1
|
Chen X, Qin Y, Gan J, Wei T, Wei X, Xiong Y, Zhang Z, Wei B. Uncovering global research frontiers in deubiquitinating enzymes and immunotherapy: A bibliometric study. Hum Vaccin Immunother 2025; 21:2483558. [PMID: 40130728 PMCID: PMC11938311 DOI: 10.1080/21645515.2025.2483558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/28/2025] [Accepted: 03/15/2025] [Indexed: 03/26/2025] Open
Abstract
Recently, immunotherapy has been a key therapeutic strategy for cancer. Deubiquitinating enzymes (DUBs), which are protein-modifying enzymes, have a crucial role in the pathogenesis of cancer, autoimmune diseases, and inflammation. DUBs influence the tumor immune microenvironment by regulating immune cell functions and key signaling pathways. Thus, the potential applications of DUBs in immunotherapy have piqued the interest of the scientific community. This study performed bibliometric analysis to comprehensively examine the research hotspots and trends in this field, providing theoretical foundations and guidance for future research. Studies associated with DUBs and immunotherapy conducted over a decade (2014 to 2024) were searched and extracted from Web of Science Collection database. The analysis was performed using CiteSpace, VOSviewer, and the Bibliometrix package in R software. Visualizations were generated for countries, institutions, authors, journals, references, and keyword co-occurrences. In total, 321 articles related to DUBs and immunotherapy were retrieved. The number of publications increased markedly since 2020. China had the highest number of publications, while the United States exerted the most influence in this field. Zhang Jinfang was the most influential author in this field. Zhejiang University was the institution with the highest number of publications. Nature was the most cited journal (807 total citations). Keyword analysis revealed that the primary research hotspots were expression, immunotherapy, ubiquitination, degradation, and cancer. This bibliometric analysis revealed the research trends and emerging frontiers in DUBs and immunotherapy, offering novel strategies for the application of DUBs in immunotherapy.
Collapse
Affiliation(s)
- Xia Chen
- Department of Geriatrics, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Yang Qin
- Department of Rheumatology and immunology, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jinfeng Gan
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor‑Targeted Drug Basic Research, Guilin Medical University, Guilin, China
| | - Tangwen Wei
- School of Public Health, Guilin Medical University, Guilin, China
| | - Xinyi Wei
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Yaling Xiong
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Zhichang Zhang
- Department of Computer, School of Intelligent Medicine China Medical University, Shenyang, Liaoning Province, China
| | - Bing Wei
- Department of Geriatrics, Affiliated Hospital of Guilin Medical University, Guilin, China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, China
| |
Collapse
|
2
|
Baldi S, Alnaggar M, AL-Mogahed M, Khalil KAA, Zhan X. Monoclonal antibody immune therapy response instrument for stratification and cost-effective personalized approaches in 3PM-guided pan cancer management. EPMA J 2025; 16:465-503. [PMID: 40438490 PMCID: PMC12106254 DOI: 10.1007/s13167-025-00403-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/06/2025] [Indexed: 06/01/2025]
Abstract
Background Immune checkpoint inhibitors (ICIs), such as anti-PD-1, anti-PD-L1, and anti-CTLA-4 therapies, have revolutionized cancer treatment by harnessing the body's immune system to eliminate cancer cells. Despite their considerable promise, the efficacy of ICIs significantly differs based on tumor types and specific patient conditions, highlighting the necessity for personalized approaches in the framework of predictive preventive personalized medicine (PPPM; 3PM). Main body This review proposes a stratification instrument within the 3PM framework to enhance the therapeutic efficacy of ICIs across Pan-cancer. Predictive approaches need to be utilized to enhance the effectiveness of ICIs. For example, biomarkers such as particular genetic alterations and metabolic pathways provide key information on patient treatment responses. To predict treatment outcomes, uncover resistance mechanisms, and tailor medications, we examine biomarkers including PDL-1 and CTLA4. Focusing on cancers like melanoma, bladder, and renal cell carcinoma, we highlight advances in combination therapies and cellular approaches to overcome resistance. We conducted an analysis of clinical trials and public datasets (TCGA, GEO) to evaluate ICI responses across number of cancer types. Survival analysis employed Kaplan-Meier curves and Cox regression. Pan-cancer analysis shows response rates ranging from 19.8% in bladder cancer to > 39% in melanoma when combination therapy is used, emphasizing the potential of 3PM to improve outcomes. By exploring resistance mechanisms and emerging therapeutic innovations, we propose a cost-effective model for better patient stratification and care. Validation of this model requires standardized biomarkers and prospective trials, promising a shift toward precision oncology. Conclusion Within the 3PM framework, this review addresses the urgent need for cost-effective stratification tools and adaptive combinatorial strategies to optimize outcomes.
Collapse
Affiliation(s)
- Salem Baldi
- Department of Medical Laboratory Diagnostics, School of Medical Technology, Shaoyang University, Shaoyang, 422000 China
- Department of Medical Laboratory Diagnostics, Al-Thawra General Hospital, Al Hudaydah, Yemen
| | - Mohammed Alnaggar
- Department of Oncology, South Hubei Cancer Hospital, Chibi, Xianning, 437000 Hubei China
| | - Maged AL-Mogahed
- Department of Urology, The First Bethune Hospital of Jilin University, Changchun, 130012 China
| | - Khalil A. A. Khalil
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 61922 Bisha, Saudi Arabia
| | - Xianquan Zhan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Jinan Key Laboratory of Cancer Multiomics, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
3
|
Yang X, Xu C, Zeng Y, Wang C, Gao Y, Ding J, Chen S, Pan Y, Zhang X, Mao Z, Shi S. Pyroptosis-Inducing Platinum(IV) Prodrugs via GSDME Pathway for Chemoimmunotherapy and Metastasis Inhibition in Triple-Negative Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e05567. [PMID: 40432601 DOI: 10.1002/advs.202505567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/29/2025] [Indexed: 05/29/2025]
Abstract
Pyroptosis has attracted significant attention for its role in cancer chemotherapy and immunotherapy. However, few drugs have been reported to induce pyroptosis via the Caspase-3/gasdermin E (GSDME) pathway. Herein, three novel PtIV prodrugs, MRP, DRP, and HRP are rationally designed by conjugating DNA methyltransferase (DNMT) inhibitor (RG108) and/or histone deacetylase (HDAC) inhibitor (PhB) to the PtIV center. These prodrugs can be easily reduced to cisplatin (CDDP) due to the high glutathione (GSH) levels in tumors, liberating the coordinated ligands. Released RG108 reactivates the GSDME gene and reduces pyroptosis in low GSDME-expressing tumor cells. Meanwhile, PhB-induced chromatin loosening enhances CDDP-DNA binding, which not only increases Caspase-3 expression, but also upregulates GSDME. HRP demonstrates superior ability to suppress tumor growth and metastasis while reducing systemic toxicity compared with CDDP. By reactivating GSDME and loosening chromatin, HRP effectively boosts tumor cell pyroptosis and exhibits the most pronounced anticancer performance. These findings highlight HRP's potential as a therapeutic agent for triple-negative breast cancer (TNBC) and offer innovative strategies for combining chemotherapy with immunotherapy. To the best of current knowledge, this is the first report of platinum complexes inducing pyroptosis via the Caspase-3/GSDME pathway in low GSDME-expressing tumor cells.
Collapse
Affiliation(s)
- Xinda Yang
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Chuansheng Xu
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Youliang Zeng
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, GBRCE for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Chunhui Wang
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Yan Gao
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Jie Ding
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Sirui Chen
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Yuheng Pan
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Xin Zhang
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Zongwan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, GBRCE for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Shuo Shi
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| |
Collapse
|
4
|
Liu P, Chen Y. Integrative Analysis of EPHX4 as a Novel Prognostic and Diagnostic Biomarker in Lung Adenocarcinoma. Int J Mol Sci 2025; 26:5095. [PMID: 40507905 PMCID: PMC12154108 DOI: 10.3390/ijms26115095] [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: 05/01/2025] [Revised: 05/22/2025] [Accepted: 05/23/2025] [Indexed: 06/16/2025] Open
Abstract
Lung adenocarcinoma (LUAD) remains a leading cause of cancer-related mortality, necessitating the identification of novel biomarkers for improved prognosis and diagnosis. This study investigates the role of epoxide hydrolase 4 (EPHX4), a member of the epoxide hydrolase family, in LUAD. Using data sourced from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases, which were subsequently validated by the Gene Expression Omnibus (GEO), we analyzed levels of EPHX4 expression, mutation, and methylation in tumors versus normal tissues. Our findings revealed a significant upregulation of EPHX4 in LUAD tissues compared to normal lung tissues (p < 0.001), correlating with poorer overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI). Furthermore, EPHX4 exhibited considerable diagnostic potential, as demonstrated by an area under the curve (AUC) of 0.854 in a Receiver Operating Characteristic (ROC) analysis. Notably, EPHX4 expression was associated with immune infiltration, specifically Th2 cells, neutrophils, and macrophages, along with immune checkpoint molecules including PD-L1, PD-L2, and TIM-3. Additionally, EPHX4 was involved in pivotal tumor-associated pathways, particularly cell cycle regulation. In conclusion, an elevated EPHX4 expression is indicative of poorer prognosis in LUAD and may play a role in immune evasion and cell cycle dysregulation, highlighting its potential as a promising biomarker for the diagnosis and prognostic prediction of LUAD.
Collapse
Affiliation(s)
- Pengze Liu
- Department of Pathology, School of Medicine, Jinan University, Guangzhou 510632, China;
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China
| | - Yutong Chen
- Department of Pathology, School of Medicine, Jinan University, Guangzhou 510632, China;
| |
Collapse
|
5
|
Vairaperumal T, Lee PT, Liu PY. Portable Point-of-Care Diagnosis Platforms and Emerging Predictive Biomarkers for Rapid Detection of Severe Dengue Viral Infection. ACS Sens 2025; 10:3302-3316. [PMID: 40165016 DOI: 10.1021/acssensors.5c00263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Dengue virus (DENV) infection is a major global public health problem, particularly in tropical and subtropical regions where Aedes mosquitoes are prevalent. The clinical spectrum of dengue ranges from mild febrile illness to severe conditions such as dengue hemorrhagic fever and dengue shock syndrome. Early prediction of dengue progress is crucial for timely therapeutic medications, which can reduce both morbidity and mortality. Traditional diagnostic methods such as serological tests and polymerase chain reactions are often time-consuming and require sophisticated infrastructure and skilled personnel. To overcome these limitations, the development of point-of-care (POC) diagnosis platforms and novel predictive biomarkers is crucial to providing rapid, real-time diagnostic tools that can be used in low-resource settings and at the patient's bedside. Predictive biomarkers enable the identification of disease risk in the early stages and can reduce hospitalization visits. This review offers a comprehensive overview of portable POC diagnosis platforms and emerging predictive biomarkers for the rapid diagnosis of severe DENV infection. Its provides an overview of its epidemiology, discusses the global burden of DENV, and explores DENV infection with different serotypes, as well as the clinical spectrum and severity of dengue. The key focus is on the latest advancements in POC diagnosis readout methods and portable POC devices for DENV diagnosis, including colorimetric assay, electrochemical method, lateral flow strip, and microfluidic chip platforms. In addition, the review article explores various emerging predictive biomarkers for the rapid detection of DENV, while also highlighting the limitations associated with protein, nucleic acid, and metabolic biomarkers. Finally, we address the current challenges, limitations, and potential future directions of POC diagnosis platforms for the diagnosis of severe DENV infection.
Collapse
Affiliation(s)
- Tharmaraj Vairaperumal
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan, ROC
| | - Po-Tseng Lee
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan, ROC
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan, ROC
| | - Ping-Yen Liu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan, ROC
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan, ROC
| |
Collapse
|
6
|
Harel M, Dahan N, Lahav C, Jacob E, Elon Y, Puzanov I, Kelly RJ, Shaked Y, Leibowitz R, Carbone DP, Gandara DR, Dicker AP. Decoding resistance to immune checkpoint inhibitors in non-small cell lung cancer: a comprehensive analysis of plasma proteomics and therapeutic implications. J Immunother Cancer 2025; 13:e011427. [PMID: 40404205 PMCID: PMC12097049 DOI: 10.1136/jitc-2024-011427] [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: 01/13/2025] [Accepted: 05/05/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have shown substantial benefit for patients with advanced non-small cell lung cancer (NSCLC). However, resistance to ICIs remains a major clinical challenge. Here, we perform a comprehensive bioinformatic analysis of plasma proteomic profiles to explore the underlying biology of treatment resistance in NSCLC. METHODS The analysis was performed on 388 "resistance-associated proteins" (RAPs) that were previously described as pretreatment plasma proteomic predictors within the PROphet computational model designed to predict ICI clinical benefit in NSCLC. Putative tissue origins of the RAPs were explored using publicly available datasets. Enrichment analyses were performed to investigate RAP-related biological processes. Plasma proteomic data from 50 healthy subjects and 272 patients with NSCLC were compared, where patients were classified as displaying clinical benefit (CB; n=76) or no CB (NCB; n=196). Therapeutic agents targeting RAPs were identified in drug and clinical trial databases. RESULTS The RAP set was significantly enriched with proteins associated with lung cancer, liver tissue, cell proliferation, extracellular matrix, invasion, and metastasis. Comparison of RAP expression in healthy subjects and patients with NSCLC revealed five distinct RAP subsets that provide mechanistic insights. The RAP subset displaying a pattern of high expression in the healthy population relative to the NSCLC population included multiple proteins associated with antitumor activities, while the subset displaying a pattern of highest expression in the NCB population included proteins associated with various hallmarks of treatment resistance. Analysis of patient-specific RAP profiles revealed inter-patient diversity of potential resistance mechanisms, suggesting that RAPs may aid in developing personalized therapeutic strategies. Furthermore, examination of drug and clinical trial databases revealed that 17.5% of the RAPs are drug targets, highlighting the RAP set as a valuable resource for drug development. CONCLUSIONS The study provides insight into the underlying biology of ICI resistance in NSCLC and highlights the potential clinical value of RAP profiles for developing personalized therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- The Roswell Park Comprehensive Cancer Center Data Bank and BioRepository, Buffalo, New York, USA
| | - Ronan J Kelly
- Department of Hematology and Oncology, Baylor University Medical Center at Dallas, Dallas, Texas, USA
| | - Yuval Shaked
- Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | | | | | - David R Gandara
- Division of Hematology/Oncology, UC Davis Comprehensive Cancer Center, Sacramento, California, USA
| | - Adam P Dicker
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Fawaz H, Numan H, El Charif MH, Charbel N, El Khoury S, Rizkallah J, El Masri A, Tfayli A, Kreidieh F. Exploring the Emerging Association Between Immune Checkpoint Inhibitors and Thrombosis. J Clin Med 2025; 14:3451. [PMID: 40429445 PMCID: PMC12112099 DOI: 10.3390/jcm14103451] [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: 03/13/2025] [Revised: 04/19/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, but their association with thrombosis presents significant clinical challenges. Patients with cancer already exhibit elevated risks for venous thromboembolism and arterial thrombosis, with treatment modalities like chemotherapy further exacerbating this risk. Emerging evidence suggests that ICIs contribute to thrombotic events through multifactorial mechanisms, including immune dysregulation, T cell activation, endothelial dysfunction, elevated tissue factor expression, and impaired fibrinolysis. Additional risk factors such as obesity, smoking, prior thrombotic events, and combination ICI therapy further increase thrombosis susceptibility. The literature reports varying incidence rates of ICI-associated thrombosis, with some studies indicating comparable risks to chemotherapy, while others highlight higher rates, particularly during the initial treatment phase. Management aligns with standard protocols for cancer-associated thrombosis, using low-molecular-weight heparin or direct oral anticoagulants, though optimal treatment duration and the role of prophylactic anticoagulation require further investigation. This review provides a comprehensive overview of the mechanisms, incidence rates, and clinical management strategies of ICI-associated thrombosis, emphasizing the importance of proactive risk assessment to optimize patient outcomes.
