1
|
Li Q, Sheng M, Chen Y, Yi Q, Yang Z, Chen T. Comprehensive immunogenomic landscape analysis unveils CD33 + myeloid cell-driven immunomodulatory signatures in melanoma development. Pathol Res Pract 2025; 270:155981. [PMID: 40300524 DOI: 10.1016/j.prp.2025.155981] [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: 02/13/2025] [Revised: 04/07/2025] [Accepted: 04/16/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND Understanding the causal relationships between immune cell populations and cancer development remains a critical challenge in tumor immunology. METHODS We employed Mendelian Randomization analysis leveraging genome-wide association studies of 612 immune cell traits and 91 cancer types to systematically evaluate causal associations. Single-cell RNA sequencing and computational deconvolution analyses were performed to characterize myeloid cell subpopulations in melanoma samples. FINDINGS Our analysis revealed significant relationships between specific immune cell subsets and cancer risk, particularly highlighting the role of CD33 + myeloid cells in melanoma pathogenesis. Single-cell RNA sequencing identified distinct CD33high myeloid subpopulations characterized by elevated expression of complement cascade components and chemokine signaling pathways. Through computational deconvolution of The Cancer Genome Atlas melanoma cohort, we demonstrated that elevated CD33high monocyte abundance correlates with increased immune dysfunction scores, reduced CD8 + T cell infiltration, and poor survival outcomes. INTERPRETATION Here we delineate the multifaceted mechanisms through which CD33 + myeloid cell populations orchestrate perturbations in the tumor-immune microenvironmental landscape, manifesting in compromised immunosurveillance and enhanced tumor progression. Our findings illuminate novel therapeutic opportunities through targeted modulation of myeloid cell function, while providing a systematic framework for understanding the complex interplay between immune cell populations and oncogenic processes.
Collapse
Affiliation(s)
- Qinke Li
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400010, China; Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Min Sheng
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yiqian Chen
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400010, China; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing 400010, China
| | - Qiang Yi
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400010, China; Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Zhu Yang
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Tong Chen
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400010, China; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing 400010, China.
| |
Collapse
|
2
|
Jayab NA, Abed A, Talaat IM, Hamoudi R. The molecular mechanism of NF-κB dysregulation across different subtypes of renal cell carcinoma. J Adv Res 2025; 72:501-514. [PMID: 39094893 DOI: 10.1016/j.jare.2024.07.030] [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: 04/17/2024] [Revised: 06/27/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND The nuclear factor kappa B (NF-κB) is a critical pathway that regulates various cellular functions, including immune response, proliferation, growth, and apoptosis. Furthermore, this pathway is tightly regulated to ensure stability in the presence of immunogenic triggers or genotoxic stimuli. The lack of control of the NF-κB pathway can lead to the initiation of different diseases, mainly autoimmune diseases and cancer, including Renal cell carcinoma (RCC). RCC is the most common type of kidney cancer and is characterized by complex genetic composition and elusive molecular mechanisms. AIM OF REVIEW The current review summarizes the mechanism of NF-κB dysregulation in different subtypes of RCC and its impact on pathogenesis. KEY SCIENTIFIC CONCEPT OF REVIEW This review highlights the prominent role of NF-κB in RCC development and progression by driving the expression of multiple genes and interplaying with different pathways, including the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway. In silico analysis of RCC cohorts and molecular studies have revealed that multiple NF-κB members and target genes are dysregulated. The dysregulation includes receptors such as TLR2, signal-transmitting members including RelA, and target genes, for instance, HIF-1α. The lack of effective regulatory mechanisms results in a constitutively active NF-κB pathway, which promotes cancer growth, migration, and survival. In this review, we comprehensively summarize the role of dysregulated NF-κB-related genes in the most common subtypes of RCC, including clear cell RCC (ccRCC), chromophobe RCC (chRCC), and papillary RCC (PRCC).
Collapse
Affiliation(s)
- Nour Abu Jayab
- Research Institute for Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Alaa Abed
- Research Institute for Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; ASPIRE Precision Medicine Research Institute Abu Dhabi, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Iman M Talaat
- Research Institute for Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, 27272 Sharjah, United Arab Emirates; Pathology Department, Faculty of Medicine, Alexandria University, 21131 Alexandria, Egypt.
| | - Rifat Hamoudi
- Research Institute for Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Center of Excellence for Precision Medicine, Research Institute of Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, 27272 Sharjah, United Arab Emirates; BIMAI-Lab, Biomedically Informed Artificial Intelligence Laboratory, University of Sharjah, 27272 Sharjah, United Arab Emirates; Division of Surgery and Interventional Science, University College London, London, United Kingdom; ASPIRE Precision Medicine Research Institute Abu Dhabi, University of Sharjah, 27272 Sharjah, United Arab Emirates.
| |
Collapse
|
3
|
Peng W, Yang Z, Yan R, Mu L, Li L, Jin S, Tan S. SUMOylation-regulated genes in colon cancer: expression patterns and clinical implications. Discov Oncol 2025; 16:837. [PMID: 40394398 PMCID: PMC12092877 DOI: 10.1007/s12672-025-02614-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025] Open
Abstract
Colon cancer (CRC) demonstrates significant heterogeneity, and identifying effective biomarkers can advance the development of precision therapies. Emerging evidence implicates SUMOylation-regulated genes as pivotal regulators of cancer-associated pathways, yet their prognostic potential and therapeutic implications in CRC remain unexplored. A comprehensive analysis of SUMOylation-regulated gene expression, clinical and prognostic value in CRC was performed using transcriptomic data from TCGA-COAD and GEO datasets. We identified 46 differentially expressed SUMOylation-regulated genes (33 upregulated, 13 downregulated) in CRC tumors versus normal tissues. Unsupervised clustering based on 216 SUMOylation-related genes stratified CRC patients into two distinct subtypes: SUMO Cluster 1 (aggressive phenotype, poor prognosis) and SUMO Cluster 2 (favorable prognosis). Cluster 1 exhibited advanced tumor stages (N-stage, p < 0.05) and may present an immunosuppressive microenvironment marked by reduced HLA/immune checkpoint gene expression, while Cluster 2 showed enhanced anti-tumor immunity (activated dendritic cells, γδ T cells). A five-gene SUMOylation-based prognostic signature (MC1R, LRRC4C, SAGE1, GJB6, HOXC5) was developed, and patients were divided into high Riskscore and low Riskscore groups with significant survival differences (log-rank p < 0.05). The nomogram integrating risk score, age, and stage demonstrated robust predictive accuracy (C-index = 0.763, AUC = 0.728-0.785). Nomoscore-high patients exhibited resistance to AMG.706 and ABT.888, suggesting therapeutic vulnerabilities. These findings highlight SUMOylation plays a critical role in CRC heterogeneity, immune modulation, and prognosis, offering a novel biomarker system for risk stratification and personalized therapy.
Collapse
Affiliation(s)
- Wen Peng
- Department of Oncology, The People's Hospital of Guizhou Province, Guiyang, China
| | - Zirong Yang
- Institute of Life Science, eBond Pharmaceutical Technology Ltd, Chengdu, China
| | - Ru Yan
- Department of Oncology, The People's Hospital of Guizhou Province, Guiyang, China
| | - Lan Mu
- Department of Oncology, The People's Hospital of Guizhou Province, Guiyang, China
| | - Lan Li
- Department of General Medicine, The People's Hospital of Guizhou Province, Guiyang, China
| | - Shan Jin
- Department of Oncology, The People's Hospital of Guizhou Province, Guiyang, China
| | - Shisheng Tan
- Department of Oncology, The People's Hospital of Guizhou Province, Guiyang, China.
| |
Collapse
|
4
|
Wei Y, Jia H, Guo X, Zhang H, Yang X, Can C, He N, Wu H, Liu W, Ma D. A novel LncRNA risk model for disulfidptosis-related prognosis prediction and response to chemotherapy in acute myeloid leukemia. Sci Rep 2025; 15:16995. [PMID: 40379722 PMCID: PMC12084325 DOI: 10.1038/s41598-025-01730-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/07/2025] [Indexed: 05/19/2025] Open
Abstract
Acute myeloid leukemia (AML), the most prevalent acute leukemia in adults, is characterized by its heterogeneity, which contributes to a poor prognosis and high recurrence rate. Recently, a unique form of cell death, called disulfidptosis, has been identified, which could transforming our understanding of and strategy for cancer treatment. Consequently, further inquiry is necessary to explore the possible link between disulfidptosis and AML. To facilitate this analysis, the researchers obtained single-cell RNA sequencing (scRNA-seq) data from AML patients using the Gene Expression Omnibus (GEO) database. By applying the Cox proportional hazards model and least absolute shrinkage and selection operator (LASSO) regression analysis, we created a signature of disulfidptosis-related long non-coding RNAs (DRLs). This predictive model was established based on six specific DRLs (AC005076.1, AP002807.1, HDAC4-AS1, L3MBTL4-AS1, LINC01694, and THAP9-AS1). The utility of this model in forecasting the prognosis of AML patients was corroborated by the receiver operating characteristic (ROC) curve. Moreover, significant variations in the biological functions and signaling pathways were discovered by gene ontology (GO) and Gene Set Enrichment Analysis (GSEA). To further investigate the relationship between immune infiltration, the study assessed variations in immune checkpoint expression and immune cell subset infiltration. Additionally, we used real-time quantitative PCR (RT-qPCR) to detect lncRNA expression in AML and healthy control to substantiate our analysis results. In conclusion, the results of this study may help discover novel therapeutic targets and prognostic biomarkers for AML, paving the way for customized precision chemotherapy.
Collapse
MESH Headings
- Humans
- RNA, Long Noncoding/genetics
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/mortality
- Prognosis
- Female
- Male
- ROC Curve
- Middle Aged
- Gene Expression Regulation, Leukemic
- Biomarkers, Tumor/genetics
- Proportional Hazards Models
- Disulfidptosis
Collapse
Affiliation(s)
- Yihong Wei
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Hexiao Jia
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Xiaodong Guo
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Hailei Zhang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Xinyu Yang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Can Can
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Na He
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Hanyang Wu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Wancheng Liu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Daoxin Ma
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
5
|
Modak S, Aktar T, Majumder D, Singha AK, Maiti D. A systematic review on leptin's role in defining cancer: special emphasis on immunomodulation, inflammation, and therapeutic interventions. Genes Immun 2025:10.1038/s41435-025-00333-7. [PMID: 40374921 DOI: 10.1038/s41435-025-00333-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/22/2025] [Accepted: 04/30/2025] [Indexed: 05/18/2025]
Abstract
Leptin, an adipokine related to obesity, is mainly known for its role in regulating energy homeostasis and appetite by working via the leptin receptor. Recently, different groups have demonstrated that apart from adipocytes, specific cell types associated with cancer and tumor microenvironments express leptin and leptin receptors. This tumor microenvironment-associated leptin-leptin receptor signaling contributes to the different hallmarks of cancer, ranging from inflammatory changes to metastasis. Eventually, it has also been reported that high serum level of leptin, a characteristic of obese people, is linked to enhanced tumor growth. On the other hand, leptin can influence both innate as well as adaptive immunity related to cancer. Overall, leptin's role in modulating cancer is controversial. So, in this review, we summarized the role of leptin in shaping different forms of cancer that are influenced by leptin-leptin receptor signaling with special emphasis on immunomodulation and inflammatory events and also discussed the possible therapeutic interventions to date. As this review work, with the collection of different updated knowledge, has summarized the role of leptin on cancer, it would be useful material to have on hand for both beginners as well as pioneers of these and related fields.
Collapse
Affiliation(s)
- Snehashish Modak
- Immunology Microbiology Lab, Tripura University, Suryamaninagar, Tripura, India
| | - Tamanna Aktar
- Immunology Microbiology Lab, Tripura University, Suryamaninagar, Tripura, India
| | - Debabrata Majumder
- Immunology Microbiology Lab, Tripura University, Suryamaninagar, Tripura, India
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC, USA
| | - Ashish Kr Singha
- Immunology Microbiology Lab, Tripura University, Suryamaninagar, Tripura, India
- Department of Human Physiology, Holy Cross College, Agartala, West Tripura, India
| | - Debasish Maiti
- Immunology Microbiology Lab, Tripura University, Suryamaninagar, Tripura, India.
| |
Collapse
|
6
|
Sureka N, Zaheer S. Regulatory T Cells in Tumor Microenvironment: Therapeutic Approaches and Clinical Implications. Cell Biol Int 2025. [PMID: 40365758 DOI: 10.1002/cbin.70031] [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: 03/10/2025] [Revised: 04/19/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025]
Abstract
Regulatory T cells (Tregs), previously referred to as suppressor T cells, represent a distinct subset of CD4+ T cells that are uniquely specialized for immune suppression. They are characterized by the constitutive expression of the transcription factor FoxP3 in their nuclei, along with CD25 (the IL-2 receptor α-chain) and CTLA-4 on their cell surface. Tregs not only restrict natural killer cell-mediated cytotoxicity but also inhibit the proliferation of CD4+ and CD8+ T-cells and suppress interferon-γ secretion by immune cells, ultimately impairing an effective antitumor immune response. Treg cells are widely recognized as a significant barrier to the effectiveness of tumor immunotherapy in clinical settings. Extensive research has consistently shown that Treg cells play a pivotal role in facilitating tumor initiation and progression. Conversely, the depletion of Treg cells has been linked to a marked delay in tumor growth and development.
Collapse
Affiliation(s)
- Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
7
|
Chen Y, Deng Q, Fu T, Huang Y, Li H, Xie J, Liao F, Zeng F, Fang X, Li R, Chen Z. Comprehensive analysis of potential biomarkers for the diagnosis and prognosis of Cervical squamous cell carcinoma - based on GEO and TCGA databases. Front Oncol 2025; 15:1524225. [PMID: 40406253 PMCID: PMC12094959 DOI: 10.3389/fonc.2025.1524225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 04/08/2025] [Indexed: 05/26/2025] Open
Abstract
Background Cervical squamous cell carcinoma (CESC) constitutes a substantial global health burden, especially in resource-limited regions. The identification of reliable biomarkers is critical for developing a clinically applicable nomogram to predict survival outcomes and evaluate immune infiltration in CESC patients. Methods This study integrated RNA-seq data from GEO and TCGA databases to identify key genes associated with CESC through differential expression analysis and machine learning techniques. Prognostic models were constructed and validated, with additional analyses exploring immune cell infiltration and gene function via GSEA and clinical correlation. Finally, key genes were validated via qRT-PCR in CESC tissues. Results A total of 112 differentially expressed genes (DEGs) were identified through differential analysis of the GEO and TCGA datasets. EFNA1, CXCL8, and PPP1R14A emerged as prognostic biomarkers for CESC, showing significant associations with survival, tumor stage, and immune infiltration. EFNA1 may drive tumor progression via the MAPK signaling pathway, CXCL8 could influence immune evasion through NOD-like receptor signaling, and PPP1R14A may contribute to tumor invasion by modulating extracellular matrix remodeling. A nomogram integrating these genes demonstrated high predictive accuracy for overall survival (AUC>0.75) and calibration plots. Decision curve analysis (DCA) was performed to assess the nomogram's clinical utility and net benefit for application in clinical practice. Additionally, it was validated by qRT-PCR, showing elevated expression in tumors versus normal tissues (P<0.05). Conclusion EFNA1, CXCL8, and PPP1R14A are promising biomarkers for CESC prognosis and immune regulation. The nomogram model provides a practical tool for personalized survival prediction, enhancing clinical decision-making for immunotherapy and risk stratification.
