, Zhang L. Yu-Ping-Feng-San improve the immunosuppression of microenvironment in hepatocellular carcinoma by promoting the maturation of DCs through the JAK2-STAT3 pathway. Sci Rep 2024; 14:31522. [PMID: 39733089 PMCID: PMC11682348 DOI: 10.1038/s41598-024-83197-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/12/2024] [Indexed: 12/30/2024] Open
Abstract
Yu-Ping-Feng-San (YPF) is a famous classical Chinese medicine formula known for its ability to boost immunity. YPF has been applied to enhance the immune status of tumor patients in clinical practice. However, there is still a lack of research on its immune regulatory effects and mechanisms in the tumor microenvironment. This study was designed to investigate the effects and mechanism of YPF on improving the immune suppression state of hepatocellular carcinoma (HCC) microenvironment. In an orthotopic mouse model of HCC, YPF improved the immune microenvironment of HCC immunosuppression, enhanced the maturation of dendritic cells (DCs), promoted the release of IL-12, and decreased the presence of JAK2, p-JAK2, STAT3, and p-STAT3 proteins in both tumor tissue and paracancerous tissues. YPF also could promote the maturation and reduce the activation of JAK2, p-JAK2, STAT3, and p-STAT3 proteins of mouse bone marrow-derived DCs induced by culture medium or tumor supernatant in vitro. Transient transfection of siRNA-STAT3 with DCs resulted in an increase in its maturation and its secretion of IL-12. On the whole, these combined effects of YPF served to ameliorate the HCC immune suppression microenvironment, which conducive to immune cells play the role of immune surveillance and killing liver cancer cells. The mechanisms of these combined effects were, at least in part, related to its inhibition of the activated JAK2-STAT3 signaling pathway in DCs within the HCC microenvironment.
Collapse
Affiliation(s)
- Fei Yao
- Central Laboratory, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, Jiangsu, China
- Clinical Pharmaceutical Laboratory of Traditional Chinese Medicine, Suzhou Academy of Wumen Chinese Medicine, Suzhou, 215000, Jiangsu, China
| | - Qin Yuan
- Central Laboratory, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, Jiangsu, China
- Clinical Pharmaceutical Laboratory of Traditional Chinese Medicine, Suzhou Academy of Wumen Chinese Medicine, Suzhou, 215000, Jiangsu, China
| | - Yichao Yan
- Central Laboratory, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, Jiangsu, China
| | - Guoqiang Liang
- Central Laboratory, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, Jiangsu, China
- Clinical Pharmaceutical Laboratory of Traditional Chinese Medicine, Suzhou Academy of Wumen Chinese Medicine, Suzhou, 215000, Jiangsu, China
| | - Liang Zhou
- Central Laboratory, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, Jiangsu, China
- Clinical Pharmaceutical Laboratory of Traditional Chinese Medicine, Suzhou Academy of Wumen Chinese Medicine, Suzhou, 215000, Jiangsu, China
| | - Heng Xu
- Central Laboratory, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, Jiangsu, China
- Clinical Pharmaceutical Laboratory of Traditional Chinese Medicine, Suzhou Academy of Wumen Chinese Medicine, Suzhou, 215000, Jiangsu, China
| | - Shaomei Gao
- Central Laboratory, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, Jiangsu, China
| | - Ting Zou
- Central Laboratory, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, Jiangsu, China
| | - Lurong Zhang
- Central Laboratory, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, Jiangsu, China.
- Clinical Pharmaceutical Laboratory of Traditional Chinese Medicine, Suzhou Academy of Wumen Chinese Medicine, Suzhou, 215000, Jiangsu, China.
| |
Collapse
|
15
|
Wang W, Zou C, Liu X, He L, Cao Z, Zhu M, Wu Y, Liu X, Ma J, Wang Y, Zhang Y, Zhang K, Wang S, Zhang W, Liu W, Lin W, Zhang Y, Guo Q, Li M, Gu J. Biomimetic Dendritic Cell-Based Nanovaccines for Reprogramming the Immune Microenvironment to Boost Tumor Immunotherapy. ACS NANO 2024; 18:34063-34076. [PMID: 39625243 DOI: 10.1021/acsnano.4c09653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2024]
Abstract
Although dendritic cell (DC)-mediated immunotherapies are effective options for immunotherapy, traditional DC vaccines are hampered by a variety of drawbacks such as insufficient antigen delivery, weak lymph node homing, and the risk of living cell transfusion. To address the above-mentioned issues, we developed a personalized DC-mimicking nanovaccine (HybridDC) that enhances antigen presentation and elicits effective antitumor immunity. The biomimetic nanovaccine contains cell membranes derived from genetically engineered DCs, and several cellular components are simultaneously anchored onto these membranes, including CC-chemokine receptor 7 (CCR7), tumor-associated antigenic (TAA) peptide/tumor-derived exosome (TEX), and relevant costimulatory molecules. Compared with previous vaccines, the HybridDC vaccine showed an increased ability to target lymphoid tissues and reshape the immune landscape in the tumor milieu. HybridDC demonstrated significant therapeutic and prophylactic efficacy in poorly immunogenic, orthotopic models of glioma. Furthermore, the HybridDC vaccine potentiates the therapeutic efficacy of immune checkpoint blockade (ICB) therapy, providing a potential combination strategy to maximize the efficacy of ICB. Specifically, HybridDC can induce long-term protective immunity in memory T cells. Overall, the HybridDC vaccine is a promising platform for personalized cancer vaccines and may offer a combinational modality to improve current immunotherapy.
Collapse
Affiliation(s)
- Weizhong Wang
- Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Cheng Zou
- Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Xiao Liu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Lei He
- Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Zhengcong Cao
- Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Maorong Zhu
- Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Yuxin Wu
- Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Xiaolin Liu
- Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Jiying Ma
- Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Yaoliang Wang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yile Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Kuo Zhang
- Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Shuning Wang
- Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Wangqian Zhang
- Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Wei Liu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Wei Lin
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yingqi Zhang
- Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Qingdong Guo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Meng Li
- Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Jintao Gu
- Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
16
|
Sosa Cuevas E, Mouret S, Vayssière G, Kerboua S, Girard P, Molens JP, Manceau M, Charles J, Saas P, Aspord C. Circulating immune landscape in melanoma patients undergoing anti-PD1 therapy reveals key immune features according to clinical response to treatment. Front Immunol 2024; 15:1507938. [PMID: 39687620 PMCID: PMC11646980 DOI: 10.3389/fimmu.2024.1507938] [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: 10/08/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction Immune checkpoint blockers (ICB) bring unprecedented clinical success, yet many patients endure immune mediated adverse effects and/or fail to respond. Predictive signatures of response to ICB and mechanisms of clinical efficacy or failure remain understudied. DC subsets, in network with conventional αβ T (Tconv), NK, γδ T and iNKT cells, harbor pivotal roles in tumor control, yet their involvement in response to ICB remained underexplored. Methods We performed an extensive longitudinal monitoring of circulating immune cells from melanoma patients treated with first-line anti-PD1, before (T0) and during treatment. We assessed the phenotypic and functional features of DC and effector cells' subsets by multi-parametric flow cytometry and ProcartaPlex® dosages. Results We revealed differences according to response to treatment and modulations of patterns during treatment, highlighting a strong link between the immune landscape and the outcome of anti-PD1 therapy. Responders exhibited higher frequencies of circulating cDC1s, CD8+ T cells, and γδ2+ T cells in central memory (CM) stage. Notably, we observed a distinct remodeling of ICP expression profile, activation status and natural cytotoxicity receptor patterns of immune subsets during treatment. Anti-PD1 modulated DCs' functionality and triggered deep changes in the functional orientation of Tconv and γδT cells. Discussion Overall, our work provides new insights into the immunological landscape sustaining favorable clinical responses or resistance to first-line anti-PD1 therapy in melanoma patients. Such exploration participates in uncovering the mechanism of action of anti-PD1, discovering innovative predictive signatures of response, and paves the way to design pertinent combination strategies to improve patient clinical benefits in the future.
