1
|
Tian Y, Wang S, Ren S, Lin X, Song L, Zhang L, Wang Z. Targeting suppressive feedback of adenosine immunometabolic pathway by tumor cell membranes-coated nanocapsules for chemo-photothermal therapy and enhanced immunotherapy. CHEMICAL ENGINEERING JOURNAL 2025; 515:163579. [DOI: 10.1016/j.cej.2025.163579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2025]
|
2
|
Wang Q, He J, Lei T, Li X, Yue S, Liu C, Hu Q. New insights into cancer immune checkpoints landscape from single-cell RNA sequencing. Biochim Biophys Acta Rev Cancer 2025; 1880:189298. [PMID: 40088992 DOI: 10.1016/j.bbcan.2025.189298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Immune checkpoint blockade (ICB) therapy represents a pivotal advancement in tumor immunotherapy by restoring the cytotoxic lymphocytes' anti-tumor activity through the modulation of immune checkpoint functions. Nevertheless, many patients experience suboptimal therapeutic outcomes, likely due to the immunosuppressive tumor microenvironment, drug resistance, and other factors. Single-cell RNA sequencing has assisted to precisely investigate the immune infiltration patterns before and after ICB treatment, enabling a high-resolution depiction of previously unrecognized functional interaction among immune checkpoints. This review addresses the heterogeneity between tumor microenvironments that respond to or resist ICB therapy, highlighting critical factors underlying the variation in immunotherapy efficacy and elucidating treatment failure. Furthermore, a comprehensive examination is provided of how specific ICBs modulate immune and tumor cells to achieve anti-tumor effects and generate treatment resistance, alongside a summary of emerging immune checkpoints identified as promising targets for cancer immunotherapy through single-cell RNA sequencing applications.
Collapse
Affiliation(s)
- Qian Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiahui He
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tianyu Lei
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaohui Li
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China
| | - Shengqin Yue
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Chao Liu
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China.
| | - Qinyong Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Renmin Hospital of Wuhan Economic and Technological Development Zone (Hannan), Wuhan 430090, China.
| |
Collapse
|
3
|
Wang Y, Tang C, Wang K, Zhang X, Zhang L, Xiao X, Lin H, Xiong L. The role of ferroptosis in breast cancer: Tumor progression, immune microenvironment interactions and therapeutic interventions. Eur J Pharmacol 2025; 996:177561. [PMID: 40154567 DOI: 10.1016/j.ejphar.2025.177561] [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: 12/29/2024] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Ferroptosis represents a distinctive and distinct form of regulated cellular death, which is driven by the accumulation of lipid peroxidation. It is distinguished by altered redox lipid metabolism and is linked to a spectrum of cellular activities, including cancer. In breast cancer (BC), with triple negative breast cancer (TNBC) being an iron-and lipid-rich tumor, inducing ferroptosis was thought to be a novel approach to killing breast tumor cells. However, in the recent past, a novel conceptual framework has emerged which posits that in addition to the promotion of tumor cell death, ferritin deposition has a potent immunosuppressive effect on the tumor immune microenvironment (TIME) via the influence on both innate and adaptive immune responses. TIME of BC includes various cell populations from both the innate and adaptive immune systems. In this review, the internal association between iron homeostasis and the progression of ferroptosis, along with the common inducers and protectors of ferroptosis in BC, are discussed in detail. Furthermore, a comprehensive analysis is conducted on the dual role of ferroptosis in immune cells and proto-oncogenic functions, along with an evaluation of the potential applications of immunogenic cell death-targeted immunotherapy in TIME of BC. It is anticipated that our review will inform future research endeavors that seek to integrate ferroptosis and immunotherapy in the management of BC.
Collapse
Affiliation(s)
- Yi Wang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Chuanyun Tang
- First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Keqin Wang
- First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiaoan Zhang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lifang Zhang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xinghua Xiao
- Department of Pathology, The First Affiliated Hospital, Nanchang University, 17 Yongwaizheng Road, Nanschang, 330066, China
| | - Hui Lin
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lixia Xiong
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
4
|
Zhang D, Zhu Y, Shen Z, Ma S, Liu S, Lu Z. Immunosenescence and immunotherapy in elderly patients with hepatocellular carcinoma. Semin Cancer Biol 2025; 111:60-75. [PMID: 40020977 DOI: 10.1016/j.semcancer.2025.02.010] [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: 12/28/2024] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 03/03/2025]
Abstract
Liver cancer, more specifically hepatocellular carcinoma (HCC), is a global health issue and one of the dominant causes of cancer death around the world. In the past few decades, remarkable advances have been achieved in the systemic therapy of HCC. Immune checkpoint inhibitors (ICIs) have become a therapy mainstay for advanced HCC and have shown promise in the neoadjuvant therapy before resection. Despite these significant advancements, the compositions and functions of the immune system occur various alterations with age, called "immunosenescence", which may affect the antitumor effects and safety of ICIs, thus raising concerns that immunosenescence may impair elderly patients' response to ICIs. Therefore, it is important to learn more about the immunosenescence characteristics of elderly patients. However, the real-world elderly HCC patients may be not accurately represented by the elderly patients included in the clinical trials, affecting the generalizability of the efficacy and safety profiles from the clinical trials to the real-world elderly patients. This review summarizes the characteristics of immunosenescence and its influence on HCC progression and immunotherapy efficacy as well as provides the latest progress in ICIs available for HCC and discusses their treatment efficacy and safety on elderly patients. In the future, more studies are needed to clarify the mechanisms of immunosenescence in HCC, and to find sensitive screening tools or biomarkers to identify the patients who may benefit from ICIs.