Collapse
Affiliation(s)
- Yufen Chen
- Department of Obstetrics and Gynecology, The First Affiliate Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Qinghua Deng
- Department of Gynaecology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Tengyue Fu
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yuxiang Huang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong, China
| | - Houlin Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong, China
| | - Jingmu Xie
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong, China
| | - Feng Liao
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong, China
| | - Feimiao Zeng
- Department of Gynaecology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xinyi Fang
- Reproductive Medical Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Ruiman Li
- Department of Obstetrics and Gynecology, The First Affiliate Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zhuming Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Sun H, Cao Z, Zhao B, Zhou D, Chen Z, Zhang B. An elevated percentage of CD4⁺CD25⁺CD127 low regulatory T cells in peripheral blood indicates a poorer prognosis in hepatocellular carcinoma after curative hepatectomy. BMC Gastroenterol 2025; 25:340. [PMID: 40335903 PMCID: PMC12060481 DOI: 10.1186/s12876-025-03940-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 04/25/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Previous studies suggest the percentage of CD4⁺CD25⁺CD127low regulatory T cells (Tregs) in peripheral blood of patients with hepatocellular carcinoma (HCC) was significantly higher than that in healthy, which may be a significant predictor of HCC clinical outcome, and we examined the utility of Tregs in predicting prognosis in HCC after curative hepatectomy. METHODS 77 diagnosed HCC patients from August 2018 to March 2023 were selected as research objects, we retrospectively analyzed whether the preoperative percentage of CD4⁺CD25⁺CD127low Tregs in peripheral blood predicts prognosis after curative hepatectomy in HCC patients. The percentage of CD4⁺CD25⁺CD127low Tregs was detected by flow cytometry. RESULTS The percentage of CD4⁺CD25⁺CD127low Tregs was significantly elevated in patients who developed recurrence and death (p < 0.050). X-tile software was used to calculate optimal cut-off value of Treg percentage (5.85%), and patients were divided into two groups with high and low Treg percentage. Patients with higher preoperative Treg percentage had a significantly poorer prognosis (p < 0.050). Cox regression demonstrated the percentage of CD4⁺CD25⁺CD127low Tregs was an independent indicator for poor prognosis after hepatectomy. The Recurrence-free survival (RFS) (the log-rank test, p < 0.001) and Overall survival (OS) (the log-rank test, p = 0.008) in patients with higher Treg percentage were significantly lower than that in patients with lower Treg percentage. The results were confirmed by the subgroup analysis. CONCLUSION The percentage of CD4⁺CD25⁺ CD127low Tregs in peripheral blood is associated with poor prognosis in HCC patients. It can be suggested as a potential prognostic indicator for HCC patients after hepatectomy and complement existing risk stratification tools. Measuring the percentage of CD4⁺CD25⁺ CD127low Tregs may contribute to the formulation of treatment strategies and the improvement of the prognosis for HCC patients.
Collapse
Affiliation(s)
- Haoran Sun
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui Province, People's Republic of China
| | - Zepeng Cao
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui Province, People's Republic of China
| | - Baochen Zhao
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui Province, People's Republic of China
| | - Dachen Zhou
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui Province, People's Republic of China
| | - Zhongbiao Chen
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui Province, People's Republic of China
| | - Bin Zhang
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui Province, People's Republic of China.
| |
Collapse
|
9
|
Sampson JF, Zhang H, Zhang D, Bi M, Hinthorne A, Syed S, Zhang Y, Chattopadhyay N, Collins S, Pogue S, Björck P, Curley M. CD38-targeted attenuated interferon alpha immunocytokine activates both innate and adaptive immune cells to drive anti-tumor activity. PLoS One 2025; 20:e0321622. [PMID: 40315226 PMCID: PMC12047799 DOI: 10.1371/journal.pone.0321622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 03/10/2025] [Indexed: 05/04/2025] Open
Abstract
Recombinant interferon alpha (IFNα) has been used to treat cancer patients for over 30 years; however, its clinical utility has been limited by a narrow therapeutic index. Given the recognized anti-tumor and immunomodulatory impacts of IFNα, the development of novel strategies to harness these attributes while minimizing associated toxicity could provide significant benefit for patients. The concept of attenuating IFNα binding affinity for its receptor was conceived to address this challenge and led to the development of CD38-targeted Attenukine™, a CD38-targeted antibody attenuated IFNα immunocytokine. In this study, we sought to delineate the effects of targeting AttenukineTM specifically to tumor cells and/or immune cells using an antibody to CD38, a cell surface glycoprotein expressed on certain tumor and immune cells, using different mouse models and anti-human or anti-mouse CD38-targeted Attenukine™. Our results demonstrate that an anti-human CD38 AttenukineTM inhibits tumor growth through direct anti-proliferative effects of IFNα on CD38 + tumor cells as well as by indirectly modulating the anti-tumor immune response. In various in vivo models leveraging syngeneic mice bearing tumors with or without CD38 expression, administration of CD38-murine AttenukineTM mediated anti-tumor efficacy with increased immune activation and intra-tumoral infiltration. These data point to a potential dual mechanism of action for CD38-targeted Attenukine™, involving both tumor- and immune-directed effects, and highlight the potential benefit of a CD38-targeted attenuated IFNα therapy to deliver the known effects of IFNαtreatment to a broad spectrum of patients, while limiting the toxicity typically associated with recombinant IFNα.
Collapse
Affiliation(s)
- James F. Sampson
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| | - Hong Zhang
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| | - Dongmei Zhang
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| | - Mingying Bi
- Discovery R and D, Teva Pharmaceutical Industries, Ltd., Redwood City, United States of America
| | - Adam Hinthorne
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| | - Sakeena Syed
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| | - Yuhong Zhang
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| | - Nibedita Chattopadhyay
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| | - Sabrina Collins
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| | - Sarah Pogue
- Discovery R and D, Teva Pharmaceutical Industries, Ltd., Redwood City, United States of America
| | - Pia Björck
- Discovery R and D, Teva Pharmaceutical Industries, Ltd., Redwood City, United States of America
| | - Michael Curley
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, United States of America
| |
Collapse
|
10
|
Chalepaki AM, Gkoris M, Chondrou I, Kourti M, Georgakopoulos-Soares I, Zaravinos A. A multi-omics analysis of effector and resting treg cells in pan-cancer. Comput Biol Med 2025; 189:110021. [PMID: 40088713 DOI: 10.1016/j.compbiomed.2025.110021] [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: 10/05/2024] [Revised: 02/09/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
Regulatory T cells (Tregs) are critical for maintaining the stability of the immune system and facilitating tumor escape through various mechanisms. Resting T cells are involved in cell-mediated immunity and remain in a resting state until stimulated, while effector T cells promote immune responses. Here, we investigated the roles of two gene signatures, one for resting Tregs (FOXP3 and IL2RA) and another for effector Tregs (FOXP3, CTLA-4, CCR8 and TNFRSF9) in pan-cancer. Using data from The Cancer Genome Atlas (TCGA), The Cancer Proteome Atlas (TCPA) and Gene Expression Omnibus (GEO), we focused on the expression profile of the two signatures, the existence of single nucleotide variants (SNVs) and copy number variants (CNVs), methylation, infiltration of immune cells in the tumor and sensitivity to different drugs. Our analysis revealed that both signatures are differentially expressed across different cancer types, and correlate with patient survival. Furthermore, both types of Tregs influence important pathways in cancer development and progression, like apoptosis, epithelial-to-mesenchymal transition (EMT) and the DNA damage pathway. Moreover, a positive correlation was highlighted between the expression of gene markers in both resting and effector Tregs and immune cell infiltration in adrenocortical carcinoma, while mutations in both signatures correlated with enrichment of specific immune cells, mainly in skin melanoma and endometrial cancer. In addition, we reveal the existence of widespread CNVs and hypomethylation affecting both Treg signatures in most cancer types. Last, we identified a few correlations between the expression of CCR8 and TNFRSF9 and sensitivity to several drugs, including COL-3, Chlorambucil and GSK1070916, in pan-cancer. Overall, these findings highlight new evidence that both Treg signatures are crucial regulators of cancer progression, providing potential clinical outcomes for cancer therapy.
Collapse
Affiliation(s)
- Anna-Maria Chalepaki
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| | - Marios Gkoris
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| | - Irene Chondrou
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus.
| | - Malamati Kourti
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus.
| | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA.
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| |
Collapse
|
11
|
Qi S, Li J, Gu X, Zhang Y, Zhou W, Wang F, Wang W. Impacts of ageing on the efficacy of CAR-T cell therapy. Ageing Res Rev 2025; 107:102715. [PMID: 40058461 DOI: 10.1016/j.arr.2025.102715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/19/2025] [Accepted: 03/02/2025] [Indexed: 03/17/2025]
Abstract
Chimeric antigen receptor T cells recognizing CD19 (19CAR-T) cell therapy has achieved robust clinical efficacy when treating some hematological malignancies, but which patient subgroups benefit mostly remains elusive. Here we summarized the data of 541 patients from 30 clinical trials who underwent 19 CAR-T therapy and analyzed the different clinical responses between young (<44 years), middle-aged (45-59 years) and elderly (>60 years) patients and found that the young patients showed a higher level of complete response (CR) rate. Therefore, we then summarize the advances of studies focusing on the effects of age on anti-tumor efficacy of CAR-T therapy and analyze the reasons for the low CR rate after CAR-T cell therapy in elderly patients with tumors, aiming to provide hints for oncologists to select the most suitable candidate for this cancer immunotherapy.
Collapse
Affiliation(s)
- Shimao Qi
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Jiaqian Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Xinyu Gu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Yalan Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Weilin Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Fengling Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Wei Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
12
|
Khan FU, Khongorzul P, Gris D, Amrani A. Role of USP7 in the regulation of tolerogenic dendritic cell function in type 1 diabetes. Cell Mol Biol Lett 2025; 30:47. [PMID: 40247205 PMCID: PMC12004606 DOI: 10.1186/s11658-025-00727-5] [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: 12/18/2024] [Accepted: 04/03/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Tolerogenic dendritic cells (toDCs) are critical for maintaining immune homeostasis and preventing autoimmune disease development, such as type 1 diabetes (T1D). We have previously shown that DCs of non-obese diabetic (NOD) mice expressing active Stat5b (Stat5b-CA.DCs) acquire toDCs signature and protect against diabetes. However, the mechanisms involved in reprogramming DCs to adopt tolerogenic or immunogenic signatures are not fully known. This study investigates for the first time the role of USP7 in DC-mediated immune regulation in T1D using a transgenic NOD mouse model expressing an active form of Stat5b (NOD.Stat5b-CA). METHODS Splenic DCs were purified from diabetes-prone NOD mice and diabetes-resistant NOD.Stat5b-CA transgenic mice and their tolerogenic and immunogenic phenotypes were analyzed by FACS. Their pro-and anti-inflammatory cytokine patterns, IRF4, IRF8, de-ubiquitin ligase USP7, and methyltransferase Ezh2 expression were assessed by FACS and Western blot. Moreover, the impact of USP7 inhibition in DCs on Th1/Th2/Th17 and Treg and diabetes onset was assessed using an in vivo DC-based transfer model. RESULTS In this study, we found that splenic Stat5b-CA.DCs expressed high levels of USP7, Ezh2, and PD-L-1/2 and contained a higher proportion of tolerogenic conventional DC2 (cDC2) subsets than immunogenic cDC1 compared to NOD mice DCs. We also found that the USP7 blockade increased Stat5b-CA.DCs maturation and proinflammatory cytokines production while decreasing anti-inflammatory cytokines and PD-L1 and PD-L2 expressions. Mechanistically, USP7 blockade in Stat5-CA.DCs promoted cDC1 over cDC2 subsets by increasing IRF8 expression in an Ezh2-dependent manner and decreasing IRF4 expression independently of Ezh2. USP7 blockade also increased Stat5b-CA.DC capacity to promote Th17 and to restrain Th2 and Treg cells. Importantly, the capacity of Stat5b-CA.DCs to protect NOD mice from diabetes were lost when treated with USP7 inhibitor. CONCLUSIONS Our findings underscore the role of the USP7/Ezh2 axis in maintaining tolerogenic DC functions that are required to tailor adaptive immune response and diabetes protection in NOD mice.
Collapse
Affiliation(s)
- Farhan Ullah Khan
- Department of Pediatrics, Immunology Division, Université de Sherbrooke Faculté de Médecine et des Sciences de la Santé, 3001, 12 th Avenue North, Sherbrooke, QC, J1H 5 N4, Canada
| | - Puregmaa Khongorzul
- Department of Pediatrics, Immunology Division, Université de Sherbrooke Faculté de Médecine et des Sciences de la Santé, 3001, 12 th Avenue North, Sherbrooke, QC, J1H 5 N4, Canada
| | - Denis Gris
- Department of Phamacology-Physiology, Université de Sherbrooke Faculté de Médecine et des Sciences de la Santé, 3001, 12 th Avenue North, Sherbrooke, QC, J1H 5 N4, Canada
| | - Abdelaziz Amrani
- Department of Pediatrics, Immunology Division, Université de Sherbrooke Faculté de Médecine et des Sciences de la Santé, 3001, 12 th Avenue North, Sherbrooke, QC, J1H 5 N4, Canada.
| |
Collapse
|
13
|
Siu LL, Postel-Vinay S, Villanueva-Vázquez R, de Velasco G, Castanon Alvarez E, Kyriakopoulos CE, Johnson M, Ouali K, McMorn S, Angell HK, Ng F, Saran S, Bayat M, Collins T, Roy A, Lambert AW, Cho S, Miller N, Petruzzelli M, Stone J, Massard C. AZD8701, an Antisense Oligonucleotide Targeting FOXP3 mRNA, as Monotherapy and in Combination with Durvalumab: A Phase I Trial in Patients with Advanced Solid Tumors. Clin Cancer Res 2025; 31:1449-1462. [PMID: 39937271 PMCID: PMC11995004 DOI: 10.1158/1078-0432.ccr-24-1818] [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: 06/12/2024] [Revised: 12/13/2024] [Accepted: 02/06/2025] [Indexed: 02/13/2025]
Abstract
PURPOSE AZD8701 uses next-generation antisense oligonucleotide (ASO) technology to selectively reduce human forkhead box P3 (FOXP3) expression in regulatory T cells, reversing their immunosuppressive function. FOXP3 ASO alone or with PD-(L)1 inhibition attenuated tumor growth in mice. We report a phase I study of AZD8701 alone or combined with durvalumab in patients with advanced solid tumors. PATIENTS AND METHODS Eligible patients had solid tumors and received prior standard-of-care treatment, including anti-PD-(L)1 therapy. Patient cohorts were treated with AZD8701 intravenously weekly at escalating doses, either alone (60-960 mg) or combined (240-720 mg) with durvalumab 1,500 mg intravenous every 4 weeks. The primary objective was safety and tolerability, with the aim of determining the MTD. RESULTS Forty-five patients received AZD8701 monotherapy, and 18 received AZD8701 with durvalumab. One dose-limiting toxicity (increased alanine aminotransferase) occurred with AZD8701 960 mg. The most common adverse events related to AZD8701 monotherapy were fatigue (22.2%), asthenia, pyrexia, and increased alanine aminotransferase (20% each); the safety profile was similar when combined with durvalumab. With AZD8701 monotherapy, 24.4% and 15.6% of the patients had stable disease for ≥16 and ≥24 weeks, respectively; one patient treated with AZD8701 720 mg and durvalumab had a partial response. FOXP3 mRNA changes were heterogeneous (8/13 patients showed a reduction), with no clear dose relationship. ASO accumulated in the tumor epithelium and stroma. CONCLUSIONS This study demonstrates the clinical feasibility of ASO therapy, with generally manageable adverse events, FOXP3 knockdown, and ASO delivery to the tumor.