Collapse
Affiliation(s)
- Eleonora Sosa Cuevas
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Stéphane Mouret
- Dermatology, Allergology & Photobiology Department, CHU Grenoble Alpes, Univ. Grenoble Alpes, Grenoble, France
| | - Guillaume Vayssière
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Siham Kerboua
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Pauline Girard
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Jean-Paul Molens
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Marc Manceau
- Department of Medicine, Clinical Investigation Center, CHU Grenoble Alpes, Univ. Grenoble Alpes, Grenoble, France
| | - Julie Charles
- Dermatology, Allergology & Photobiology Department, CHU Grenoble Alpes, Univ. Grenoble Alpes, Grenoble, France
| | - Philippe Saas
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Caroline Aspord
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| |
Collapse
|
17
|
Sun H, Lv X, Zhang D, Shen Y, Lu H. The 1400 metabolite-mediated relationship between 91 inflammatory cytokines and migraine: An exploratory two-step Mendelian randomization study. Eur J Clin Invest 2024; 54:e14316. [PMID: 39279254 DOI: 10.1111/eci.14316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Inflammatory cytokines and migraines have been associated in previous research, but the underlying mechanisms of action are still elusive. The biological functions of metabolites are crucial in the onset of migraine. Our goals were to clarify the cause-and-effect connection between inflammatory cytokines and migraines and explore the potential mediating function of metabolites. METHODS Utilizing summary-level data from genome-wide association studies (GWAS), we conducted two-sample Mendelian randomization (MR) analyses to evaluate the possible causal connection between inflammatory cytokines and migraines. A two-step MR analysis was employed to further investigate the potential mediating pathways of metabolites. RESULTS MR analysis identified a total of 9 inflammatory cytokines that were genetically associated with migraines, and we subsequently identified 21 mediated relationships, with 20 metabolites (13 metabolites, 7 ratios) acting as potential mediators between 8 inflammatory cytokines and migraine. The 9 inflammatory cytokines were beta-nerve growth factor levels (β-NGF), T-cell surface glycoprotein CD5 levels (CD5), T-cell surface glycoprotein CD6 isoform levels (CD6), C-X-C motif chemokine 11 levels (CXCL11), interleukin-4 levels (IL-4), oncostatin-M levels (OSM), signalling lymphocytic activation molecule levels (SLAM), C-C motif chemokine 25 levels (CCL25) and monocyte chemoattractant protein-1 levels (MCP-1). CONCLUSION Our research findings provide evidence for both a causal connection between inflammatory cytokines and migraines, as well as a metabolite-mediated pathway. These biomarkers facilitate the detection, diagnosis and treatment of migraines while offering fresh perspectives on their underlying mechanisms.
Collapse
Affiliation(s)
- Huiqi Sun
- First school of clinical medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xutong Lv
- First school of clinical medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Dongbin Zhang
- Department of Anesthesiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yue Shen
- First school of clinical medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Hongxiu Lu
- Department of Anesthesiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
18
|
Li X, Gao ML, Wang SS, Hu Y, Hou D, Liu PN, Xiang H. Nanoscale covalent organic framework-mediated pyroelectrocatalytic activation of immunogenic cell death for potent immunotherapy. SCIENCE ADVANCES 2024; 10:eadr5145. [PMID: 39612337 PMCID: PMC11606443 DOI: 10.1126/sciadv.adr5145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/25/2024] [Indexed: 12/01/2024]
Abstract
The conventional molecular immunogenic cell death (ICD) inducers suffer from poor biocompatibility and unsatisfactory efficacy. Here, a biocompatible nanosized covalent organic framework (nCOF)-based pyroelectric catalyst (denoted as TPAD-COF NPs) is designed for pyroelectric catalysis-activated in situ immunotherapy. TPAD-COF NPs confine organic pyroelectric molecules to rigid TPAD-COF NPs to substantially reduce aggregation and enhance biocompatibility, thus improving pyroelectrocatalytic efficiency. After tumor internalization, TPAD-COF NPs facilitate photothermal tumor ablation under near-infrared (NIR) laser exposure, resulting in effective ICD induction. In addition, TPAD-COF NPs effectively catalyze the conversion of temperature changes to pyroelectric changes, which subsequently react with adjacent O2 to generate reactive oxygen species, thus triggering robust ICD activation. In vivo evaluation using mouse models confirmed that TPAD-COF NPs evidently inhibited the proliferation of primary and distant tumors and prevented lung metastasis under NIR laser illumination. Therefore, this study opens an avenue for designing nCOF-based catalysts for pyroelectric catalysis-activated in situ immunotherapy.
Collapse
Affiliation(s)
- Xingguang Li
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237 China
| | - Meng-Lu Gao
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237 China
| | - Shan-Shan Wang
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237 China
| | - Yizhi Hu
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237 China
| | - Dongzhi Hou
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237 China
| | - Pei-Nian Liu
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237 China
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Huijing Xiang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| |
Collapse
|
19
|
Li X, Yao X, Wen J, Chen Q, Zhu Z, Zhang X, Wang S, Lan W, Huang Y, Tang S, Zhou X, Han X, Zhang T. The application of sphingomyelin in mediating the causal role of the T-cell surface glycoprotein CD5 in Crohn's disease: A two-step Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40513. [PMID: 39560554 PMCID: PMC11576039 DOI: 10.1097/md.0000000000040513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/25/2024] [Indexed: 11/20/2024] Open
Abstract
To examine the possible causative association between Crohn disease (CD) and the T-cell surface glycoprotein CD5 and to ascertain whether sphingomyelin (SM) functions as a mediator. We conducted a two-step Mendelian randomization (MR) study to further explore the pathogenesis of Crohn and its related targets. MR study was performed on CD5 and CD using summary-level data from a genome-wide association study. Additionally, by employing a two-step MR study method, we determined that SM might mediate the causal effect of CD5 on CD. There was a favorable correlation between the surface glycoprotein CD5 on T cells and vulnerability to CD, and SM mediated the causal effect of CD5 on CD (the mediating effect accounts for 9.2%). Our study revealed that CD5 and CD are causally related, with SM mediating a small fraction of the impact (approximately 9.2%). The mediating function of SM in the link between CD5 and CD is anticipated to be realized through the regulation of immune cell transportation, apoptosis of intestinal barrier cells, and maintenance of the intestinal microenvironment.
Collapse
Affiliation(s)
- Xiao Li
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xin Yao
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Jieying Wen
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Qiaoling Chen
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Ziming Zhu
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xinyue Zhang
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Song Wang
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Weixuan Lan
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Yunsi Huang
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Shanneng Tang
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xuan Zhou
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xuedong Han
- The graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Tao Zhang
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
20
|
Yao L, Hatami M, Ma W, Skutella T. Vaccine-based immunotherapy and related preclinical models for glioma. Trends Mol Med 2024; 30:965-981. [PMID: 39013724 DOI: 10.1016/j.molmed.2024.06.009] [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] [Received: 02/19/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 07/18/2024]
Abstract
Glioma, the most common primary malignant tumor in the central nervous system (CNS), lacks effective treatments, and >60% of cases are glioblastoma (GBM), the most aggressive form. Despite advances in immunotherapy, GBM remains highly resistant. Approaches that target tumor antigens expedite the development of immunotherapies, including personalized tumor-specific vaccines, patient-specific target selection, dendritic cell (DC) vaccines, and chimeric antigen receptor (CAR) and T cell receptor (TCR) T cells. Recent studies show promising results in treating GBM and lower-grade glioma (LGG), fostering hope for future immunotherapy. This review discusses tumor vaccines against glioma, preclinical models in immunological research, and the role of CD4+ T cells in vaccine-induced antitumor immunity. We also summarize clinical approaches, challenges, and future research for creating more effective vaccines.
Collapse
Affiliation(s)
- Longping Yao
- Institute for Anatomy and Cell Biology, Heidelberg Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Maryam Hatami
- Institute for Anatomy and Cell Biology, Heidelberg Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China; State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Thomas Skutella
- Institute for Anatomy and Cell Biology, Heidelberg Medical Faculty, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
21
|
Welch BM, Parikh SA, Kay NE, Medina KL. Profound deficiencies in mature blood and bone marrow progenitor dendritic cells in Chronic Lymphocyticcytic Leukemia patients. RESEARCH SQUARE 2024:rs.3.rs-4953853. [PMID: 39399662 PMCID: PMC11469369 DOI: 10.21203/rs.3.rs-4953853/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Chronic lymphocytic leukemia (CLL) patients are immunocompromised and highly vulnerable to serious recurrent infections. Conventional dendritic cells (cDCs) and plasmacytoid DCs (pDCs) are principal sensors of infection and are essential in orchestrating innate and adaptive immune responses to resolve infection. This study identified significant deficiencies in six functionally distinct DC subsets in blood of untreated CLL (UT-CLL) patients and selective normalization of pDCs in response to acalabrutinib (a Bruton tyrosine kinase inhibitor) therapy. DCs are continuously replenished from hematopoiesis in bone marrow (BM). Four BM developmental intermediates that give rise to cDCs and pDCs were examined and significant reductions of these were identified in UT-CLL patients supporting a precursor/progeny relationship. The deficiencies in blood DCs and BM DC progenitors were significantly associated with alterations in the Flt3/FL signaling pathway critical to DC development and function. Regarding clinical parameter, cDC subset deficiencies are associated with adverse prognostic indicators of disease progression, including IGHV mutation, CD49d, CD38, and ZAP-70 status. Importantly, UT-CLL patients with shared DC subset deficiencies had shorter time-to-first treatment (TTFT), uncovering a profound alteration in innate immunity with the potential to instruct therapeutic decision-making.