Collapse
Affiliation(s)
- Dengyong Zhang
- Department of General Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Yan Zhu
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhengchao Shen
- Department of General Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, China
| | - Shuoshuo Ma
- Department of General Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Sihua Liu
- Department of General Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China
| | - Zheng Lu
- Department of General Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, China.
| |
Collapse
|
5
|
Li J, Xu N, Hu L, Xu J, Huang Y, Wang D, Chen F, Wang Y, Jiang J, Hong Y, Ye H. Chaperonin containing TCP1 subunit 5 as a novel pan-cancer prognostic biomarker for tumor stemness and immunotherapy response: insights from multi-omics data, integrated machine learning, and experimental validation. Cancer Immunol Immunother 2025; 74:224. [PMID: 40423850 PMCID: PMC12116413 DOI: 10.1007/s00262-025-04071-7] [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: 03/23/2025] [Accepted: 04/28/2025] [Indexed: 05/28/2025]
Abstract
BACKGROUND Chaperonin containing TCP1 subunit 5 (CCT5), a vital component of the molecular chaperonin complex, has been implicated in tumorigenesis, cancer stemness maintenance, and therapeutic resistance. Nevertheless, its comprehensive roles in pan-cancer progression, underlying biological functions, and potential as a predictor of immunotherapy response remains poorly understood. METHODS We performed a comprehensive multi-omics pan-cancer analysis of CCT5 across 33 cancer types, integrating bulk RNA-seq, single-cell RNA-seq (scRNA-seq), and spatial transcriptomics data. CCT5 expression patterns, prognostic relevance, stemness association, and immune microenvironment relationships were evaluated. A novel CCT5-based signature (CCT5.Sig) was developed using machine learning on 23 immune checkpoint blockade (ICB) cohorts (n = 1394) spanning eight cancer types. Model performance was assessed using AUC metrics and survival analyses. RESULTS CCT5 was significantly overexpressed in tumor tissues and primarily localized to malignant and cycling cells. High CCT5 expression correlated with poor prognosis in multiple cancers and was enriched in oncogenic, cell cycle, and DNA damage repair pathways. CCT5 expression was positively associated with mRNAsi, mDNAsi, and CytoTRACE scores, indicating a role in stemness maintenance. Furthermore, CCT5-high tumors exhibited immune-cold phenotypes, with reduced TILs and CD8⁺ T cell activity. The CCT5.Sig model, based on genes co-expressed with CCT5, achieved superior predictive accuracy for ICB response (AUC = 0.82 in validation and 0.76 in independent testing), outperforming existing pan-cancer signatures. CONCLUSION This study reveals the multifaceted oncogenic roles of CCT5 and highlights its potential as a pan-cancer biomarker for prognosis and immunotherapy response. The machine learning-derived CCT5.Sig model provides a robust tool for patient stratification and may inform personalized immunotherapy strategies.
Collapse
Affiliation(s)
- Jiajun Li
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute and Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Nuo Xu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute and Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Leyin Hu
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, 305000, Zhejiang, China
| | - Jiayue Xu
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yifan Huang
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Deqi Wang
- Department of Gastroenterology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Feng Chen
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yi Wang
- Department of Gastroenterology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Jiani Jiang
- Department of Gastroenterology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yanggang Hong
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Huajun Ye
- Department of Gastroenterology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
6
|
She H, Li TR, Zhao G, Yi L, Liu Q, Liu ZC, Pei HY, Li X, Zuo D, Mao Q, Li Y. Aberrant PLAC8 expression characterizes glioblastoma with temozolomide resistance and an immunosuppressive microenvironment. Cancer Lett 2025; 625:217805. [PMID: 40398706 DOI: 10.1016/j.canlet.2025.217805] [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: 03/01/2025] [Revised: 05/08/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025]
Abstract
Glioblastoma (GBM), Isocitrate Dehydrogenase-wildtype (IDH-WT) represents the most prevalent and clinically aggressive subtype of adult diffuse gliomas, typically associated with poor prognosis. Temozolomide (TMZ) remains the first-line chemotherapeutic agent for GBM; however, the emergence of TMZ resistance represents a major therapeutic obstacle in clinical practice. This study identifies placenta-specific 8 (PLAC8) as a novel mediator of TMZ resistance in IDH-WT GBM. Elevated PLAC8 expression was strongly correlated with poorer survival rates, higher tumor grades in glioma, establishing it as an independent prognostic factor. Notably, consistent upregulation of PLAC8 was observed in both TMZ-resistant GBM cells and TMZ-treated patients, suggesting its potential as a biomarker for TMZ resistance. Mechanistic studies revealed that PLAC8 regulates TMZ sensitivity in GBM cells through the AKT-mTOR signaling pathway. Additionally, integrated bioinformatics and clinical analyses demonstrated that PLAC8 expression positively correlates with immune cell infiltration while promoting an immunosuppressive tumor microenvironment and modulating immunotherapy-related biomarkers, suggesting its potential as a predictive biomarker for immunotherapy response. In conclusion, PLAC8 represents a promising biomarker and therapeutic target for overcoming TMZ resistance and guiding immunotherapy in GBM. This study provides valuable insights for the development of personalized treatment strategies aimed at improving patient outcomes.