Collapse
Affiliation(s)
| | | | - Rafael Villanueva-Vázquez
- Institut Català d'Oncologia, Early Drug Development Unit, Medical Oncology Department, ICO-Hospitalet, Barcelona, Spain
| | | | | | | | - Melissa Johnson
- Lung Cancer Research, Sarah Cannon Research Institute at Tennessee Oncology, Nashville, Tennessee
| | - Kaïssa Ouali
- Drug Development Department, Institut Gustave Roussy, Villejuif, France
| | | | | | | | | | | | | | | | | | - Song Cho
- AstraZeneca, Gaithersburg, Maryland
| | | | | | | | - Christophe Massard
- DITEP, Institut Gustave Roussy, Villejuif, France
- Faculty of Medicine, Paris Saclay University, Paris, France
- Molecular Radiotherapy Unit 1030, National Institute of Health and Medical Research (INSERM), Paris, France
| |
Collapse
|
14
|
Chen F, Cai Y, Chen X, Chen C, Fang Q, Liu J, Zhang Y, Zhou J. The role of hypoxia-senescence co-related molecular subtypes and prognostic characteristics in hepatocellular carcinoma. Sci Rep 2025; 15:12390. [PMID: 40216977 PMCID: PMC11992139 DOI: 10.1038/s41598-025-97604-0] [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: 10/26/2024] [Accepted: 04/07/2025] [Indexed: 04/14/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is known for its high invasiveness, high fatality rate. Both hypoxia and senescence play crucial roles in the initiation and progression of cancer, yet their prognostic implications in HCC are yet to be fully understood. The hypoxia-senescence co-related genes (HSCRGs) were screened from public databases. Transcriptome data and clinical information were obtained from patients with HCC using the Cancer Genome Atlas, GSE76427, and International Cancer Genome Consortium (ICGC). The random forest tree algorithm was used to identify the characteristic genes of the disease, and the genes were verified by related experiments. SVM algorithm was used to classify HCC patients based on HSCRGs. The prediction model based on HSCRGs was established by LASSO, univariate and multivariate COX regression analysis. We used the ICGC for outside validation. The risk score model was analyzed from subgroup analysis, immune infiltration, and functional strength. The expression patterns of key prognostic genes in tumor microenvironment were decoded by single cell analysis. A total of 184 HSCRGs were identified. The expression pattern and functional characteristics of MLH1 gene in HCC were verified. Two HCC subtypes were identified based on HSCRGs. Then, a prediction model based on HSCRGs was established, and risk score was identified as an independent prognostic indicator of HCC. A new nomogram is constructed and shows good prediction ability. We further determined that the level of infiltration of immune cells and the expression of immune checkpoints are significantly affected by the risk score. The immune microenvironment was different between the two risk groups. The high-risk group was dominated by immunosuppressed cells, and the prognosis was poor. Single-cell analysis revealed the expression of seven key prognostic genes in the tumor microenvironment. Finally, qPCR results further verified the expression levels of seven prognostic genes. HSCRGs are of great significance in the prognosis prediction, risk stratification and targeted therapy of patients with HCC.
Collapse
Affiliation(s)
- Fuqing Chen
- Department of Hepatobiliary Surgery, Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361004, Fujian Province, People's Republic of China
| | - Yifan Cai
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361004, Fujian Province, People's Republic of China
| | - Xiangmei Chen
- Department of Hepatobiliary Surgery, Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361004, Fujian Province, People's Republic of China
| | - Changzhou Chen
- Department Minimally Invasive and Interventional Oncology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361004, Fujian Province, People's Republic of China
| | - Qinliang Fang
- Department of Hepatobiliary Surgery, Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361004, Fujian Province, People's Republic of China
| | - Jianming Liu
- Department of Hepatobiliary Surgery, Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361004, Fujian Province, People's Republic of China
| | - Yibin Zhang
- Department of Hepatobiliary Surgery, Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361004, Fujian Province, People's Republic of China
| | - Jianyin Zhou
- Department of Hepatobiliary Surgery, Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361004, Fujian Province, People's Republic of China.
| |
Collapse
|
15
|
Pan Y, Zhou H, Sun Z, Zhu Y, Zhang Z, Han J, Liu Y, Wang Q. Regulatory T cells in solid tumor immunotherapy: effect, mechanism and clinical application. Cell Death Dis 2025; 16:277. [PMID: 40216744 PMCID: PMC11992189 DOI: 10.1038/s41419-025-07544-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 01/12/2025] [Accepted: 03/17/2025] [Indexed: 04/14/2025]
Abstract
The tumor-immune response is mobilized to suppress tumorigenesis, while the immune microenvironment and lymph node microenvironment are formed gradually during tumor progression. In fact, tumor surface antigens are not easily recognized by antigen-presenting cells. So it is hard for the immune system to kill the newly formed tumor cells effectively. In a normal immune environment, immune function is always suppressed to maintain the stability of the body, and regulatory T cells play an important role in maintaining immune suppression. However, during tumorigenesis, the suppression of regulatory T cell immune functions is more likely to contribute to tumor cell proliferation and migration leading directly to tumor progression. Therefore, focusing on the role of regulatory T cells in tumor immunity could improve tumor immunotherapy outcomes in the clinic. Regulatory T cells are more mature in hematologic system tumors than in solid tumors. However, there are continuing efforts to apply regulatory T cells for immunotherapy in solid tumors. This review describes the role of regulatory T cells in solid tumor immunotherapy from the perspective of prognosis, immune microenvironment remodeling, and current clinical applications. This summary could help us better understand the mechanisms of regulatory T cells in solid tumor immunotherapy and further expand their clinical application.
Collapse
Affiliation(s)
- Yan Pan
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
- Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, 451162, China
| | - Hanqiong Zhou
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
- Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, 451162, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Yichen Zhu
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
- Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, 451162, China
| | - Zhe Zhang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
- Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, 451162, China
| | - Jing Han
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
- Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, 451162, China
| | - Yang Liu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| | - Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
- Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, 451162, China.
| |
Collapse
|
16
|
Nielsen AT, Saqi IK, Justesen TF, Madsen MT, Gögenur I, Orhan A. The prognostic impact of tumor mutations and tumor-infiltrating lymphocytes in patients with localized pMMR colorectal cancer - A systematic review and meta-analysis. Crit Rev Oncol Hematol 2025; 211:104714. [PMID: 40188978 DOI: 10.1016/j.critrevonc.2025.104714] [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: 02/09/2025] [Revised: 03/21/2025] [Accepted: 03/28/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Tumor mutations and the composition of the tumor microenvironment have prognostic and therapeutic significance in colorectal cancer (CRC). However, immunotherapy remains a challenge for patients with proficient mismatch repair (pMMR) CRC. In this paper, the association between tumor-infiltrating lymphocytes (TILs) and tumor mutations on survival outcomes in patients with localized pMMR CRC was examined. METHODS A systematic review of the literature and a meta-analysis were conducted in accordance with the PRISMA guidelines. The literature search was conducted in PubMed, Embase, Cochrane Library, and Web of Science. The outcomes of interest were overall survival, disease-free survival, and cancer-specific survival. The risk of bias was assessed through the Newcastle-Ottawa Scale and the quality of the cumulative evidence was evaluated through the modified GRADE approach. FINDINGS In total, 8498 articles were screened for eligibility and 44 articles were included in the meta-analysis with 33,704 patients in total. Patients with high infiltration of any TILs showed significantly improved overall survival (HR = 0.57, 95 % CI: 0.49-0.67, I2: 0 %), especially for the subgroup of CD3 + (HR = 0.52, 95 % CI: 0.38-0.71, I2: 0 %) and CD8 + (HR = 0.60, 95 % CI: 0.37-0.99, I2: 10 %) TILs. Patients with BRAF mutation (HR = 2.68, 95 % CI: 1.47-4.89, I2: 83 %) and KRAS mutation (HR = 1.25, 95 % CI: 1.18-1.33, I2: 0 %) showed decreased overall survival. INTERPRETATION High infiltration of TILs, especially CD3 + and CD8 + , was associated with significantly improved survival, while BRAF and KRAS mutations were correlated with worse survival outcomes for patients with non-metastatic pMMR CRC.
Collapse
Affiliation(s)
- Amalie Thomsen Nielsen
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Køge, Denmark.
| | - Ida Kolukisa Saqi
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Køge, Denmark
| | | | | | - Ismail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Køge, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Adile Orhan
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Køge, Denmark
| |
Collapse
|
17
|
Ryba-Stanisławowska M. Unraveling Th subsets: insights into their role in immune checkpoint inhibitor therapy. Cell Oncol (Dordr) 2025; 48:295-312. [PMID: 39325360 PMCID: PMC11996958 DOI: 10.1007/s13402-024-00992-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2024] [Indexed: 09/27/2024] Open
Abstract
T helper (Th) cell subsets play pivotal roles in regulating immune responses within the tumor microenvironment, influencing both tumor progression and anti-tumor immunity. Among these subsets, Th1 cells promote cytotoxic responses through the production of IFN-γ, while Th2 cells and regulatory T cells (Tregs) exert immunosuppressive effects that support tumor growth. Th9 and Th17 cells have context-dependent roles, contributing to both pro-inflammatory and regulatory processes in tumor immunity. Tumor antigen-specific T cells within the tumor microenvironment often exhibit a dysfunctional phenotype due to increased expression of inhibitory receptors such as CTLA-4 and PD-1, leading to reduced antitumor activity. Monoclonal antibodies that block these inhibitory signals-collectively known as immune checkpoint inhibitors (ICIs)-can reactivate these T cells, enhancing their ability to target and destroy cancer cells. Recent advancements have highlighted the critical role of T helper subsets in modulating responses to ICIs, with their interactions remaining a focus of ongoing research. Both positive and negative effects of ICIs have been reported in relation to Th cell subsets, with some effects depending on the type of tumor microenvironment. This review summarizes the crucial roles of different T helper cell subsets in tumor immunity and their complex relationship with immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Monika Ryba-Stanisławowska
- Department of Medical Immunology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 1, Gdańsk, 80-211, Poland.
| |
Collapse
|
18
|
Beltzung F, Le VL, Molnar I, Boutault E, Darcha C, Le Loarer F, Kossai M, Saut O, Biau J, Penault-Llorca F, Chautard E. Leveraging Deep Learning for Immune Cell Quantification and Prognostic Evaluation in Radiotherapy-Treated Oropharyngeal Squamous Cell Carcinomas. J Transl Med 2025; 105:104094. [PMID: 39826685 DOI: 10.1016/j.labinv.2025.104094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
The tumor microenvironment plays a critical role in cancer progression and therapeutic responsiveness, with the tumor immune microenvironment (TIME) being a key modulator. In head and neck squamous cell carcinomas (HNSCCs), immune cell infiltration significantly influences the response to radiotherapy (RT). A better understanding of the TIME in HNSCCs could help identify patients most likely to benefit from combining RT with immunotherapy. Standardized, cost-effective methods for studying TIME in HNSCCs are currently lacking. This study aims to leverage deep learning (DL) to quantify immune cell densities using immunohistochemistry in untreated oropharyngeal squamous cell carcinoma (OPSCC) biopsies of patients scheduled for curative RT and assess their prognostic value. We analyzed 84 pretreatment formalin-fixed paraffin-embedded tumor biopsies from OPSCC patients. Immunohistochemistry was performed for CD3, CD8, CD20, CD163, and FOXP3, and whole slide images were digitized for analysis using a U-Net-based DL model. Two quantification approaches were applied: a cell-counting method and an area-based method. These methods were applied to stained regions. The DL model achieved high accuracy in detecting stained cells across all biomarkers. Strong correlations were found between our DL pipeline, the HALO Image Analysis Platform, and the open-source QuPath software for estimating immune cell densities. Our DL pipeline provided an accurate and reproducible approach for quantifying immune cells in OPSCC. The area-based method demonstrated superior prognostic value for recurrence-free survival, when compared with the cell-counting method. Elevated densities of CD3, CD8, CD20, and FOXP3 were associated with improved recurrence-free survival, whereas CD163 showed no significant prognostic association. These results highlight the potential of DL in digital pathology for assessing TIME and predicting patient outcomes.
Collapse
Affiliation(s)
- Fanny Beltzung
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Department of Pathology, Hôpital Haut-Lévêque, CHU de Bordeaux, Pessac, France.
| | - Van-Linh Le
- MONC team, Center INRIA at University of Bordeaux, Talence, France; Bordeaux Mathematics Institute (IMB), UMR CNRS 5251, University of Bordeaux, Talence, France; Department of Data and Digital Health, Bergonié Institute, Bordeaux, France
| | - Ioana Molnar
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Clinical Research Division, Clinical Research & Innovation Division, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Erwan Boutault
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France
| | - Claude Darcha
- Department of Pathology, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - François Le Loarer
- Department of Pathology, Bergonié Institute, Bordeaux, France; Bordeaux Institute of Oncology (BRIC U1312), INSERM, Université de Bordeaux, Institut Bergonié, Bordeaux, France
| | - Myriam Kossai
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Department of Pathology, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Olivier Saut
- MONC team, Center INRIA at University of Bordeaux, Talence, France; Bordeaux Mathematics Institute (IMB), UMR CNRS 5251, University of Bordeaux, Talence, France
| | - Julian Biau
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Department of Radiation Therapy, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Frédérique Penault-Llorca
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Department of Pathology, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Emmanuel Chautard
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Department of Pathology, Centre Jean PERRIN, Clermont-Ferrand, France
| |
Collapse
|
19
|
Marr B, Jo D, Jang M, Lee SH. Cytokines in Focus: IL-2 and IL-15 in NK Adoptive Cell Cancer Immunotherapy. Immune Netw 2025; 25:e17. [PMID: 40342841 PMCID: PMC12056295 DOI: 10.4110/in.2025.25.e17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/02/2025] [Accepted: 03/06/2025] [Indexed: 05/11/2025] Open
Abstract
NK cell adoptive cell therapy (ACT) has emerged as a promising strategy for cancer immunotherapy, offering advantages in scalability, accessibility, efficacy, and safety. Ex vivo activation and expansion protocols, incorporating feeder cells and cytokine cocktails, have enabled the production of highly functional NK cells in clinically relevant quantities. Advances in NK cell engineering, including CRISPR-mediated gene editing and chimeric Ag receptor technologies, have further enhanced cytotoxicity, persistence, and tumor targeting. Cytokine support post-adoptive transfer, particularly with IL-2 and IL-15, remains critical for promoting NK cell survival, proliferation, and anti-tumor activity despite persistent challenges such as regulatory T cell expansion and cytokine-related toxicities. This review explores the evolving roles of IL-2 and IL-15 in NK cell-based ACT, evaluating their potential and limitations, and highlights strategies to optimize these cytokines for effective cancer immunotherapy.
Collapse
Affiliation(s)
- Bryan Marr
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Donghyeon Jo
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Mihue Jang
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul 02792, Korea
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Korea
| | - Seung-Hwan Lee
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, Faculty of Medicine and Centre for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
20
|
Di Spirito A, Balkhi S, Vivona V, Mortara L. Key immune cells and their crosstalk in the tumor microenvironment of bladder cancer: insights for innovative therapies. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002304. [PMID: 40177538 PMCID: PMC11964778 DOI: 10.37349/etat.2025.1002304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Bladder cancer (BC) is a heterogeneous disease associated with high mortality if not diagnosed early. BC is classified into non-muscle-invasive BC (NMIBC) and muscle-invasive BC (MIBC), with MIBC linked to poor systemic therapy response and high recurrence rates. Current treatments include transurethral resection with Bacillus Calmette-Guérin (BCG) therapy for NMIBC and radical cystectomy with chemotherapy and/or immunotherapy for MIBC. The tumor microenvironment (TME) plays a critical role in cancer progression, metastasis, and therapeutic efficacy. A comprehensive understanding of the TME's complex interactions holds substantial translational significance for developing innovative treatments. The TME can contribute to therapeutic resistance, particularly in immune checkpoint inhibitor (ICI) therapies, where resistance arises from tumor-intrinsic changes or extrinsic TME factors. Recent advancements in immunotherapy highlight the importance of translational research to address these challenges. Strategies to overcome resistance focus on remodeling the TME to transform immunologically "cold" tumors, which lack immune cell infiltration, into "hot" tumors that respond better to immunotherapy. These strategies involve disrupting cancer-microenvironment interactions, inhibiting angiogenesis, and modulating immune components to enhance anti-tumor responses. Key mechanisms include cytokine involvement [e.g., interleukin-6 (IL-6)], phenotypic alterations in macrophages and natural killer (NK) cells, and the plasticity of cancer-associated fibroblasts (CAFs). Identifying potential therapeutic targets within the TME can improve outcomes for MIBC patients. This review emphasizes the TME's complexity and its impact on guiding novel therapeutic approaches, offering hope for better survival in MIBC.