Collapse
Affiliation(s)
- Baustin M. Welch
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Neil E. Kay
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
- Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA
| | - Kay L. Medina
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
22
|
Nie W, He Y, Mi X, He S, Chen J, Zhang Y, Wang B, Zheng S, Qian Z, Gao X. Immunostimulatory CKb11 gene combined with immune checkpoint PD-1/PD-L1 blockade activates immune response and simultaneously overcomes the immunosuppression of cancer. Bioact Mater 2024; 39:239-254. [PMID: 38832303 PMCID: PMC11145080 DOI: 10.1016/j.bioactmat.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/05/2024] [Accepted: 05/05/2024] [Indexed: 06/05/2024] Open
Abstract
Immunosuppression tumor microenvironment (TME) seriously impedes anti-tumor immune response, resulting in poor immunotherapy effect of cancer. This study develops a folate-modified delivery system to transport the plasmids encoding immune stimulatory chemokine CKb11 and PD-L1 inhibitors to tumor cells, resulting in high CKb11 secretion from tumor cells, successfully activating immune cells and increasing cytokine secretion to reshape the TME, and ultimately delaying tumor progression. The chemokine CKb11 enhances the effectiveness of tumor immunotherapy by increasing the infiltration of immune cells in TME. It can cause high expression of IFN-γ, which is a double-edged sword that inhibits tumor growth while causing an increase in the expression of PD-L1 on tumor cells. Therefore, combining CKb11 with PD-L1 inhibitors can counterbalance the suppressive impact of PD-L1 on anti-cancer defense, leading to a collaborative anti-tumor outcome. Thus, utilizing nanotechnology to achieve targeted delivery of immune stimulatory chemokines and immune checkpoint inhibitors to tumor sites, thereby reshaping immunosuppressive TME for cancer treatment, has great potential as an immunogene therapy in clinical applications.
Collapse
Affiliation(s)
- Wen Nie
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, PR China
| | - Yihong He
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, PR China
| | - Xue Mi
- Department of Pharmacy, West China Second University Hospital of Sichuan University, 610041, Chengdu, PR China
| | - Shi He
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, PR China
| | - Jing Chen
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, PR China
| | - Yunchu Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, PR China
| | - Bilan Wang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, 610041, Chengdu, PR China
| | - Songping Zheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, PR China
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, PR China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, PR China
| |
Collapse
|
23
|
Wang Y, Wang Y, Wang X, Sun W, Yang F, Yao X, Pan T, Li B, Chu J. Label-free active single-cell encapsulation enabled by microvalve-based on-demand droplet generation and real-time image processing. Talanta 2024; 276:126299. [PMID: 38788384 DOI: 10.1016/j.talanta.2024.126299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/01/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024]
Abstract
Droplet microfluidics-based single-cell encapsulation is a critical technology that enables large-scale parallel single-cell analysis by capturing and processing thousands of individual cells. As the efficiency of passive single-cell encapsulation is limited by Poisson distribution, active single-cell encapsulation has been developed to theoretically ensure that each droplet contains one cell. However, existing active single-cell encapsulation technologies still face issues related to fluorescence labeling and low throughput. Here, we present an active single-cell encapsulation technique by using microvalve-based drop-on-demand technology and real-time image processing to encapsulate single cells with high throughput in a label-free manner. Our experiments demonstrated that the single-cell encapsulation system can encapsulate individual polystyrene beads with 96.3 % efficiency and HeLa cells with 94.9 % efficiency. The flow speed of cells in this system can reach 150 mm/s, resulting in a corresponding theoretical encapsulation throughput of 150 Hz. This technology has significant potential in various biomedical applications, including single-cell omics, secretion detection, and drug screening.
Collapse
Affiliation(s)
- Yiming Wang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, 230027, China; Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, 230027, China
| | - Yousu Wang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, 230027, China; Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, 230027, China
| | - Xiaojie Wang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, 230027, China; Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, 230027, China
| | - Wei Sun
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, 230027, China; Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, 230027, China
| | - Fengrui Yang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, University of Science and Technology of China, Hefei, 230026, China
| | - Xuebiao Yao
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, University of Science and Technology of China, Hefei, 230026, China
| | - Tingrui Pan
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, 230027, China; Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China
| | - Baoqing Li
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, 230027, China; Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, 230027, China.
| | - Jiaru Chu
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, 230027, China; Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, 230027, China
| |
Collapse
|
24
|
Lei PJ, Fraser C, Jones D, Ubellacker JM, Padera TP. Lymphatic system regulation of anti-cancer immunity and metastasis. Front Immunol 2024; 15:1449291. [PMID: 39211044 PMCID: PMC11357954 DOI: 10.3389/fimmu.2024.1449291] [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: 06/14/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer dissemination to lymph nodes (LN) is associated with a worse prognosis, increased incidence of distant metastases and reduced response to therapy. The LN microenvironment puts selective pressure on cancer cells, creating cells that can survive in LN as well as providing survival advantages for distant metastatic spread. Additionally, the presence of cancer cells leads to an immunosuppressive LN microenvironment, favoring the evasion of anti-cancer immune surveillance. However, recent studies have also characterized previously unrecognized roles for tumor-draining lymph nodes (TDLNs) in cancer immunotherapy response, including acting as a reservoir for pre-exhausted CD8+ T cells and stem-like CD8+ T cells. In this review, we will discuss the spread of cancer cells through the lymphatic system, the roles of TDLNs in metastasis and anti-cancer immune responses, and the therapeutic opportunities and challenges in targeting LN metastasis.
Collapse
Affiliation(s)
- Pin-Ji Lei
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Cameron Fraser
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Dennis Jones
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Jessalyn M. Ubellacker
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Timothy P. Padera
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
25
|
Han Z, Li T, Zhang H, Liang K, You M, Xu M, Bai F, Zhang T. A case of response to combination treatment with TSA-DC-CTL immunotherapy and osimertinib in EGFR mutated advanced lung adenocarcinoma. Mol Cancer 2024; 23:163. [PMID: 39123231 PMCID: PMC11313023 DOI: 10.1186/s12943-024-02070-3] [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: 03/07/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND This study details a case of a patient with advanced lung adenocarcinoma harboring an exon 19 deletion in the EGFR gene. METHOD A 46-year-old female patient was diagnosed with stage IVb left lung adenocarcinoma, with multiple bone and lymph node metastases. Following the identification of tumor-specific antigen peptides, the patient received a combination treatment of immunotherapy (TSA-DC-CTL) and oral osimertinib. Peripheral blood circulating immune cells and circulating tumor cells (CTCs) were monitored before and after treatment. PET-CT and CT scans were used to assess the tumor response to treatment. RESULTS A significant increase in total lymphocyte percentage and decrease in the number of CTCs in the patient was observed. Imaging studies showed a notable reduction in tumor metastases. CONCLUSION This report demonstrates the safety and efficacy of TSA-DC-CTL cell immunotherapy combined with osimertinib in the treatment of a patient with advanced lung adenocarcinoma with an EGFR exon 19 deletions. This study describes a promising new treatment option for patients with advanced lung cancer with EGFR mutations.
Collapse
Affiliation(s)
- Zhiyi Han
- Department of Surgery, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Tao Li
- Department of Medical Oncology, Affiliated Tumor Hospital of Nantong University & Nantong Tumor Hospital, Nantong, 226361, China
| | - Heng Zhang
- Healthy Cell Biotechnology LLC, Beijing, 100036, China
| | - Kai Liang
- Healthy Cell Biotechnology LLC, Beijing, 100036, China
| | - Mingcong You
- Healthy Cell Biotechnology LLC, Beijing, 100036, China
| | - Mengdi Xu
- Healthy Cell Biotechnology LLC, Beijing, 100036, China
| | - Fan Bai
- Healthy Cell Biotechnology LLC, Beijing, 100036, China
| | - Tongmei Zhang
- Department of Medical Oncology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic & Laboratory for Clinical Medicine, Capital Medical University, Beijing, 101149, China.
| |
Collapse
|
26
|
Chang IYF, Tsai HC, Chen CH, Chen HC, Huang CW, Cox GF, Huang FM, Lin YY, Chen KT, Lin YJ, Wei KC. CAN008 prolongs overall survival in patients with newly diagnosed GBM characterized by high tumor mutational burden. Biomed J 2024; 47:100660. [PMID: 37741340 PMCID: PMC11340566 DOI: 10.1016/j.bj.2023.100660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/31/2023] [Accepted: 09/15/2023] [Indexed: 09/25/2023] Open
Abstract
BACKGROUND A previous phase 1 dose-escalation study in Taiwan indicated CAN008 (asunercept) with standard concurrent chemoradiotherapy (CCRT) improved progression-free survival (PFS) in newly diagnosed glioblastoma (GBM) patients. This study evaluates the efficacy of CAN008 in promoting overall survival (OS) and identifies genetic alterations associated with treatment responses. METHODS We compared OS of 5-year follow-ups from 9 evaluable CAN008 cohort patients (6 received high-dose and 3 received low-dose) to a historical Taiwanese GBM cohort with 164 newly diagnosed patients. CAN008 treatment response-associated genetic alterations were identified by whole-exome sequencing and comparing variant differences between response groups. Associations among patient survival, tumor mutational burden (TMB), and genetic alterations were analyzed using CAN008 cohort and TCGA-GBM dataset. RESULTS OS for high-dose CAN008 patients at 2 and 5 years was 83% and 67%, respectively, and 40.1% and 8.8% for the historical GBM cohort, respectively. Better OS was observed in the high-dose CAN008 cohort (without reaching the median survival) than the historical GBM cohort (median OS: 20 months; p = 0.0103). Five high-dose CAN008 patients were divided into good and poor response groups based on their PFS. A higher variant count and TMB were observed in good response patients, whereas no significant association was observed between TMB and patient survival in the newly diagnosed TCGA-GBM dataset, suggesting TMB may modulate patient CAN008 response. CONCLUSION CAN008 combined with standard CCRT treatment prolonged the PFS and OS of newly diagnosed GBM patients compared to standard therapy alone. Higher treatment efficacy was associated with higher TMB.