Collapse
Affiliation(s)
- Han She
- Department of Anesthesiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Tian-Ran Li
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Guozhi Zhao
- Department of Urology Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Liang Yi
- Department of Neurosurgery, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Qing Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Zheng-Chao Liu
- Department of Anesthesiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Hao-Yu Pei
- Department of Anesthesiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Xunjia Li
- Department of Nephrology, The First Affiliated Hospital of Chongqing University of Chinese Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Deyu Zuo
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing University of Chinese Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China; Department of Research and Development, Chongqing Precision Medical Industry Technology Research Institute, Chongqing, 400000, China.
| | - Qingxiang Mao
- Department of Anesthesiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China.
| | - Yong Li
- Department of Anesthesiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China.
| |
Collapse
|
7
|
Yamamoto T, Ito T, Suzuki T, Mizuno K, Yokoyama S, Yamamoto K, Imai N, Ishizu Y, Honda T, Kawashima H. Steroid initiation dose and duration of steroid reduction for immune checkpoint inhibitor-induced liver injury. Hepatol Res 2025. [PMID: 40369781 DOI: 10.1111/hepr.14200] [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: 02/10/2025] [Revised: 03/23/2025] [Accepted: 04/13/2025] [Indexed: 05/16/2025]
Abstract
AIM Treatment for severe immune checkpoint inhibitor (ICI)-induced liver injury (≥ Grade 3) requires prednisolone (PSL) administration and interruption of the underlying malignancy treatment. If the liver injury improves steadily, a lower initial dose of PSL is beneficial. We aimed to investigate the relationship between the initial dose of PSL and the response to PSL or the duration of PSL dose reduction. METHODS We retrospectively collected clinical data of patients treated with ICIs at Nagoya University Hospital between September 2014 and December 2023. Patients who received PSL for severe ICI-induced liver injury were divided according to the starting dose into group A (0.8 mg/kg/day) and group B (1.0 mg/kg/day). The time to improvement in liver injury and the reduction of the PSL dose to 10 mg/day were compared between the groups. RESULTS Overall, 1271 patients were treated with ICIs, of whom 80 experienced severe ICI-induced liver injury. Of the patients, 29 did not receive steroids, and five used PSL doses of <0.5 mg/kg/day. There were no significant differences in the baseline characteristics or laboratory data between the groups. The time to dose reduction to 10 mg/day PSL was significantly shorter in group A than in group B. The time to improvement in liver injury did not differ between the two groups. CONCLUSION In some patients with severe ICI-induced liver injury, a dose of 0.8 mg/kg/day of PSL is sufficient to achieve a therapeutic effect and shorten the time required to reduce the dose to 10 mg/day.
Collapse
Affiliation(s)
- Takafumi Yamamoto
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takanori Ito
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takaya Suzuki
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuyuki Mizuno
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
| | - Shinya Yokoyama
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenta Yamamoto
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norihiro Imai
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoji Ishizu
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Honda
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
8
|
Wu D, Liu N, Dong H, Zhou K, Du L, Li Y, Chao Y, Ma F. Efficacy and safety of neoadjuvant systemic therapy in resectable hepatocellular carcinoma: a Systematic Review and meta-analysis. Front Oncol 2025; 15:1504917. [PMID: 40416873 PMCID: PMC12098073 DOI: 10.3389/fonc.2025.1504917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 04/18/2025] [Indexed: 05/27/2025] Open
Abstract
Background Neoadjuvant systemic therapy has been shown to benefit patients with solid tumors such as breast cancer and colorectal cancer, but its application in hepatocellular carcinoma (HCC) is still in the exploratory stage, with no established effective regimen. This systematic review and meta-analysis aims to investigate the efficacy and safety of neoadjuvant systemic therapy in patients with resectable HCC. Methods The clinical trials of resectable HCC neoadjuvant systemic therapy in PubMed, Embase and the Cochrane Library were systematically searched. A meta-analysis was performed using STATA/MP18.0 software, and the effect size was calculated using either a fixed effects model or a random effects model, and 95% confidence intervals (CIs) were calculated. Subgroup analysis was performed according to the neoadjuvant systemic therapy regimen. Results This meta-analysis included 328 patients from 15 studies. In patients with resectable HCC, the pooled pathologic complete response (pCR) rate was 15% (95%CI: 10%-21%), the major pathologic response (MPR) rate was 28% (95%CI: 21%-35%), the incidence of grade 3-4 treatment-related adverse events (TRAEs) was 11% (95% CI: 4%-20%), the objective response rate (ORR) was 27% (95% CI: 20%-35%), the surgical resection rate was 84% (95%CI: 75%-92%), and the delay rate was 0.00% (95% CI: 0%-4%). The results of subgroup analysis showed that the efficacy of targeted therapy combined with immunotherapy is superior to dual ICI (immune checkpoint inhibitor) combination therapy and ICI monotherapy, while the safety of the ICI monotherapy was the highest, superior to the dual ICIs and the targeted therapy combined with immunotherapy. Conclusion Neoadjuvant systemic therapy shows preliminarily beneficial outcomes in resectable HCC treatment. However, future large-scale and multicenter randomized controlled trials are needed to confirm this conclusion. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024562257.
Collapse
Affiliation(s)
| | | | | | | | - Lei Du
- Department of Hepatobiliary Surgery, Xianyang Central Hospital, Xianyang, Shaanxi, China
| | | | | | | |
Collapse
|
9
|
Zhou J, Gao M, Zhang S, Guo WW, He W, Zhang M, Chen X, Dongzhi C, Li X, Yuan Y, Ma W. PP1A Modulates the Efficacy of Lenvatinib Plus ICIs Therapy by Inhibiting Ferroptosis in Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2501730. [PMID: 40344394 DOI: 10.1002/advs.202501730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/09/2025] [Indexed: 05/11/2025]
Abstract
Advanced hepatocellular carcinoma (HCC) is characterized by poor prognosis, primarily due to limited therapeutic options and resistance to treatment. Although the combination of tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors (ICIs) has shown promising potential, the underlying mechanisms remain inadequately understood. Here, serine/threonine-specific protein phosphatase (PP1A) is upregulated in Lenvatinib-resistant HCC cells and correlates with poor prognosis. Functional experiments revealed that PP1A promotes HCC progression both in vitro and in vivo. Transcriptomic analysis and ferroptosis metabolite profiling (e.g., ROS, Fe2⁺, lipid-ROS, and GSH) demonstrated that PP1A inhibits Lenvatinib-induced ferroptosis by dephosphorylating Keap1 at site 104. This disruption of the Keap1-Nrf2 interaction enhances the transcription of ferroptosis-related markers and immune checkpoint PD-L1. Notably, single-cell sequencing and co-culture experiments revealed that PP1A knockdown alleviates T cell exhaustion and immune evasion, thereby improving antitumor immunity. In vivo experiments further demonstrated that PP1A knockdown significantly enhances the efficacy of Lenvatinib-ICIs combination therapy. Overall, our findings highlight PP1A as a critical regulator of ferroptosis and antitumor immunity, suggesting its potential as a predictive biomarker and therapeutic target for improving outcomes in advanced HCC.