Collapse
Affiliation(s)
- Anna Di Spirito
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Sahar Balkhi
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Veronica Vivona
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
21
|
Gambardella V, Ong M, Rodriguez-Ruiz ME, Machiels JP, Sanmamed MF, Galvao V, Spreafico A, Renouf DJ, Luen SJ, Galot R, Doger de Spéville B, Calvo E, Naing A, Curdt S, Kolben TM, Rossmann E, Tanos T, Smart K, Amann M, Xie Y, Xu L, Gomez Alcaide E, Städler N, Justies N, Boetsch C, Karanikas V, Schnetzler G, Rohrberg KS. Safety and Antitumor Activity of a Novel aCD25 Treg Depleter RG6292 as a Single Agent and in Combination with Atezolizumab in Patients with Solid Tumors. CANCER RESEARCH COMMUNICATIONS 2025; 5:422-432. [PMID: 39983024 PMCID: PMC11891644 DOI: 10.1158/2767-9764.crc-24-0638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 02/23/2025]
Abstract
PURPOSE Therapeutic depletion of immunosuppressive regulatory T cells (Treg) may overcome resistance to cancer immunotherapies. RG6292 is an anti-CD25 antibody that preferentially depletes Tregs while preserving effector T-cell functions in preclinical models. The safety, pharmacokinetics, pharmacodynamics, and antitumor efficacy of selective Treg depletion by RG6292 administered as monotherapy or in combination with atezolizumab were evaluated in two phase I studies. PATIENTS AND METHODS Adult patients with advanced solid tumors were administered intravenous RG6292, given every 3 weeks alone (study 1: NCT04158583, n = 76) or with 1,200 mg atezolizumab every 3 weeks (study 2: NCT04642365, n = 49). Both studies included dose escalation and expansion parts to determine the maximum tolerated dose and recommended phase II dose. RESULTS RG6292 was well tolerated. Pruritus and rash were the most frequent adverse events and were manageable with supportive treatment. Serum RG6292 levels increased dose proportionally, independent of the atezolizumab combination. RG6292 induced a sustained dose-dependent depletion of peripheral Tregs with no apparent effect on other immune cells. Evidence of intratumoral Treg reduction (≥50% vs. baseline) was observed at RG6292 doses of 35 to 100 mg. The maximum tolerated dose was 165 mg every 3 weeks, and the recommended phase II dose was proposed as 70 mg every 3 weeks. Objective responses were limited to three partial responses in patients receiving RG6292 combined with atezolizumab. CONCLUSIONS RG6292 induced a dose-dependent peripheral blood and measurable intratumoral Treg depletion in concordance with the proposed mode of action; however, clinical efficacy as a single agent or combined with atezolizumab was insufficient to warrant further exploration in this population. SIGNIFICANCE RG6292 (vopikitug) targets CD25 (IL-2Rα) and mediates regulatory T-cell depletion while not interfering with IL-2 signaling. Peripheral and intratumoral Treg depletion was shown in two phase I studies. However, RG6292 alone or in combination with atezolizumab was insufficient to reverse and rescue from established resistance mechanisms in solid tumors.
Collapse
Affiliation(s)
| | - Michael Ong
- The Ottawa Hospital Cancer Centre, Ottawa, Canada
| | | | - Jean-Pascal Machiels
- Department of Medical Oncology, Institut Roi Albert II, Cliniques universitaires Saint-Luc, and Institut de Recherche Clinique et Expérimentale, UCLouvain, Brussels, Belgium
| | - Miguel F. Sanmamed
- Department of Medical Oncology, Clinica Universidad de Navarra, Pamplona, Spain
| | - Vladimir Galvao
- Vall d/Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Anna Spreafico
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | | | - Stephen J. Luen
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Rachel Galot
- Department of Medical Oncology, Institut Roi Albert II, Cliniques universitaires Saint-Luc, and Institut de Recherche Clinique et Expérimentale, UCLouvain, Brussels, Belgium
| | | | - Emiliano Calvo
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Aung Naing
- MD Anderson Cancer Center, Houston, Texas
| | - Samira Curdt
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Development, Penzberg, Germany
| | - Theresa Maria Kolben
- Roche Innovation Center Munich, Roche Pharmaceutical Research and Development, Penzberg, Germany
| | - Eva Rossmann
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Tamara Tanos
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Kevin Smart
- Roche Innovation Centre Welwyn, Roche Pharmaceutical Research and Early Development, Welwyn, United Kingdom
| | - Maria Amann
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, Schlieren, Switzerland
| | - Yuying Xie
- F. Hoffmann-La Roche Ltd., Mississauga, Canada
| | - Linxinyu Xu
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Enrique Gomez Alcaide
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Nicolas Städler
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Nicole Justies
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Christophe Boetsch
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Vaios Karanikas
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, Schlieren, Switzerland
| | - Gabriel Schnetzler
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Kristoffer S. Rohrberg
- Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Shi J, Zhao L, Wang K, Lin J, Shen J. Disulfidptosis classification of pancreatic carcinoma reveals correlation with clinical prognosis and immune profile. Hereditas 2025; 162:26. [PMID: 39987145 PMCID: PMC11846472 DOI: 10.1186/s41065-025-00381-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/27/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Disulfidptosis, a novel form of metabolism-related regulated cell death, is a promising intervention for cancer therapeutic intervention. Although aberrant expression of long-chain noncoding RNAs (lncRNAs) expression has been associated with pancreatic carcinoma (PC) development, the biological properties and prognostic potential of disulfidptosis-related lncRNAs (DRLs) remain unclear. METHODS We obtained RNA-seq data, clinical data, and genomic mutations of PC from the TCGA database, and then determined DRLs. We developed a risk score model and analyzed the role of risk score in the predictive ability, immune cell infiltration, immunotherapy response, and drug sensitivity. RESULTS We finally established a prognostic model including three DRLs (AP005233.2, FAM83A-AS1, and TRAF3IP2-AS1). According to Kaplan-Meier curve analysis, the survival time of patients in the low-risk group was significantly longer than that in the high-risk group. Based on enrichment analysis, significant associations between metabolic processes and differentially expressed genes were assessed in two risk groups. In addition, we observed significant differences in the tumor immune microenvironment landscape. Tumor Immune Dysfunction and Rejection (TIDE) analysis showed no statistically significant likelihood of immune evasion in both risk groups. Patients exhibiting both high risk and high tumor mutation burden (TMB) had the poorest survival times, while those falling into the low risk and low TMB categories showed the best prognosis. Moreover, the risk group identified by the 3-DRLs profile showed significant drug sensitivity. CONCLUSIONS Our proposed 3-DRLs-based feature could serve as a promising tool for predicting the prognosis, immune landscape, and treatment response of PC patients, thus facilitating optimal clinical decision-making.
Collapse
Affiliation(s)
- Jiangmin Shi
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to, Ningbo University), Ningbo, Zhejiang Province, 315040, P.R. China
| | - Liang Zhao
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to, Ningbo University), Ningbo, Zhejiang Province, 315040, P.R. China
| | - Kai Wang
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to, Ningbo University), Ningbo, Zhejiang Province, 315040, P.R. China
| | - Jieqiong Lin
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to, Ningbo University), Ningbo, Zhejiang Province, 315040, P.R. China
| | - Jianwei Shen
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to, Ningbo University), Ningbo, Zhejiang Province, 315040, P.R. China.
| |
Collapse
|
23
|
García-Díaz N, Solli E, Hajjar E, Cornillot-Clément S, Landskron J, Ahmad R, Wei Q, Taskén K. MAPK and STAT3 Inhibitors Modulate FoxP3 Expression and Regulatory T Cell Function. Eur J Immunol 2025; 55:e202451225. [PMID: 39955647 DOI: 10.1002/eji.202451225] [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: 04/26/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/17/2025]
Abstract
Regulatory T cells (Tregs) are a subset of T cells defined by the expression of Forkhead box protein P3 (FoxP3) playing a crucial role in regulating effector T cell activity. Tregs accumulate in the tumor microenvironment facilitating tumor growth. Thus, targeting FoxP3+ Tregs could improve cancer immunotherapies. Here, we conducted a high-throughput, phenotypic screening of a drug repurposing library to identify compounds downregulating FoxP3 expression in human primary T cells. We identified the tyrosine kinase inhibitor bosutinib and the STAT3 inhibitor nifuroxazide effectively downregulating FoxP3 expression. To identify more potent compounds, structural analogs of these two compounds were searched and validated. These analogs were found to reduce FoxP3 expression in a similar- or more potent manner than the original hits. All compounds inhibited Treg suppressive functions and reduced the expression of Treg activation markers. Importantly, bosutinib disrupted FAK and CaMKII signaling more potently in Tregs, whilst nifuroxazide and its analog NA16 targeted STAT3 protein levels more effectively in Tregs. Additionally, bosutinib and NA16 targeted effector Tregs more effectively than other Treg subsets. In summary, bosutinib, nifuroxazide, and their analogs inhibited FoxP3 expression, Treg suppressive abilities, and Treg activation effectively, which could serve as tools for the improvement of current cancer immunotherapies.
Collapse
Affiliation(s)
- Nuria García-Díaz
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Elise Solli
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ehsan Hajjar
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Selma Cornillot-Clément
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Johannes Landskron
- Centre for Molecular Medicine, Nordic EMBL Partnership, University of Oslo, Oslo, Norway
| | - Rafi Ahmad
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Biotechnology, University of Inland Norway, Hamar, Norway
| | - Qian Wei
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Kjetil Taskén
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Centre for B-cell Malignancies, University of Oslo, Oslo, Norway
| |
Collapse
|
24
|
Härm J, Fan YT, Brenner D. Navigating the metabolic landscape of regulatory T cells: from autoimmune diseases to tumor microenvironments. Curr Opin Immunol 2025; 92:102511. [PMID: 39674060 DOI: 10.1016/j.coi.2024.102511] [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: 06/16/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/16/2024]
Abstract
Regulatory T cells (Tregs) are essential for maintaining immune homeostasis, playing crucial roles in modulating autoimmune conditions and contributing to the suppressive tumor microenvironment. Their cellular metabolism governs their generation, stability, proliferation, and suppressive function. Enhancing Treg metabolism to boost their suppressive function offers promising therapeutic potential for alleviating inflammatory symptoms in autoimmune diseases. Conversely, inhibiting Treg metabolism to reduce their suppressive function can enhance the efficacy of traditional immunotherapy in cancer patients. This review explores recent advances in targeting Treg metabolism in autoimmune diseases and the metabolic adaptations of Tregs within the tumor microenvironment that increase their immunosuppressive function.
Collapse
Affiliation(s)
- Janika Härm
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology and Genetics, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Yu-Tong Fan
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology and Genetics, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Dirk Brenner
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology and Genetics, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis (ORCA), Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
25
|
Zhang X, Qian X, Zhao Y, Ye M, Li L, Chu J. Identification and analysis of prognostic ion homeostasis characteristics in kidney renal clear cell carcinoma. Heliyon 2025; 11:e41736. [PMID: 39897849 PMCID: PMC11782977 DOI: 10.1016/j.heliyon.2025.e41736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 01/04/2025] [Accepted: 01/05/2025] [Indexed: 02/04/2025] Open
Abstract
Kidney renal clear cell carcinoma (KIRC), a prevalent primary malignant tumor within the urinary system, is characterized by significant heterogeneity. It has been shown that ion channels are important targets for anti-tumor therapy. In this study, we screened 70 selected KIRC related ion homeostasis genes with significant differential expression. We established diagnostic and prognostic models for 15 genes (PDK4, JPH4, ATP1A3, CCL7, CYP27B1, ABCB6, TNFSF11, MCHR1, TNNI3, ANGPTL3, Ednrb, SAA1, Chrna9, TMPRSS6, CCL14) by LASSO regression in the TCGA-KIRC cohort. We also provided a nomogram based on ion homeostasis for clinicians to explore the combined effect of the risk model on clinical variables. Patients in the low-risk group have a significant survival advantage. The potential clinical benefit of our predicted 15 gene signatures in clinical strategies was validated by Calibration Curves and DCA curves. Ultimately, the immune microenvironment and enrichment pathways were analyzed among individuals categorized as high-risk and low-risk. The predictable ion homeostasis-associated 15 gene signature established in this study predicts overall survival outcomes in patients with KIRC, to some extent helping clinicians to select personalized treatment regimens.
Collapse
Affiliation(s)
| | | | - Yong Zhao
- Department of Urology, Shanghai Baoshan Luodian Hospital, Baoshan District, Shanghai, 201908, China
| | - Maofei Ye
- Department of Urology, Shanghai Baoshan Luodian Hospital, Baoshan District, Shanghai, 201908, China
| | - Liyang Li
- Department of Urology, Shanghai Baoshan Luodian Hospital, Baoshan District, Shanghai, 201908, China
| | - Jian Chu
- Department of Urology, Shanghai Baoshan Luodian Hospital, Baoshan District, Shanghai, 201908, China
| |
Collapse
|
26
|
Hugdahl E, Aziz S, Klingen TA, Akslen LA. Prognostic value of immune biomarkers in melanoma loco-regional metastases. PLoS One 2025; 20:e0315284. [PMID: 39883679 PMCID: PMC11781691 DOI: 10.1371/journal.pone.0315284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 11/24/2024] [Indexed: 02/01/2025] Open
Abstract
The prognosis for patients with melanoma loco-regional metastases is very heterogenous. Adjuvant PD-L1-inhibitors have improved clinical outcome for this patient group, but the prognostic impact of tumour PD-L1 expression and number of tumour infiltrating lymphocytes (TILs) is still largely unknown. Here, we investigated the impact on survival for CD3, CD8, FOXP3 and PD-L1 TIL counts and tumour PD-L1 expression in melanoma loco-regional metastases. In a patient series of loco-regional metastases from nodular melanomas (n = 78; n = 26 skin metastases, n = 52 lymph node metastases), expression of PD-L1 in tumour cells and the number of CD3, CD8, FOXP3 and PD-L1 positive TILs were determined by immunohistochemistry on tissue microarray (TMA) slides. Due to limited tumour tissue in the paraffin blocks, 67 of the 78 cases were included for tissue microarrays. Low FOXP3 TIL count and negative tumour PD-L1 expression (cut off 1%) were both significantly associated with reduced survival in lymph node metastases. Low FOXP3 TIL count was significantly associated with low CD8, CD3 and PD-L1 TIL counts. Negative tumour PD-L1 expression was significantly associated with low CD8 and PD-L1 TIL count, large lymph node metastasis tumour size and presence of necrosis in lymph node metastases. Our findings demonstrate for the first time the negative prognostic value of low FOXP3 TIL count and confirm a negative prognostic value of negative tumour PD-L1 expression in melanoma lymph node metastases.