Collapse
Affiliation(s)
- Hassan Fawaz
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (H.F.); (H.N.); (M.H.E.C.); (N.C.); (S.E.K.); (A.E.M.); (A.T.)
| | - Hasan Numan
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (H.F.); (H.N.); (M.H.E.C.); (N.C.); (S.E.K.); (A.E.M.); (A.T.)
| | - Mohamad Hadi El Charif
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (H.F.); (H.N.); (M.H.E.C.); (N.C.); (S.E.K.); (A.E.M.); (A.T.)
| | - Nicole Charbel
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (H.F.); (H.N.); (M.H.E.C.); (N.C.); (S.E.K.); (A.E.M.); (A.T.)
| | - Sacha El Khoury
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (H.F.); (H.N.); (M.H.E.C.); (N.C.); (S.E.K.); (A.E.M.); (A.T.)
| | - Joe Rizkallah
- Department of Diagnostic Radiology, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon;
| | - Amal El Masri
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (H.F.); (H.N.); (M.H.E.C.); (N.C.); (S.E.K.); (A.E.M.); (A.T.)
| | - Arafat Tfayli
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (H.F.); (H.N.); (M.H.E.C.); (N.C.); (S.E.K.); (A.E.M.); (A.T.)
| | - Firas Kreidieh
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (H.F.); (H.N.); (M.H.E.C.); (N.C.); (S.E.K.); (A.E.M.); (A.T.)
| |
Collapse
|
8
|
Wang HL, Li S, Ma CC, Zheng XH, Wu HY, Chang CX, Yang ZH, Wang JW, Pan FM, Zhao B. Characterization and prognostic of CD8 + TIM3 + CD101 + T cells in glioblastoma multiforme. Cell Biosci 2025; 15:60. [PMID: 40375334 DOI: 10.1186/s13578-025-01390-1] [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: 10/03/2024] [Accepted: 04/04/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a pervasive and aggressive malignant brain tumor. In the tumor immune microenvironment, CD8 + TIM3 + CD101 + T cells (CCT cells) play a pivotal role in tumor progression and immune evasion. This study aimed to characterize differentially expressed genes (DEGs) in CCT cells, establish a prognostic model for GBM, and explore clinical implications. METHODS Analysis of data from TCGA, CGGA, and GEO databases included whole-genome expression profiles, clinical data, single nucleotide mutations, and single-cell RNA sequencing. DEGs were identified, and cell trajectories were constructed using Seurat, Monocle 2, and CellChat packages. Functional enrichment analysis was conducted with clusterProfiler, and a prognostic model was developed. Immune infiltration and drug sensitivity analyses were performed to evaluate therapeutic implications. RESULTS Eight distinct cell types were distinguished, encompassing T cells, macrophages, neurons, mural cells, endothelial cells, oligodendrocytes, fibroblasts, and B cells. Comparative analysis revealed differences in these cell types between GBM samples with new adjuvant therapy and initial diagnosis controls. Pseudotime analysis indicated CD8 + TIM3 + CD101-T cells as precursors to CCT cells, unveiling unique gene expression patterns during this transition. The prognostic model, incorporating 22 gene features via LASSO regression, demonstrated strong predictive ability through Receiver Operating Characteristic (ROC) curves. Analysis of 28 immune cell types revealed differences between high-risk and low-risk groups, providing insights into GBM's immune evasion mechanisms. Drug sensitivity analysis proposed potential therapeutic strategies for high-risk patients. CONCLUSION This study offers an in-depth understanding of CCT cells in GBM, introducing a novel prognostic model and suggesting promising therapeutic approaches.
Collapse
Affiliation(s)
- Hong-Liang Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, No 678 Furong Road, Hefei Economic and Technological Development Zone, Hefei, 230000, People's Republic of China
| | - Sai Li
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, No 678 Furong Road, Hefei Economic and Technological Development Zone, Hefei, 230000, People's Republic of China
| | - Chun-Chun Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, No 678 Furong Road, Hefei Economic and Technological Development Zone, Hefei, 230000, People's Republic of China
| | - Xiang-Hu Zheng
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, No 678 Furong Road, Hefei Economic and Technological Development Zone, Hefei, 230000, People's Republic of China
| | - Hao-Yuan Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, No 678 Furong Road, Hefei Economic and Technological Development Zone, Hefei, 230000, People's Republic of China
| | - Chen-Xi Chang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, No 678 Furong Road, Hefei Economic and Technological Development Zone, Hefei, 230000, People's Republic of China
| | - Zhi-Hao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, No 678 Furong Road, Hefei Economic and Technological Development Zone, Hefei, 230000, People's Republic of China
| | - Jia-Wei Wang
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, People's Republic of China.
| | - Fa-Ming Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China.
- The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Bing Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, No 678 Furong Road, Hefei Economic and Technological Development Zone, Hefei, 230000, People's Republic of China.
| |
Collapse
|
9
|
Zhang X, Zhao C, Liu T. Integrative analysis of the expression profile and prognostic values of SENP gene family in hepatocellular carcinoma. Discov Oncol 2025; 16:752. [PMID: 40358846 PMCID: PMC12075750 DOI: 10.1007/s12672-025-02598-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 05/06/2025] [Indexed: 05/15/2025] Open
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) stands as the fourth leading cause of cancer-related deaths worldwide. SUMO-specific peptidases, known as SENPs, emerge as critical players, regulating tumorigenesis and progression of numerous cancer types. Despite this, the specific impact of SENPs in HCC remains unclear. Hence, our study aimed to reveal the immune and prognostic implications of SENPs in HCC. METHODS The gene expression of SENP in various cancers was examined using open-access databases including TCGA, GTEx, and CPTAC. In order to investigate the prognostic potential of the SENP family, Kaplan-Meier analysis was used. To clarify the underlying biological mechanisms, gene set enrichment analysis (GSEA) was carried out. cBioPortal database was used to evaluate genetic mutation profiles. For insight into the relationship between SENP genes and tumor immunity, various algorithms were used. RESULTS Our findings showed that SENP1, SENP2, SENP3, SENP5, SENP6, and SENP7 expression levels were significantly higher in HCC tumor tissues compared to normal tissues. In HCC patients, elevated SENP1 and SENP5 expression has been associated with tumor development and poor outcomes. Our immune infiltration patterns results also showed significant correlations between SENP5 expression and neutrophil (cor = 0.346, p < 0.001), myeloid dendritic cell (cor = 0.491, p < 0.001), macrophage (cor = 0.465, p < 0.001), and memory B cell (cor = 0.336, p < 0.001) infiltration in HCC, whereas SENP1 expression was associated with none of these infiltrations. CONCLUSIONS The prognostic and immunogenetic value of SENP1 and SENP5 in HCC was demonstrated in this study. Therefore, these two genes have the potential to function as separate prognostic biomarkers and offer promise as immunotherapeutic targets in the fight against HCC.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| | - Chenglei Zhao
- Department of Computed Tomography, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Tianyi Liu
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| |
Collapse
|
10
|
Graser CJ, McDonald TO, Catalano PJ, Sonpavde G, Michor F. Early dynamics of clinical and laboratory parameters predict primary refractory disease in patients with metastatic urothelial carcinoma receiving atezolizumab. J Immunother Cancer 2025; 13:e011740. [PMID: 40341029 PMCID: PMC12060874 DOI: 10.1136/jitc-2025-011740] [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: 02/04/2025] [Accepted: 03/31/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND In patients with metastatic urothelial carcinoma (mUC) receiving programmed cell death ligand 1 (PD-L1) inhibitors, it is critically important to identify primary refractory patients very early to enable modification of therapy before clinical progression and decline of performance status. We hypothesized that baseline and early-on-treatment (EOT) parameters may help identify patients likely to have primary refractory disease. METHODS We considered baseline and EOT variables measured up to 5 weeks after initiating therapy in the phase 3 clinical trial IMvigor211, which compared atezolizumab versus chemotherapy, in muC patients who had progressed on platinum-based chemotherapy. We used least absolute shrinkage and selection operator-regularized logistic regression models to predict the risk of primary refractory disease employing clinical and laboratory variables. RESULTS 902 patients were evaluable for analysis. Our baseline model achieves an area under the curve (AUC) of 0.730, 0.717 for the atezolizumab group and 0.696 for the chemotherapy group. The AUC increases to 0.848 overall with EOT parameters, 0.871 for the atezolizumab group and 0.788 for the chemotherapy group. The EOT model suggests that 33.7% of patients receiving atezolizumab may benefit from switching to chemotherapy, reducing their risk of primary refractoriness from 67.1% to 51.5%. CONCLUSIONS Our prediction model employs readily available and routinely measured clinical and laboratory factors, such as urine-specific gravity, presence of liver metastases, and total protein and erythrocyte counts. It robustly identifies patients with early primary refractory disease to atezolizumab before clinical progression and may inform therapeutic decisions. Validation in larger independent cohorts and other treatments is required.
Collapse
Affiliation(s)
- Christopher J Graser
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T H Chan School of Public Health, Cambridge, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Boston, Massachusetts, USA
| | - Thomas O McDonald
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T H Chan School of Public Health, Cambridge, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Boston, Massachusetts, USA
- Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Paul J Catalano
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T H Chan School of Public Health, Cambridge, Massachusetts, USA
| | - Guru Sonpavde
- University of Central Florida, Orlando, Florida, USA
- AdventHealth Cancer Institute, Orlando, Florida, USA
| | - Franziska Michor
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T H Chan School of Public Health, Cambridge, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Boston, Massachusetts, USA
- Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- The Eli and Edythe L Broad Institute, Cambridge, Massachusetts, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
11
|
von Bubnoff D, Schmitt C, Goldinger SM, Schadendorf D, Kähler KC, Hafner C, Kramer N, Fröhlich W, Dummer R, Berking C, Schliep S, Kirchberger MC, Heinzerling L. Prognostic and predictive value of IDO expression in metastatic melanoma treated with Ipilimumab. PLoS One 2025; 20:e0321937. [PMID: 40334245 PMCID: PMC12058187 DOI: 10.1371/journal.pone.0321937] [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: 06/27/2024] [Accepted: 03/11/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND The tumor microenvironment is crucial for prognosis and response to immunotherapy in several tumor entities. METHODS In a multicenter retrospective study, a total of 86 tumor samples from patients with metastatic melanoma were evaluated for baseline expression of indoleamine 2,3-dioxygenase (IDO) and programmed death ligand 1 (PD-L1). Expression patterns of IDO and PD-L1 on tumor cells and antigen-presenting cells (APCs) as determined by immunohistochemical (IHC) staining of paraffin-embedded tissue sections were correlated with response to ipilimumab and overall survival (OS). Statistical analysis was performed using the Spearman correlation, the Mann-Whitney test and Kaplan-Meier estimator. RESULTS IDO expression in tumor cells or APCs was not predictive for treatment response. The median OS was 26 months in IDO-positive and IDO-negative patients, regardless of IDO expression in tumor cells or APCs. A correlation of IHC expression scores of IDO and PD-L1 could not be documented. CONCLUSION The exact role of IDO in creating an immunosuppressive tumor environment and its reversal needs to be further elucidated.
Collapse
Affiliation(s)
| | - Christina Schmitt
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
| | - Simone M. Goldinger
- Department of Dermatology, University Hospital of Zurich, Zürich, Switzerland
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Katharina C. Kähler
- Department of Dermatology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Christian Hafner
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Nora Kramer
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
| | - Waltraud Fröhlich
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, Zürich, Switzerland
| | - Carola Berking
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Stefan Schliep
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Michael C. Kirchberger
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Lucie Heinzerling
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| |
Collapse
|
12
|
Fang CF, Li Y, Yang C, Fang H, Li C. Bioinformatics analysis of intrinsic drivers of immune dysregulation in multiple myeloma to elucidate immune phenotypes and discover prognostic gene signatures. Sci Rep 2025; 15:15662. [PMID: 40325058 PMCID: PMC12053621 DOI: 10.1038/s41598-025-00074-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 04/24/2025] [Indexed: 05/07/2025] Open
Abstract
Multiple myeloma (MM) progression is driven by immune dysregulation within the tumor microenvironment (TME). However, myeloma-intrinsic mechanisms underlying immune dysfunction remain poorly defined, and current immunotherapies show limited efficacy. Using RNA-seq data from 859 MM patients (MMRF-CoMMpass), we integrated xCELL, CIBERSORT, and ESTIMATE algorithms to deconvolute immune-stromal dynamics. Consensus clustering identified immune subtypes, followed by differential gene analysis and LASSO-Cox regression to construct a prognostic model validated in an independent cohort (GSE19784, N = 328). Immune Subtype Classification: Two subgroups emerged: Multiple myeloma-associated immune-related cluster 1 (N = 482): Immune-dysfunctional TME with Th2 cell enrichment, preadipocyte accumulation, and CXCL family suppression, linked to poor survival (P < 0.001). Multiple myeloma-associated immune-related cluster 2 (N = 377): Immune-active TME with cytotoxic CD8 + T/NK cell infiltration and favorable outcomes. Prognostic Gene Signature: Ten immune-related genes (UBE2T, E2F2, EXO1, SH2D2A, DRP2, WNT9A, SHROOM3, TMC8, CDCA7, and GPR132) predicted survival (The One-year AUC = 0.682 and The Over 5-years AUC = 0.714). We define a myeloma-intrinsic immune classification system and a 10-gene prognostic index, offering a framework for risk-stratified immunotherapy. Integration with flow cytometry could optimize precision treatment in MM.