Collapse
Affiliation(s)
- Ian Yi-Feng Chang
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Hong-Chieh Tsai
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Chia-Hua Chen
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Hsiu-Chi Chen
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Wen Huang
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan
| | | | | | - You-Yu Lin
- Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei, Taiwan
| | - Ko-Ting Chen
- School of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan; Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Ya-Jui Lin
- School of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan; Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Kuo-Chen Wei
- School of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan; Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan; Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan.
| |
Collapse
|
27
|
Wang C, Chen L, Fu D, Liu W, Puri A, Kellis M, Yang J. Antigen presenting cells in cancer immunity and mediation of immune checkpoint blockade. Clin Exp Metastasis 2024; 41:333-349. [PMID: 38261139 PMCID: PMC11374820 DOI: 10.1007/s10585-023-10257-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024]
Abstract
Antigen-presenting cells (APCs) are pivotal mediators of immune responses. Their role has increasingly been spotlighted in the realm of cancer immunology, particularly as our understanding of immunotherapy continues to evolve and improve. There is growing evidence that these cells play a non-trivial role in cancer immunity and have roles dependent on surface markers, growth factors, transcription factors, and their surrounding environment. The main dendritic cell (DC) subsets found in cancer are conventional DCs (cDC1 and cDC2), monocyte-derived DCs (moDC), plasmacytoid DCs (pDC), and mature and regulatory DCs (mregDC). The notable subsets of monocytes and macrophages include classical and non-classical monocytes, macrophages, which demonstrate a continuum from a pro-inflammatory (M1) phenotype to an anti-inflammatory (M2) phenotype, and tumor-associated macrophages (TAMs). Despite their classification in the same cell type, each subset may take on an immune-activating or immunosuppressive phenotype, shaped by factors in the tumor microenvironment (TME). In this review, we introduce the role of DCs, monocytes, and macrophages and recent studies investigating them in the cancer immunity context. Additionally, we review how certain characteristics such as abundance, surface markers, and indirect or direct signaling pathways of DCs and macrophages may influence tumor response to immune checkpoint blockade (ICB) therapy. We also highlight existing knowledge gaps regarding the precise contributions of different myeloid cell subsets in influencing the response to ICB therapy. These findings provide a summary of our current understanding of myeloid cells in mediating cancer immunity and ICB and offer insight into alternative or combination therapies that may enhance the success of ICB in cancers.
Collapse
Affiliation(s)
- Cassia Wang
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lee Chen
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Doris Fu
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Wendi Liu
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Anusha Puri
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Manolis Kellis
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jiekun Yang
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
28
|
Zhang Y, Zhang C, He J, Lai G, Li W, Zeng H, Zhong X, Xie B. Comprehensive analysis of single cell and bulk RNA sequencing reveals the heterogeneity of melanoma tumor microenvironment and predicts the response of immunotherapy. Inflamm Res 2024; 73:1393-1409. [PMID: 38896289 DOI: 10.1007/s00011-024-01905-5] [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: 03/26/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Tumor microenvironment (TME) heterogeneity is an important factor affecting the treatment response of immune checkpoint inhibitors (ICI). However, the TME heterogeneity of melanoma is still widely characterized. METHODS We downloaded the single-cell sequencing data sets of two melanoma patients from the GEO database, and used the "Scissor" algorithm and the "BayesPrism" algorithm to comprehensively analyze the characteristics of microenvironment cells based on single-cell and bulk RNA-seq data. The prediction model of immunotherapy response was constructed by machine learning and verified in three cohorts of GEO database. RESULTS We identified seven cell types. In the Scissor+ subtype cell population, the top three were T cells, B cells and melanoma cells. In the Scissor- subtype, there are more macrophages. By quantifying the characteristics of TME, significant differences in B cells between responders and non-responders were observed. The higher the proportion of B cells, the better the prognosis. At the same time, macrophages in the non-responsive group increased significantly. Finally, nine gene features for predicting ICI response were constructed, and their predictive performance was superior in three external validation groups. CONCLUSION Our study revealed the heterogeneity of melanoma TME and found a new predictive biomarker, which provided theoretical support and new insights for precise immunotherapy of melanoma patients.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Cong Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Jing He
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Guichuan Lai
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Wenlong Li
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Haijiao Zeng
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Xiaoni Zhong
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China.
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China.
| | - Biao Xie
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China.
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
29
|
Jiang Y, Yu Z, Zheng H, Zhou X, Zhou M, Geng X, Zhu Y, Huang S, Gong Y, Guo L. An immune biomarker associated with EMT serves as a predictor for prognosis and drug response in bladder cancer. Aging (Albany NY) 2024; 16:10813-10831. [PMID: 38980253 PMCID: PMC11272103 DOI: 10.18632/aging.205927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/22/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Bladder cancer (BLCA), which develops from the upper endometrial of the bladder, is the sixth most prevalent cancer across the globe. WDHD1 (WD repeat and HMG-box DNA binding protein 1 gene) directly affects signaling, the cell cycle, and the development of the cell skeleton. Uncertainty surrounds WDHD1's function in BLCA immunity and prognosis, though. MATERIALS AND METHODS Using weighed gene co-expression network analysis (WGCNA), initially, we first identified 32 risk factors in genes with differential expression for this investigation. Then, using a variety of bioinformatic techniques and experimental validation, we examined the connections between WDHD1 and BLCA expression, clinical pathological traits, WDHD1-related proteins, upper-skin-intermediate conversion (EMT), immune cell immersion, convergence factors, immune markers, and drug sensitivity. RESULT The findings demonstrated that we constructed a 32-gene risk-predicting model where WDHD1 was elevated as a representative gene expression in BLCA and related to a range of clinical traits. Furthermore, high WDHD1 expression was a standalone predictor associated with a worse survival rate. The most commonly recruited cells and their evolutionary patterns were highlighted to better comprehend WDHD1's function in cancer. High WDHD1 expression was associated with many aspects of immunology. Finally, the study found that individuals with high expression of WDHD1 were drug-sensitive to four different broad-spectrum anti-cancer drugs. CONCLUSION These results describe dynamic changes in the tumor microenvironment in BLCA and provide evidence for the hypothesis that WDHD1 is a novel biomarker of tumor development. WDHD1 may therefore be a useful target for the detection and management of BLCA.
Collapse
Affiliation(s)
- Yike Jiang
- Department of Ultrasonography, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, China
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330000, China
| | - Zichuan Yu
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330000, China
| | - Hao Zheng
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330000, China
| | - Xuanrui Zhou
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330000, China
| | - Minqin Zhou
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330000, China
| | - Xitong Geng
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330000, China
| | - Yanting Zhu
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330000, China
| | - Shuhan Huang
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330000, China
| | - Yiyang Gong
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330000, China
| | - Liangyun Guo
- Department of Ultrasonography, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, China
| |
Collapse
|
30
|
Gerashchenko T, Frolova A, Patysheva M, Fedorov A, Stakheyeva M, Denisov E, Cherdyntseva N. Breast Cancer Immune Landscape: Interplay Between Systemic and Local Immunity. Adv Biol (Weinh) 2024; 8:e2400140. [PMID: 38727796 DOI: 10.1002/adbi.202400140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/16/2024] [Indexed: 07/13/2024]
Abstract
Breast cancer (BC) is one of the most common malignancies in women worldwide. Numerous studies in immuno-oncology and successful trials of immunotherapy have demonstrated the causal role of the immune system in cancer pathogenesis. The interaction between the tumor and the immune system is known to have a dual nature. Despite cytotoxic lymphocyte activity against transformed cells, a tumor can escape immune surveillance and leverage chronic inflammation to maintain its own development. Research on antitumor immunity primarily focuses on the role of the tumor microenvironment, whereas the systemic immune response beyond the tumor site is described less thoroughly. Here, a comprehensive review of the formation of the immune profile in breast cancer patients is offered. The interplay between systemic and local immune reactions as self-sustaining mechanism of tumor progression is described and the functional activity of the main cell populations related to innate and adaptive immunity is discussed. Additionally, the interaction between different functional levels of the immune system and their contribution to the development of the pro- or anti-tumor immune response in BC is highlighted. The presented data can potentially inform the development of new immunotherapy strategies in the treatment of patients with BC.
Collapse
Affiliation(s)
- Tatiana Gerashchenko
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Anastasia Frolova
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Researc, Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
- Tomsk State University, 36 Lenin Ave., Tomsk, 634050, Russia
| | - Marina Patysheva
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Anton Fedorov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Marina Stakheyeva
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Researc, Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Evgeny Denisov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Nadezda Cherdyntseva
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Researc, Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
- Tomsk State University, 36 Lenin Ave., Tomsk, 634050, Russia
| |
Collapse
|
31
|
Knutson KL. Regulation of Tumor Dendritic Cells by Programmed Cell Death 1 Pathways. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1397-1405. [PMID: 38621195 PMCID: PMC11027937 DOI: 10.4049/jimmunol.2300674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/18/2024] [Indexed: 04/17/2024]
Abstract
The advent of immune checkpoint blockade therapy has revolutionized cancer treatments and is partly responsible for the significant decline in cancer-related mortality observed during the last decade. Immune checkpoint inhibitors, such as anti-programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1), have demonstrated remarkable clinical successes in a subset of cancer patients. However, a considerable proportion of patients remain refractory to immune checkpoint blockade, prompting the exploration of mechanisms of treatment resistance. Whereas much emphasis has been placed on the role of PD-L1 and PD-1 in regulating the activity of tumor-infiltrating T cells, recent studies have now shown that this immunoregulatory axis also directly regulates myeloid cell activity in the tumor microenvironment including tumor-infiltrating dendritic cells. In this review, I discuss the most recent advances in the understanding of how PD-1, PD-L1, and programmed cell death ligand 2 regulate the function of tumor-infiltrating dendritic cells, emphasizing the need for further mechanistic studies that could facilitate the development of novel combination immunotherapies for improved cancer patient benefit.