Collapse
Affiliation(s)
- Emilia Hugdahl
- Department of Clinical Medicine, Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway
| | - Sura Aziz
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Tor A. Klingen
- Department of Clinical Medicine, Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway
- Department of Pathology, Vestfold Hospital, Tønsberg, Norway
| | - Lars A. Akslen
- Department of Clinical Medicine, Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
27
|
Luan F, Cui Y, Huang R, Yang Z, Qiao S. Comprehensive pan-cancer analysis reveals NTN1 as an immune infiltrate risk factor and its potential prognostic value in SKCM. Sci Rep 2025; 15:3223. [PMID: 39863609 PMCID: PMC11762998 DOI: 10.1038/s41598-025-85444-x] [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: 08/25/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Netrin-1 (NTN1) is a laminin-related secreted protein involved in axon guidance and cell migration. Previous research has established a significant connection between NTN1 and nervous system development. In recent years, mounting evidence indicates that NTN1 also plays a crucial role in tumorigenesis and tumor progression. For instance, inhibiting Netrin-1 has been shown to suppress tumor growth and epithelial-mesenchymal transition (EMT) characteristics in endometrial cancer. To further elucidate the influence of genes on tumors, we utilized a variety of machine learning techniques and found that NTN1 is strongly linked to multiple cancer types, suggesting it as a potential therapeutic target. This study aimed to elucidate the role of NTN1 in pan-cancer using multi-omics data and explore its potential as a prognostic biomarker in SKCM. Analysis of the TCGA, GTEx, and UALCAN databases revealed significant differences in NTN1 expression at both the mRNA and protein levels. Prognostic value was evaluated through univariate Cox regression and Kaplan-Meier methods. Mutation and methylation analyses were conducted using the cBioPortal and SMART databases. We identified genes interacting with and correlated to NTN1 through STRING and GEPIA2, respectively. Subsequently, we performed GO and KEGG enrichment analyses. The results suggested that NTN1 might be involved in crucial biological processes and pathways related to cancer development and progression, including cell adhesion, axon guidance, immune response, and various signaling pathways. We then explored the correlation between NTN1 and immune infiltration as well as immunotherapy using the ESTIMATE package, TIMER2.0, TISIDB, TIDE, TIMSO, and TCIA. The relationship between NTN1 and tumor heterogeneity, stemness, DNA methyltransferases, and MMR genes was also examined. Lastly, we constructed a nomogram based on NTN1 in SKCM and investigated its association with drug sensitivity. NTN1 expression was significantly associated with tumor immune infiltration, molecular subtypes, and clinicopathological features in various cancers. Genetic analysis revealed that Deep deletions were the most common type of NTN1 alteration. Additionally, a positive correlation was observed between NTN1 CNAs and its expression levels. In most cancers, NTN1 showed positive correlations with immune and stromal scores, as well as with specific immune cell populations. Its predictive value for immunotherapy response was comparable to that of tumor mutational burden. Furthermore, NTN1 exhibited positive correlations with tumor heterogeneity, stemness, DNA methyltransferase genes, and MMR genes. In SKCM, NTN1 was identified as an independent risk factor and demonstrated potential associations with multiple drugs. NTN1 exhibits substantial clinical utility as a prognostic marker and indicator of immune response across various tumor types. This comprehensive analysis provides insights into its potential implications in pan-cancer research.
Collapse
Affiliation(s)
- Fuxiang Luan
- The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Yuying Cui
- The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Ruizhe Huang
- The First Clinical College of Changsha Medical University, Changsha, China
| | - Zhuojie Yang
- Academy of medical sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Shishi Qiao
- The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
28
|
Gwin WR, Salazar LG, Dai JY, Higgins D, Coveler AL, Childs JS, Blancas R, Dang Y, Reichow J, Slota M, Lu H, Disis ML. A Phase II Study of Denileukin Diftitox in Patients with Advanced Treatment Refractory Breast Cancer. Vaccines (Basel) 2025; 13:117. [PMID: 40006664 PMCID: PMC11860294 DOI: 10.3390/vaccines13020117] [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: 10/19/2024] [Revised: 12/05/2024] [Accepted: 12/17/2024] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Regulatory T cells (Treg) suppress immunity in the tumor microenvironment, are linked to poor prognosis across breast cancer subtypes, and suppress the cytolytic function of cytotoxic CD8+ T cells. Denileukin diftitox, a diphtheria toxin (DT)/IL-2 fusion protein, targets and depletes Tregs. This Phase II study aimed to assess the safety of denileukin diftitox and its effect on Tregs and tumor growth in patients with advanced breast cancer. Methods: This single-arm Phase II study of denileukin diftitox enrolled patients with refractory stage IV breast cancer. Patients received denileukin diftitox 18 mcg/kg/day IV for Days 1-5 every 21 days for up to six cycles. Toxicity was assessed using CTCAE v3.0 and tumor response was evaluated per RECIST criteria. Blood samples were collected to analyze Tregs by flow cytometry and anti-DT antibodies by ELISA. Results: Fifteen patients with stage IV breast cancer were enrolled. Four patients completed all planned denileukin diftitox infusions and achieved stable disease (27%, 95% CI [0.08, 0.55]). Two patients (13%) discontinued due to toxicity, and nine patients (60%) discontinued due to progressive disease. Eleven patients experienced at least one grade 3 or 4 adverse event. Although there was a general reduction in peripheral blood Tregs, the difference in CD4+CD25+FOXP3+ Tregs levels post-treatment was not statistically significant (p = 0.10). Six patients (40%) achieved ≥25% reductions in Tregs. A significant increase in anti-DT IgG antibodies was observed post-treatment (p < 0.005). Conclusions: Denileukin diftitox demonstrated moderate toxicity in this advanced breast cancer cohort. Denileukin diftitox modulated regulatory T cells. However, the majority of patients experienced disease progression in the study.
Collapse
Affiliation(s)
- William R. Gwin
- Cancer Vaccine Institute, University of Washington, 850 Republican Street, Box 358050, Seattle, WA 98195, USA; (L.G.S.); (A.L.C.); (J.S.C.); (Y.D.)
- Fred Hutchinson Cancer Center, 1100 Fairview Ave. N., Seattle, WA 98109, USA;
| | - Lupe G. Salazar
- Cancer Vaccine Institute, University of Washington, 850 Republican Street, Box 358050, Seattle, WA 98195, USA; (L.G.S.); (A.L.C.); (J.S.C.); (Y.D.)
| | - James Y. Dai
- Fred Hutchinson Cancer Center, 1100 Fairview Ave. N., Seattle, WA 98109, USA;
| | - Doreen Higgins
- Cancer Vaccine Institute, University of Washington, 850 Republican Street, Box 358050, Seattle, WA 98195, USA; (L.G.S.); (A.L.C.); (J.S.C.); (Y.D.)
| | - Andrew L. Coveler
- Cancer Vaccine Institute, University of Washington, 850 Republican Street, Box 358050, Seattle, WA 98195, USA; (L.G.S.); (A.L.C.); (J.S.C.); (Y.D.)
- Fred Hutchinson Cancer Center, 1100 Fairview Ave. N., Seattle, WA 98109, USA;
| | - Jennifer S. Childs
- Cancer Vaccine Institute, University of Washington, 850 Republican Street, Box 358050, Seattle, WA 98195, USA; (L.G.S.); (A.L.C.); (J.S.C.); (Y.D.)
| | - Rosie Blancas
- Breastlink Medical Group, 230 S Main St, #100, Orange County, CA 92868, USA
| | - Yushe Dang
- Cancer Vaccine Institute, University of Washington, 850 Republican Street, Box 358050, Seattle, WA 98195, USA; (L.G.S.); (A.L.C.); (J.S.C.); (Y.D.)
| | - Jessica Reichow
- Cancer Vaccine Institute, University of Washington, 850 Republican Street, Box 358050, Seattle, WA 98195, USA; (L.G.S.); (A.L.C.); (J.S.C.); (Y.D.)
| | - Meredith Slota
- Cancer Vaccine Institute, University of Washington, 850 Republican Street, Box 358050, Seattle, WA 98195, USA; (L.G.S.); (A.L.C.); (J.S.C.); (Y.D.)
| | - Hailing Lu
- Seattle Genetics (Pfizer), 21823 30th DR SE, Bothell, WA 98021, USA
| | - Mary L. Disis
- Cancer Vaccine Institute, University of Washington, 850 Republican Street, Box 358050, Seattle, WA 98195, USA; (L.G.S.); (A.L.C.); (J.S.C.); (Y.D.)
- Fred Hutchinson Cancer Center, 1100 Fairview Ave. N., Seattle, WA 98109, USA;
| |
Collapse
|
29
|
Zilberg C, Ferguson AL, Lyons JG, Gupta R, Damian DL. The tumor immune microenvironment in primary cutaneous melanoma. Arch Dermatol Res 2025; 317:273. [PMID: 39825956 PMCID: PMC11742903 DOI: 10.1007/s00403-024-03758-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 12/04/2024] [Accepted: 12/20/2024] [Indexed: 01/20/2025]
Abstract
Melanoma is an immunogenic tumor. The melanoma tumor immune microenvironment (TIME) is made up of a heterogenous mix of both immune and non-immune cells as well as a multitude of signaling molecules. The interactions between tumor cells, immune cells and signaling molecules affect tumor progression and therapeutic responses. Understanding the composition and function of the TIME in primary cutaneous melanoma is useful for prognostication and therapeutic decisions. This review provides an overview of the components of the TIME in primary cutaneous melanoma, and their influence on clinical outcomes.
Collapse
Affiliation(s)
- Catherine Zilberg
- The University of Sydney, NSW , Camperdown, 2050, Australia.
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Missenden Rd, Camperdown, NSW, 2050, Australia.
| | - Angela L Ferguson
- The University of Sydney, NSW , Camperdown, 2050, Australia
- Centenary Institute, The University of Sydney, Missenden Rd, Camperdown, NSW, 2050, Australia
| | - J Guy Lyons
- Centenary Institute, The University of Sydney, Missenden Rd, Camperdown, NSW, 2050, Australia
- Department of Dermatology, The University of Sydney at Royal Prince Alfred Hospital, Missenden Rd, NSW , Camperdown, 2050, Australia
| | - Ruta Gupta
- The University of Sydney, NSW , Camperdown, 2050, Australia
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Missenden Rd, Camperdown, NSW, 2050, Australia
| | - Diona L Damian
- Department of Dermatology, The University of Sydney at Royal Prince Alfred Hospital, Missenden Rd, NSW , Camperdown, 2050, Australia
- Melanoma Institute Australia, 40 Rocklands Rd, NSW, Wollstonecraft, 2065, Australia
| |
Collapse
|
30
|
Fusco C, Di Rella F, Liotti A, Colamatteo A, Ferrara AL, Gigantino V, Collina F, Esposito E, Donzelli I, Porcellini A, Feola A, Micillo T, Perna F, Garziano F, Maniscalco GT, Varricchi G, Mottola M, Zuccarelli B, De Simone B, di Bonito M, Matarese G, Accurso A, Pontillo M, Russo D, Insabato L, Spaziano A, Cantone I, Pezone A, De Rosa V. CD4 +FOXP3Exon2 + regulatory T cell frequency predicts breast cancer prognosis and survival. SCIENCE ADVANCES 2025; 11:eadr7934. [PMID: 39813341 PMCID: PMC11734725 DOI: 10.1126/sciadv.adr7934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 12/13/2024] [Indexed: 01/18/2025]
Abstract
CD4+FOXP3+ regulatory T cells (Tregs) suppress immune responses to tumors, and their accumulation in the tumor microenvironment (TME) correlates with poor clinical outcome in several cancers, including breast cancer (BC). However, the properties of intratumoral Tregs remain largely unknown. Here, we found that a functionally distinct subpopulation of Tregs, expressing the FOXP3 Exon2 splicing variants, is prominent in patients with hormone receptor-positive BC with poor prognosis. Notably, a comprehensive examination of the TCGA validated FOXP3E2 as an independent prognostic marker in all other BC subtypes. We found that FOXP3E2 expression underlies BCs with defective mismatch repair and a stem-like signature and highlights pathways involved in tumor survival. Last, we found that the TME induces FOXP3E2 through the CXCL12/CXCR4 axis and confirmed the higher immunosuppressive capacity of FOXP3E2+ Tregs derived from patients with BC. Our study suggests that FOXP3E2+ Tregs might be used as an independent biomarker to predict BC prognosis and survival and to develop super-targeted immunotherapies.
Collapse
Affiliation(s)
- Clorinda Fusco
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
| | - Francesca Di Rella
- Oncologia Clinica Sperimentale di Senologia, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Antonietta Liotti
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “G. Salvatore”, IEOS-CNR, Napoli, Italy
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Alessandra Colamatteo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
| | - Anne Lise Ferrara
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Vincenzo Gigantino
- Unità di Anatomia Patologica, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Francesca Collina
- Unità di Anatomia Patologica, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Emanuela Esposito
- Chirurgia Oncologica di Senologia, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Ivana Donzelli
- Chirurgia Oncologica di Senologia, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Antonio Porcellini
- Dipartimento di Biologia, Complesso Universitario di Monte Sant’Angelo, Università di Napoli “Federico II”, Napoli 80126, Italy
| | - Antonia Feola
- Dipartimento di Biologia, Complesso Universitario di Monte Sant’Angelo, Università di Napoli “Federico II”, Napoli 80126, Italy
| | - Teresa Micillo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
| | - Francesco Perna
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Federica Garziano
- U.O.C Biochimica Clinica Azienda Ospedaliera Specialistica dei Colli Monaldi-Cotugno-C.T.O. Presidio Monaldi, Napoli, Italy
| | - Giorgia Teresa Maniscalco
- Clinica Neurologica e Unità Stroke, Centro Sclerosi Multipla, Ospedale “A.Cardarelli”, Napoli 80131, Italy
| | - Gilda Varricchi
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “G. Salvatore”, IEOS-CNR, Napoli, Italy
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Maria Mottola
- U.O.C Medicina Trasfusionale, Azienda Ospedaliera Specialistica dei Colli, Napoli 80131, Italy
| | - Bruno Zuccarelli
- U.O.C Medicina Trasfusionale, Azienda Ospedaliera Specialistica dei Colli, Napoli 80131, Italy
| | - Bruna De Simone
- Dipartimento di Sanità Pubblica, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Maurizio di Bonito
- Unità di Anatomia Patologica, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “G. Salvatore”, IEOS-CNR, Napoli, Italy
| | - Antonello Accurso
- Dipartimento di Chirurgia Generale, Oncologica, Bariatrica e Metabolica, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Martina Pontillo
- Dipartimento di Chirurgia Generale, Oncologica, Bariatrica e Metabolica, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Daniela Russo
- Unità di Anatomia Patologica, Dipartimento di Scienze Biomediche Avanzate, Facoltà di Medicina, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Luigi Insabato
- Unità di Anatomia Patologica, Dipartimento di Scienze Biomediche Avanzate, Facoltà di Medicina, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Alessandra Spaziano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
| | - Irene Cantone
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “G. Salvatore”, IEOS-CNR, Napoli, Italy
| | - Antonio Pezone
- Dipartimento di Biologia, Complesso Universitario di Monte Sant’Angelo, Università di Napoli “Federico II”, Napoli 80126, Italy
| | - Veronica De Rosa
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “G. Salvatore”, IEOS-CNR, Napoli, Italy
| |
Collapse
|
31
|
Mohd Faizal NF, Shai S, Savaliya BP, Karen-Ng LP, Kumari R, Kumar R, Vincent-Chong VK. A Narrative Review of Prognostic Gene Signatures in Oral Squamous Cell Carcinoma Using LASSO Cox Regression. Biomedicines 2025; 13:134. [PMID: 39857718 PMCID: PMC11759772 DOI: 10.3390/biomedicines13010134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/28/2024] [Accepted: 01/01/2025] [Indexed: 01/27/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most common malignancies of the head and neck squamous cell carcinoma (HNSCC). HNSCC is recognized as the eighth most commonly occurring cancer globally in men. It is essential to distinguish between cancers arising in the head and neck regions due to significant differences in their etiologies, treatment approaches, and prognoses. As the Cancer Genome Atlas (TCGA) dataset is available in HNSCC, the survival analysis prognosis of OSCC patients based on the TCGA dataset for discovering gene expression-based prognostic biomarkers is limited. To address this paucity, we aimed to provide comprehensive evidence by recruiting studies that have reported new biomarkers/signatures to establish a prognostic model to predict the survival of OSCC patients. Using PubMed search, we have identified 34 studies that have been using the least absolute shrinkage and selection operator (LASSO)-based Cox regression analyses to establish signature prognosis that related to different pathways in OSCC from the past 4 years. Our review was focused on summarizing these signatures and implications for targeted therapy using FDA-approved drugs. Furthermore, we conducted an analysis of the LASSO Cox regression gene signatures. Our findings revealed 13 studies that correlated a greater number of regulatory T cells (Tregs) cells in protective gene signatures with increased recurrence-free and overall survival rates. Conversely, two studies displayed an opposing trend in cases of OSCC. We will also explore how the dysregulation of these signatures impacts immune status, promoting tumor immune evasion or, conversely, enhancing immune surveillance. Overall, this review will provide new insight for future anti-cancer therapies based on the potential gene that is associated with poor prognosis in OSCC.