Collapse
Affiliation(s)
- Chuan-Feng Fang
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, 150001, People's Republic of China.
| | - Yan Li
- Department of Anesthesia, The Fourth Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, 150001, People's Republic of China
| | - Chun Yang
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, 150001, People's Republic of China
| | - Hua Fang
- Department of Medical Oncology, Fuxing Hospital of the Capital Medical University, Xicheng District, Beijing, 10001, People's Republic of China
| | - Chen Li
- Department of Bioengineering, the Hebei Agriculture University, Baoding City, Hebei Provence, 071001, People's Republic of China
| |
Collapse
|
13
|
Wei X, Wei W, Liu H, Yi J, Wang M, Xu W, Zhao M, Zhao M, Wang R, Jin S. GIMAP8 could serve as a potential prognostic factor for lung adenocarcinoma and is closely related to immunity. Sci Rep 2025; 15:15465. [PMID: 40316818 PMCID: PMC12048572 DOI: 10.1038/s41598-025-99894-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: 07/21/2024] [Accepted: 04/23/2025] [Indexed: 05/04/2025] Open
Abstract
GTPase IMAP family member 8 (GIMAP8) plays a key role in pathophysiology of several malignancies. The objective of this current research endeavor was to investigate the prognosis value of GIMAP8 in lung adenocarcinoma and examine how it relates to immunity. Expression profiles associated with GIMAP8 and related clinical details were acquired from The Cancer Genome Atlas database, and we conducted survival analysis, enrichment analysis and immune infiltration studies. Additionally, we evaluated the effect of GIMAP8 on radiation resistance of tumor by in vivo and in vitro experiments. Our results showed that lung adenocarcinoma tumor tissues exhibited lower GIMAP8 levels compared to nearby normal tissues. Furthermore, decreased GIMAP8 expression strongly correlated with poorer OS. The expression of GIMAP8 is closely related to the formation of radiation resistance in tumor cells. GSEA identified multiple signaling pathways linked to GIMAP8, including immune-related, chemokine, cell adhesion molecule, and NF-κB signaling pathways. GIMAP8 expression strongly correlated with the expression of immune checkpoint molecules, tumor mutational burden, tumor neoantigen burden, immune cells, and tumor immune microenvironment. GIMAP8 was found to have an inhibitory effect on lung adenocarcinoma and was closely related to the immune response. Moreover, GIMAP8 may also influence radiation resistance in tumors.
Collapse
Affiliation(s)
- Xinfeng Wei
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Wei Wei
- Department of Radiation Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Hongmei Liu
- Nanguan Hospital of Bethune Second Hospital of Jilin University, Changchun, China
| | - Junxuan Yi
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, China
- Cancer Center, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Guangzhou, China
| | - Mingwei Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Weiqiang Xu
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Mingqi Zhao
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Mengdie Zhao
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Rong Wang
- Department of Radiation Oncology, Zhongshan City People's Hospital, No. 2, Sunwen East Road, Zhongshan, Guangdong, China.
| | - Shunzi Jin
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
14
|
Nejat Dehkordi A, Maddahi M, Vafa P, Ebrahimi N, Aref AR. Salivary biomarkers: a promising approach for predicting immunotherapy response in head and neck cancers. Clin Transl Oncol 2025; 27:1887-1920. [PMID: 39377974 DOI: 10.1007/s12094-024-03742-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 09/21/2024] [Indexed: 04/27/2025]
Abstract
Head and neck cancers, including cancers of the mouth, throat, voice box, salivary glands, and nose, are a significant global health issue. Radiotherapy and surgery are commonly used treatments. However, due to treatment resistance and disease recurrence, new approaches such as immunotherapy are being explored. Immune checkpoint inhibitors (ICIs) have shown promise, but patient responses vary, necessitating predictive markers to guide appropriate treatment selection. This study investigates the potential of non-invasive biomarkers found in saliva, oral rinses, and tumor-derived exosomes to predict ICI response in head and neck cancer patients. The tumor microenvironment significantly impacts immunotherapy efficacy. Oral biomarkers can provide valuable information on composition, such as immune cell presence and checkpoint expression. Elevated tumor mutation load is also associated with heightened immunogenicity and ICI responsiveness. Furthermore, the oral microbiota may influence treatment outcomes. Current research aims to identify predictive salivary biomarkers. Initial studies indicate that tumor-derived exosomes and miRNAs present in saliva could identify immunosuppressive pathways and predict ICI response. While tissue-based markers like PD-L1 have limitations, combining multiple oral fluid biomarkers could create a robust panel to guide treatment decisions and advance personalized immunotherapy for head and neck cancer patients.
Collapse
Affiliation(s)
| | - Moein Maddahi
- Faculty of Density, Yeditepe University, Istanbul, Turkey
| | - Parinaz Vafa
- Faculty of Density, Yeditepe University, Istanbul, Turkey
| | - Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Amir Reza Aref
- Mass General Cancer Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
15
|
Aden D, Zaheer S, Sureka N, Trisal M, Chaurasia JK, Zaheer S. Exploring immune checkpoint inhibitors: Focus on PD-1/PD-L1 axis and beyond. Pathol Res Pract 2025; 269:155864. [PMID: 40068282 DOI: 10.1016/j.prp.2025.155864] [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: 08/31/2024] [Revised: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025]
Abstract
Immunotherapy emerges as a promising approach, marked by recent substantial progress in elucidating how the host immune response impacts tumor development and its sensitivity to various treatments. Immune checkpoint inhibitors have revolutionized cancer therapy by unleashing the power of the immune system to recognize and eradicate tumor cells. Among these, inhibitors targeting the programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have garnered significant attention due to their remarkable clinical efficacy across various malignancies. This review delves into the mechanisms of action, clinical applications, and emerging therapeutic strategies surrounding PD-1/PD-L1 blockade. We explore the intricate interactions between PD-1/PD-L1 and other immune checkpoints, shedding light on combinatorial approaches to enhance treatment outcomes and overcome resistance mechanisms. Furthermore, we discuss the expanding landscape of immune checkpoint inhibitors beyond PD-1/PD-L1, including novel targets such as CTLA-4, LAG-3, TIM-3, and TIGIT. Through a comprehensive analysis of preclinical and clinical studies, we highlight the promise and challenges of immune checkpoint blockade in cancer immunotherapy, paving the way for future advancements in the field.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India.
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Monal Trisal
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| |
Collapse
|
16
|
Wang J, Chen Q, Shan Q, Liang T, Forde P, Zheng L. Clinical development of immuno-oncology therapeutics. Cancer Lett 2025; 617:217616. [PMID: 40054657 PMCID: PMC11930610 DOI: 10.1016/j.canlet.2025.217616] [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: 11/05/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
Immuno-oncology (IO) is one of the fastest growing therapeutic areas within oncology. IO agents work indirectly via the host's adaptive and innate immune system to recognize and eradicate tumor cells. Despite checkpoint inhibitors being only introduced to the market since 2011, they have become the second most approved product category. Current Food and Drug Administration (FDA)-approved classes of IO agents include: immune checkpoint inhibitors (ICIs), chimeric antigen receptor T-cell therapy (CAR-T), bi-specific T-cell engager (BiTE) antibody therapy, T-cell receptor (TCR) engineered T cell therapy, tumor-infiltrating lymphocyte (TIL) therapy, cytokine therapy, cancer vaccine therapy, and oncolytic virus therapy. Cancer immunotherapy has made progress in multiple cancer types including melanoma, non-small cell lung cancer (NSCLC), renal cell carcinoma (RCC), and urothelial carcinoma; however, several cancers remain refractory to immunotherapy. Future directions of IO include exploration in the neoadjuvant/perioperative setting, combination strategies, and optimizing patient selection through improved biomarkers.
Collapse
Affiliation(s)
- Jianxin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Qi Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Qiang Shan
- Department of General Surgery, Haining People's Hospital, Haining, 314400, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Patrick Forde
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA; The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Mays Cancer Center at the University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Lei Zheng
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA; The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Mays Cancer Center at the University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
17
|
Proppe L, Jagomast T, Beume S, Köster F, Bräutigam K, Rody A, Perner S, Hemptenmacher F, Ribbat-Idel J, Hanker LC. Prognostic and clinical heterogeneity of PD1 and PD-L1- immunohistochemical scores in endometrial cancers. Arch Gynecol Obstet 2025; 311:1395-1405. [PMID: 39849119 PMCID: PMC12033092 DOI: 10.1007/s00404-024-07862-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/26/2024] [Indexed: 01/25/2025]
Abstract
INTRODUCTION PD1/PD-L1 inhibition (ICi) has recently become a new standard of care for patients with advanced MMR-deficient (MMRd) endometrial cancers. Nevertheless, response to immunotherapy is more complex than the presence of a single biomarker and therefore it remains challenging to predict patients response to ICi beyond MMRd tumors. Elevated PD-L1 expression (CPS ≥ 1) is often used as a prognostic marker as well as a predictive biomarker of response to ICi in different tumor types. In a retrospective, patient derived study, we analyzed PD1- and PD-L1 staining and correlated the results of different scores to clinical data to evaluate the prognostic impact of these scores. MATERIALS AND METHODS Immunohistochemical analysis of the receptor PD1 and the receptor ligand PD-L1 were performed on TMAs of primary paraffin‑embedded tumor samples. All patients were treated for primary endometrial cancer in the Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Campus-Lübeck, Germany between the years 2006-2018. The evaluation and determination of the tumor proportion scoring (TPS), the combined positive score (CPS) and the immune cell scoring (IC) was automatically assessed semi-quantitatively, and results were correlated with clinicopathological characteristics and survival. RESULTS 130 samples were evaluable and 64% showed a positivity (IC > 0) for the receptor PD1 and 56% for the receptor ligand PD-L1. Patients with a PD1 IC Score ≥ 1 showed a significant longer disease-free survival of 140 months (95% confidence interval (CI): 124-158) compared to patients with a lower IC < 1 for PD1 of 89 months (95% confidence interval (CI): 69-110); p = 0.017). Furthermore, the disease-free survival for patients with a CPS ≥ 5 for PD1 was longer (153.7 months (95% confidence interval (CI): 134-173.6) vs. 98.6 months (95% confidence interval (CI): 83-114); p = 0.036). Additionally, a PD1 CPS ≥ 5 showed a better overall survival but the result was not statistically significant. No difference in survival was found between patients with PD-L1 higher or lower than CPS 5. CONCLUSION In this study we pointed out that there are significant clinical differences among several immunohistochemical scoring systems. In our trial, a PD1-positivity with CPS ≥ 5 and IC ≥ 1 were significantly associated to a better disease-free survival while there was no association with TPS. The PD1-IC scoring was associated with MMRd while the TPS scoring was not. Therefore, PD1-IC could be more appropriate for endometrial carcinomas compared to TPS and could also add prognostic information beside the more established PD-L1-staining. Further prospective studies are needed for a validation of these scores in combination with other biomarkers.
Collapse
Affiliation(s)
- L Proppe
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Campus-Lübeck, Lübeck, Germany.
- Department of Gynecology and Gynecologic Oncology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - T Jagomast
- Institute of Pathology, University of Luebeck, Luebeck, Germany
| | - S Beume
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Campus-Lübeck, Lübeck, Germany
| | - F Köster
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Campus-Lübeck, Lübeck, Germany
| | - K Bräutigam
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Campus-Lübeck, Lübeck, Germany
| | - A Rody
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Campus-Lübeck, Lübeck, Germany
| | - S Perner
- Institute of Pathology, University of Luebeck, Luebeck, Germany
| | - F Hemptenmacher
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Campus-Lübeck, Lübeck, Germany
| | - J Ribbat-Idel
- Institute of Pathology, University of Luebeck, Luebeck, Germany
| | - L C Hanker
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Campus-Lübeck, Lübeck, Germany
- Department of Gynecology and Obstetrics, University Medical Center, Muenster, Germany
| |
Collapse
|
18
|
Luo SQ, Dai L, Zhou YJ, He T, Wang FJ, Jin XR, Wang Q. Multiple primary tumors patient developed microsatellite stable gastric cancer after cadonilimab treatment for liver cancer: A case report. World J Clin Oncol 2025; 16:102418. [PMID: 40290697 PMCID: PMC12019263 DOI: 10.5306/wjco.v16.i4.102418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/22/2025] [Accepted: 02/27/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Multiple primary malignant tumors refer to the occurrence of two or more primary malignant tumors in the same organ or multiple organs or tissues at the same time or successively in the same patient, and can occur anywhere in the body. The treatment guidelines for patients with multiple primary malignant tumors are currently controversial. CASE SUMMARY A 51-year-old male patient with liver cancer and portal hypertension received 42 months of co-treatment with atezolizumab and bevacizumab. After that, the disease was rated stable disease. The patient was then diagnosed with gastric cancer. Since the patient was not sensitive to anti-programmed death ligand 1 immunosuppressive agents, a co-treatment with oxaliplatin, tegafur, apatinib, and cadonilimab was selected after multidisciplinary consultation and the patient's agreement. After four cycles of treatment, partial response and stable disease were observed in gastric and liver cancers, respectively. Surgical treatment was performed considering the high-risk factors of gastrointestinal bleeding in patients with gastroesophageal varices. Postoperative pathology showed that the Tumor Regression Grade was 1. Moreover, the genetic testing of postoperative tumor specimens indicated negative programmed death ligand 1 and microsatellite stability. In addition, the latest follow-up indicated an 8 and 40-month progression-free survival in gastric and liver cancer patients, respectively. Currently, the patient is receiving postoperative immunotherapy with cadonilimab. CONCLUSION Cadonilimab not only treats microsatellite stability gastric cancer patients but can also be used for liver cancer treatment.
Collapse
Affiliation(s)
- Si-Qi Luo
- Department of Clinical Medicine, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Li Dai
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Yong-Jin Zhou
- Department of Clinical Medicine, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Tong He
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Fang-Jie Wang
- Department of Emergency, The People’s Hospital of Xishui, Zunyi 564600, Guizhou Province, China
| | - Xiang-Ren Jin
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Qian Wang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| |
Collapse
|
19
|
Gao LL, Gao DN, Yuan HT, Chen WQ, Yang J, Peng JQ. Combining anti-PD-1 antibodies with surufatinib for gastrointestinal neuroendocrine carcinoma: Two cases report and review of literature. World J Clin Oncol 2025; 16:102297. [PMID: 40290678 PMCID: PMC12019270 DOI: 10.5306/wjco.v16.i4.102297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/20/2024] [Accepted: 02/27/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Gastrointestinal neuroendocrine carcinoma (GI NEC) has a low incidence rate and poor prognosis. Most patients already have metastatic disease when they are diagnosed. Platinum chemotherapy is the main means of treating metastatic GI NECs. There is a lack of effective treatment methods after chemotherapy failure. Therefore, Therefore, selecting appropriate posterior-line treatment programs to improve the prognosis of patients is urgently needed. CASE SUMMARY A 64-year-old female was diagnosed with stage IV NEC of the rectum due to abdominal pain and rectal bleeding. After multiline chemotherapy, the condition progressed, and the patient was treated with a combination of camrelizumab and surufatinib. The efficacy evaluation revealed partial remission (PR) and stable conditions, with the expression of the tumor marker neuron-specific enolase (NSE) returning to normal. The adverse reactions were controllable, and the overall condition was good, with weight gain achieved in the past four years. Another 51-year-old female experienced recurrence and metastasis of a duodenal NEC after surgery. After multiline chemotherapy, she received sintilimab combined with surufatinib. The curative effect fluctuated between PR and stability. During treatment, she recovered from immune-related diabetes and later died due to deterioration of her condition. During the treatment, the patient's NSE level returned to normal. CONCLUSION The combination of antiangiogenic targeted drugs and immunotherapy provides a new therapeutic approach for the treatment of metastatic GI-NECs.