Collapse
Affiliation(s)
- Jitong Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Meng Gao
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Shikun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Wing-Wa Guo
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Wenzhi He
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Minghe Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Xi Chen
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Cairang Dongzhi
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Xiaomian Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Yufeng Yuan
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, P. R. China
| | - Weijie Ma
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| |
Collapse
|
10
|
Marino R, Hassan AT, Fagenson A, Tabrizian P. Liver transplantation for hepatocellular carcinoma following immunotherapy. Curr Opin Organ Transplant 2025:00075200-990000000-00182. [PMID: 40326429 DOI: 10.1097/mot.0000000000001228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
PURPOSE OF REVIEW To explore the emerging use of immune checkpoint inhibitors (ICIs) in hepatocellular carcinoma (HCC) patients eligible for liver transplantation (LT), particularly as bridging and downstaging therapies. This review also addresses the clinical challenges of integrating ICIs into transplant protocols, including graft rejection, immune-related toxicities, and gaps in evidence. RECENT FINDINGS ICIs have shown potential as bridging and downstaging therapies before LT, with multicentric studies reporting 75.6% successful downstaging, 85% 3-year post-LT survival, and 7.2% rejection-related mortality. A washout interval >94 days and older age have been identified as protective factors against allograft rejection. Combining locoregional therapies with ICIs has proven effective in the EMERALD-1 and LEAP-012 trials, which demonstrated improved progression-free survival (15.0 and 14.6 months, respectively) with ICI-TACE combinations. Similarly, the STAR-FIT phase II trial, combining TACE, SBRT, and avelumab, showed a 42% complete response rate and 12% conversion to curative therapy. Toxicity and rejection risk remain major challenges. SUMMARY ICIs represent a promising tool for expanding transplant eligibility in HCC, but their integration into LT pathways remains complex. Safety concerns, particularly regarding timing and immune modulation, require careful evaluation. Prospective studies and biomarker development are needed to guide clinical decision-making. Novel therapies such as CAR-T cells may offer more targeted approaches in the future.
Collapse
Affiliation(s)
- Rebecca Marino
- Liver Transplant and Hepatobiliary Surgery, Recanati-Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy, New York, New York, USA
| | | | | | | |
Collapse
|
11
|
Wu Z, Shan Q, Jiang Y, Huang W, Wang Z, Zhuang Y, Liu J, Li T, Yang Z, Li C, Wei T, Wen C, Cui W, Qiu Z, Liu X, Wang Z. Irreversible electroporation combined with PD-L1/IL-6 dual blockade promotes anti-tumor immunity via cDC2/CD4 +T cell axis in MHC-I deficient pancreatic cancer. Cancer Lett 2025; 617:217620. [PMID: 40068706 DOI: 10.1016/j.canlet.2025.217620] [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: 12/13/2024] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 03/15/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a "cold" solid tumor with frequent Major Histocompatibility Complex I (MHC-I) deficiency, thereby making it resistant to type-1-conventional dendritic cell (cDC1)-CD8+T cell mediated anti-tumor immunity. Current studies have demonstrated the emerging compensatory role of MHC-II-mediated antigen presentation and CD4+T cell activation in anti-tumor immunity against MHC-I-deficient tumors. However, the underlying mechanism of the compensatory immune response by CD4+T cells in cancer ablation therapy remains to be elucidate. In clinical samples and murine models, we observed that irreversible electroporation (IRE) ablation therapy promoted immune infiltration and the conversion of CD4+T cells into anti-tumor IFN-γ+Th1 cells and Th17 cells in MHC-I low-expressed PDAC using scRNA-seq and flow-cytometry analyses. Furthermore, we found that PD-L1 blockade predominantly enhanced the activation of CD11b+CD103-type-2 conventional dendritic cells (cDC2s) and their antigen presentation to CD4+T cells after ablation, stimulating the anti-tumor immune response through the tumor antigen-specific IFN-γ+Th1-NK cell axis. Elevated plasma levels of IL-6 in pancreatic cancer patients receiving ablation therapy are significant indicators for impaired prognosis. IL-6 and PD-L1 dual blockade could significantly augment the ratio of IFN-γ+Th1 in CD4+T cells to boost the anti-tumor immunity of NK cells, leading to prolonged survival of mouse bearing pancreatic cancer. Collectively, we have elucidated that PD-L1 blockade activates the cDC2-CD4+T cell axis after IRE therapy, thereby playing a pivotal compensatory anti-tumor role in MHC-I low-expressed pancreatic cancer. Moreover, a combination strategy involving dual-target blockade of PD-L1/IL-6 along with ablation therapy could emerge as a novel therapeutic approach for MHC-I deficient tumors.