Collapse
Affiliation(s)
- Nur Fatinazwa Mohd Faizal
- Oral Cancer Research & Coordinating Centre (OCRCC), Faculty of Dentistry, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (N.F.M.F.); (L.P.K.-N.)
| | - Saptarsi Shai
- Baylor College of Medicine, Texas Children’s Hospital, Houston, TX 77030, USA;
| | - Bansi P. Savaliya
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55901, USA;
| | - Lee Peng Karen-Ng
- Oral Cancer Research & Coordinating Centre (OCRCC), Faculty of Dentistry, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (N.F.M.F.); (L.P.K.-N.)
| | - Rupa Kumari
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Rahul Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Vui King Vincent-Chong
- Center for Oral Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
32
|
Chen X, Agustinus AS, Li J, DiBona M, Bakhoum SF. Chromosomal instability as a driver of cancer progression. Nat Rev Genet 2025; 26:31-46. [PMID: 39075192 DOI: 10.1038/s41576-024-00761-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 07/31/2024]
Abstract
Chromosomal instability (CIN) refers to an increased propensity of cells to acquire structural and numerical chromosomal abnormalities during cell division, which contributes to tumour genetic heterogeneity. CIN has long been recognized as a hallmark of cancer, and evidence over the past decade has strongly linked CIN to tumour evolution, metastasis, immune evasion and treatment resistance. Until recently, the mechanisms by which CIN propels cancer progression have remained elusive. Beyond the generation of genomic copy number heterogeneity, recent work has unveiled additional tumour-promoting consequences of abnormal chromosome segregation. These mechanisms include complex chromosomal rearrangements, epigenetic reprogramming and the induction of cancer cell-intrinsic inflammation, emphasizing the multifaceted role of CIN in cancer.
Collapse
Affiliation(s)
- Xuelan Chen
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Albert S Agustinus
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Pharmacology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Jun Li
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Melody DiBona
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samuel F Bakhoum
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
33
|
Ferkel SAM, Holman EA, Sojwal RS, Rubin SJS, Rogalla S. Tumor-Infiltrating Immune Cells in Colorectal Cancer. Neoplasia 2025; 59:101091. [PMID: 39642846 DOI: 10.1016/j.neo.2024.101091] [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: 11/07/2024] [Accepted: 11/18/2024] [Indexed: 12/09/2024]
Abstract
Colorectal cancer encompasses a heterogeneous group of malignancies that differ in pathophysiological mechanisms, immune response and infiltration, therapeutic response, and clinical prognosis. Numerous studies have highlighted the clinical relevance of tumor-infiltrating immune cells among different types of colorectal tumors yet vary in cell type definitions and cell identification strategies. The distinction of immune signatures is particularly challenging when several immune subtypes are involved but crucial to identify novel intercellular mechanisms within the tumor microenvironment. In this review, we compile human and non-human studies on tumor-infiltrating immune cells and provide an overview of immune subtypes, their pathophysiological functions, and their prognostic role in colorectal cancer. We discuss how differentiating immune signatures can guide the development of immunotherapeutic targets and personalized treatment regimens. We analyzed comprehensive human protein biomarker profiles across the entire immune spectrum to improve interpretability and application of tumor studies and to ultimately enhance immunotherapy and advance precision medicine for colorectal cancer patients.
Collapse
Affiliation(s)
- Sonia A M Ferkel
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Elizabeth A Holman
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Raoul S Sojwal
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Samuel J S Rubin
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Stephan Rogalla
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA.
| |
Collapse
|
34
|
Chen L, Tan H, Geng R, Li Y, Wang Y, Li T. Immune Signatures of Solid Tumor Patients Treated With Immune Checkpoint Inhibitors: An Observational Study. Thorac Cancer 2025; 16:e15493. [PMID: 39582218 PMCID: PMC11729440 DOI: 10.1111/1759-7714.15493] [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: 09/10/2024] [Revised: 10/18/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024] Open
Abstract
PURPOSE Our study aimed to comprehensively describe the features of peripheral blood multiple immune cell phenotypes in solid tumor patients during pretreatment and after immunotherapy, providing a more convenient approach for studying the prognosis of immunotherapy in different solid tumor patients. METHODS We prospectively recruited patients with advanced solid tumors from Peking Union Medical College Hospital (PUMCH) between February 2023 and April 2024. Using multicolor flow cytometry, our study comprehensively observed and described the signatures of peripheral blood lymphocyte subsets including activation, proliferation, function, naïve memory, and T cell exhaustion immune cell subsets in this population of pretreatment and after immunotherapy. RESULTS Our study enrolled 59 advanced solid tumor patients with immunotherapy and 59 healthy controls were matched by age and gender. The results demonstrated a marked upregulation in the expression of lymphocyte activation markers CD38 and HLA-DR, as well as exhaustion and proliferation markers PD-1 and Ki67, in solid tumor patients compared to healthy controls. After immune checkpoint blockade (ICB) treatment, mainly the expression of Ki67CD4+T and HLA-DRCD38CD4+T, was significantly upregulated compared to pretreatment levels (p = 0.017, p = 0.019, respectively). We further found that gynecological tumors with better prognoses had higher baseline activation levels of CD4+ T cells compared to other solid tumors with poorer prognoses. CONCLUSION Our study elucidated the characteristics of different lymphocyte subsets in the peripheral blood of solid tumor patients. Further research revealed changes in the phenotypes of different lymphocyte subsets after ICIs treatment, with the activated phenotype of CD4+ T cells playing a crucial role in the antitumor effect. This lays the groundwork for further exploration of prognostic biomarkers and predictive models for cancer patients with immunotherapy.
Collapse
Affiliation(s)
- Ling Chen
- Department of Infectious DiseasesPeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Hourui Tan
- Department of Medical OncologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ruixuan Geng
- Department of International Medical ServicesPeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yifan Li
- Department of Gynecologic OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yingyi Wang
- Department of Medical OncologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Taisheng Li
- Department of Infectious DiseasesPeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
35
|
Dorjkhorloo G, Shiraishi T, Erkhem-Ochir B, Sohda M, Okami H, Yamaguchi A, Shioi I, Komine C, Nakazawa N, Shibasaki Y, Okada T, Osone K, Sano A, Sakai M, Ogawa H, Katayama A, Oyama T, Yokobori T, Shirabe K, Saeki H. High levels of fibrotic tumor components are associated with recurrence and intratumoral immune status in advanced colorectal cancer patients. Sci Rep 2024; 14:30735. [PMID: 39730445 DOI: 10.1038/s41598-024-80489-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/19/2024] [Indexed: 12/29/2024] Open
Abstract
The importance of collagen and elastin remains incompletely understood concerning tumor immunity in cancer tissues. This study explored the clinical significance of collagen and elastin deposition on tumor immunity in advanced colorectal cancer patients. The collagen and elastin contents were assessed simultaneously using elastic van Gieson (EVG) histochemical staining. Immunohistochemical staining was performed to measure the immune cell markers CD3, CD8, CD86, and CD163 in surgically resected primary tumors from 78 pT4 colorectal cancer patients. High collagen, elastin, and EVG scores are associated with aggressive characteristics and short disease-free survival. A high EVG score was identified as an independent predictor of poor disease-free survival. Furthermore, tumors with high collagen and EVG scores exhibited significantly fewer intratumoral CD3 + and CD8 + cells. Evaluating tumor fibrosis using the classical and straightforward EVG staining method could be a reliable predictor of recurrence in high-risk colorectal cancer patients with tumor immune tolerance.
Collapse
Affiliation(s)
- Gendensuren Dorjkhorloo
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Takuya Shiraishi
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Bilguun Erkhem-Ochir
- Research Program for Omics-based Medical Science, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Makoto Sohda
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan.
| | - Haruka Okami
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Arisa Yamaguchi
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Ikuma Shioi
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Chika Komine
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Nobuhiro Nakazawa
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Yuta Shibasaki
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Takuhisa Okada
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Katsuya Osone
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Akihiko Sano
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Makoto Sakai
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hiroomi Ogawa
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Ayaka Katayama
- Department of Diagnostic Pathology, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Tetsunari Oyama
- Department of Diagnostic Pathology, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Takehiko Yokobori
- Research Program for Omics-based Medical Science, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan.
| | - Ken Shirabe
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hiroshi Saeki
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
36
|
Velázquez-Enríquez JM, Cerna R, Beltrán-Ramírez O, Piña-Vázquez C, Villa-Treviño S, Vásquez-Garzón VR. DCLK1 is Overexpressed and Associated with Immune Cell Infiltration in Hepatocellular Carcinoma. Biochem Genet 2024; 62:4280-4302. [PMID: 38294590 DOI: 10.1007/s10528-024-10667-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 01/02/2024] [Indexed: 02/01/2024]
Abstract
Recent research has shown that Doublecortin-like kinase 1 (DCLK1) is overexpressed in different types of cancer. It has recently been described as a cancer stem cells (CSCs) marker, is associated with carcinogenesis, and positively correlates with infiltration of multiple immune cell types in some cancers. However, studies focused on assessing DCLK1 expression in HCC are limited, and the role of DCLK1 in HCC tumor immunity remains to be determined. In this study, we used a modified model of the resistant hepatocyte (MRHM) to evaluate DCLK1 expression in HCC. Furthermore, DCLK1 expression in HCC was analyzed using TIMER 2.0, UALCAN, GEPIA, GEO, and HPA web-based tools. Correlations between DCLK1 expression and clinicopathological factors in patients were analyzed using the UALCAN web-based tool. Finally, correlations between DCLK1 and immune infiltrates were investigated using the TIMER 2.0 and TISIDB web-based tools. The results showed that DCLK1 is significantly overexpressed during progression of the HCC carcinogenic process in the MRHM. DCLK1 is overexpressed in HCC according to multiple publics web-based tools, and its overexpression is associated with cancer stage. Furthermore, DCLK1 expression was correlated with infiltration levels of multiple immune cells, immunomodulatory factors, immunoinhibitors, MHC molecules, chemokines, receptors, and immune cell-specific markers. These results suggest that DCLK1 is a potential prognostic biomarker that determines cancer progression and correlates with immune cell infiltration in HCC.
Collapse
Affiliation(s)
- Juan Manuel Velázquez-Enríquez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, C.P. 68020, Oaxaca, México
| | - Renata Cerna
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, C.P. 07360, Ciudad de México, México
| | - Olga Beltrán-Ramírez
- Universidad Estatal de Sonora, Unidad Académica Navojoa, Boulevard Manlio Fabio Beltrones 810, Colonia Bugambilias, C.P. 85875, Navojoa, Sonora, México
| | - Carolina Piña-Vázquez
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, C.P. 07360, Ciudad de México, México
| | - Saúl Villa-Treviño
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, C.P. 07360, Ciudad de México, México
| | - Verónica Rocío Vásquez-Garzón
- CONACYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, C.P. 68020, Oaxaca, México.
| |
Collapse
|
37
|
Rachunek-Medved K, Krauß S, Daigeler A, Adams C, Eckert F, Ganser K, Gonzalez-Menendez I, Quintanilla-Martinez L, Kolbenschlag J. Acute remote ischemic conditioning enhances (CD3+)- but not (FoxP3+)-T-cell invasion in the tumor center and increases IL 17 and TNF-alpha expression in a murine melanoma model. Front Immunol 2024; 15:1501885. [PMID: 39650654 PMCID: PMC11621216 DOI: 10.3389/fimmu.2024.1501885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/29/2024] [Indexed: 12/11/2024] Open
Abstract
Introduction Hypoxia can drive tumor progression, suppress anti-tumor immunity, and reduce the effectiveness of radiotherapy and chemotherapy. This study aimed to assess the impact of remote ischemic conditioning (RIC) on tumor oxygenation (sO2) and the anti-tumor immune response. Material and methods Fourteen B16-Ova tumor-bearing C57BL/6N mice received six 5-minute RIC cycles, while another fourteen underwent anesthesia only. Pimonidazole was administered 1.5 hours before sacrifice. Blood flow, sO2, and hemoglobin levels were measured in the non-ischemic hind limb and tumor. Tumor hypoxia was assessed using pimonidazole and CA IX immunohistochemistry, and T cell infiltration by CD3 and FoxP3 staining. Serum levels of 23 cytokines were analyzed using a multiplex immunoassay. Results Isoflurane anesthesia caused a slight intraindividual increase in blood flow (p = 0.07) and sO2 (p = 0.06) of the hind limb and a sole increase in tumor sO2 (p = 0.035), whereas RIC improved sO2 of the tumor in relation to the hind limb (p=0.03). The median of the tumor oxygen saturation reached 51.4% in the control group and 62.7% in the RIC group (p = 0.09), exhibiting a slight tendency towards better oxygenation in the RIC group. Pimonidazole (p=0.24) and CA IX hypoxia score (p=0.48) did not reveal statistically significant differences between the two groups. In RIC-treated tumors, the number of CD3 (p=0.006), but not FoxP3- positive cells (p = 0.84), in the tumor core was significantly higher compared to the control group. In the RIC group, the mean fluorescence intensity (MFI) of IL-17 was significantly higher (p=0.035), and TNF-α was trend-wise higher (p=0.063) compared to the control group. Conclusion Both isoflurane anesthesia and RIC have an impact on microcirculation. The application of RIC counteracted some of the effects of isoflurane, primarily in healthy tissue, and led to a significant improvement in relative tumor tissue oxygenation compared to the non-ischemic hind limb. RIC selectively enhanced immune infiltration within the tumor center, probably by previously activated tumor infiltrating T cells, while having no significant impact on T-regulatory cells. RIC appears to impact the cytokine profile, as indicated by elevated MFIs of TNF-α and IL-17.
Collapse
Affiliation(s)
- Katarzyna Rachunek-Medved
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Sabrina Krauß
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Adrien Daigeler
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Constantin Adams
- Department of Paediatrics, University Hospital Tuebingen, Tuebingen, Germany
| | - Franziska Eckert
- Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
| | - Katrin Ganser
- Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
| | - Irene Gonzalez-Menendez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, University Hospital Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, Tuebingen, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, University Hospital Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, Tuebingen, Germany
| | - Jonas Kolbenschlag
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
38
|
Inés SM, Celia MO, Lasarte JJ, Teresa L. Optimizing protocols for human regulatory T isolation, expansion, and characterization. Methods Cell Biol 2024; 191:59-77. [PMID: 39824564 DOI: 10.1016/bs.mcb.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2025]
Affiliation(s)
- Sánchez-Moreno Inés
- Program in Immunology and Immunotherapy, Cima Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Martín-Otal Celia
- Program in Immunology and Immunotherapy, Cima Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Juan José Lasarte
- Program in Immunology and Immunotherapy, Cima Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Lozano Teresa
- Program in Immunology and Immunotherapy, Cima Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), Navarra Institute for Health Research (IDISNA), Pamplona, Spain.
| |
Collapse
|
39
|
Qixin Y, Jing H, Jiang H, Xueyang L, Lu Y, Yuehua L. Transcriptome-based network analysis related to regulatory T cells infiltration identified RCN1 as a potential biomarker for prognosis in clear cell renal cell carcinoma. BioData Min 2024; 17:51. [PMID: 39543725 PMCID: PMC11566375 DOI: 10.1186/s13040-024-00404-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Regulatory T cells (Tregs) play a critical role in shaping the immunosuppressive microenvironment within tumors. Investigating the role of Tregs in Clear cell renal cell carcinoma (ccRCC) is crucial for identifying prognostic markers and therapeutic targets for ccRCC. METHODS Weighted gene co-expression network analysis (WGCNA) was utilized to pinpoint modules related to Treg infiltration in TCGA-KIRC samples. Following this, consensus clustering was employed to derive two clusters associated with Treg infiltration in ccRCC. A prognostic model was then developed using the gene module associated with Treg infiltration. We then evaluated the ability of the prognostic model to predict ccRCC overall survival and demonstrated that RCN1 can be used as a target to predict ccRCC prognosis. RESULTS We deduce that the two clusters associated with Treg infiltration exhibit distinct compositions of the immune microenvironment, pathway activations, prognosis, and drug sensitivities commonly utilized in ccRCC treatment. Furthermore, a 7-gene model risk score, developed based on ccRCC Treg infiltration, proved to be a reliable prognostic marker in both training and validation cohorts. Additionally, survival analysis indicated that RCN1 serves as a reliable prognostic factor for ccRCC. Single-cell sequencing analysis revealed that RCN1 is predominantly expressed in tumor cells. A pan-cancer analysis highlighted that RCN1 is linked with poor prognosis and the activation of inflammatory response pathways across various cancers. CONCLUSION We developed a prognostic model associated with Treg infiltration, which facilitates the clinical categorization of ccRCC progression. Moreover, our findings underscore the significant potential of RCN1 as a ccRCC biomarker.