Collapse
Affiliation(s)
- Lou-Lu Gao
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Dong-Ni Gao
- Department of Oncology, Shandong Public Health Clinical Center, Jinan 250100, Shandong Province, China
| | - Hong-Tu Yuan
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong Province, China
| | - Wen-Qiang Chen
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Jing Yang
- Department of Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong Province, China
| | - Jie-Qiong Peng
- Department of Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong Province, China
| |
Collapse
|
20
|
Alsaafeen BH, Ali BR, Elkord E. Combinational therapeutic strategies to overcome resistance to immune checkpoint inhibitors. Front Immunol 2025; 16:1546717. [PMID: 40342408 PMCID: PMC12058545 DOI: 10.3389/fimmu.2025.1546717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/31/2025] [Indexed: 05/11/2025] Open
Abstract
Over the past few years, immune checkpoint inhibitors resulted in magnificent and durable successes in treating cancer; however, only a minority of patients respond favorably to the treatment due to a broad-spectrum of tumor-intrinsic and tumor-extrinsic factors. With the recent insights gained into the mechanisms of resistance, combination treatment strategies to overcome the resistance and enhance the therapeutic potential of immune checkpoint inhibitors are emerging and showing promising results in both pre-clinical and clinical settings. This has been derived through multiple interconnected mechanisms such as enhancing tumor immunogenicity, improving neoantigen processing and presentation in addition to augmenting T cell infiltration and cytotoxic potentials. In the clinical settings, several avenues of combination treatments involving immune checkpoint inhibitors were associated with considerable improvement in the therapeutic outcome in terms of patient's survival and tumor growth control. This, in turn, increased the spectrum of cancer patients benefiting from the unprecedented and durable effects of immune checkpoint inhibitors leading to their adoption as a first-line treatment for certain cancers. Moreover, the significance of precision medicine in cancer immunotherapy and the unmet demand to develop more personalized predictive biomarkers and treatment strategies are also highlighted in this review.
Collapse
Affiliation(s)
- Besan H. Alsaafeen
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bassam R. Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Eyad Elkord
- Department of Biosciences and Bioinformatics & Suzhou Municipal Key Lab of Biomedical Sciences and Translational Immunology, School of Science, Xi’an Jiaotong-Liverpool University, Suzhou, China
- College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
- Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, Manchester, United Kingdom
| |
Collapse
|
21
|
Li H, Chang Y, Jin T, Zhang M. Progress of PD-1/PD-L1 immune checkpoint inhibitors in the treatment of triple-negative breast cancer. Cancer Cell Int 2025; 25:139. [PMID: 40211301 PMCID: PMC11987362 DOI: 10.1186/s12935-025-03769-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 03/28/2025] [Indexed: 04/13/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly heterogeneous cancer with substantial recurrence potential. Currently, surgery and chemotherapy are the main treatments for this disease. However, chemotherapy is often limited by several factors, including low bioavailability, significant systemic toxicity, inadequate targeting, and multidrug resistance. Immune checkpoint inhibitors (ICIs), including those targeting programmed death protein-1 (PD-1) and its ligand (PD-L1), have been proven effective in the treatment of various tumours. In particular, in the treatment of TNBC with PD-1/PD-L1 inhibitors, both monotherapy and combination chemotherapy, as well as targeted drugs and other therapeutic strategies, have broad therapeutic prospects. In addition, these inhibitors can participate in the tumour immune microenvironment (TIME) through blocking PD-1/PD-L1 binding, which can improve immune efficacy. This article provides an overview of the use of PD-1/PD-L1 inhibitors in the treatment of TNBC and the progress of multiple therapeutic studies. To increase the survival of TNBC patients, relevant biomarkers for predicting the efficacy of PD-1/PD-L1 inhibitor therapy have been explored to identify new strategies for the treatment of TNBC.
Collapse
Affiliation(s)
- Hongshu Li
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Gong Yuan Road No. 977, Yanji, 133002, P. R. China
- Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, P. R. China
| | - Ying Chang
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Gong Yuan Road No. 977, Yanji, 133002, P. R. China
- Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, P. R. China
| | - Tiefeng Jin
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Gong Yuan Road No. 977, Yanji, 133002, P. R. China.
- Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, P. R. China.
| | - Meihua Zhang
- Department of Ultrasound Medicine, Yanbian University Hospital, Yanji, 133000, P. R. China.
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Gong Yuan Road No. 977, Yanji, 133002, P. R. China.
- Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, P. R. China.
| |
Collapse
|
22
|
Wei J, Zhuang Y, Jiang C, Chen L, Yuan B, Zhao Y, Li H, Mao JH, Hang B, Ye C, Wang L, Wang P. Cohort-based pan-cancer analysis and experimental studies reveal ISG15 gene as a novel biomarker for prognosis and immunotherapy efficacy prediction. Cancer Immunol Immunother 2025; 74:168. [PMID: 40208307 PMCID: PMC11985735 DOI: 10.1007/s00262-025-04026-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025]
Abstract
ISG15, an interferon-stimulated ubiquitin-like protein, plays a multifaceted role in tumorigenesis and immune regulation. This study comprehensively evaluates ISG15 as a prognostic biomarker and predictor of immunotherapy response through pan-cancer bioinformatics analysis and experimental validation. By integrating multiomics data from TCGA, GEO, and clinical cohorts, we found that ISG15 is significantly overexpressed in multiple cancers and generally correlates with poor prognosis. Elevated ISG15 expression is associated with increased immune checkpoint gene expression, particularly PD-L1, and immune infiltration, notably M2-like tumor-associated macrophages. Immunohistochemistry and multiplexed immunofluorescence confirmed a strong positive correlation between ISG15, PD-L1, and M2-TAM infiltration in lung and gastric cancer samples. Functional analysis at the single-cell level revealed significant associations between ISG15 and tumor proliferation, angiogenesis, and immune suppression. Immunotherapy cohort analysis demonstrated that tumors with high ISG15 expression responded favorably to PD-L1 inhibitors but exhibited resistance to CTLA-4 blockade, findings further validated in lung cancer patients receiving anti-PD-1 therapy. These results suggest that ISG15 is a promising biomarker for prognosis and immunotherapy response prediction across cancers. Its integration into clinical decision-making may enhance personalized treatment strategies, improve immunotherapy outcomes, and provide new insights into the tumor immune microenvironment, cancer progression, and potential therapeutic targets for future drug development.
Collapse
Affiliation(s)
- Jingjing Wei
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yingjia Zhuang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Ultrasound, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Chengfei Jiang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Lingyan Chen
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Binbin Yuan
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Yue Zhao
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Happi Li
- Saratoga High School, 20300 Herriman Ave, Saratoga, CA, USA
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Bo Hang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| | - Chunping Ye
- Department of Obstetrics and Gynecology, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Lei Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| | - Pin Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China.
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
23
|
Shi K, Zhao Y, Ye H, Zhu X, Chen Z. Targeting DKK3 to remodel tumor immune microenvironment and enhance cancer immunotherapy. BMC Cancer 2025; 25:645. [PMID: 40205566 PMCID: PMC11984186 DOI: 10.1186/s12885-025-14075-2] [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: 09/13/2024] [Accepted: 04/02/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer immunotherapy such as immune checkpoint blockade (ICB) therapy has made important breakthroughs in cancer treatment, however, currently only parts of cancer patients benefit from ICB therapy. The suppressive tumor immune microenvironment (TIME) impedes the treatment response of immunotherapy, indicating the necessity to explore new treatment targets. Here, we reported a new potential immunotherapeutic target, Dickkopf-3 (DKK3), for cancer treatment. DKK3 expression is up-regulated in the tumors from multiple cancer types, and high DKK3 expression is associated with worse survival outcome across different cancers. We observed that DKK3 directly inhibits the activation of CD8+ T cells and the Th1 differentiation of CD4+ T cells ex vivo. Also, by establishing four different mouse cancer models, we found that DKK3 blockade triggers effective anti-tumor effects and improve the survival of tumor-bearing mice in vivo. DKK3 blockade also remodels the suppressive TIME of different cancer types, including the increased infiltration of CD8+ T cells, IFN-γ+CD8+ T cells, Th1 cells, and decreased infiltration of M2 macrophages and MDSCs in the TIME. Moreover, we found that combined blockade of DKK3 and PD-1 induces synergistic tumor-control effect in our mouse cancer model. Therefore, our study reveals the impact of DKK3 in the TIME and cancer progression, which suggests that DKK3 is a novel and promising immunotherapeutic target for enhanced cancer immunotherapy.
Collapse
Affiliation(s)
- Kai Shi
- Department of Thoracic Surgery, Huai'an Hospital of Huai'an City & Huai'an Cancer Hospital & The Affiliated Huai'an Hospital of Jiangsu College of Nursing, Huai'an, Jiangsu Province, China
| | - Yan Zhao
- Department of Thoracic Surgery, Huai'an Hospital of Huai'an City & Huai'an Cancer Hospital & The Affiliated Huai'an Hospital of Jiangsu College of Nursing, Huai'an, Jiangsu Province, China
| | - Hao Ye
- Department of Thoracic Surgery, Huai'an Hospital of Huai'an City & Huai'an Cancer Hospital & The Affiliated Huai'an Hospital of Jiangsu College of Nursing, Huai'an, Jiangsu Province, China
| | - Xiaoming Zhu
- Department of Thoracic Surgery, Huai'an Hospital of Huai'an City & Huai'an Cancer Hospital & The Affiliated Huai'an Hospital of Jiangsu College of Nursing, Huai'an, Jiangsu Province, China
| | - Zhenghai Chen
- Department of Thoracic Surgery, Huai'an Hospital of Huai'an City & Huai'an Cancer Hospital & The Affiliated Huai'an Hospital of Jiangsu College of Nursing, Huai'an, Jiangsu Province, China.
| |
Collapse
|
24
|
Peng M, Wang M, Yang X, Wang Y, Xie L, An W, Ge F, Yang C, Wang K. Prediction of PD-L1 expression in NSCLC patients using PET/CT radiomics and prognostic modelling for immunotherapy in PD-L1-positive NSCLC patients. Clin Radiol 2025; 86:106915. [PMID: 40375402 DOI: 10.1016/j.crad.2025.106915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 05/18/2025]
Abstract
AIM To develop a positron emission tomography/computed tomography (PET/CT)-based radiomics model for predicting programmed cell death ligand 1 (PD-L1) expression in non-small cell lung cancer (NSCLC) patients and estimating progression-free survival (PFS) and overall survival (OS) in PD-L1-positive patients undergoing first-line immunotherapy. MATERIALS AND METHODS We retrospectively analysed 143 NSCLC patients who underwent pretreatment 18F-fluorodeoxyglucose (18F-FDG) PET/CT scans, of whom 86 were PD-L1-positive. Clinical data collected included gender, age, smoking history, Tumor-Node-Metastases (TNM) staging system, pathologic types, laboratory parameters, and PET metabolic parameters. Four machine learning algorithms-Bayes, logistic, random forest, and Supportsupport vector machine (SVM)-were used to build models. The predictive performance was validated using receiver operating characteristic (ROC) curves. Univariate and multivariate Cox analyses identified independent predictors of OS and PFS in PD-L1-positive expression patients undergoing immunotherapy, and a nomogram was created to predict OS. RESULTS A total of 20 models were built for predicting PD-L1 expression. The clinical combined PET/CT radiomics model based on the SVM algorithm performed best (area under curve for training and test sets: 0.914 and 0.877, respectively). The Cox analyses showed that smoking history independently predicted PFS. SUVmean, monocyte percentage and white blood cell count were independent predictors of OS, and the nomogram was created to predict 1-year, 2-year, and 3-year OS based on these three factors. CONCLUSION We developed PET/CT-based machine learning models to help predict PD-L1 expression in NSCLC patients and identified independent predictors of PFS and OS in PD-L1-positive patients receiving immunotherapy, thereby aiding precision treatment.
Collapse
Affiliation(s)
- M Peng
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang, China.
| | - M Wang
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang, China.
| | - X Yang
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang, China.
| | - Y Wang
- Scientific Research Center Department, Beijing General Electric Company, No.2 Yongchang North Road, Yizhuang Economic and Technological Development Zone, Daxing District, Beijing, 102200, China.
| | - L Xie
- MR Research China, GE Healthcare, Beijing, 100176, China.
| | - W An
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - F Ge
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang, China.
| | - C Yang
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang, China.
| | - K Wang
- PET-CT/MRI Department, Harbin Medical University Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang, China.
| |
Collapse
|
25
|
Su L, Xu R, Ren Y, Zhao S, Song L, Meng C, Liu W, Zhou X, Du Z. 5-Methylcytosine methylation predicts cervical cancer prognosis, shaping immune cell infiltration. J Int Med Res 2025; 53:3000605251328301. [PMID: 40219803 PMCID: PMC12033582 DOI: 10.1177/03000605251328301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 02/19/2025] [Indexed: 04/14/2025] Open
Abstract
BackgroundEpigenetics, encompassing DNA and RNA modifications, has emerged as a prominent area of research in the post-genomic era. Numerous studies have elucidated the impact of epigenetics on tumor regulation. However, the methylation patterns of 5-methylcytosine in cervical cancer as well as its role in the tumor microenvironment and immunotherapy remain poorly understood.MethodsUtilizing a comprehensive dataset encompassing samples from 306 patients with cervical cancer from The Cancer Genome Atlas and Gene Expression Omnibus repositories, we conducted an in-depth analysis to evaluate the potential association between the modification patterns of 5-methylcytosine and the infiltration of cells within the tumor microenvironment, taking into account 11 regulators of 5-methylcytosine modification. Subsequently, we employed stepwise regression and Least Absolute Shrinkage and Selection Operator Cox regression to quantify 5-methylcytosine modification patterns in patients with cervical squamous cell carcinoma and endocervical adenocarcinoma, yielding the 5-methylcytosine score. Our study explored the link between the 5-methylcytosine score and clinical characteristics as well as prognostic outcomes in patients with cervical squamous cell carcinoma and endocervical adenocarcinoma.ResultsA comprehensive analysis of 306 patients with cervical cancer revealed two distinct 5-methylcytosine modification patterns, henceforth labeled as 5-methylcytosine clusters A and B. These clusters exhibited distinct immunological profiles and biological attributes, with 5-methylcytosine cluster A exhibiting a higher degree of immune cell infiltration. Utilizing univariate Cox regression analysis, we identified 367 genes regulated by 5-methylcytosine that were significantly correlated with patient prognosis. This analysis further stratified the samples into three distinct genomic subtypes. Survival analyses indicated that patients belonging to gene cluster C exhibited more favorable survival outcomes than those belonging to gene clusters A and B. Intriguingly, most 5-methylcytosine regulatory factors had higher expression levels in gene cluster B than in gene cluster A. Gene set enrichment analysis of a single sample revealed elevated immune cell infiltration within gene cluster B, indicating a stronger immune response in this cluster. The 5-methylcytosine score feature was utilized to determine the 5-methylcytosine modification pattern in cervical cancer, revealing that patients with low 5-methylcytosine scores exhibited better survival rates, whereas those with high scores had increased mutation frequencies and better treatment responses.ConclusionsThis research underscores the key role of 5-methylcytosine modification patterns in cervical cancer. Analysis of these patterns will deepen our understanding of the cervical cancer tumor microenvironment, paving the way for the development of more refined and effective immunotherapy strategies.