Collapse
Affiliation(s)
- Zhuozhuo Wu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Qungang Shan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Yuyue Jiang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Wei Huang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Ziyin Wang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Yaping Zhuang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Jingjing Liu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Tiankuan Li
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Ziyu Yang
- Faculty of Medical Imaging Technology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Chaojie Li
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China; Department of Radiology, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, No.149, South Chongqing Road, Shanghai, 200025, China
| | - Tao Wei
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, China
| | - Chenlei Wen
- Department of Pancreatic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Zilong Qiu
- Songjiang Research Institute, Institute of Autism & MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China, No.748, Middle Zhongshan Road, Shanghai, 200025, China
| | - Xiaoyu Liu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China; Faculty of Medical Imaging Technology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Zhongmin Wang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China; Faculty of Medical Imaging Technology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China; Department of Radiology, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, No.149, South Chongqing Road, Shanghai, 200025, China
| |
Collapse
|
12
|
Jia H, Bian Y, Yuan J, Zhang Y, Zhang S. The Potential Role of C4 MYH11+ Fibroblasts and the MDK-SDC2 Ligand-Receptor Pair in Lung Adenocarcinoma: Implications for Prognosis and Therapeutic Strategies. Transl Oncol 2025; 55:102364. [PMID: 40121996 PMCID: PMC11982484 DOI: 10.1016/j.tranon.2025.102364] [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: 11/05/2024] [Revised: 03/09/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) posed a significant threat to global human health. This study employed single-cell RNA sequencing (scRNA-seq) to analyze transcriptomic data from nine LUAD patients at different stages of tumor infiltration, aiming to elucidate the tumor microenvironment and key biological processes of LUAD. METHODS In this study, we processed the scRNA-seq data using the Seurat package and sequentially applied principal component analysis followed by the Harmony package to effectively correct for batch effects, identifying 105,725 high-quality cells. Through cell clustering and gene expression profiling, we identified critical cell subpopulations and gene expression patterns in LUAD patients. RESULTS Our analysis revealed that the C4 MYH11+ Fibroblasts subtype was primarily involved in biological processes related to muscle function. Further investigations uncovered the MDK-SDC2 ligand-receptor pair as a critical regulator of tumor cell invasion, proliferation, and migration, driving LUAD progression. Additionally, we developed a gene-based prognostic model that effectively predicted patient survival, providing valuable clinical insights. CONCLUSION This study provided a comprehensive atlas of the LUAD tumor microenvironment, highlighted the role of the C4 MYH11+ Fibroblasts in tumor progression. It also proposed the MDK-SDC2 ligand-receptor pair as a novel mechanism, addressing a significant gap in this area of research. And presented a gene-based prognostic model as a novel perspective for research into immunotherapy and drug sensitivity in LUAD.
Collapse
Affiliation(s)
- Hongling Jia
- Department of Thoracic Surgery, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.; The first clinical medical college of Shandong university of Traditional Chinese Medicine, Jinan, China
| | - Yanjie Bian
- Xinxiang Medical University, Xinxiang, China
| | - Jie Yuan
- Sijing Town Community Healthcare Center, Shanghai, China
| | - Yi Zhang
- Department of Thoracic Surgery, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China..
| | - Shengyi Zhang
- Department of Thoracic Surgery, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China..
| |
Collapse
|
13
|
Ma Z, Wang Y, Wang W, Wei C, Liu Z, Li Z, Ye Y, Mao Y, Yuan Y, Huang Z, Zhang J, Cao Y, Mao X, Zhang Y, Jin X, Yin J, Li G, Zheng L, Liu Z, Li X, Liang X, Liu Z. Targeting VSIG4 + tissue-resident macrophages enhances T cell cytotoxicity and immunotherapy efficacy in cancer. Dev Cell 2025:S1534-5807(25)00249-7. [PMID: 40339578 DOI: 10.1016/j.devcel.2025.04.011] [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: 10/23/2024] [Revised: 02/23/2025] [Accepted: 04/16/2025] [Indexed: 05/10/2025]
Abstract
Tissue-resident macrophage (TRM) is crucial for organ development and homeostasis. However, the role of TRM-derived tumor-associated macrophage (TAM) subpopulations in cancer remains unclear. Using single-cell RNA sequencing and lineage tracing, we reported a TRM-derived TAM subpopulation, characterized by VSIG4 overexpression in testicular cancer. Macroscopically, such subpopulation was also found in tumors such as hepatocellular carcinoma, lung cancer, and glioblastoma. It was associated with poor prognosis and the suppression of CD8+ T-cell-dependent immunity via VSIG4. Notably, VSIG4 promoted immunosuppressive effects through direct or indirect modes, including interacting with receptors on CD8+ T cells or inducing transcription of IL-11 in TAMs. More importantly, MEF2C was identified as a key transcription factor that maintained VSIG4 expression and determined the biological behaviors of VSIG4+ TAMs. In preclinical models, targeting VSIG4+ TAMs via VSIG4 or MEF2C demonstrated a favorable effect of enhancing the efficacy of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Zikun Ma
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China.
| | - Yuzhao Wang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Weikai Wang
- BGI Research, Chongqing 401329, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen Wei
- BGI Research, Chongqing 401329, China
| | - Zhenhua Liu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Zhiyong Li
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Yunlin Ye
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Yize Mao
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Pancreatobiliary Surgery, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Yunfei Yuan
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Zhenkun Huang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Ji Zhang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Yun Cao
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Xiaopeng Mao
- Department of Urology, the First Affiliated Hospital of Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, China
| | - Yan Zhang
- BGI Research, Shenzhen 518083, China
| | - Xin Jin
- BGI Research, Shenzhen 518083, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Jianhua Yin
- BGI Research, Shenzhen 518083, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Guibo Li
- BGI Research, Chongqing 401329, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Limin Zheng
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiangdong Li
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China.
| | - Xiaoyu Liang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China.
| | - Zhuowei Liu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China; Sun Yat-sen University Cancer Center Gansu Hospital, Lanzhou 730050, China.