Collapse
Affiliation(s)
- Yang Qixin
- Department of Urology, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, P.R. China
| | - Huang Jing
- Department of Rehabilitation, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, P.R. China
| | - He Jiang
- Department of Urology, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, P.R. China
| | - Liu Xueyang
- Department of Urology, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, P.R. China
| | - Yu Lu
- Department of Urology, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, P.R. China
| | - Li Yuehua
- Department of Urology, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, P.R. China.
| |
Collapse
|
40
|
Ding G, Wang T, Sun F, Liu M, Tang G, Yu S, Chu Y, Ma J, Cui Y, Wu G, Wu J. Multi-omics analysis of Prolyl 3-hydroxylase 1 as a prognostic biomarker for immune infiltration in ccRCC. NPJ Precis Oncol 2024; 8:256. [PMID: 39516330 PMCID: PMC11549470 DOI: 10.1038/s41698-024-00748-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The formation of human collagen requires the presence of Prolyl 3-hydroxylase 1 (P3H1), but the regulatory mechanism of P3H1 remained insufficiently understood. Our study aimed to identify the role of P3H1 in clear cell renal cell carcinoma (ccRCC). P3H1 expression in ccRCC was validated using multiple databases and in vitro experiments. We performed a correlation analysis of P3H1 with drug sensitivity, immune checkpoints, and immune cell infiltration using transcriptome and single-cell sequencing. Drawing upon the Encyclopedia of RNA Interactomes database, we selected P3H1 as the focal point of our investigation, meticulously uncovering the intricate network of microRNAs and lncRNAs that potentially orchestrate ceRNA mechanisms. This study employs a multidimensional approach integrating vitro assays and multi-omics bioinformatics analyses to investigate P3H1's impact on ccRCC prognosis, immune modulation, immune checkpoints, ceRNA regulatory network, drug sensitivity, and therapeutic responses, aiming to uncover new insights into its therapeutic potential and inform future clinical strategies.
Collapse
Affiliation(s)
- Guixin Ding
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Tianqi Wang
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Fengze Sun
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Ming Liu
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Gonglin Tang
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Shengqiang Yu
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Yongli Chu
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Department of Scientific Research, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Jian Ma
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Yuanshan Cui
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China.
| | - Gang Wu
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China.
| | - Jitao Wu
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China.
| |
Collapse
|
41
|
Huo MH, Adeerjiang Y, Abulitipu A, Khan U, Li XX, Zhang L, Tian Y, Jiang S, Xu CC, Chao XZ, Yang YF, Zhang JX, Du GL. Th17/Treg cell balance in patients with papillary thyroid carcinoma: a new potential biomarker and therapeutic target. Front Oncol 2024; 14:1325575. [PMID: 39534095 PMCID: PMC11554530 DOI: 10.3389/fonc.2024.1325575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Papillary thyroid carcinoma (PTC) is the most common subtype of thyroid carcinoma. The most effective treatment for PTC is surgical resection, and patients who undergo surgery have good survival outcomes, but some patients have distant metastasis or even multiorgan metastases at the time of initial diagnosis. Distant metastasis is associated with poorer prognosis and a higher mortality rate. Helper T lymphocyte 17 (Th17) cells and regulatory T lymphocytes (Tregs) play different roles in PTC, and the Th17/Treg balance is closely related to the progression of PTC. Th17 cells play anticancer roles, whereas Tregs play cancer-promoting roles. A Th17/Treg imbalance promotes tumor progression and accelerates invasive behaviors such as tumor metastasis. Th17/Treg homeostasis can be regulated by the TGF-β/IL-2 and IL-6 cytokine axes. Immune checkpoint inhibitors contribute to Treg/Th17 cell homeostasis. For PTC, monoclonal antibodies against CTLA-4, PD-1 and PD-L1 inhibit the activation of Tregs, reversing the Th17/Treg cell imbalance and providing a new option for the prevention and treatment of PTC. This article reviews the role of Tregs and Th17 cells in PTC and their potential targets, aiming to provide better treatment options for PTC.
Collapse
Affiliation(s)
- Meng-Han Huo
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Gastroenterology and Endocrinology, Tianjin Haihe Hospital, Tianjin, China
| | - Yilinuer Adeerjiang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ayiguzhali Abulitipu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Umair Khan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xin-Xi Li
- Department of Endocrine Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Lei Zhang
- Department of Endocrine Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Ye Tian
- Department of Endocrine Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Sheng Jiang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Can-Can Xu
- First Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xian-Zhen Chao
- First Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Ye-Fan Yang
- First Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Jin-Xia Zhang
- First Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Guo-Li Du
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Endocrinology, Bayingolin Mongolian Autonomous Prefecture People's Hospital, Kuerle, China
| |
Collapse
|
42
|
Shang S, Hu L, Wu C, Wu J, Chen M, Zhu G, Xu WY, Zhang Y, Sun G, Wei Z. Unveiling the pan-cancer landscape of S100A16: A comprehensive analysis of prognostic significance, drug sensitivity, and immunomodulatory roles. Medicine (Baltimore) 2024; 103:e39998. [PMID: 39465820 PMCID: PMC11479482 DOI: 10.1097/md.0000000000039998] [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: 01/24/2024] [Accepted: 09/19/2024] [Indexed: 10/29/2024] Open
Abstract
Accumulating evidence supports the notion that S100A16 exhibits differential expression in many human cancers, affecting cellular functions associated with tumorigenesis through various signaling pathways. While extensive research has been conducted on S100A16 in specific cancer types, a comprehensive evaluation of its role across diverse cancers remains lacking. To explore the prognostic significance, drug sensitivity, and immunomodulatory roles of S100A16, a thorough analysis was conducted at a pan-cancer level using multiple databases. Our findings revealed high expression of S100A16 RNA in various human cancers. Importantly, this elevated expression was linked to disease prognosis and drug sensitivity across a spectrum of cancers. Genetic alterations in S100A16 were characterized across multiple cancer types, and a confirmed correlation was observed in the prognosis of skin cutaneous melanoma (SKCM). Furthermore, our study demonstrated a significant association between S100A16 expression and the infiltrating levels of diverse cell types in the tumor microenvironment (TME), suggesting its potential as a prognosis predictor for immunotherapy. Novel collections of miRNAs, such as has-miR-423-5p, has-miR-769-5p, has-miR-151a-3p, and has-miR-550a-5p, targeting S100A16 at a pan-cancer level were predicted through various databases. These findings contribute to a comprehensive understanding the role of S100A16 in prognosis prediction, chemotherapy, and immunotherapy, providing valuable insights for identifying novel targets in cancer treatment.
Collapse
Affiliation(s)
- Shixuan Shang
- Department of Pathology, The First People’s Hospital of Jiangxia District, Wuhan, China
| | - Ling Hu
- Department of Medical Examination, Wuhan Red Cross Hospital, Wuhan, China
| | - Chengbi Wu
- Department of Medical Laboratory, Wuhan Red Cross Hospital, Wuhan, China
| | - Jin Wu
- Department of Pathology, The First People’s Hospital of Jiangxia District, Wuhan, China
| | - Meixiang Chen
- Department of Pathology, The First People’s Hospital of Jiangxia District, Wuhan, China
| | - Guoqiang Zhu
- Department of Medicine, Singlera Genomics (Shanghai) Ltd., Shanghai, China
| | - Wang-yang Xu
- Department of Medicine, Singlera Genomics (Shanghai) Ltd., Shanghai, China
| | - Yunzhi Zhang
- Department of Medicine, Singlera Genomics (Shanghai) Ltd., Shanghai, China
| | - Ge Sun
- Department of Medicine, Singlera Genomics (Shanghai) Ltd., Shanghai, China
| | - Zexiong Wei
- Department of Oncology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
43
|
Wu B, Tapadar S, Ruan Z, Sun CQ, Arnold RS, Johnston A, Olugbami JO, Arunsi U, Gaul DA, Petros JA, Kobayashi T, Duda DG, Oyelere AK. A Novel Liver Cancer-Selective Histone Deacetylase Inhibitor Is Effective against Hepatocellular Carcinoma and Induces Durable Responses with Immunotherapy. ACS Pharmacol Transl Sci 2024; 7:3155-3169. [PMID: 39416967 PMCID: PMC11475281 DOI: 10.1021/acsptsci.4c00358] [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: 06/11/2024] [Revised: 08/16/2024] [Accepted: 08/27/2024] [Indexed: 10/19/2024]
Abstract
Hepatocellular carcinoma (HCC) progression is facilitated by gene-silencing chromatin histone hypoacetylation due to histone deacetylase (HDAC) activation. However, inhibiting HDACs-an effective treatment for lymphomas-has shown limited success in solid tumors. We report the discovery of a class of HDAC inhibitors (HDACi) that demonstrates exquisite selective cytotoxicity against human HCC cells. The lead compound STR-V-53 (3) showed a favorable safety profile in mice and robustly suppressed tumor growth in orthotopic xenograft models of HCC. When combined with the anti-HCC drug sorafenib, STR-V-53, showed greater in vivo efficacy. Moreover, STR-V-53 combined with anti-PD1 therapy increased the CD8+ to regulatory T-cell (Treg) ratio and survival in an orthotopic HCC model in immunocompetent mice. This combination therapy resulted in durable responses in 40% of the mice. Transcriptomic analysis revealed that STR-V-53 primed HCC cells to immunotherapy through HDAC inhibition, impaired glucose-regulated transcription, impaired DNA synthesis, upregulated apoptosis, and stimulated the immune response pathway. Collectively, our data demonstrate that the novel HDACi STR-V-53 is an effective anti-HCC agent that can induce profound responses when combined with standard immunotherapy.
Collapse
Affiliation(s)
- Bocheng Wu
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, Georgia 30332-0400, United States
| | - Subhasish Tapadar
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, Georgia 30332-0400, United States
- Sophia
Bioscience, Inc., 311
Ferst Drive NW, Ste. L1325A, Atlanta, Georgia 30332, United States
| | - Zhiping Ruan
- Edwin
L. Steele Laboratories for Tumor Biology, Department of Radiation
Oncology, Harvard Medical School & Massachusetts
General Hospital, Boston, Massachusetts 02114, United States
- Department
of Medical Oncology, The First Affiliated
Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Carrie Q. Sun
- Department
of Urology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Rebecca S. Arnold
- Department
of Urology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Alexis Johnston
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, Georgia 30332-0400, United States
| | - Jeremiah O. Olugbami
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, Georgia 30332-0400, United States
| | - Uche Arunsi
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, Georgia 30332-0400, United States
| | - David A. Gaul
- Sophia
Bioscience, Inc., 311
Ferst Drive NW, Ste. L1325A, Atlanta, Georgia 30332, United States
| | - John A. Petros
- Department
of Urology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Tatsuya Kobayashi
- Edwin
L. Steele Laboratories for Tumor Biology, Department of Radiation
Oncology, Harvard Medical School & Massachusetts
General Hospital, Boston, Massachusetts 02114, United States
| | - Dan G. Duda
- Edwin
L. Steele Laboratories for Tumor Biology, Department of Radiation
Oncology, Harvard Medical School & Massachusetts
General Hospital, Boston, Massachusetts 02114, United States
| | - Adegboyega K. Oyelere
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, Georgia 30332-0400, United States
- Parker
H.
Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, United States
| |
Collapse
|
44
|
Tang B, Chen Y, Jiang Y, Fang M, Gao Q, Ren X, Yao L, Huang G, Chen J, Zhang X, Li R, Zhao S, Gao M, Luo R, Qi M, Li F, Zheng F, Lee M, Tao X, Duan R, Guo J, Chi Z, Cui C. Toripalimab in combination with HBM4003, an anti-CTLA-4 heavy chain-only antibody, in advanced melanoma and other solid tumors: an open-label phase I trial. J Immunother Cancer 2024; 12:e009662. [PMID: 39366752 PMCID: PMC11459314 DOI: 10.1136/jitc-2024-009662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND HBM4003 is a novel anti-CTLA-4 heavy chain-only antibody, designed to enhance Treg ablation and antibody-dependent cell-mediated cytotoxicity while ensuring a manageable safety profile. This phase I trial investigated the safety, pharmacokinetics, immunogenicity and preliminary efficacy of HBM4003 plus with anti-PD-1 antibody toripalimab in patients with advanced solid tumors, especially focusing on melanoma. METHODS The multicenter, open-label phase I trial was divided into two parts: dose-escalation phase (part 1) and dose-expansion phase (part 2). In part 1, HBM4003 was administered at doses of 0.03, 0.1, 0.3 mg/kg in combination with toripalimab with fixed dosage of 240 mg every 3 weeks. The recommended phase II dose (RP2D) was used in the expansion phase. Primary endpoints were safety and RP2D in part 1 and objective response rate (ORR) in part 2. Biomarkers based on cytokines and multiplex immunofluorescence staining were explored. RESULTS A total of 40 patients received study treatment, including 36 patients treated with RP2D of HBM4003 0.3 mg/kg plus toripalimab 240 mg every 3 week. 36 participants (90.0%) experienced at least one treatment-related adverse event (TRAE), of which 10 (25.0%) patients experienced grade ≥3 TRAEs and 5 (12.5%) experienced immune-mediated adverse events (irAEs) with maximum severity of grade 3. No grade 4 or 5 irAEs occurred. Efficacy analysis set included 32 melanoma patients treated with RP2D and with available post-baseline imaging data. The ORRs of anti-PD-1/PD-L1 treatment-naïve subgroup and anti-PD-1/PD-L1 treatment-failed subgroup were 33.3% and 5.9%, respectively. In mucosal melanoma, the ORR of the two subgroups were 40.0% and 10.0%, respectively. Baseline high Treg/CD4+ratio in the tumor serves as an independent predictive factor for the efficacy of immunotherapy. CONCLUSIONS HBM4003 0.3 mg/kg plus toripalimab 240 mg every 3 week demonstrated manageable safety in solid tumors and no new safety signal. Limited data demonstrated promising antitumor activity, especially in PD-1 treatment-naïve mucosal melanoma. TRIAL REGISTRATION NUMBER NCT04727164.
Collapse
Affiliation(s)
- Bixia Tang
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Yu Chen
- Department of Medical Oncology, Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China
| | - Yu Jiang
- Department of Head and Neck Oncology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China
| | - Meiyu Fang
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Quanli Gao
- Immunotherapy Department, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Li Yao
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Gang Huang
- Central South University (Hunan Cancer Hospital), Changsha, Hunan, China
| | - Jing Chen
- Union Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoshi Zhang
- Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Rongqing Li
- Tumor Radiotherapy Department, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | | | | | | | - Meng Qi
- Harbour BioMed, Shanghai, China
| | - Feng Li
- Harbour BioMed, Shanghai, China
| | | | | | | | - Rong Duan
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Jun Guo
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhihong Chi
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Chuanliang Cui
- Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
45
|
Semancik CS, Zhao N, Koestler DC, Boerwinkle E, Bressler J, Buchsbaum RJ, Kelsey KT, Platz EA, Michaud DS. DNA Methylation-Derived Immune Cell Proportions and Cancer Risk in Black Participants. CANCER RESEARCH COMMUNICATIONS 2024; 4:2714-2723. [PMID: 39324671 PMCID: PMC11484294 DOI: 10.1158/2767-9764.crc-24-0257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/31/2024] [Accepted: 09/24/2024] [Indexed: 09/27/2024]
Abstract
SIGNIFICANCE This study describes associations between immune cell types and cancer risk in a Black population; elevated regulatory T-cell proportions that were associated with increased overall cancer and lung cancer risk, and elevated memory B-cell proportions that were associated with increased prostate and all cancer risk.