Collapse
Affiliation(s)
- Luyang Su
- Physical Examination Center, Hebei General Hospital, Shijiazhuang, Hebei-China
| | - Ren Xu
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, Hebei-China
| | - Yanan Ren
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, Hebei-China
| | - Shixia Zhao
- Physical Examination Center, Hebei General Hospital, Shijiazhuang, Hebei-China
| | - Liyun Song
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, Hebei-China
| | - Cuiqiao Meng
- Physical Examination Center, Hebei General Hospital, Shijiazhuang, Hebei-China
| | - Weilan Liu
- Physical Examination Center, Hebei General Hospital, Shijiazhuang, Hebei-China
| | - Xuan Zhou
- Department of Obstetrics and Gynecology, Affiliated Hospital of Chengde Medical University, Chengde, Hebei-China
| | - Zeqing Du
- Department of Obstetrics and Gynecology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei-China
| |
Collapse
|
26
|
Wu YT, Gao M, Cheng KY, Li L, Wang BQ, He YN, Zhang Y, Liu XY, Du RL, Li GQ, Liang YX, Zhang JF, Zhang XD, Liu Y. VRK2 promotes colorectal cancer growth and impedes immunotherapy and 5-FU treatment efficacy. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167729. [PMID: 39978443 DOI: 10.1016/j.bbadis.2025.167729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/31/2024] [Accepted: 02/13/2025] [Indexed: 02/22/2025]
Abstract
Vaccinia-Related Kinase 2 (VRK2), a member of the vaccinia virus-related protein kinase family, is crucial in regulating apoptosis and tumor cell growth signaling pathways. Despite its established roles in various cancers, investigations into its functions in colorectal cancer have been relatively limited. Utilizing The Cancer Genome Atlas and Genotype-Tissue Expression databases, this study assesses VRK2 expression across 33 cancer types, highlighting significant upregulation and diagnostic relevance, particularly in colorectal cancer, where it marks poor prognosis. VRK2's influence extends across multiple cancer-related signaling pathways, with focused experiments confirming its vital role in the E2F signaling pathway through transcriptomic sequencing and dual-luciferase reporter assays. Deletion of VRK2 markedly inhibits proliferation, cell cycle progression, migration, and tumorigenesis in colorectal cancer cells, whereas overexpression enhances these oncogenic traits. Additionally, VRK2 expression correlates with genomic instability and the tumor microenvironment, influencing antitumor immunity and response to immunotherapy. Importantly, our analysis reveals that VRK2 modulates the chemosensitivity of tumor cells, specifically enhancing resistance to the chemotherapeutic agent 5-FU. These findings underscore VRK2's multifaceted role in promoting colorectal cancer development and suggest its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yu-Tong Wu
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Meng Gao
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Kun-Yang Cheng
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Le Li
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Bai-Qi Wang
- Department of Radiation Oncology, The Second Affiliated Hospital University of South China Clinical Research Center, For Prevention and Treatment of Breast & Thyroid Disease In Hunan Province, Hengyang, Hunan, China
| | - Ya-Nan He
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China
| | - Yue Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China
| | - Xue-Yi Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China
| | - Run-Lei Du
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China
| | - Guo-Qing Li
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yue-Xiu Liang
- Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions & Department of Gynecology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Jian-Feng Zhang
- Xuancheng Institutes of Food and Drug Control, Xuancheng, China
| | - Xiao-Dong Zhang
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Yi Liu
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|
27
|
Mitra S, Jang HJ, Kuncheria A, Kang SW, Choi JM, Shim JS, Lee C, Ranchod P, Jindra P, Ramineni M, Patel M, Ripley RT, Groth SS, Blackmon SH, Burt BM, Lee HS. Soluble mesothelin-related peptide as a prognosticator in pleural mesothelioma patients receiving checkpoint immunotherapy. J Thorac Cardiovasc Surg 2025; 169:1082-1095.e4. [PMID: 39395787 PMCID: PMC11949723 DOI: 10.1016/j.jtcvs.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/27/2024] [Accepted: 10/02/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Immune checkpoint therapy (ICT) has significantly impacted the treatment of malignant pleural mesothelioma (MPM). Despite some promising results from combination therapies, nearly half of MPM patients do not benefit, underscoring the urgent need for reliable predictive biomarkers. This study assesses the prognostic value of serum soluble mesothelin-related peptide (SMRP) and PD-L1 levels in MPM patients receiving ICT. METHODS We conducted a retrospective analysis of 125 MPM patients treated with ICT by measuring pre-ICT serum levels of SMRP and PD-L1. We also examined the correlation of these serum levels with tumor mRNA expressions of mesothelin and PD-L1. Both univariable and multivariable Cox regression analyses were used to determine independent prognosticators for overall survival (OS). A prospective ICT clinical trial and our historical cohort were included for validation. RESULTS Seventy-seven patients (62%) were treated with either anti-PD-(L)1 monotherapy, and the remaining 38% received combination ICT. Higher pre-ICT SMRP levels were observed in epithelioid MPM compared to nonepithelioid MPM. Serum PD-L1 levels did not differ significantly between the different histologic groups. Univariable analysis identified durable clinical benefit, development of immune-related adverse events, and SMRP levels as significantly associated with OS. Multivariable analysis confirmed SMRP as an independent prognostic factor, with lower levels (≤1.35 nmol/L) correlating with improved OS. The association of high SMRP with worse prognosis was validated in the prospective ICT clinical trial cohort and not in our historical cohort treated without ICT. CONCLUSIONS SMRP is a promising serum biomarker for predicting survival in MPM patients treated with ICT and warrants prospective investigation.
Collapse
Affiliation(s)
- Sonali Mitra
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Hee-Jin Jang
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Allen Kuncheria
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Sung Wook Kang
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Jong Min Choi
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Ji Seon Shim
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Claire Lee
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Priyanka Ranchod
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Peter Jindra
- Immune Evaluation Laboratory, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Maheshwari Ramineni
- Department of Pathology, Baylor College of Medicine, Houston, Tex; Dan L Ducan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Tex
| | - Meera Patel
- Dan L Ducan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Tex; Division of Hemato-Oncology, Department of Medicine, Baylor College of Medicine, Houston, Tex
| | - R Taylor Ripley
- Dan L Ducan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Tex; David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Shawn S Groth
- Dan L Ducan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Tex; David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Shanda H Blackmon
- Dan L Ducan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Tex; David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Bryan M Burt
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex; Division of Thoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, Calif.
| | - Hyun-Sung Lee
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex; Division of Hemato-Oncology, Department of Medicine, Baylor College of Medicine, Houston, Tex; David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex.
| |
Collapse
|
28
|
Omero F, Speranza D, Murdaca G, Cavaleri M, Marafioti M, Cianci V, Berretta M, Casciaro M, Gangemi S, Santarpia M. The Role of Eosinophils, Eosinophil-Related Cytokines and AI in Predicting Immunotherapy Efficacy in NSCLC Cancer. Biomolecules 2025; 15:491. [PMID: 40305195 PMCID: PMC12024677 DOI: 10.3390/biom15040491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
Immunotherapy and chemoimmunotherapy are standard treatments for non-oncogene-addicted advanced non-small cell lung cancer (NSCLC). Currently, a limited number of biomarkers, including programmed death-ligand 1 (PD-L1) expression, microsatellite instability (MSI), and tumor mutational burden (TMB), are used in clinical practice to predict benefits from immune checkpoint inhibitors (ICIs). It is therefore necessary to search for novel biomarkers that could be helpful to identify patients who respond to immunotherapy. In this context, research efforts are focusing on different cells and mechanisms involved in anti-tumor immune response. Herein, we provide un updated literature review on the role of eosinophils in cancer development and immune response, and the functions of some cytokines, including IL-31 and IL-33, in eosinophil activation. We discuss available data demonstrating a correlation between eosinophils and clinical outcomes of ICIs in lung cancer. In this context, we underscore the role of absolute eosinophil count (AEC) and tumor-associated tissue eosinophilia (TATE) as promising biomarkers able to predict the efficacy and toxicities from immunotherapy. The role of eosinophils and cytokines in NSCLC, treated with ICIs, is not yet fully understood, and further research may be crucial to determine their role as biomarkers of response. Artificial intelligence, through the analysis of big data, could be exploited in the future to elucidate the role of eosinophils and cytokines in lung cancer.
Collapse
Affiliation(s)
- Fausto Omero
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy; (F.O.); (D.S.); (M.C.); (M.M.); (M.S.)
| | - Desirèe Speranza
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy; (F.O.); (D.S.); (M.C.); (M.M.); (M.S.)
| | - Giuseppe Murdaca
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
| | - Mariacarmela Cavaleri
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy; (F.O.); (D.S.); (M.C.); (M.M.); (M.S.)
| | - Mariapia Marafioti
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy; (F.O.); (D.S.); (M.C.); (M.M.); (M.S.)
| | - Vincenzo Cianci
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Section of Legal Medicine, University of Messina, Via Consolare Valeria, 1, 98125 Messina, Italy;
| | - Massimiliano Berretta
- Medical Oncology Unit, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| | - Marco Casciaro
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (M.C.); (S.G.)
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (M.C.); (S.G.)
| | - Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy; (F.O.); (D.S.); (M.C.); (M.M.); (M.S.)
| |
Collapse
|
29
|
Nueraihemaiti N, Dilimulati D, Baishan A, Hailati S, Maihemuti N, Aikebaier A, Paerhati Y, Zhou W. Shared Genomic Features Between Lung Adenocarcinoma and Type 2 Diabetes: A Bioinformatics Study. BIOLOGY 2025; 14:331. [PMID: 40282196 PMCID: PMC12025072 DOI: 10.3390/biology14040331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a common histopathological variant of non-small cell lung cancer. Individuals with type 2 diabetes (T2DM) face an elevated risk of developing LUAD. We examined the common genomic characteristics between LUAD and T2DM through bioinformatics analysis. METHODS We acquired the GSE40791, GSE25724, GSE10072, and GSE71416 datasets. Differentially expressed genes (DEGs) were identified through R software, particularly its version 4.1.3 and analyzed via gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Subsequently, we analyzed the relationship between immune cell infiltration and DEGs. we constructed a protein-protein interaction network using STRING and visualized it with Cytoscape. Moreover, gene modules were identified utilizing the MCODE plugin, and hub genes were selected through the CytoHubba plugin. Additionally, we evaluated the predictive significance of hub genes using receiver operating characteristic curves and identified the final central hub genes. Finally, we forecasted the regulatory networks of miRNA and transcription factors for the central hub genes. RESULTS A total of 748 DEGs were identified. Analysis of immune infiltration showed a notable accumulation of effector-memory CD8 T cells, T follicular helper cells, type 1 T helper cells, activated B cells, natural killer cells, macrophages, and neutrophils in both LUAD and T2DM. Moreover, these DEGs were predominantly enriched in immune-related pathways, including the positive regulation of I-κB kinase/NF-κB signaling, positive regulation of immunoglobulin production, cellular response to interleukin-7, and cellular response to interleukin-4. The TGF-β signaling pathway was significantly important among them. Additionally, seven hub genes were identified, including ATR, RFC4, MCM2, NUP155, NUP107, NUP85, and NUP37. Among them, ATR, RFC4, and MCM2 were identified as pivotal hub genes. Additionally, hsa-mir147a, hsa-mir16-5p, and hsa-mir-1-3p were associated with LUAD and T2DM. SP1 (specific protein 1) and KDM5A (lysine-specific demethylase 5A) regulated MCM2, ATR, and RFC4. CONCLUSIONS Our study elucidates the common mechanisms of immune response, TGF-β signaling pathway, and natural killer cells in LUAD and T2DM, and identifies ATR, RFC4, and MCM2 as key potential biomarkers and therapeutic targets for the comorbidity of these two conditions.
Collapse
Affiliation(s)
- Nuerbiye Nueraihemaiti
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Dilihuma Dilimulati
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Alhar Baishan
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Sendaer Hailati
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Nulibiya Maihemuti
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Alifeiye Aikebaier
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Yipaerguli Paerhati
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Wenting Zhou
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| |
Collapse
|
30
|
Gao Y, Huo Y, Wang L, Ruan J, Chen L, Li H, Hong G. Relative expression orderings based prediction of treatment response to Anti-PD-1 immunotherapy in advanced melanoma. Sci Rep 2025; 15:10235. [PMID: 40133499 PMCID: PMC11937249 DOI: 10.1038/s41598-025-94931-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Programmed cell death protein 1 (PD-1) plays a critical role in immune tolerance and evasion within the tumor microenvironment, and anti-PD-1 immunotherapy has shown efficacy in treating advanced melanoma. However, response rates vary significantly among patients, necessitating the identification of reliable biomarkers to predict treatment efficacy. Based on within-sample relative expression orderings, we analyzed RNA sequencing data from melanoma patients to construct a predictive model comprising gene pairs associated with treatment response. The model's performance was validated across multiple independent datasets and assessed for correlations with immune infiltration and survival outcomes. The constructed 15-pair model achieved a prediction accuracy of 100% in training datasets and 89.47% in validation sets. Validation in melanoma patients lacking treatment response data revealed significant differences between predicted responders and non-responders across datasets, with the model being an independent prognostic factor. Increased immune cell infiltration was observed in responders, correlating with higher expression levels of key immune checkpoint genes. The relative expression orderings-based model shows promise as a tool for predicting responses to anti-PD-1 therapy in melanoma patients, supporting personalized treatment strategies.