| |
Collapse
|
14
|
Ellert-Miklaszewska A, Pilanc P, Poleszak K, Roura AJ, Cyranowski S, Ghosh M, Baluszek S, Pasierbinska M, Gielniewski B, Swatler J, Hovorova Y, Wojnicki K, Kaminska B. 7aaRGD - a novel SPP1/integrin signaling-blocking peptide reverses immunosuppression and improves anti-PD-1 immunotherapy outcomes in experimental gliomas. J Exp Clin Cancer Res 2025; 44:132. [PMID: 40281508 PMCID: PMC12032770 DOI: 10.1186/s13046-025-03393-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) present clinical benefits in many cancer patients but invariably fail in glioblastoma (GBM), the most common and deadly primary brain tumor. The lack of ICIs efficacy in GBM is attributed to the accumulation of tumor-reprogrammed glioma-associated myeloid cells (GAMs) that create a "cold" immunosuppressive tumor microenvironment (TME), impeding the infiltration and activation of effector T cells. GBM-derived αvβ3/αvβ5-integrin ligands, including SPP1, were shown to mediate the emergence of GAMs. We hypothesized that a combination strategy aiming to block the reprogramming of GAMs using a synthetic 7aaRGD peptide that targets SPP1/integrin signaling might overcome resistance to ICIs and reinvigorate anti-tumor immunity. METHODS Matrigel invasion assay was used to test the efficacy of 7aaRGD in glioma-microglia co-cultures. We determined the impact of 7aaRGD, administered as a monotherapy or combined with PD-1 blockade, on tumor growth, GAMs accumulation and phenotypes, arginase-1 levels and neovasculature in experimental gliomas. The effects of treatments on the tumor immune landscape were dissected using multiparameter flow cytometry, immunocytochemistry, cytokine profiling and RNA-seq analysis of sorted GAMs followed by CITE-seq based data deconvolution. RESULTS 7aaRGD efficiently blocked microglia-dependent invasion of human and mouse glioma cells in vitro. Intratumorally delivered 7aaRGD alone did not reduce tumor growth in orthotopic gliomas but prevented the emergence of immunosuppressive GAMs and led to normalization of peritumoral blood vessels. Combining 7aaRGD with anti-PD-1 antibody resulted in reduced tumor growth, with an increase in the number of proliferating, interferon-ɣ producing CD8+T cells and depletion of regulatory T cells. Transcriptomic profiles of myeloid cells were altered by the combined treatment, reflecting the restored "hot" inflammatory TME and boosted immunotherapy responses. Intratumoral administration of 7aaRGD similarly modified the phenotypes of GAMs in human U87-MG gliomas in immunocompromised mice. Exploration of transcriptomic datasets revealed that high expression of integrin receptor coding genes in pre-treatment biopsies was associated with a poorer response to immune check-point blockade in patients with several types of cancers. CONCLUSIONS We demonstrate that combining the blockade of SPP1/integrin signaling with ICIs modifies innate immunity and reinvigorates adaptive antitumor responses, which paves the way to improve immunotherapy outcomes in GBM.
Collapse
Affiliation(s)
| | - Paulina Pilanc
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Katarzyna Poleszak
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Adria-Jaume Roura
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Salwador Cyranowski
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Mitrajit Ghosh
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Szymon Baluszek
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Maria Pasierbinska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bartłomiej Gielniewski
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Julian Swatler
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Yuliana Hovorova
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Kamil Wojnicki
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland.
| |
Collapse
|
15
|
Lynch SM, Richards CE, Ui Mhaonaigh A, Lynam-Lennon N, Eustace AJ, Allott EH, Robson T, Marcone S. Translating Basic Science Discoveries into Clinical Advances: Highlights from the EACR-AACR-IACR 2024 Conference in Celebration of Irish Association for Cancer Research's 60th Anniversary. Cancers (Basel) 2025; 17:1420. [PMID: 40361346 PMCID: PMC12071098 DOI: 10.3390/cancers17091420] [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: 03/21/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025] Open
Abstract
The EACR-AACR-IACR 2024 Basic and Translational Research Conference, held in Dublin, Ireland, from 27th-29th February, 2024, marked a significant milestone as part of the 60th anniversary celebrations of the Irish Association for Cancer Research (IACR). Organized in collaboration with the European Association for Cancer Research (EACR) and the American Association for Cancer Research (AACR), this prestigious event brought together leading experts in oncology research from around the world. The conference provided a platform for cutting-edge discussions on the latest advancements in immunotherapy, drug combinations, cell-based therapies, liquid biopsies, epigenetics, tumour microenvironment, and novel drug targets. With keynote lectures from esteemed researchers such as Kevan Shokat, Jerome Galon, Suzanne Topalian, and Scott Lowe, the conference facilitated knowledge exchange and fostered international collaboration in the pursuit of improved cancer treatments. The report highlights the key sessions, research breakthroughs, and discussions that shaped this landmark event.
Collapse
Affiliation(s)
- Seodhna M. Lynch
- Personalised Medicine Centre, School of Medicine, Ulster University, C-TRIC Building, Altnagelvin Area Hospital, Glenshane Road, Londonderry BT47 6SB, UK;
| | - Cathy E. Richards
- School of Dentistry, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
| | - Aisling Ui Mhaonaigh
- Department of Surgery, Trinity Translational Medicine Institute, Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, D08 NHY1 Dublin, Ireland; (A.U.M.); (N.L.-L.)
| | - Niamh Lynam-Lennon
- Department of Surgery, Trinity Translational Medicine Institute, Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, D08 NHY1 Dublin, Ireland; (A.U.M.); (N.L.-L.)