Collapse
Affiliation(s)
- Christopher S. Semancik
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Tufts University, Boston, Massachusetts.
| | - Naisi Zhao
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Tufts University, Boston, Massachusetts.
| | - Devin C. Koestler
- The University of Kansas Cancer Center, Kansas City, Kansas.
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas.
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas.
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas.
| | - Jan Bressler
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas.
| | - Rachel J. Buchsbaum
- Division of Hematology and Oncology, Tufts Medical Center, Boston, Massachusetts.
| | - Karl T. Kelsey
- Department of Epidemiology, Brown University, Providence, Rhode Island.
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island.
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland.
| | - Dominique S. Michaud
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Tufts University, Boston, Massachusetts.
- Department of Epidemiology, Brown University, Providence, Rhode Island.
| |
Collapse
|
46
|
Mani NL, Weinberg SE, Chaudhuri S, Montauti E, Tang A, Iyer R, Fang D. Acidity induces durable enhancement of T reg cell suppressive functions for tumor immune evasion. Mol Immunol 2024; 174:57-68. [PMID: 39213947 PMCID: PMC11681611 DOI: 10.1016/j.molimm.2024.08.004] [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: 03/31/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
The microenvironment within solid tumors often becomes acidic due to various factors associated with abnormal metabolism and cellular activities, including increased lactate production as a result of dysregulated tumor glycolysis. Recently, we have identified multiple tumor microenvironment (TME) factors that potentiate regulatory T (Treg) cell function in evading anti-tumor immunosurveillance. Despite the strong correlation between lactate and acidity, the potential roles of acidity in intratumoral Treg cell adaptation and underlying molecular mechanisms have gone largely unstudied. In this study, we demonstrate that acidity significantly enhances immunosuppressive functions of nTreg cells, but not iTreg cells, without altering the expression of either FoxP3 or the cell surface receptors CD25, CTLA4, or GITR in these cells. Surprisingly, the addition of lactate, often considered a major contributor to increased acidity of the TME, completely abolished the acidity-induced enhancement of nTreg suppressive functions. Consistently, metabolic flux analyses showed elevated basal mitochondrial respiratory capacity and ATP-coupled respiration in acidity-treated nTreg cells without altering glycolytic capacity. Genome-wide transcriptome and metabolomics analyses revealed alterations in multiple metabolic pathways, particularly the one-carbon folate metabolism pathway, with reduced SAM, folate, and glutathione, in nTreg cells exposed to low pH conditions. Addition of a one-carbon metabolic contributor, formate, diminished the acidity-induced enhancement in nTreg cell suppressive functions, but neither SAM nor glutathione could reverse the phenotype. Remarkably, in vitro transient treatment of nTreg cells resulted in sustained enhancement of their functions, as evidenced by more vigorous tumor growth observed in mice adoptively receiving acidity-treated nTreg cells. Further analysis of intratumoral infiltrated T cells confirmed a significant reduction in CD8+ T cell frequency and their granzyme B production. In summary, our study elucidates how acidity-mediated metabolic reprogramming leads to sustained Treg-mediated tumor immune evasion.
Collapse
Affiliation(s)
- Nikita L Mani
- Department of Pathology, Northwestern University, USA; Center for Human Immunobiology, Northwestern University, USA
| | - Samuel E Weinberg
- Department of Pathology, Northwestern University, USA; Center for Human Immunobiology, Northwestern University, USA.
| | | | - Elena Montauti
- Department of Pathology, Northwestern University, USA; Department of Medicine Hematology and Oncology, University of California San Francisco, USA
| | - Amy Tang
- Department of Pathology, Northwestern University, USA; Center for Human Immunobiology, Northwestern University, USA
| | - Radhika Iyer
- Department of Pathology, Northwestern University, USA; Center for Human Immunobiology, Northwestern University, USA
| | - Deyu Fang
- Department of Pathology, Northwestern University, USA; Center for Human Immunobiology, Northwestern University, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern Medicine, USA.
| |
Collapse
|
47
|
Degirmenci M, Diniz G, Kahraman DS, Sahbazlar M, Koral L, Varol U, Uslu R. Investigating the Correlation Between Long-Term Response in Patients with Metastatic HER2+ Breast Cancer and the Activity of Regulatory T Cells: A Retrospective Study. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:645-655. [PMID: 39355199 PMCID: PMC11444060 DOI: 10.2147/bctt.s470570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/12/2024] [Indexed: 10/03/2024]
Abstract
Background Trastuzumab is commonly utilized in the management of metastatic HER2-positive breast cancer. Our main goal was to examine the clinical outcomes and immune markers of patients who received trastuzumab and chemotherapy treatment. Methods Between 1995 and 2012, a total of 98 patients diagnosed with metastatic HER2-positive breast cancer were retrospectively analyzed at Ege University Hospital and Tepecik Training and Research Hospital. The clinicopathological characteristics and clinical outcomes of the patients were assessed, and the associations between response rates, survival and the immune profiles of tumor infiltrating lymphocytes were statistically evaluated. Results The average age of patients at the time of diagnosis was 50.1±10.3 (ranging from 30 to 79) years. The mean follow-up period for all patients was 97.9±53.8 months. Among the patients, complete response was observed in 24.5%, partial response in 61.2%, and stable disease in 8.2% of cases. The average progression-free survival was 50.3±26.9 months (ranging from 1 to 163 months), and the average overall survival was 88.8±59.4 months (ranging from 12 to 272 months). After analyzing all cases, it was found that patients who were younger (p=0.006), exhibited higher CD3-positivity (p=0.041), presented with higher FOXP3-positivity (p=0.025), showed complete or at least partial response to treatment (p=0.008), and experienced a long-term response to trastuzumab (and chemotherapy) treatment had longer survival (p=0.001). Conclusion Patients with HER2-positive breast cancer, who initially respond positively to palliative trastuzumab and chemotherapy treatment, can achieve long-term tumor remission lasting for several years.
Collapse
Affiliation(s)
- Mustafa Degirmenci
- Department of Medical Oncology, Health Sciences University, Izmir, Turkey
| | - Gulden Diniz
- Department of Pathology, Izmir Democracy University, Izmir, Turkey
| | | | - Mustafa Sahbazlar
- Department of Medical Oncology, Celal Bayar University, Manisa, Turkey
| | - Lokman Koral
- Department of Medical Oncology, Canakkale Onsekiz Mart University, Canakkale, Turkey
| | - Umut Varol
- Department of Medical Oncology, Izmir Democracy university, Izmir, Turkey
| | - Ruchan Uslu
- Department of Medical Oncology, izmir Medicana Hospital, Izmir, Turkey
| |
Collapse
|
48
|
Zhang J, Zhan J, Guan Z, Lin X, Li T, Li M, Zhang C, Zhong L. The prognostic value of Th17/Treg cell in cervical cancer: a systematic review and meta-analysis. Front Oncol 2024; 14:1442103. [PMID: 39324000 PMCID: PMC11422014 DOI: 10.3389/fonc.2024.1442103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Background The prognostic significance of Treg and Th17 cells, as well as their ratio (Th17/Treg), in cervical cancer remains a topic of debate. Our study aimed to clarify their association with patient survival and clinical outcomes in cervical cancer through a comprehensive meta-analysis. Materials and methods We conducted a comprehensive search in PubMed, Embase, and Web of Science to identify eligible studies. Studies related to cervical cancer and involving Treg cells or Th17 cells were included. For prognostic analysis, we collected Hazard Ratio values of patient survival. For studies focusing on clinical characteristics, we selected mean and standard deviation values for further analysis. This study was registered at PROSPERO (ID:CRD42024546507). Results Out of the 2949 records initially retrieved, we ultimately included 21 studies in our analysis. High levels of Treg cells were found to be correlated with shorter survival in patients with cervical cancer. Subgroup analysis revealed that the prognostic effect of Treg cells on cervical cancer was not influenced by their source or definition. However, analyses of different survival measures indicated that only Overall Survival showed a correlation with Treg cell levels. Additionally, Treg cells were associated with clinical staging. High-grade Th17 cells were associated with lymphatic metastases and advanced clinical stage. The Th17/Treg ratio was found to be elevated in both cervical intraepithelial neoplasia and cervical cancer patients compared to controls. Discussion Despite limitations such as heterogeneity among selected studies and inadequate subgroup analyses, our study contributes to a deeper understanding of the significance of Treg cells in the onset and progression of cervical cancer. It also provides valuable insights for future research in immunotherapy. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024546507.
Collapse
Affiliation(s)
- Jingwei Zhang
- Department of Gynecology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Jijie Zhan
- Department of Gynecology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Ziting Guan
- Department of Gynecology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Xinmei Lin
- Department of Gynecology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Tian Li
- Department of Gynecology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Miao Li
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Department of Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Changlin Zhang
- Department of Gynecology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Li Zhong
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Department of Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
49
|
Suto A, Minaguchi T, Qi N, Fujieda K, Itagaki H, Tenjimbayashi Y, Shikama A, Tasaka N, Akiyama A, Nakao S, Nakahashi-Oda C, Kobayashi Y, Shibuya A, Satoh T. CD25 +FOXP3 +CD45RA - regulatory T-cell infiltration as a prognostic biomarker for endometrial carcinoma. BMC Cancer 2024; 24:1100. [PMID: 39232704 PMCID: PMC11373268 DOI: 10.1186/s12885-024-12851-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Regulatory T (Treg) cells reportedly play crucial roles in tumor angiogenesis as well as antitumor immunity. In order to explore their therapeutic potential, we investigated the precise prognostic impact of Treg markers in endometrial carcinoma. METHODS We performed multiplexed immunofluorescence and quantitative image analyses of CD25, FOXP3, CTLA4, and CD45RA in tumor specimens from 176 consecutive patients treated at our institution for primary endometrial carcinomas. Bioinformatics analyses were further conducted to corroborate the findings. RESULTS High CD25+, FOXP3+, and CD25+FOXP3+CD45RA- stromal cell counts correlated with better overall survival (OS) (p = 0.00019, 0.028 and 0.0012) and MSI-high (p = 0.015, 0.016 and 0.047). High CD45RA+ stromal cell count was associated with superficial myometrial invasion (p = 0.0038). Bioinformatics survival analysis by Kaplan-Meier plotter showed that high CD25, FOXP3, CTLA4, and CD45RA mRNA expressions correlated with better OS (p = 0.046, 0.00042, 0.000044, and 0.0022). Univariate and multivariate analyses with various clinicopathologic prognostic factors indicated that high CD25+ or CD25+FOXP3+CD45RA- stromal cell count was significant and independent for favorable OS (p = 0.0053 and 0.0015). We subsequently analyzed the correlations between the multiplexed immunofluorescence results and treatment-free interval (TFI) after primary chemotherapy in recurrent cases, finding no significant associations. Further analysis revealed that high ratio of CD25+ : CD8+ cell count or CD25+FOXP3+CD45RA- : CD8+ cell count correlated with longer TFI (p = 0.021 and 0.021). CONCLUSION The current observations suggest that the balance between CD25+ or CD25+FOXP3+CD45RA- cells and CD8+ cells, corresponding to promoting or inhibiting effect on tumor angiogenesis, affect tumor chemosensitivity leading to prognostic significance. CD25+FOXP3+CD45RA- effector Treg tumor infiltration may serve as a useful prognostic biomarker and a potential target for immunotherapeutic manipulation of tumor chemosensitivity by novel management for advanced/recurrent endometrial carcinomas.
Collapse
Affiliation(s)
- Asami Suto
- Department of Obstetrics and Gynecology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575, Ibaraki, Japan
| | - Takeo Minaguchi
- Department of Obstetrics and Gynecology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575, Ibaraki, Japan.
| | - Nan Qi
- Doctoral Program in Obstetrics and Gynecology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kaoru Fujieda
- Department of Obstetrics and Gynecology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575, Ibaraki, Japan
| | - Hiroya Itagaki
- Department of Obstetrics and Gynecology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575, Ibaraki, Japan
| | - Yuri Tenjimbayashi
- Department of Obstetrics and Gynecology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575, Ibaraki, Japan
| | - Ayumi Shikama
- Department of Obstetrics and Gynecology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575, Ibaraki, Japan
| | - Nobutaka Tasaka
- Department of Obstetrics and Gynecology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575, Ibaraki, Japan
| | - Azusa Akiyama
- Department of Obstetrics and Gynecology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575, Ibaraki, Japan
| | - Sari Nakao
- Department of Obstetrics and Gynecology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575, Ibaraki, Japan
| | - Chigusa Nakahashi-Oda
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yusuke Kobayashi
- Department of Obstetrics and Gynecology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575, Ibaraki, Japan
| | - Akira Shibuya
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Toyomi Satoh
- Department of Obstetrics and Gynecology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, 305-8575, Ibaraki, Japan
| |
Collapse
|
50
|
Ortega MA, Jiménez-Álvarez L, Fraile-Martinez O, Garcia-Montero C, De León-Oliva D, Toledo-Lobo MDV, Palacios E, Granado P, Esteban A, Guijarro LG, Pekarek L, Asúnsolo Á, López-González L, Bujan J, García-Honduvilla N, Álvarez-Mon M, Saez MA, Díaz-Pedrero R. Elevated tissue expression of RANKL and RANK is associated with poorer survival rates in pancreatic cancer patients. Histol Histopathol 2024; 39:1133-1140. [PMID: 38230588 DOI: 10.14670/hh-18-700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Pancreatic cancer is a highly lethal malignancy with a growing incidence reported worldwide. Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer, which is often diagnosed at advanced stages, making its prognosis and medical management difficult. The identification of histopathological biomarkers has allowed a more precise stratification of pancreatic cancer patients, providing additional information about their prognosis and offering possible therapeutic targets to be explored. The prognostic value of the receptor activator of nuclear factor-kappa B (RANK) and its ligand (RANKL) has been evaluated in breast and prostate tumors, however, their usefulness has not been assessed in pancreatic cancer. In the present work, we analyzed the relationship between the protein expression of RANK and RANKL with the survival of 41 patients with pancreatic cancer followed for 60 months, by performing immunohistochemistry and Kaplan-Meier curves. Our results demonstrate a direct association of high expression levels of RANK and RANKL with poorer survival of pancreatic cancer patients in comparison to those with low/medium and null expression levels of both markers. Further studies should be conducted to explore the carcinogenic role of both components in this type of tumor, as well as additional promising translational uses.
Collapse
Affiliation(s)
- Miguel A Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Madrid, Spain.
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Cancer Registry and Pathology Department, Principe de Asturias University Hospital, Alcala de Henares, Madrid, Spain
| | - Laura Jiménez-Álvarez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Madrid, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Teaching Hospital, Alcala de Henares, Madrid, Spain
| | - Oscar Fraile-Martinez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Madrid, Spain.
| | - Cielo Garcia-Montero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Madrid, Spain
| | - Diego De León-Oliva
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Madrid, Spain
| | - María Del Val Toledo-Lobo
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Unit of Cell Biology, Department of Biomedicine and Biotechnology, University of Alcala, Alcala de Henares, Madrid, Spain
| | | | | | | | - Luis G Guijarro
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Unit of Biochemistry and Molecular Biology, Department of System Biology (CIBEREHD), University of Alcalá, Alcala de Henares, Spain
| | - Leonel Pekarek
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Oncology Service, Guadalajara University Hospital, Guadalajara, Spain
| | - Ángel Asúnsolo
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Madrid, Spain
- Department of Epidemiology and Biostatistics, Graduate School of Public Health and Health Policy, University of New York, New York, NY, United States
| | - Laura López-González
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Madrid, Spain
| | - Julia Bujan
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Madrid, Spain
| | - Natalio García-Honduvilla
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Madrid, Spain
| | - Melchor Álvarez-Mon
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service and Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, Alcala de Henares, Madrid, Spain
| | - Miguel A Saez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Madrid, Spain
- Pathological Anatomy Service, Central University Hospital of Defence-UAH Madrid, Alcala de Henares, Madrid, Spain
| | - Raúl Díaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Teaching Hospital, Alcala de Henares, Madrid, Spain
- Department of Epidemiology and Biostatistics, Graduate School of Public Health and Health Policy, University of New York, New York, NY, United States
| |
Collapse
|