Collapse
Affiliation(s)
- Yaru Gao
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, 341000, China
| | - Yue Huo
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, 341000, China
| | - Lingli Wang
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou, 341000, China
| | - Jiayi Ruan
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou, 341000, China
| | - Lanzhen Chen
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou, 341000, China
| | - Hongdong Li
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou, 341000, China.
| | - Guini Hong
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
31
|
Nome RV, Flatebø Ø, Bøe SL, Klaasen RA, Aamdal E, Normann M, Bolstad N, Warren DJ. A simple automated assay format for measuring multiple immune checkpoint inhibitors. J Pharm Biomed Anal 2025; 255:116657. [PMID: 39756153 DOI: 10.1016/j.jpba.2024.116657] [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: 08/27/2024] [Revised: 12/19/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have improved survival rates in oncology, but there is a rising concern for immune-related adverse health outcomes. Monitoring drug serum concentration would enable tailored dosing, however this strategy has not yet been evaluated for ICIs. Fully automated analyte capture assays with time-resolved fluorometry using protein A as tracer, were developed for three different ICIs; the cytotoxic T lymphocyte Antigen-4 (CTLA4) inhibitor ipilimumab (Yervoy; Bristol-Myers Squibb) and the Programmed Death-1 (PD-1) inhibitors nivolumab (Opdivo; Bristol-Myers Squibb) and pembrolizumab (Keytruda; Merck). Drug trough levels were measured in serum samples from ICI-treated patients. Measuring ranges were 1-100 mg/L for all three drugs. Automation allowed for 110 samples to be analyzed in < 4 h. Median drug trough-levels after 5-7 weeks of treatment were 20 (range <1.0-45) mg/L for ipilimumab (n = 113), 60 (range 14-75) mg/L) for nivolumab (n = 21) and 19 (range 7.4-39) mg/L for pembrolizumab (n = 20). Routine drug concentration monitoring for ipilimumab, nivolumab and pembrolizumab is feasible using fully automated analyte capture assays constructed with commercially available reagents. The large drug serum concentration ranges in samples from real-world patients, should be further investigated to assess the clinical relevance of ICI concentration monitoring.
Collapse
Affiliation(s)
- Ragnhild V Nome
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway.
| | | | - Sigurd Leinæs Bøe
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Rolf Anton Klaasen
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Elin Aamdal
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | | | - Nils Bolstad
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - David John Warren
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
32
|
Jin L, Yang Z, Tang W, Yu P, Chen R, Xu Y, Zhang J. The evolving landscape of genetic biomarkers for immunotherapy in primary and metastatic breast cancer. Front Oncol 2025; 15:1522262. [PMID: 40182039 PMCID: PMC11966456 DOI: 10.3389/fonc.2025.1522262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/30/2025] [Indexed: 04/05/2025] Open
Abstract
Background Major advances have been achieved in the characterization of primary breast cancer genomic profiles. Limited information is available on the genomic profile of tumors originating from different metastatic locations in recurrent/metastatic (R/M) breast cancer, especially in Asian patients. This study aims to decipher the mutational profiles of primary and R/M breast cancer in Chinese patients using next-generation sequencing. Methods A total of 563 breast cancer patients were enrolled, and 590 tumor tissues and matched peripheral blood samples were collected and subjected to targeted sequencing with a panel of 1,021 cancer-related genes. The mutation spectrum, DNA damage response (DDR) genes, commonly altered signal pathways, and immunotherapy-related markers were compared between primary and R/M breast cancer. The molecular differences between our cohort and the Memorial Sloan Kettering Cancer Center (MSKCC) dataset were also explored. Results A total of 361 samples from primary and 229 samples from R/M breast cancer were analyzed. BRCA2, ATRX, and ATM were more frequently observed in R/M lesions among the 36 DDR genes. An ESR1 mutation and PD-L1 and PD-L2 amplification were enriched in R/M breast cancer (all p<0.05). Compared with the MSKCC dataset, we recruited more patients diagnosed at age 50 or younger and more patients with triple-negative breast cancer (TNBC) subtypes. The TNBC patients in our dataset had a higher percentage of PD-L1 amplification in metastasis tumors (p<0.05). Conclusions This study revealed the distinctive mutational features of primary and R/M tumors in Chinese breast cancer patients, which are different from those from Western countries. The enrichment of PD-L1 amplification in metastatic TNBC indicates the necessity to re-biopsy metastatic tumors for immunotherapy.
Collapse
Affiliation(s)
- Liang Jin
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zijian Yang
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Wei Tang
- Department of Breast Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pengli Yu
- Medical Department, Geneplus-Beijing, Beijing, China
| | - Rongrong Chen
- Medical Department, Geneplus-Beijing, Beijing, China
| | - Yan Xu
- Department of Breast and Thyroid Surgery, Daping Hospital, Army Military Medical University, Chongqing, China
| | - Jun Zhang
- Department of Thyroid and Breast Surgery, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| |
Collapse
|
33
|
Hao Y, Wang X, Ni Z, Ma Y, Wang J, Su W. Analysis of ferritinophagy-related genes associated with the prognosis and regulatory mechanisms in non-small cell lung cancer. Front Med (Lausanne) 2025; 12:1480169. [PMID: 40124684 PMCID: PMC11925780 DOI: 10.3389/fmed.2025.1480169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
Lung cancer remains a major global health issue, with non-small cell lung cancer (NSCLC) constituting approximately 85% of cases. Ferritinophagy, a pivotal autophagic process in ferroptosis, plays an essential role in tumor initiation and progression. However, the specific contributions of ferritinophagy-related genes (FRGs) to NSCLC pathogenesis remain incompletely understood. In this study, weighted gene co-expression network analysis (WGCNA) was employed to identify key modular genes associated with FRG scores. Genes overlapping between these modules and differentially expressed genes (DEGs) were selected for further investigation. Prognostic genes were identified through univariate Cox regression and least absolute shrinkage and selection operator (LASSO) analysis, with subsequent validation using quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) on both clinical samples and the TCGA-NSCLC dataset. A nomogram incorporating clinicopathological features and risk scores was developed to predict patient outcomes. Further analyses focused on functional enrichment, drug sensitivity, and the immune microenvironment. Cross-referencing 2,142 key modular genes with 2,764 DEGs revealed 600 candidate genes. Univariate Cox regression and LASSO analysis of these candidates identified eight prognostic genes: KLK8, MFI2, B3GNT3, MYRF, CREG2, GLB1L3, AHNAK2, and NLRP10. Two distinct risk groups exhibited significant survival differences. Both the risk score and pathological N stage were found to be independent prognostic factors, forming the basis for the nomogram. Notable correlations were observed between certain immune cells, prognostic genes, and immune responses, affecting the efficacy of immunotherapy and drug sensitivity. qRT-PCR confirmed that, except for NLRP10, all prognostic genes exhibited expression patterns consistent with TCGA-NSCLC data. This study highlights the significant role of FRGs in NSCLC prognosis and regulation, offering novel insights for personalized treatment strategies.
Collapse
Affiliation(s)
- Yuan Hao
- Clinical Trials Center, Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China
| | - Xin Wang
- Clinical Trials Center, Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China
| | - Zerong Ni
- Clinical Trials Center, Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China
| | - Yuhui Ma
- Department of Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Tongji Medical College Huazhong University Science of and Technology, Taiyuan, China
| | - Jing Wang
- Department of Pathology, Shanxi Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Wen Su
- Department of Immunology, Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China
| |
Collapse
|
34
|
Tajima K, Shimodate Y, Mouri H, Nagahisa Y, Nishina S, Mizuno M. Complete Remission with Nivolumab Monotherapy of Advanced Gastric Neuroendocrine Carcinoma. Intern Med 2025; 64:717-723. [PMID: 40024686 PMCID: PMC11949651 DOI: 10.2169/internalmedicine.3585-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/23/2024] [Indexed: 03/04/2025] Open
Abstract
A 77-year-old man with large-cell gastric neuroendocrine carcinoma underwent palliative total gastrectomy after 10 cycles of carboplatin and etoposide chemotherapy for severe tumor bleeding. Despite a histological grade 0 therapeutic effect, the remnant lesions were treated with paclitaxel and ramucirumab, but progression occurred after two cycles. Nivolumab monotherapy was then initiated, resulting in complete remission after three cycles. Nivolumab was discontinued because of immune-related psoriasis, but complete remission persisted for at least 16 months. This is the first reported case of nivolumab monotherapy achieving complete remission in gastric neuroendocrine carcinoma with the patient surviving 28 months.
Collapse
Affiliation(s)
- Koichiro Tajima
- Department of Gastroenterology and Hepatology, Kurashiki Central Hospital, Japan
| | - Yuichi Shimodate
- Department of Gastroenterology and Hepatology, Kurashiki Central Hospital, Japan
| | - Hirokazu Mouri
- Department of Gastroenterology and Hepatology, Kurashiki Central Hospital, Japan
| | | | | | - Motowo Mizuno
- Department of Gastroenterology and Hepatology, Kurashiki Central Hospital, Japan
| |
Collapse
|
35
|
Harvanik P, Šemeláková M, Solárová Z, Solár P. Novel factors of cisplatin resistance in epithelial ovarian tumours. Adv Med Sci 2025; 70:94-102. [PMID: 39880191 DOI: 10.1016/j.advms.2025.01.005] [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: 07/18/2024] [Revised: 10/11/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025]
Abstract
Ovarian tumours are these days one of the biggest oncogynecological problems. In addition to surgery, the treatment of ovarian cancer includes also chemotherapy in which platinum preparations are one of the most used chemotherapeutic drugs. The principle of antineoplastic effects of cisplatin (cis-diamminedichloroplatinum(II), CDDP) is its binding to the DNA and the formation of adducts. While DNA adducts induce the process of apoptosis, or inhibit the process of DNA replication, which prevents further division of tumour cells, various molecular mechanisms can reverse this process. On the other hand, with increasing scientific knowledge, it is becoming clearer that chemotherapy resistance is a very complex process. In this regard, factors and the amount of their expression may regulate the effect of resistance to chemotherapy. This review focuses on new molecular mechanisms and factors such as mitochondrial dynamics, epithelial-mesenchymal transition (EMT), cluster of differentiation, exosomes and others, that could be involved in the emergence of CDDP resistance.
Collapse
Affiliation(s)
- Pavol Harvanik
- Department of Medical Biology, Faculty of Medicine, Pavol Jozef Šafárik University, Košice, Slovak Republic
| | - Martina Šemeláková
- Department of Medical Biology, Faculty of Medicine, Pavol Jozef Šafárik University, Košice, Slovak Republic
| | - Zuzana Solárová
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, Košice, Slovak Republic
| | - Peter Solár
- Department of Medical Biology, Faculty of Medicine, Pavol Jozef Šafárik University, Košice, Slovak Republic.
| |
Collapse
|
36
|
Dash UC, Nayak V, Navani HS, Samal RR, Agrawal P, Singh AK, Majhi S, Mogare DG, Duttaroy AK, Jena AB. Understanding the molecular bridges between the drugs and immune cell. Pharmacol Ther 2025; 267:108805. [PMID: 39908660 DOI: 10.1016/j.pharmthera.2025.108805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/11/2025] [Accepted: 01/21/2025] [Indexed: 02/07/2025]
Abstract
The interactions of drugs with the host's immune cells determine the drug's efficacy and adverse effects in patients. Nonsteroidal Anti-Inflammatory Drugs (NSAID), such as corticosteroids, NSAIDs, and immunosuppressants, affect the immune cells and alter the immune response. Molecularly, drugs can interact with immune cells via cell surface receptors, changing the antigen presentation by modifying the co-stimulatory molecules and interacting with the signaling pathways of T cells, B cells, Natural killer (NK) cells, mast cells, basophils, and macrophages. Immunotoxicity, resulting from drug-induced changes in redox status, generation of Reactive Oxygen Species (ROS)/Reactive Nitrogen Species (RNS), and alterations in antioxidant enzymes within immune cells, leads to immunodeficiency. This, in turn, causes allergic reactions, autoimmune diseases, and cytokine release syndrome (CRS). The treatment options should include the evaluation of immune status and utilization of the concept of pharmacogenomics to minimize the chances of immunotoxicity. Many strategies in redox, like targeting the redox pathway or using redox-active agents, are available for the modulation of the immune system and developing drugs. Case studies highlight significant drug-immune cell interactions and patient outcomes, underscoring the importance of understanding these complexities. The future direction focuses on the drugs to deliver antiviral therapy, new approaches to immunomodulation, and modern technologies for increasing antidote effects with reduced toxicity. In conclusion, in-depth knowledge of the interaction between drugs and immune cells is critical to protect the patient from the adverse effects of the drug and improve therapeutic outcomes of the treatment process. This review focuses on the multifaceted interactions of drugs and their consequences at the cellular levels of immune cells.
Collapse
Affiliation(s)
- Umesh Chandra Dash
- School of Biotechnology, Campus 11, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha 751024, India
| | - Vinayak Nayak
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, India
| | - Hiten Shanker Navani
- Biological Materials Laboratory, CSIR- Central Leather Research Institute, Adyar, Chennai 600020, India
| | - Rashmi Rekha Samal
- CSIR-Institute of Minerals & Materials Technology, Bhubaneswar 751 013, India
| | - Palak Agrawal
- Unit de Microbiologie Structurale, Institut Pasteur, Paris, France
| | - Anup Kumar Singh
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India
| | - Sanatan Majhi
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India
| | - Devraj Ganpat Mogare
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway.
| | - Atala Bihari Jena
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India
| |
Collapse
|
37
|
Zhuang AB, Xi Z, Cheng YX, Zhang CH, Li WG. Current status and future perspectives of immunotherapy for abdominal liposarcoma: From basic research to clinical practice. Shijie Huaren Xiaohua Zazhi 2025; 33:81-88. [DOI: 10.11569/wcjd.v33.i2.81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/06/2024] [Accepted: 12/17/2024] [Indexed: 02/28/2025] Open
Abstract
Liposarcoma is a highly heterogeneous type of soft tissue sarcoma originating from adipose tissue, characterized by complex biological behavior and invasiveness. Traditional treatments have shown limited efficacy in high-grade and metastatic liposarcoma, with unsatisfactory patient outcomes. In recent years, the breakthroughs of immunotherapy in various solid tumors have sparked interest in its potential application to liposarcoma. This review systematically examines the progress in basic research and clinical practice of immunotherapy for liposarcoma, discussing the tumor immune microenvironment, mechanisms of immune evasion, the application of immune checkpoint inhibitors, combination therapy strategies, the challenges faced, as well as the future direction, with an aim to provide a theoretical basis for personalized treatment of liposarcoma, promote the development of novel immunotherapy strategies, and ultimately improve patient prognosis and quality of life.
Collapse
Affiliation(s)
- Ao-Bo Zhuang
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Zhe Xi
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Ying-Xue Cheng
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Chen-He Zhang
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Wen-Gang Li
- Department of Hepatobiliary and Pancreatic Surgery, Xiang'an Hospital of Xiamen University, Xiamen 361102, Fujian Province, China
- Cancer Research Center of Xiamen University, Xiamen 361005, Fujian Province, China
| |
Collapse
|
38
|
Lu R, Wang Z, Tian W, Shi W, Chu X, Zhou R. A retrospective study of radiotherapy combined with immunotherapy for patients with baseline brain metastases from non-small cell lung cancer. Sci Rep 2025; 15:7036. [PMID: 40016281 PMCID: PMC11868486 DOI: 10.1038/s41598-025-91863-7] [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: 07/15/2024] [Accepted: 02/24/2025] [Indexed: 03/01/2025] Open
Abstract
This multi-center retrospective study aimed to evaluate the safety and efficacy of first-line immunotherapy in non-small-cell lung cancer (NSCLC) patients with brain metastases (BM). The study included 138 patients treated with immune checkpoint inhibitors (ICIs), either alone or in combination with brain radiotherapy (BRT), from 2020 to October 2023. Intracranial overall response rate (iORR), overall response rate (ORR), progression-free survival (PFS), intracranial progression-free survival (iPFS), overall survival (OS) and treatment-related toxicities were evaluated. Although patients receiving ICIs plus BRT showed a trend toward longer OS compared with ICI alone, the difference was not statistically significant (P = 0.201). Among 82 patients with available data, the iORR was 49.1% (35-63) in the ICIs alone group, and 75.9% (56-90) in the ICIs + BRT group. Notably, in patients requiring corticosteroids or mannitol, combination therapy was associated with a better prognosis (P = 0.05). We found that the iORR of patients treated with ICIs + BRT was improved and did not increase the incidence of serious adverse events (SAEs). Besides, the combination of ICIs and BRT improved the survival rate of subgroups of patients using corticosteroids.