- Department of Biology, Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 NPY6 Maynooth, Ireland
| | - Alex J. Eustace
- Life Science Institute, School of Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland;
| | - Emma H. Allott
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK;
| | - Tracy Robson
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
| | - Simone Marcone
- UCD School of Biology and Environmental Science, University College Dublin, D04 N2E5 Dublin, Ireland;
| |
Collapse
|
16
|
Guan J, Gong X, Zeng H, Zhang W, Qin Q, Gou H, Liu X, Song B. Gastrointestinal tumor personalized immunotherapy: an integrated analysis from molecular genetics to imaging biomarkers. Therap Adv Gastroenterol 2025; 18:17562848251333527. [PMID: 40297204 PMCID: PMC12035075 DOI: 10.1177/17562848251333527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
The immunotherapy landscape for gastrointestinal (GI) tumors is rapidly evolving. There is an urgent need for reliable biomarkers capable of predicting treatment outcomes to optimize therapeutic strategies and enhance patient prognosis. This review presents a comprehensive overview of biomarkers associated with the immunotherapy response of GI tumors, covering advances in molecular genetics, histopathological markers, and imaging. Key molecular biomarkers, such as microsatellite instability, tumor mutational burden, and programmed death-ligand 1 expression, remain critical for identifying patients likely to benefit from immune checkpoint inhibitors. The significance of tumor-infiltrating lymphocytes, notably the CD8+ T cell to regulatory T cell ratio, as a predictor of immunotherapy response is explored. In addition, advanced imaging techniques, including computed tomography (CT), magnetic resonance imaging, and positron emission tomography-CT, facilitate the noninvasive evaluation of tumor biology and therapeutic response. By bridging molecular and imaging data, this integrated strategy enhances precision in patient selection, treatment monitoring, and adaptive therapy design. Future studies should aim to validate these biomarkers in larger, multicenter cohorts and focus on clinical translation to advance precision medicine in GI oncology.
Collapse
Affiliation(s)
- Jian Guan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’s Armed Police Forces, Leshan, China
| | - Xiaoling Gong
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hanjiang Zeng
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’s Armed Police Forces, Leshan, China
| | - Qing Qin
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongfeng Gou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xijiao Liu
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya, Hainan, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya, Hainan, China
| |
Collapse
|
17
|
Wang Y, Chen Z, Liang K, Wang W, Hu Z, Mao Y, Liang X, Jiang L, Liu Z, Ma Z. AGO2 mediates immunotherapy failure via suppressing tumor IFN-gamma response-dependent CD8 + T cell immunity. Cell Rep 2025; 44:115445. [PMID: 40106436 DOI: 10.1016/j.celrep.2025.115445] [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: 08/22/2024] [Revised: 11/28/2024] [Accepted: 02/27/2025] [Indexed: 03/22/2025] Open
Abstract
Interferon-gamma (IFN-γ), a cytokine essential for activating cellular immune responses, plays a crucial role in cancer immunosurveillance and the clinical success of immune checkpoint blockade therapy. In this study, we show that Argonaute 2 (AGO2), a key mediator in small RNA-guided gene regulation, inversely correlates with tumor responsiveness to IFN-γ and the efficacy of immunotherapy. Mechanistically, IFN-γ upregulates miR-1246 expression in tumor cells, enhancing its interaction with AGO2. This miR-1246-AGO2 complex disrupts IFN-γ-mediated signal transducer and activator of transcription 1 (STAT1) phosphorylation by stabilizing protein tyrosine phosphatase non-receptor 6 (PTPN6) mRNA, thereby suppressing the expression of downstream C-X-C motif chemokine ligands (CXCLs), IFN-stimulated genes (ISGs), and human leukocyte antigen (HLA) molecules, which collectively contribute to tumor immune evasion. In preclinical cancer models, inhibiting AGO2 with BCI-137 or targeting miR-1246 with its antagomir re-sensitizes tumor cells to IFN-γ, leading to the enhanced recruitment, activation, and cytotoxicity of CD8+ T cells and ultimately improving immunotherapy efficacy.
Collapse
Affiliation(s)
- Yuzhao Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zibin Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ke Liang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Weikai Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhihao Hu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yize Mao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Pancreatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xiaoyu Liang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Lijuan Jiang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Zhuowei Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Sun Yat-sen University Cancer Center Gansu Hospital, Lanzhou 730050, China.
| | - Zikun Ma
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
18
|
Wang SL, Chan TA. Navigating established and emerging biomarkers for immune checkpoint inhibitor therapy. Cancer Cell 2025; 43:641-664. [PMID: 40154483 DOI: 10.1016/j.ccell.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/19/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have improved outcomes of patients with many different cancers. These antibodies target molecules such as programmed cell death 1 (PD-1) or cytotoxic T lymphocyte associated protein 4 (CTLA-4) which normally function to limit immune activity. Treatment with ICIs reactivates T cells to destroy tumor cells in a highly specific manner, which in some patients, results in dramatic remissions and durable disease control. Over the last decade, much effort has been directed at characterizing factors that drive efficacy and resistance to ICI therapy. Food and Drug Administration (FDA)-approved biomarkers for ICI therapy have facilitated more judicious treatment of cancer patients and transformed the field of precision oncology. Yet, adaptive immunity against cancers is complex, and newer data have revealed the potential utility of other biomarkers. In this review, we discuss the utility of currently approved biomarkers and highlight how emerging biomarkers can further improve the identification of patients who benefit from ICIs.