Collapse
|
32
|
Song MS, Nam JH, Noh KE, Lim DS. Dendritic Cell-Based Immunotherapy: The Importance of Dendritic Cell Migration. J Immunol Res 2024; 2024:7827246. [PMID: 38628676 PMCID: PMC11019573 DOI: 10.1155/2024/7827246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/19/2024] Open
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that are crucial for maintaining self-tolerance, initiating immune responses against pathogens, and patrolling body compartments. Despite promising aspects, DC-based immunotherapy faces challenges that include limited availability, immune escape in tumors, immunosuppression in the tumor microenvironment, and the need for effective combination therapies. A further limitation in DC-based immunotherapy is the low population of migratory DC (around 5%-10%) that migrate to lymph nodes (LNs) through afferent lymphatics depending on the LN draining site. By increasing the population of migratory DCs, DC-based immunotherapy could enhance immunotherapeutic effects on target diseases. This paper reviews the importance of DC migration and current research progress in the context of DC-based immunotherapy.
Collapse
Affiliation(s)
- Min-Seon Song
- Department of Bioconvergence, Graduate School and Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Ji-Hee Nam
- Department of Bioconvergence, Graduate School and Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Kyung-Eun Noh
- Department of Bioconvergence, Graduate School and Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Dae-Seog Lim
- Department of Bioconvergence, Graduate School and Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| |
Collapse
|
33
|
Heras-Murillo I, Adán-Barrientos I, Galán M, Wculek SK, Sancho D. Dendritic cells as orchestrators of anticancer immunity and immunotherapy. Nat Rev Clin Oncol 2024; 21:257-277. [PMID: 38326563 DOI: 10.1038/s41571-024-00859-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2024] [Indexed: 02/09/2024]
Abstract
Dendritic cells (DCs) are a heterogeneous group of antigen-presenting innate immune cells that regulate adaptive immunity, including against cancer. Therefore, understanding the precise activities of DCs in tumours and patients with cancer is important. The classification of DC subsets has historically been based on ontogeny; however, single-cell analyses are now additionally revealing a diversity of functional states of DCs in cancer. DCs can promote the activation of potent antitumour T cells and immune responses via numerous mechanisms, although they can also be hijacked by tumour-mediated factors to contribute to immune tolerance and cancer progression. Consequently, DC activities are often key determinants of the efficacy of immunotherapies, including immune-checkpoint inhibitors. Potentiating the antitumour functions of DCs or using them as tools to orchestrate short-term and long-term anticancer immunity has immense but as-yet underexploited therapeutic potential. In this Review, we outline the nature and emerging complexity of DC states as well as their functions in regulating adaptive immunity across different cancer types. We also describe how DCs are required for the success of current immunotherapies and explore the inherent potential of targeting DCs for cancer therapy. We focus on novel insights on DCs derived from patients with different cancers, single-cell studies of DCs and their relevance to therapeutic strategies.
Collapse
Affiliation(s)
- Ignacio Heras-Murillo
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Irene Adán-Barrientos
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Galán
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Stefanie K Wculek
- Innate Immune Biology Laboratory, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
34
|
Manoutcharian K, Gevorkian G. Are we getting closer to a successful neoantigen cancer vaccine? Mol Aspects Med 2024; 96:101254. [PMID: 38354548 DOI: 10.1016/j.mam.2024.101254] [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: 10/24/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/16/2024]
Abstract
Although significant advances in immunotherapy have revolutionized the treatment of many cancer types over the past decade, the field of vaccine therapy, an important component of cancer immunotherapy, despite decades-long intense efforts, is still transmitting signals of promises and awaiting strong data on efficacy to proceed with regulatory approval. The field of cancer vaccines faces standard challenges, such as tumor-induced immunosuppression, immune response in inhibitory tumor microenvironment (TME), intratumor heterogeneity (ITH), permanently evolving cancer mutational landscape leading to neoantigens, and less known obstacles: neoantigen gain/loss upon immunotherapy, the timing and speed of appearance of neoantigens and responding T cell clonotypes and possible involvement of immune interference/heterologous immunity, in the complex interplay between evolving tumor epitopes and the immune system. In this review, we discuss some key issues related to challenges hampering the development of cancer vaccines, along with the current approaches focusing on neoantigens. We summarize currently well-known ideas/rationales, thus revealing the need for alternative vaccine approaches. Such a discussion should stimulate vaccine researchers to apply out-of-box, unconventional thinking in search of new avenues to deal with critical, often yet unaddressed challenges on the road to a new generation of therapeutics and vaccines.
Collapse
Affiliation(s)
- Karen Manoutcharian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), CDMX, Apartado Postal 70228, Cuidad Universitaria, Mexico DF, CP, 04510, Mexico.
| | - Goar Gevorkian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), CDMX, Apartado Postal 70228, Cuidad Universitaria, Mexico DF, CP, 04510, Mexico.
| |
Collapse
|
35
|
Peng S, Lin A, Jiang A, Zhang C, Zhang J, Cheng Q, Luo P, Bai Y. CTLs heterogeneity and plasticity: implications for cancer immunotherapy. Mol Cancer 2024; 23:58. [PMID: 38515134 PMCID: PMC10956324 DOI: 10.1186/s12943-024-01972-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024] Open
Abstract
Cytotoxic T lymphocytes (CTLs) play critical antitumor roles, encompassing diverse subsets including CD4+, NK, and γδ T cells beyond conventional CD8+ CTLs. However, definitive CTLs biomarkers remain elusive, as cytotoxicity-molecule expression does not necessarily confer cytotoxic capacity. CTLs differentiation involves transcriptional regulation by factors such as T-bet and Blimp-1, although epigenetic regulation of CTLs is less clear. CTLs promote tumor killing through cytotoxic granules and death receptor pathways, but may also stimulate tumorigenesis in some contexts. Given that CTLs cytotoxicity varies across tumors, enhancing this function is critical. This review summarizes current knowledge on CTLs subsets, biomarkers, differentiation mechanisms, cancer-related functions, and strategies for improving cytotoxicity. Key outstanding questions include refining the CTLs definition, characterizing subtype diversity, elucidating differentiation and senescence pathways, delineating CTL-microbe relationships, and enabling multi-omics profiling. A more comprehensive understanding of CTLs biology will facilitate optimization of their immunotherapy applications. Overall, this review synthesizes the heterogeneity, regulation, functional roles, and enhancement strategies of CTLs in antitumor immunity, highlighting gaps in our knowledge of subtype diversity, definitive biomarkers, epigenetic control, microbial interactions, and multi-omics characterization. Addressing these questions will refine our understanding of CTLs immunology to better leverage cytotoxic functions against cancer.
Collapse
Affiliation(s)
- Shengkun Peng
- Department of Radiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Aimin Jiang
- Department of Urology, Changhai hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and ImmunologySchool of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South University, Hunan, China.
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| | - Yifeng Bai
- Department of Oncology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
36
|
Wu TH, Lu YJ, Chiang MR, Chen PH, Lee YS, Shen MY, Chiang WH, Liu YC, Chuang CY, Amy Lin HC, Hu SH. Lung metastasis-Harnessed in-Situ adherent porous organic nanosponge-mediated antigen capture for A self-cascaded detained dendritic cells and T cell infiltration. Biomaterials 2024; 305:122443. [PMID: 38160627 DOI: 10.1016/j.biomaterials.2023.122443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/06/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
The infiltration of cytotoxic T lymphocytes promises to suppress the most irresistible metastatic tumor for immunotherapy, yet immune privilege and low immunogenic responses in these aggressive clusters often restrict lymphocyte recruitment. Here, an in situ adherent porous organic nanosponge (APON) doubles as organ selection agent and antigen captor to overcome immune privilege is developed. With selective organ targeting, the geometric effect of APON composed of disc catechol-functionalized covalent organic framework (COF) boosts the drug delivery to lung metastases. Along with a self-cascaded immune therapy, the therapeutic agents promote tumor release of damage-associated molecular patterns (DAMPs), and then, in situ deposition of gels to capture these antigens. Furthermore, APON with catechol analogs functions as a reservoir of antigens and delivers autologous DAMPs to detain dendritic cells, resulting in a sustained enhancement of immunity. This disc sponges (APON) at lung metastasis as antigen reservoirs and immune modulators effectively suppress the tumor in 60 days and enhanced the survival rate.