Collapse
Affiliation(s)
- Ruoyu Lu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Ziqi Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Wentao Tian
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Wen Shi
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Xianjing Chu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China.
| | - Rongrong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China.
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, People's Republic of China.
| |
Collapse
|
39
|
Gan Y, Yuan Z, Weng J, Huang M, Li T, Wu Y, Lin K, Han J, Li X, Liu H, Wan Z, Li Z, Chen Z, Cui J, Luo Y, Huang M, Yu H, Lin J. Transcriptomic profile of RNA pseudouridine modification as a biomarker for cellular senescence associated with survival outcomes in colorectal cancer. BMC Biol 2025; 23:61. [PMID: 40016751 PMCID: PMC11866714 DOI: 10.1186/s12915-025-02170-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 02/18/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is considered as an age-related disease, and cellular senescence (CS) plays a crucial role in cancer development and progression. Previous studies have shown the role of epigenetic changes in aging and cancer development, but the role of RNA pseudouridine (Ψ) modification in aging and cancer remains to be explored. RESULTS Using bulk RNA sequencing, CRC cells with low Ψ writers expression levels have higher CS levels. We developed the Psi Score for assessing the transcriptomic profile of RNA Ψ modification regulation and found that the Psi Score correlates with CS. Furthermore, Psi-related senescence may be mediated by mTOR, TGF-β, TNF-α, and inflammatory response signaling pathways. Meanwhile, Psi Score could predict the anti-cancer treatment outcomes of anti-aging interventions and could be used to predict the response to immunotherapy. CONCLUSIONS Overall, these findings reveal that RNA Ψ modification connected aging and cancer and provided novel insights into biomarker-guided cancer regimens.
Collapse
Affiliation(s)
- Yingguo Gan
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Ze Yuan
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Ministry of Education, Guangzhou, China
| | - Jingrong Weng
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Mingzhe Huang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Tuoyang Li
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Yuanhui Wu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Kaixin Lin
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Junyi Han
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Xuan Li
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Haotian Liu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Zixiao Wan
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Ministry of Education, Guangzhou, China
| | - Ziming Li
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Ministry of Education, Guangzhou, China
| | - Zhenghua Chen
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Ji Cui
- Departments of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Yanxin Luo
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China
| | - Meijin Huang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China.
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Ministry of Education, Guangzhou, China.
| | - Huichuan Yu
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China.
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Ministry of Education, Guangzhou, China.
| | - Jinxin Lin
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, China.
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Ministry of Education, Guangzhou, China.
| |
Collapse
|
40
|
Ooyama T, Hirayama M, Seki Y, Iwamoto A, Yoshida R, Nakayama H. Pretreatment nutritional indices are associated with survival and T-cell exhaustion in recurrent or metastatic oral squamous cell carcinoma patients treated with immune checkpoint inhibitors: a retrospective cohort study. Int J Oral Maxillofac Surg 2025:S0901-5027(25)00011-6. [PMID: 39939190 DOI: 10.1016/j.ijom.2025.01.010] [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: 05/03/2024] [Revised: 12/28/2024] [Accepted: 01/17/2025] [Indexed: 02/14/2025]
Abstract
Pretreatment immune dynamics and nutritional status are important predictors of survival outcomes in various malignancies. This study was performed to evaluate the relationships between survival outcomes and the pretreatment nutritional indices - Onodera's prognostic nutritional index (OPNI) and neutrophil-to-lymphocyte ratio (NLR) - in 42 patients with recurrent or metastatic oral squamous cell carcinoma (OSCC) who underwent treatment with immune checkpoint inhibitors (ICI). Additionally, the relationships between these nutritional indices and T-cell exhaustion in the peripheral blood of the patients were analysed. As a result, the Kaplan-Meier method revealed that lower OPNI was significantly associated with poorer overall survival (OS) and progression-free survival (PFS) (both P < 0.001). Likewise, the results of the multivariate analysis showed that a low OPNI was independently associated with poor 5-year OS (hazard ratio 4.36, P = 0.008) and PFS (hazard ratio 4.04, P = 0.010). Patients with a low OPNI had a significantly higher frequency of PD-1+ CD8+ T-cells than those with a high OPNI (P = 0.009). These findings demonstrate that pretreatment OPNI is a valuable independent prognostic indicator of OS and PFS in OSCC patients following treatment with ICI. The OPNI might reflect T-cell exhaustion in the peripheral blood of OSCC patients.
Collapse
Affiliation(s)
- T Ooyama
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - M Hirayama
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| | - Y Seki
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - A Iwamoto
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - R Yoshida
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - H Nakayama
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
41
|
Qin X, Xu W, Wu J, Li M. Integration of single-cell and bulk RNA-sequencing data to construct and validate a signature based on NK cell marker genes to predict immunotherapy response and prognosis in colorectal cancer. Discov Oncol 2025; 16:134. [PMID: 39920524 PMCID: PMC11805743 DOI: 10.1007/s12672-025-01842-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/20/2025] [Indexed: 02/09/2025] Open
Abstract
We aimed to create a NK cell marker genes-based signature to predict immunotherapy response and prognosis in colorectal cancer. We integrated scRNA-seq data from four Gene Expression Omnibus (GEO) samples and performed Weighted gene correlation network analysis (WGCNA) based on 587 the Cancer Genome Atlas (TCGA) colorectal cancer samples to uncover NK cell-related genes. We identified 1080 NK cell-related core genes and 276 NK cell-related feature genes based on WGCNA and clustering and annotation of scRNA-seq data, respectively. Six key NK cell-related prognostic signature genes were obtained by univariate and LASSO regression analyses, including ADAM8, CTSD, CCL4, IL2RB, TTC38, and PLEK. Two validation cohorts from the GEO dataset, comprising 124 and 201 samples respectively, were used. The signature was significantly associated with overall survival and correlated with immune cell infiltration, immune and stromal scores, and immune checkpoint genes. Furthermore, the signature was associated with the homologous recombination deficiency (HRD) and T-cell receptor (TCR) scores. In conclusion, our study proposes a new prognostic signature based on NK cell marker genes, which may serve as a potential tool to predict overall survival and immunotherapy response for CRC patients.
Collapse
Affiliation(s)
- Xiaoyu Qin
- Department of Gastroenterology, Shanghai Pudong New Area Gongli Hospital, Shanghai, 200135, China
| | - Wenjuan Xu
- Department of General Surgery, Shanghai Punan Hospital, Pudong New District, Shanghai, 200125, China
| | - Jinxiu Wu
- Department of General Surgery, Shanghai Punan Hospital, Pudong New District, Shanghai, 200125, China
| | - Ming Li
- Department of General Surgery, Shanghai Punan Hospital, Pudong New District, Shanghai, 200125, China.
| |
Collapse
|
42
|
Wu L, Zhi X, Xie S, Li K, Chen M, Li G, Wu Q, Jiao S, Wang J, Liu T. Immunological characteristics of peripheral T cells as prognostic markers for Camrelizumab and Apatinib combination therapy in advanced squamous non-small-cell lung cancer. Mol Immunol 2025; 178:87-96. [PMID: 39870014 DOI: 10.1016/j.molimm.2025.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 01/29/2025]
Abstract
PURPOSE To determine the characteristic changes of peripheral blood T cells and identify potential biomarkers that associated with the clinical efficacy of combined immunotherapy and anti-angiogenic therapy in patients with advanced squamous non-small cell lung cancer (NSCLC). METHODS We performed a comprehensive immunological assessment of peripheral blood mononuclear cell samples from advanced squamous NSCLC patients before and after combination of immunotherapy (Camrelizumab) and anti-angiogenic therapy (Apatinib) using spectral flow cytometry. Correlations between these immunological features and clinical efficacy were analyzed. RESULTS Our findings revealed that, following two treatment cycles, the concentration of type 1 T helper (Th1) cells in the peripheral circulation was significantly higher in the responder group than in the non-responder group, correlating with a statistically significant improvement in survival outcomes. Post-treatment, CD137 expression within Th1 cells in the responders, whereas TIM-3 expression was significantly reduced. In the validation cohort, elevated CD4+ CXCR3+ CD137+ cells in the peripheral blood were associated with a positive clinical reaction to the treatment and extended survival. CONCLUSIONS Our findings suggest that peripheral blood circulating CD4+ CXCR3+ CD137+ cells serve as biomarkers of response to combined immunotherapy and anti-angiogenic therapy in patients with advanced squamous NSCLC, providing potential guidance for improving clinical outcomes.
Collapse
Affiliation(s)
- Liangliang Wu
- Laboratory of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China; Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaoyu Zhi
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China
| | - Shengzhi Xie
- Department of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Keren Li
- Hepato-Pancereato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Man Chen
- Department of laboratory medicine, Hebei Yanda Lu Daopei Hospital, Langfang 065201, China
| | - Gong Li
- Department of Radiation Oncology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Qiyan Wu
- Laboratory of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China; Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shunchang Jiao
- Department of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Jinliang Wang
- Department of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Tianyi Liu
- Laboratory of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China; Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
43
|
Li J, Zhang Y, Hu L, Ye H, Yan X, Li X, Li Y, Ye S, Wu B, Li Z. T-cell Receptor Repertoire Analysis in the Context of Transarterial Chemoembolization Synergy with Systemic Therapy for Hepatocellular Carcinoma. J Clin Transl Hepatol 2025; 13:69-83. [PMID: 39801788 PMCID: PMC11712086 DOI: 10.14218/jcth.2024.00238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/03/2024] [Accepted: 10/25/2024] [Indexed: 01/16/2025] Open
Abstract
T-cell receptor (TCR) sequencing provides a novel platform for insight into and characterization of intricate T-cell profiles, advancing the understanding of tumor immune heterogeneity. Recently, transarterial chemoembolization (TACE) combined with systemic therapy has become the recommended regimen for advanced hepatocellular carcinoma. The regulation of the immune microenvironment after TACE and its impact on tumor progression and recurrence has been a focus of research. By examining and tracking fluctuations in the TCR repertoire following combination treatment, novel perspectives on the modulation of the tumor microenvironment post-TACE and the underlying mechanisms governing tumor progression and recurrence can be gained. Clarifying the distinctive metrics and dynamic alterations of the TCR repertoire within the context of combination therapy is imperative for understanding the mechanisms of anti-tumor immunity, assessing efficacy, exploiting novel treatments, and further advancing precision oncology in the treatment of hepatocellular carcinoma. In this review, we initially summarized the fundamental characteristics of TCR repertoire and depicted immune microenvironment remodeling after TACE. Ultimately, we illustrated the prospective applications of TCR repertoires in TACE combined with systemic therapy.
Collapse
Affiliation(s)
- Jie Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Luqi Hu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Heqing Ye
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Xingli Yan
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Xin Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Yifan Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Shuwen Ye
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Bailu Wu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Zhen Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
44
|
Ring BZ, Cronister CT, Ring HZ, Ross DT, Seitz RS. Immune infiltrate populations within distinct tumor immune microenvironments predictive of immune checkpoint treatment outcome. Sci Rep 2025; 15:3126. [PMID: 39856115 PMCID: PMC11760962 DOI: 10.1038/s41598-024-83915-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 12/18/2024] [Indexed: 01/27/2025] Open
Abstract
Understanding the dynamic tumor immune microenvironment (TIME) is important in guiding immunotherapy. We have previously validated signatures predictive of checkpoint inhibitor efficacy which distinguish immunomodulatory, mesenchymal stem-like, and mesenchymal phenotypes. Here we use twenty tumor types (7162 samples) to identify potentially conserved immune biology within these TIME spaces, genes co-expressed across distinct cell types involved these immune processes, and the association of these signatures with ICI response. One signature, which contained multiple B-cell markers, was associated with immunotherapy efficacy in three cohorts, including IMvigor210. This signature of potentially conserved B-cell biology in co-infiltrated immune cell ecosystems had a more consistent association with outcome than comparable single cell type models and likely reflects a complex immunological response involving multilayered relationships between distinct immune effector cell types. These signatures were most highly expressed in tumors with prominent immune cell invasion, however there was consistent identification of infiltrate presence in relatively immune restricted cases. This suggests that these immune population signatures may identify conserved immune cell type co-infiltrate physiology of the TIME that best captures immune physiology with potential clinical utility.
Collapse
Affiliation(s)
| | | | - Huijun Z Ring
- Department of Medicine, Stanford University, Stanford, CA, USA
| | | | | |
Collapse
|
45
|
Sun Z, Hu M, Huang X, Song M, Chen X, Bei J, Lin Y, Chen S. Predictive value of dendritic cell-related genes for prognosis and immunotherapy response in lung adenocarcinoma. Cancer Cell Int 2025; 25:13. [PMID: 39810206 PMCID: PMC11730157 DOI: 10.1186/s12935-025-03642-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Patients with lung adenocarcinoma (LUAD) receiving drug treatment often have an unpredictive response and there is a lack of effective methods to predict treatment outcome for patients. Dendritic cells (DCs) play a significant role in the tumor microenvironment and the DCs-related gene signature may be used to predict treatment outcome. Here, we screened for DC-related genes to construct a prognostic signature to predict prognosis and response to immunotherapy in LUAD patients. METHODS DC-related biological functions and genes were identified using single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing. DCs-related gene signature (DCRGS) was constructed using integrated machine learning algorithms. Expression of key genes in clinical samples was examined by real-time q-PCR. Performance of the prognostic model, DCRGS, for the prognostic evaluation, was assessed using a multiple time-dependent receiver operating characteristic (ROC) curve, the R package, "timeROC", and validated using GEO datasets. RESULTS Analysis of scRNA-seq data showed that there is a significant upregulation of LGALS9 expression in DCs isolated from malignant pleural effusion samples. Leveraging the Coxboost and random survival forest combination algorithm, we filtered out six DC-related genes on which a prognostic prediction model, DCRGS, was established. A high predictive capability nomogram was constructed by combining DCRGS with clinical features. We found that patients with a high-DCRGS score had immunosuppression, activated tumor-associated pathways, and elevated somatic mutational load and copy number variant load. In contrast, patients in the low-DCRGS subgroup were resistant to chemotherapy but sensitive to the CTLA-4 immune checkpoint inhibitor and targeted therapy. CONCLUSION We have innovatively established a deep learning-based prediction model, DCRGS, for the prediction of the prognosis of patients with LUAD. The model possesses a strong prognostic prediction performance with high accuracy and sensitivity and could be clinically useful to guide the management of LUAD. Furthermore, the findings of this study could provide an important reference for individualized clinical treatment and prognostic prediction of patients with LUAD.