Collapse
Affiliation(s)
- Ting-Hsien Wu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Yu-Jen Lu
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Tao-Yuan 33305, Taiwan; The College of Medicine, Chang Gung University, Tao-Yuan 33302, Taiwan
| | - Min-Ren Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Pin-Hua Chen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Yu-Sheng Lee
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Ming-Yin Shen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300044, Taiwan; Department of Surgery, China Medical University Hsinchu Hospital, Hsinchu County, 30272, Taiwan
| | - Wen-Hsuan Chiang
- Department of Chemical Engineering, National Chung Hsing University, Taichung, 402, Taiwan
| | - Yu-Chen Liu
- Laboratory for Human Immunology (Single Cell Genomics), WPI Immunology Frontier Research Center, Osaka University, Osaka, 565-0871, Japan
| | - Chun-Yu Chuang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Hsiao-Chun Amy Lin
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Shang-Hsiu Hu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300044, Taiwan.
| |
Collapse
|
37
|
Hu Q, Wang S, Ma L, Sun Z, Liu Z, Deng S, Zhou J. Radiological assessment of immunotherapy effects and immune checkpoint-related pneumonitis for lung cancer. J Cell Mol Med 2024; 28:e17895. [PMID: 37525480 PMCID: PMC10902575 DOI: 10.1111/jcmm.17895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/02/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) therapy have revolutionized advanced lung cancer care. Interestingly, the host responses for patients received ICIs therapy are distinguishing from those with cytotoxic drugs, showing potential initial transient worsening of disease burden, pseudoprogression and delayed time to treatment response. Thus, a new imaging criterion to evaluate the response for immunotherapy should be developed. ICIs treatment is associated with unique adverse events, including potential life-threatening immune checkpoint inhibitor-related pneumonitis (ICI-pneumonitis) if treated patients are not managed promptly. Currently, the diagnosis and clinical management of ICI-pneumonitis remain challenging. As the clinical manifestation is often nonspecific, computed tomography (CT) scan and X-ray films play important roles in diagnosis and triage. This article reviews the complications of immunotherapy in lung cancer and illustrates various radiologic patterns of ICI-pneumonitis. Additionally, it is tried to differentiate ICI-pneumonitis from other pulmonary pathologies common to lung cancer such as radiation pneumonitis, bacterial pneumonia and coronavirus disease of 2019 (COVID-19) infection in recent months. Maybe it is challenging to distinguish radiologically but clinical presentation may help.
Collapse
Affiliation(s)
- Qiongjie Hu
- Department of Radiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Shaofang Wang
- Department of Radiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Li Ma
- Department of Orthopedics, Songzi HospitalRenmin Hospital of Wuhan UniversityWuhanChina
| | - Ziyan Sun
- Department of Radiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zilin Liu
- Department of OrthopedicsRenmin Hospital of Wuhan UniversityWuhanChina
| | - Shuang Deng
- Department of OrthopedicsRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jianlin Zhou
- Department of OrthopedicsRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
38
|
Zhu X, Zheng W, Wang X, Li Z, Shen X, Chen Q, Lu Y, Chen K, Ai S, Zhu Y, Guan W, Yao S, Liu S. Enhanced Photodynamic Therapy Synergizing with Inhibition of Tumor Neutrophil Ferroptosis Boosts Anti-PD-1 Therapy of Gastric Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307870. [PMID: 38233204 DOI: 10.1002/advs.202307870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/19/2023] [Indexed: 01/19/2024]
Abstract
For tumor treatment, the ultimate goal in tumor therapy is to eliminate the primary tumor, manage potential metastases, and trigger an antitumor immune response, resulting in the complete clearance of all malignant cells. Tumor microenvironment (TME) refers to the local biological environment of solid tumors and has increasingly become an attractive target for cancer therapy. Neutrophils within TME of gastric cancer (GC) spontaneously undergo ferroptosis, and this process releases oxidized lipids that limit T cell activity. Enhanced photodynamic therapy (PDT) mediated by di-iodinated IR780 (Icy7) significantly increases the production of reactive oxygen species (ROS). Meanwhile, neutrophil ferroptosis can be triggered by increased ROS generation in the TME. In this study, a liposome encapsulating both ferroptosis inhibitor Liproxstatin-1 and modified photosensitizer Icy7, denoted LLI, significantly inhibits tumor growth of GC. LLI internalizes into MFC cells to generate ROS causing immunogenic cell death (ICD). Simultaneously, liposome-deliver Liproxstatin-1 effectively inhibits the ferroptosis of tumor neutrophils. LLI-based immunogenic PDT and neutrophil-targeting immunotherapy synergistically boost the anti-PD-1 treatment to elicit potent TME and systemic antitumor immune response with abscopal effects. In conclusion, LLI holds great potential for GC immunotherapy.
Collapse
Affiliation(s)
- Xudong Zhu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Wenxuan Zheng
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xingzhou Wang
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Zhiyan Li
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xiaofei Shen
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Qi Chen
- China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, 210008, China
| | - Yanjun Lu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Kai Chen
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Shichao Ai
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yun Zhu
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Wenxian Guan
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Shankun Yao
- State Key Laboratory of Coordination Chemistry, Coordination Chemistry Institute, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Song Liu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| |
Collapse
|
39
|
Guo H, Wang T, Yu J, Shi Z, Liang M, Chen S, He T, Yan H. Vitreous Olink proteomics reveals inflammatory biomarkers for diagnosis and prognosis of traumatic proliferative vitreoretinopathy. Front Immunol 2024; 15:1355314. [PMID: 38455059 PMCID: PMC10917961 DOI: 10.3389/fimmu.2024.1355314] [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: 12/13/2023] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
Background The aim of this study was to identify inflammatory biomarkers in traumatic proliferative vitreoretinopathy (TPVR) patients and further validate the expression curve of particular biomarkers in the rabbit TPVR model. Methods The Olink Inflammation Panel was used to compare the differentially expressed proteins (DEPs) in the vitreous of TPVR patients 7-14 days after open globe injury (OGI) (N = 19) and macular hole patients (N = 22), followed by correlation analysis between DEPs and clinical signs, protein-protein interaction (PPI) analysis, area under the receiver operating characteristic curve (AUC) analysis, and function enrichment analysis. A TPVR rabbit model was established and expression levels of candidate interleukin family members (IL-6, IL-7, and IL-33) were measured by enzyme-linked immunosorbent assay (ELISA) at 0, 1, 3, 7, 10, 14, and 28 days after OGI. Results Forty-eight DEPs were detected between the two groups. Correlation analysis showed that CXCL5, EN-RAGE, IL-7, ADA, CD5, CCL25, CASP8, TWEAK, and IL-33 were significantly correlated with clinical signs including ocular wound characteristics, PVR scoring, PVR recurrence, and final visual acuity (R = 0.467-0.699, p < 0.05), and all with optimal AUC values (0.7344-1). Correlations between DEP analysis and PPI analysis further verified that IL-6, IL-7, IL-8, IL-33, HGF, and CXCL5 were highly interactive (combined score: 0.669-0.983). These DEPs were enriched in novel pathways such as cancer signaling pathway (N = 14, p < 0.000). Vitreous levels of IL-6, IL-7, and IL-33 in the rabbit TPVR model displayed consistency with the trend in Olink data, all exhibiting marked differential expression 1 day following the OGI. Conclusion IL-7, IL-33, EN-RAGE, TWEAK, CXCL5, and CD5 may be potential biomarkers for TPVR pathogenesis and prognosis, and early post-injury may be an ideal time for TPVR intervention targeting interleukin family biomarkers.
Collapse
Affiliation(s)
- Haixia Guo
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Tian Wang
- Shaanxi Eye Hospital, Xi’an People’s Hospital (Xi’an Fourth Hospital), Affiliated People’s Hospital of Northwest University, Xi’an, Shaanxi, China
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Jinguo Yu
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhemin Shi
- Department of Histology and Developmental Biology, Tianjin Medical University, Tianjin, China
| | - Minghui Liang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Ocular Trauma, Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| | - Siyue Chen
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Ocular Trauma, Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Tiangeng He
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Ocular Trauma, Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
40
|
Shapir Itai Y, Barboy O, Salomon R, Bercovich A, Xie K, Winter E, Shami T, Porat Z, Erez N, Tanay A, Amit I, Dahan R. Bispecific dendritic-T cell engager potentiates anti-tumor immunity. Cell 2024; 187:375-389.e18. [PMID: 38242085 DOI: 10.1016/j.cell.2023.12.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/24/2023] [Accepted: 12/05/2023] [Indexed: 01/21/2024]
Abstract
Immune checkpoint inhibition treatment using aPD-1 monoclonal antibodies is a promising cancer immunotherapy approach. However, its effect on tumor immunity is narrow, as most patients do not respond to the treatment or suffer from recurrence. We show that the crosstalk between conventional type I dendritic cells (cDC1) and T cells is essential for an effective aPD-1-mediated anti-tumor response. Accordingly, we developed a bispecific DC-T cell engager (BiCE), a reagent that facilitates physical interactions between PD-1+ T cells and cDC1. BiCE treatment promotes the formation of active dendritic/T cell crosstalk in the tumor and tumor-draining lymph nodes. In vivo, single-cell and physical interacting cell analysis demonstrates the distinct and superior immune reprogramming of the tumors and tumor-draining lymph nodes treated with BiCE as compared to conventional aPD-1 treatment. By bridging immune cells, BiCE potentiates cell circuits and communication pathways needed for effective anti-tumor immunity.
Collapse
Affiliation(s)
- Yuval Shapir Itai
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Oren Barboy
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ran Salomon
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Akhiad Bercovich
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ken Xie
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Eitan Winter
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Tamar Shami
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ziv Porat
- Flow Cytometry Unit, Life Science Core Facility, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Neta Erez
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Amos Tanay
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ido Amit
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Rony Dahan
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
41
|
Zhang W, Xu X, Zhang R, Tian Y, Ma X, Wang X, Jiang Y, Man C. Stress-Induced Immunosuppression Inhibits Regional Immune Responses in Chicken Adipose Tissue Partially through Suppressing T Cells by Up-Regulating Steroid Metabolism. Animals (Basel) 2024; 14:225. [PMID: 38254394 PMCID: PMC10812502 DOI: 10.3390/ani14020225] [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: 11/21/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Lipid metabolism plays an important role in maintaining lipid homeostasis and regulating immune functions. However, the regulations and mechanisms of lipid metabolism on the regional immune function of avian adipose tissue (AT) have not been reported. In this study, qRT-PCR was used to investigate the changes and relationships of different lipid metabolism pathways in chicken AT during stress-induced immunosuppression (SIIS) inhibiting immune response to Newcastle disease virus vaccine, then the miRNA regulation patterns of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) gene and its potential applications were further identified. The results showed that AT actively responded to SIIS, and ATGL, CPT1A and HMGCR were all the key genes involved in the processes of SIIS inhibiting the immune responses. SIIS significantly inhibited the natural and specific immune phases of the primary immune response and the initiation phase of the secondary immune response in AT by suppressing T cells by up-regulating steroid anabolism. Moreover, steroid metabolism could play dual roles in regulating the regional immune functions of AT. The miR-29a/c-3p-HMGCR network was a potential regulation mechanism of steroid metabolism in AT, and serum circulating miR-29a/c-3p had the potential as molecular markers. The study can provide valuable references for an in-depth investigation of the regional immune functions regulated by lipid metabolism in AT.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chaolai Man
- College of Life Science and Technology, Harbin Normal University, Harbin 150025, China; (W.Z.); (X.X.); (R.Z.); (Y.T.); (X.M.); (X.W.); (Y.J.)
| |
Collapse
|
42
|
Li Y, Wu Y, Fang Z, Zhang Y, Ding H, Ren L, Zhang L, Gong Q, Gu Z, Luo K. Dendritic Nanomedicine with Boronate Bonds for Augmented Chemo-Immunotherapy via Synergistic Modulation of Tumor Immune Microenvironment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307263. [PMID: 37743633 DOI: 10.1002/adma.202307263] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/02/2023] [Indexed: 09/26/2023]
Abstract
Unsatisfied tumor accumulation of chemotherapeutic drugs and a complicated immunosuppressive microenvironment diminish the immune response rate and the therapeutic effect. Surface modification of these drugs with target ligands can promote their cellular internalization, but the modified drugs may be subjected to unexpected immune recognition and clearance. Herein, a phenylboronic acid (PBA) group-shieldable dendritic nanomedicine that integrates an immunogenic cell death (ICD)-inducing agent (epirubicin, Epi) and an indoleamine 2,3-dioxgenase 1 (IDO1) inhibitor (NLG919) is reported for tumor chemo-immunotherapy. This NLG919-loaded Epi-conjugated PEGylated dendrimers bridged with boronate bonds (NLG919@Epi-DBP) maintains a stable nanostructure during circulation. Under a moderate acidic condition, the PBA group exposes to the sialic acid residue on the tumor cell membrane to enhance the internalization and penetration of NLG919@Epi-DBP. At pH 5.0, NLG919@Epi-DBP rapidly disassembles to release the incorporated Epi and NLG919. Epi triggers robust ICD of tumor cells that evokes strong immune response. In addition, inhibition of the IDO1 activity downregulates the metabolism of L-tryptophan to kynurenine, leading to a reduction in the recruitment of immunosuppressive cells and modulation of the tumor immune microenvironment. Collectively, this promising strategy has been demonstrated to evoke robust immune response as well as remodel the immunosuppressive microenvironment for an enhanced chemo-immunotherapeutic effect.
Collapse
Affiliation(s)
- Yunkun Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yahui Wu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zaixiang Fang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuxin Zhang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Haitao Ding
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Long Ren
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lu Zhang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, 361021, China
| | - Zhongwei Gu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
43
|
Sun Y, Li Y, Jiang C, Liu C, Song Y. SLC7A2-Mediated Lysine Catabolism Inhibits Immunosuppression in Triple Negative Breast Cancer. Crit Rev Eukaryot Gene Expr 2024; 34:31-43. [PMID: 38842202 DOI: 10.1615/critreveukaryotgeneexpr.2024052503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Breast cancer is one of the most common malignant tumors worldwide. SLC7A2 is abnormally expressed in multiple cancers. However, its potential in triple negative breast cancer (TNBC) is still unclear. The purpose of this study was to investigate the roles of SLC7A2 and its underlying molecular mechanisms in TNBC. mRNA expression was detected by RT-qPCR. Protein expression was detected by western blot. Co-localization of ACOX1 and TCF1 was determined using FISH assay. Histone crotonylation was performed using in vitro histone crotonylation assay. Functional analysis was performed using CCK-8 and flow cytometry assays. Xenograft assay was conducted to further verify the role of SLC7A2 in TNBC. CD8A expression was detected using immunohistochemistry. We found that SLC7A2 is downregulated in TNBC tumors. Low levels are associated with advanced stages and lymph node metastasis. SLC7A2 expression is positively correlated with CD8A. SLC7A2-mediated lysine catabolism drives the activation of CD8+ T cells. Moreover, SLC7A2 promotes histone crotonylation via upregulating ACOX1. It also promotes interaction between ACOX1 and TCF1, thus promoting antitumor T cell immunity. Additionally, overexpression of SLC7A2 activates CD8+ T cells and enhances the chemosensitivity of anti-PD-1 therapies in vivo. In conclusion, SLC7A2 may function as an antitumor gene in TNBC by activating antitumor immunity, suggesting SLC7A2/ACOX1/TCF1 signaling as a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yuanyuan Sun
- Tianjin Medical University Cancer Institute & Hospital
| | - Yaqing Li
- Department of Breast Pathology and Lab, Tianjin Medical University Cancer Institute & Hospital; National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Chengying Jiang
- Department of Breast Pathology and Lab, Tianjin Medical University Cancer Institute & Hospital; National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Chenying Liu
- Department of Breast Pathology and Lab, Tianjin Medical University Cancer Institute & Hospital; National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Yuanming Song
- Department of Breast Pathology and Lab, Tianjin Medical University Cancer Institute & Hospital; National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| |
Collapse
|
44
|
Dallmann J, Freitag J, Jung C, Khinvasara K, Merz L, Peters D, Schork M, Beck J. CIMT 2023: report on the 20th Annual Meeting of the Association for Cancer Immunotherapy. IMMUNO-ONCOLOGY TECHNOLOGY 2023; 20:100397. [PMID: 37876518 PMCID: PMC10590812 DOI: 10.1016/j.iotech.2023.100397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
The Association for Cancer Immunotherapy (CIMT) celebrated the 20th anniversary of the CIMT Annual Meeting. CIMT2023 was held 3-5 May 2023 in Mainz, Germany. 1051 academic and clinical professionals from over 30 countries attended the meeting and discussed the latest advances in cancer immunology and immunotherapy research. This report summarizes the highlights of CIMT2023.
Collapse
Affiliation(s)
- J. Dallmann
- Immunotherapies & Preclinical Research, BioNTech SE, Mainz
| | - J. Freitag
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - C. Jung
- BioNTech Cell & Gene Therapies GmbH, Mainz
| | - K. Khinvasara
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - L. Merz
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - D. Peters
- Immunotherapies & Preclinical Research, BioNTech SE, Mainz
| | - M. Schork
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - J.D. Beck
- Immunotherapies & Preclinical Research, BioNTech SE, Mainz
| |
Collapse
|
45
|
Dombroski JA, Antunovic M, Schaffer KR, Hurley PJ, King MR. Activation of Dendritic Cells Isolated from the Blood of Patients with Prostate Cancer by Ex Vivo Fluid Shear Stress Stimulation. Curr Protoc 2023; 3:e933. [PMID: 38047658 PMCID: PMC11178276 DOI: 10.1002/cpz1.933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Prostate cancer is one of the most common cancers among men in the United States and a leading cause of cancer-related death in men. Treatment options for patients with advanced prostate cancer include hormone therapies, chemotherapies, radioligand therapies, and immunotherapies. Provenge (sipuleucel-T) is an autologous cancer-vaccine-based immunotherapy approved for men with asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer (mCRPC). Administration of sipuleucel-T involves leukapheresis of patient blood to isolate antigen-presenting cells (APCs), including dendritic cells (DCs), and subsequent incubation of isolated APCs with both an antigen, prostatic acid phosphatase (PAP), and granulocyte macrophage-colony stimulating factor (GM-CSF) before their infusion back into the patient. Although sipuleucel-T has been shown to improve overall survival, other meaningful outcomes, such as prostate-specific antigen (PSA) levels and radiographic response, are inconsistent. This lack of robust response may be due to limited ex vivo activation of DCs using current protocols. Earlier studies have shown that many cell types can be activated ex vivo by external forces such as fluid shear stress (FSS). We hypothesize that novel fluid shear stress technologies and methods can be used to improve ex vivo efficacy of prostate cancer DC activation in prostate cancer. Herein, we report a new protocol for activating DCs from patients with prostate cancer using ex vivo fluid shear stress. Ultimately, the goal of these studies is to improve DC activation to expand the efficacy of therapies such as sipuleucel-T. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Sample collection and DC isolation Basic Protocol 2: Determination and application of fluid shear stress Basic Protocol 3: Flow cytometry analysis of DCs after FSS stimulation.