Collapse
Affiliation(s)
- Zihao Sun
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Mengfei Hu
- Department of Internal Medicine, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230000, China
| | - Xiaoning Huang
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Minghan Song
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Xiujing Chen
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Jiaxin Bei
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
| | - Yiguang Lin
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
- Research & Development Division, Guangzhou Anjie Biomedical Technology Co., Ltd., Guangzhou, 510535, China.
| | - Size Chen
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangzhou, 510080, China.
- Key Laboratory of Monitoring Adverse Reactions Associated with CAR-T Cell Therapy, Guangzhou, 510080, China.
| |
Collapse
|
46
|
Glaviano A, Lau HSH, Carter LM, Lee EHC, Lam HY, Okina E, Tan DJJ, Tan W, Ang HL, Carbone D, Yee MYH, Shanmugam MK, Huang XZ, Sethi G, Tan TZ, Lim LHK, Huang RYJ, Ungefroren H, Giovannetti E, Tang DG, Bruno TC, Luo P, Andersen MH, Qian BZ, Ishihara J, Radisky DC, Elias S, Yadav S, Kim M, Robert C, Diana P, Schalper KA, Shi T, Merghoub T, Krebs S, Kusumbe AP, Davids MS, Brown JR, Kumar AP. Harnessing the tumor microenvironment: targeted cancer therapies through modulation of epithelial-mesenchymal transition. J Hematol Oncol 2025; 18:6. [PMID: 39806516 PMCID: PMC11733683 DOI: 10.1186/s13045-024-01634-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025] Open
Abstract
The tumor microenvironment (TME) is integral to cancer progression, impacting metastasis and treatment response. It consists of diverse cell types, extracellular matrix components, and signaling molecules that interact to promote tumor growth and therapeutic resistance. Elucidating the intricate interactions between cancer cells and the TME is crucial in understanding cancer progression and therapeutic challenges. A critical process induced by TME signaling is the epithelial-mesenchymal transition (EMT), wherein epithelial cells acquire mesenchymal traits, which enhance their motility and invasiveness and promote metastasis and cancer progression. By targeting various components of the TME, novel investigational strategies aim to disrupt the TME's contribution to the EMT, thereby improving treatment efficacy, addressing therapeutic resistance, and offering a nuanced approach to cancer therapy. This review scrutinizes the key players in the TME and the TME's contribution to the EMT, emphasizing avenues to therapeutically disrupt the interactions between the various TME components. Moreover, the article discusses the TME's implications for resistance mechanisms and highlights the current therapeutic strategies toward TME modulation along with potential caveats.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Hannah Si-Hui Lau
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lukas M Carter
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Donavan Jia Jie Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Wency Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Michelle Yi-Hui Yee
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Xiao Zi Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Lina H K Lim
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Ruby Yun-Ju Huang
- School of Medicine and Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, 23538, Lübeck, Germany
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Experimental Therapeutics (ET) Graduate Program, University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Tullia C Bruno
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Bin-Zhi Qian
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, W12 0BZ, UK
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Salem Elias
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Saurabh Yadav
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Minah Kim
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Caroline Robert
- Department of Cancer Medicine, Inserm U981, Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif, France
- Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, Paris, France
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Tao Shi
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, NY, USA
| | - Simone Krebs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironment Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Matthew S Davids
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jennifer R Brown
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| |
Collapse
|
47
|
Chadokiya J, Chang K, Sharma S, Hu J, Lill JR, Dionne J, Kirane A. Advancing precision cancer immunotherapy drug development, administration, and response prediction with AI-enabled Raman spectroscopy. Front Immunol 2025; 15:1520860. [PMID: 39850874 PMCID: PMC11753970 DOI: 10.3389/fimmu.2024.1520860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 11/25/2024] [Indexed: 01/25/2025] Open
Abstract
Molecular characterization of tumors is essential to identify predictive biomarkers that inform treatment decisions and improve precision immunotherapy development and administration. However, challenges such as the heterogeneity of tumors and patient responses, limited efficacy of current biomarkers, and the predominant reliance on single-omics data, have hindered advances in accurately predicting treatment outcomes. Standard therapy generally applies a "one size fits all" approach, which not only provides ineffective or limited responses, but also an increased risk of off-target toxicities and acceleration of resistance mechanisms or adverse effects. As the development of emerging multi- and spatial-omics platforms continues to evolve, an effective tumor assessment platform providing utility in a clinical setting should i) enable high-throughput and robust screening in a variety of biological matrices, ii) provide in-depth information resolved with single to subcellular precision, and iii) improve accessibility in economical point-of-care settings. In this perspective, we explore the application of label-free Raman spectroscopy as a tumor profiling tool for precision immunotherapy. We examine how Raman spectroscopy's non-invasive, label-free approach can deepen our understanding of intricate inter- and intra-cellular interactions within the tumor-immune microenvironment. Furthermore, we discuss the analytical advances in Raman spectroscopy, highlighting its evolution to be utilized as a single "Raman-omics" approach. Lastly, we highlight the translational potential of Raman for its integration in clinical practice for safe and precise patient-centric immunotherapy.
Collapse
Affiliation(s)
- Jay Chadokiya
- Department of Surgery, Stanford School of Medicine, Stanford University Medical Center, Stanford, CA, United States
| | - Kai Chang
- Department of Electrical Engineering, Stanford University,
Stanford, CA, United States
| | - Saurabh Sharma
- Department of Surgery, Stanford School of Medicine, Stanford University Medical Center, Stanford, CA, United States
| | - Jack Hu
- Pumpkinseed Technologies, Palo Alto, CA, United States
| | | | - Jennifer Dionne
- Pumpkinseed Technologies, Palo Alto, CA, United States
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, United States
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, United States
| | - Amanda Kirane
- Department of Surgery, Stanford School of Medicine, Stanford University Medical Center, Stanford, CA, United States
| |
Collapse
|
48
|
Cai H, Tian S, Liu A, Xie G, Zhang H, Wu X, Wan J, Li S. Relationship between CTF1 gene expression and prognosis and tumor immune microenvironment in glioma. Eur J Med Res 2025; 30:17. [PMID: 39780198 PMCID: PMC11715937 DOI: 10.1186/s40001-024-02192-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: 05/01/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE This study aimed to evaluate CTF1 expression in glioma, its relationship to patient prognosis and the tumor immune microenvironment, and effects on glioma phenotypes to identify a new therapeutic target for treating glioma precisely. METHODS We initially assessed the expression of CTF1, a member of the IL-6 family, in glioma, using bioinformatics tools and publicly available databases. Furthermore, we examined the correlation between CTF1 expression and tumor prognosis, DNA methylation patterns, m6A-related genes, potential biological functions, the immune microenvironment, and genes associated with immune checkpoints. We also explored potential associations with drug sensitivity. To assess the impact on glioma cell proliferation and apoptosis, we employed various assays, including the Cell Counting Kit-8, colony formation assay, and flow cytometry. RESULTS CTF1 gene and protein expression were significantly elevated in glioma tissues, and correlated with malignancy and poor prognosis. CTF1 was an independent prognostic factor and negatively associated with DNA methylation. The involvement of CTF1 in m6A modifications contributed to glioma progression. Enrichment analysis revealed immune response pathways linked with CTF1 in glioma, including natural killer cell cytotoxicity, NOD-like receptor signaling, Toll-like receptor signaling, antigen processing, chemokine signaling, and cytokine receptor interactions. CTF1 expression correlated positively with pathways related to apoptosis, inflammation, proliferation, and epithelial-mesenchymal transition, and PI3K-AKT-mTOR signaling. Additionally, CTF1 expression was positively associated with macrophage, eosinophil, and neutrophil contents and immune checkpoint-related genes, but negatively associated with sensitivity to 14 drugs. In vitro experiments confirmed that CTF1 knockdown inhibited glioma cell proliferation and promoted apoptosis. CONCLUSION This study identifies CTF1 as a significant independent prognostic factor that is closely associated with the tumor immune microenvironment in glioma. Additionally, reduced expression of CTF1 suppresses the proliferation and induces apoptosis of glioma cells in vitro. Consequently, CTF1 is a potentially promising novel therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Hongqing Cai
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, People's Republic of China
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shen Tian
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, People's Republic of China
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Angsi Liu
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, People's Republic of China
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guanchao Xie
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, People's Republic of China
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, No 678 Furong Road, Economic and Technological Development Zone, Hefei, 230000, Anhui, People's Republic of China
| | - Hongsheng Zhang
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, People's Republic of China
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, No 678 Furong Road, Economic and Technological Development Zone, Hefei, 230000, Anhui, People's Republic of China
| | - Xiaogang Wu
- Department of Neurosurgery, No. 901 Hospital of the Chinese People's Liberation Army Logistic Support Force, No 424 Changjiang West Road, Shushan District, Hefei, Anhui, 230000, People's Republic of China.
| | - Jinghai Wan
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, People's Republic of China.
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, No 678 Furong Road, Economic and Technological Development Zone, Hefei, 230000, Anhui, People's Republic of China.
| | - Sai Li
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, People's Republic of China.
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, No 678 Furong Road, Economic and Technological Development Zone, Hefei, 230000, Anhui, People's Republic of China.
| |
Collapse
|
49
|
Liu Z, Zhang C, Xiao J, He Y, Liang H, Huang J, Cai Z, Yi Z, Chen M, Li Y, Zhang J, liu F, Ren P, Li H, Chen J, Fan B, Hu J, Zu X, Deng D. TBX3 shapes an immunosuppressive microenvironment and induces immunotherapy resistance. Theranostics 2025; 15:1966-1986. [PMID: 39897553 PMCID: PMC11780534 DOI: 10.7150/thno.103175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/28/2024] [Indexed: 02/04/2025] Open
Abstract
Background: Identifying biomarkers that predict immunotherapy efficacy and discovering new targets for combination therapies are critical elements for improving the prognosis of bladder cancer (BLCA) patients. Methods: Firstly, we explored the expression patterns of TBX3 in normal and pan-cancer tissues and the correlation between TBX3 and the immune microenvironment using data from multiple public databases. Then, we combined various techniques, including bulk RNA sequencing, single-cell RNA sequencing, high-throughput cytokine arrays, functional experiments, ProcartaPlex multiplex immunoassays and TissueFAXS panoramic tissue quantification assays, to demonstrate that TBX3 shapes an immunosuppressive tumor microenvironment (TME) in BLCA. Results: We identified TBX3 as a key factor associated with the immunosuppressive microenvironment in BLCA through a systematic multi-omics analysis. We found that TBX3 is primarily expressed in malignant cells, where TBX3high tumor cells increase the secretion of TGFβ1, which promotes the infiltration of cancer-associated fibroblasts (CAFs), thereby forming an immunosuppressive microenvironment. We further demonstrated that TBX3 enhances TGFβ1 expression by binding to the TGFβ1 promoter, and blocking TGFβ1 counteracts the immunosuppressive effects of TBX3. Moreover, TBX3 reduced the cancer-killing efficiency of CD8+ T cells by decreasing the proportion of GZMB+ CD8+ T cells, and knocking down TBX3 combined with anti-PD-1 treatment increased CD8+ T cell infiltration and reduced CAFs in vivo. We also validated the inverse relationship between TBX3+ malignant cells and CD8+ T cells and the positive relationship with CAFs in tissue microarrays. Lastly, we found that TBX3 predicted immunotherapy efficacy in our real-world immunotherapy cohort and multiple public cohorts. Conclusion: In summary, TBX3 promotes BLCA progression and immunotherapy resistance by inducing an immunosuppressive microenvironment, and targeting TBX3 could enhance the efficacy of immunotherapy for BLCA.
Collapse
Affiliation(s)
- Zhi Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- Department of Urology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, China
| | - Chunyu Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiatong Xiao
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Yunbo He
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Haisu Liang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Jinliang Huang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Zhiyong Cai
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenglin Yi
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Mingfeng Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Yixiao Li
- Department of Urology, The second people's Hospital of Hunan province, Changsha, China
| | - Jun Zhang
- Department of Imaging, The first people's Hospital of Kaili city, Kaili, China
| | - Fenglian liu
- Department of Urology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, China
| | - Peng Ren
- Department of Urology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, China
| | - Huihuang Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Jinbo Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Benyi Fan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Jiao Hu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- Department of Urology, The First Affiliated Hospital of Hunan Normal University, Hunan Normal University, Changsha, China
| | - Dingshan Deng
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
50
|
Chen Y, Luo Y, Liu Y, Luo D, Liu A. Dual efficacy of tocilizumab in managing PD-1 inhibitors-induced myocardial inflammatory injury and suppressing tumor growth with PD-1 inhibitors: a preclinical study. Cancer Immunol Immunother 2025; 74:52. [PMID: 39752010 PMCID: PMC11699076 DOI: 10.1007/s00262-024-03899-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/16/2024] [Indexed: 01/04/2025]
Abstract
The combined use of tocilizumab (TCZ) and immune checkpoint inhibitors (ICIs) in cancer treatment is gaining attention, but preclinical studies are lacking. Our study aims to investigate the synergistic anti-tumor effect of TCZ combined with ICIs and its role in treating immune-related adverse events (irAEs). The clinical significance of high interleukin-6 (IL-6) expression in tumor patients was analyzed from the Cancer Genome Atlas (TCGA) database. The expression levels of IL-6 were compared before and during the onset of ICIs-associated myocarditis patients. ICIs-related myocardial inflammatory injury and therapeutic lung cancer models were constructed in C57BL/6 J mice using murine-derived programmed death-1 (PD-1) inhibitors alone or in combination with TCZ. Possible inflammatory mechanisms were proposed and validated. The anti-tumor effects and mechanisms of both drugs in combination were assessed. Patients with high IL-6 expression had a poor prognosis, and those with ICIs-associated myocarditis exhibited elevated IL-6 from baseline. In the PD-1 inhibitors-associated myocardial inflammatory injury mouse model, the levels of IL-6 in the blood and cardiac tissues were significantly elevated. TCZ ameliorated immune myocardial inflammatory injury by inhibiting the IL-6/janus kinase 2 (JAK2)/signal transducer and activator of the transcription 3 (STAT3) pathway. The group treated with PD-1 inhibitors combined with TCZ showed significantly slower tumor growth than that treated with PD-1 inhibitors alone. TCZ resisted tumor growth by inhibiting the IL-6-JAK2-STAT3 pathway. By targeting the IL-6-JAK2-STAT3 pathway, TCZ can alleviate PD-1 inhibitors-associated myocardial inflammatory injury mediated by M1-polarized macrophages and plays a synergistic anti-tumor role by inhibiting lung cancer cell proliferation.
Collapse
Affiliation(s)
- Yanxin Chen
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Department of Radiotherapy, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, Hunan Province, China
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Yuxi Luo
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Yunwei Liu
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Anwen Liu
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China.
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China.
- Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|