Collapse
Affiliation(s)
- Jenna A. Dombroski
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| | - Monika Antunovic
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kerry R. Schaffer
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt-Ingram Cancer Center, Nashville, TN, United States
| | - Paula J. Hurley
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt-Ingram Cancer Center, Nashville, TN, United States
| | - Michael R. King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
46
|
Liao ZX, Hsu SH, Tang SC, Kempson I, Yang PC, Tseng SJ. Potential targeting of the tumor microenvironment to improve cancer virotherapy. Pharmacol Ther 2023; 250:108521. [PMID: 37657673 DOI: 10.1016/j.pharmthera.2023.108521] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 09/03/2023]
Abstract
In 2015, oncolytic virotherapy was approved for clinical use, and in 2017, recombinant adeno-associated virus (AAV) delivery was also approved. However, systemic administration remains challenging due to the limited number of viruses that successfully reach the target site. Although the US Food and Drug Administration (FDA) permits the use of higher doses of AAV to achieve greater rates of transduction, most AAV still accumulates in the liver, potentially leading to toxicity there and elsewhere. Targeting the tumor microenvironment is a promising strategy for cancer treatment due to the critical role of the tumor microenvironment in controlling tumor progression and influencing the response to therapies. Newly discovered evidence indicates that administration routes focusing on the tumor microenvironment can promote delivery specificity and transduction efficacy within the tumor. Here, we review approaches that involve modifying viral surface features, modulating the immune system, and targeting the physicochemical characteristics in tumor microenvironment to regulate therapeutic delivery. Targeting tumor acidosis presents advantages that can be leveraged to enhance virotherapy outcomes and to develop new therapeutic approaches that can be integrated with standard treatments.
Collapse
Affiliation(s)
- Zi-Xian Liao
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei 10051, Taiwan
| | - Shiue-Cheng Tang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Ivan Kempson
- Future Industries Institute, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Pan-Chyr Yang
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 10051, Taiwan
| | - S Ja Tseng
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 10051, Taiwan; National Taiwan University YongLin Institute of Health, National Taiwan University, Taipei 10051, Taiwan; Program in Precision Health and Intelligent Medicine, Graduate School of Advanced Technology, National Taiwan University, Taipei 10051, Taiwan.
| |
Collapse
|
47
|
Kumari S, Sahu S, Singh B, Gupta S, Kureel AK, Srivastava A, Rikhari D, Srivastava S, Rai AK. HIF-1α regulates the expression of the non-conventional isoform of the cd5 gene in T cells under the hypoxic condition: A potential mechanism for CD5 neg/low phenotype of infiltrating cells in solid tumors. Cell Immunol 2023; 391-392:104755. [PMID: 37544247 DOI: 10.1016/j.cellimm.2023.104755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/08/2023]
Abstract
CD5, a T-cell receptor (TCR) negative regulator, is reduced on the surface of CD8+ lymphocytes in the tumor microenvironment (TME). Reduced surface CD5 expression (sCD5) occurs due to the preferential transcription of HERV-E derived exon E1B, i.e., anon-conventional formofthe cd5gene instead of its conventional exon E1A. A tumor employs several mechanisms to evade anti-tumor response, and hypoxia is one such mechanism that prevails in the TME and modulates the infiltrated T lymphocytes. We identified hypoxia response elements (HREs) upstream of E1B. We showed binding of HIF-1α onto these HREs and increased E1B mRNA expression in hypoxic T cells. This results in decreased sCD5 expression and increased cytoplasmic accumulation in T cells. We also validated our study in a solid tumor, i.e., colorectal cancer (CRC) patient samples. This hypoxia-driven mechanism reduces the surface CD5 expression on infiltrated T-cells in solid tumors.
Collapse
Affiliation(s)
- Smita Kumari
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India
| | - Srishti Sahu
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India
| | - Bharat Singh
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India
| | - Swarnima Gupta
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India
| | - Amit Kumar Kureel
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India
| | - Ankit Srivastava
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India
| | - Deeksha Rikhari
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India
| | - Sameer Srivastava
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India
| | - Ambak Kumar Rai
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad (M.N.N.I.T. Allahabad), Prayagraj, Uttar Pradesh 211004, India.
| |
Collapse
|
48
|
Gong Y, Ke Y, Yu Z, Pan J, Zhou X, Jiang Y, Zhou M, Zeng H, Geng X, Hu G. Identified RP2 as a prognostic biomarker for glioma, facilitating glioma pathogenesis mainly via regulating tumor immunity. Aging (Albany NY) 2023; 15:8155-8184. [PMID: 37602882 PMCID: PMC10497014 DOI: 10.18632/aging.204962] [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: 04/03/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023]
Abstract
Glioma is the most common primary intracranial tumor in the central nervous system, with a high degree of malignancy and poor prognosis, easy to recur, difficult to cure. The mutation of Retinitis Pigmentosa 2 (RP2) can cause retinitis pigmentosa, it is a prognostic factor of osteosarcoma, however, its role in glioma remains unclear. Based on the data from TCGA and GTEx, we identified RP2 as the most related gene for glioma by WGCNA, and used a series of bioinformatics analyses including LinkedOmics, GSCA, CTD, and so on, to explore the expression of RP2 in glioma and the biological functions it is involved in. The results showed that RP2 was highly expressed in glioma, and its overexpression could lead to poor prognosis. In addition, the results of enrichment analysis showed that RP2 was highly correlated with cell proliferation and immune response. And then, we found significant enrichment of Macrophages among immune cells. Furthermore, our experiments have confirmed that Macrophages can promote the development of glioma by secreting or influencing the secretion of some cytokines. Moreover, we investigated the influence of RP2 on the immunotherapy of glioma and the role of m6A modification in the influence of RP2 on glioma. Ultimately, we determined that RP2 is an independent prognostic factor that is mainly closely related to immune for glioma.
Collapse
Affiliation(s)
- Yiyang Gong
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Yun Ke
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Zichuan Yu
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Jingying Pan
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Xuanrui Zhou
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Yike Jiang
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Minqin Zhou
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Hong Zeng
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Xitong Geng
- Second College of Clinical Medicine, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Guowen Hu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| |
Collapse
|
49
|
Lang M, Krump C, Meshcheryakova A, Tam-Amersdorfer C, Schwarzenberger E, Passegger C, Connolly S, Mechtcheriakova D, Strobl H. Microenvironmental and cell intrinsic factors governing human cDC2 differentiation and monocyte reprogramming. Front Immunol 2023; 14:1216352. [PMID: 37539048 PMCID: PMC10395083 DOI: 10.3389/fimmu.2023.1216352] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/14/2023] [Indexed: 08/05/2023] Open
Abstract
cDC2s occur abundantly in peripheral tissues and arise from circulating blood cDC2s. However, the factors governing cDC2 differentiation in tissues, especially under inflammatory conditions, remained poorly defined. We here found that psoriatic cDC2s express the efferocytosis receptor Axl and exhibit a bone morphogenetic protein (BMP) and p38MAPK signaling signature. BMP7, strongly expressed within the lesional psoriatic epidermis, cooperates with canonical TGF-β1 signaling for inducing Axl+cDC2s from blood cDC2s in vitro. Moreover, downstream induced p38MAPK promotes Axl+cDC2s at the expense of Axl+CD207+ Langerhans cell differentiation from blood cDC2s. BMP7 supplementation allowed to model cDC2 generation and their further differentiation into LCs from CD34+ hematopoietic progenitor cells in defined serum-free medium. Additionally, p38MAPK promoted the generation of another cDC2 subset lacking Axl but expressing the non-classical NFkB transcription factor RelB in vitro. Such RelB+cDC2s occurred predominantly at dermal sites in the inflamed skin. Finally, we found that cDC2s can be induced to acquire high levels of the monocyte lineage identity factor kruppel-like-factor-4 (KLF4) along with monocyte-derived DC and macrophage phenotypic characteristics in vitro. In conclusion, inflammatory and psoriatic epidermal signals instruct blood cDC2s to acquire phenotypic characteristics of several tissue-resident cell subsets.
Collapse
Affiliation(s)
- Magdalena Lang
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Corinna Krump
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Anastasia Meshcheryakova
- Insitute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Carmen Tam-Amersdorfer
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Elke Schwarzenberger
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Christina Passegger
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Sally Connolly
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Diana Mechtcheriakova
- Insitute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Herbert Strobl
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
50
|
Nierengarten MB. Immunotherapy nonresponders may lack crucial immune cells. Cancer 2023; 129:1951-1952. [PMID: 37300811 DOI: 10.1002/cncr.34883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
|