Collapse
Affiliation(s)
- Stephen L Wang
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; National Center for Regenerative Medicine, Cleveland, OH, USA.
| |
Collapse
|
19
|
Hu H, Zhang M. PD-1 involvement in CD8+ tumor-infiltrating lymphocytes in patients with colonic-derived peritoneal adenocarcinoma. Braz J Med Biol Res 2025; 58:e14467. [PMID: 40243819 PMCID: PMC11996165 DOI: 10.1590/1414-431x2025e14467] [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: 09/02/2024] [Accepted: 03/03/2025] [Indexed: 04/18/2025] Open
Abstract
Immune checkpoint blockade with anti-programmed cell death protein 1 (PD-1) antibody has become a hot topic for the treatment of human malignancies. Here, we aimed to investigate whether the percentage of PD-1 in CD8+ tumor-infiltrating lymphocytes correlates with the progression of colonic-derived peritoneal adenocarcinoma (PA). Peripheral blood and tissue samples from 40 patients with colonic-derived PA were collected and subjected to multicolor flow cytometry analysis of the percentage of peripheral PD-1+CD8+ T cells. The multiple immunofluorescence method was used to detect the positive percentages of PD-1 and CD8 in the tissues. The enrolled patients were divided into groups by recurrence interval (less than 6 months, greater than two years) and differentiation grade (low, well/moderate). In the colonic-derived PA tissues, the percentages of cells positive for PD-1, CD8, and PD-1+CD8+ were higher in the paracancer tissues compared with cancerous tissues. PD-1+CD8+ T cells had an increased presence in peripheral blood than in tissues. Our data also indicated that colonic-derived PA patients with less than a six-month recurrence interval presented higher levels of PD-1 in CD8+ tumor-infiltrating lymphocytes in than the two-year recurrence group. The level of PD-1+CD8+T cells in the tissue correlated with the clinical outcome of colonic-derived PA. Higher percentages of PD-1+CD8+T cells correlated with a shorter progression-free survival (PFS). PD-1 in CD8+ tumor-infiltrating lymphocytes may have a good predictive value for immunotherapy of colonic-derived PA and act as the prognostic factor for PFS.
Collapse
Affiliation(s)
- Huihui Hu
- Department of Clinical Laboratory, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Man Zhang
- Department of Clinical Laboratory, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, China
- Clinical Laboratory Medicine, Peking University Ninth School of Clinical Medicine, Beijing, China
| |
Collapse
|
20
|
Su P, Han Y, Yi J, Hou Y, Xiao Y. Research status and frontiers in liver cancer immunotherapy: a bibliometric perspective on highly cited literature. Front Oncol 2025; 15:1587252. [PMID: 40276056 PMCID: PMC12018336 DOI: 10.3389/fonc.2025.1587252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 03/14/2025] [Indexed: 04/26/2025] Open
Abstract
Background Liver cancer is one of the major causes of cancer-related death in the world. As a breakthrough therapy, immunotherapy had significantly improved the prognosis of patients. However, the current research status and research hotspots in the field of liver cancer immunotherapy still lack systematic review. Based on the bibliometric analysis of highly cited papers, this study intended to reveal the current research status, research hotspots and future research trends in this field. Objective The purpose of this study was to analyze the national/regional contributions, authors and institutions cooperation network, keywords clustering and keywords burst analysis of highly cited papers on liver cancer immunotherapy through bibliometrics, so as to clarify the research frontier and development direction, and provide objective data support for future research direction and clinical practice. Methods The highly cited papers on liver cancer immunotherapy from the Web of Science core collection up to February 23, 2025 were retrieved, and 232 studies were included. CiteSpace was used to build a knowledge map, analyze the distribution of years, countries, authors, institutions and cooperation networks, and identify research hotspots and emerging trends through keyword clustering and burst detection. Results The number of highly cited papers continued to increase from 2014 and reached a peak in 2022. China and the United States had the highest number of publications and the centrality of cooperation networks. The author with the highest number of papers was Llovet, Josep M, whose research direction mainly focused on immune checkpoint inhibitor combination therapy and molecular typing. The author with the highest cooperation network centrality was Duda, Dan G, whose research team focused on tumor microenvironment regulation. Harvard University and the University of Barcelona played an important central role in the institutional collaboration. Keywords analysis showed that immune checkpoint inhibitors, tumor microenvironment and combination therapy were the core of liver cancer immunotherapy. Burst keywords such as cell lung cancer, pembrolizumab, advanced melanoma, blockade, lymphocytes, etc. had revealed the research frontier of liver cancer immunotherapy research. Conclusion The research on liver cancer immunotherapy had made multi-dimensional progress, with China and the United States leading the global cooperation. The main research directions were the combination strategy of immunization, the regulation of tumor microenvironment and the exploration of novel targets. In the future, it is necessary to optimize treatment resistance solutions, integrate interdisciplinary resources, and promote the development of precision and personalized treatment.
Collapse
Affiliation(s)
- Pan Su
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital, Central South University, Changsha, China
| | - Yeqiong Han
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital, Central South University, Changsha, China
| | - Jindong Yi
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Hou
- Department of Pulmonology, Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Xiao
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- International Joint Research Center of Minimally Invasive Endoscopic Technology Equipment & Standards, Xiangya Hospital, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
21
|
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
|
22
|
Zhang C, Wu P, Li D, Zhou J, Lin C, Gu X, Shang D, Ma R, Liu J, Zhang G, Wang P, Che Y, Zeng Q, Peng J, Zhao B, Sun N, He J. Efficacy and safety of anti-PD-1 versus anti-PD-L1 in perioperative immunotherapy: A comprehensive reanalysis of randomized controlled trials. MED 2025:100669. [PMID: 40233750 DOI: 10.1016/j.medj.2025.100669] [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: 11/29/2024] [Revised: 12/16/2024] [Accepted: 03/13/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND Perioperative anti-programmed cell death 1 (PD-1)/PD ligand 1 (PD-L1) immune checkpoint inhibitors improve outcomes, but optimal selection between agents remains debated. We compared the efficacy and safety of anti-PD-1 versus anti-PD-L1 in neoadjuvant/adjuvant settings. METHODS PubMed, Embase, Cochrane CENTRAL, and major oncology conferences (up to May 20, 2024) were systematically searched for randomized trials comparing anti-PD-1/PD-L1 with standard perioperative therapy. Data extraction followed PRISMA guidelines, including trial characteristics, efficacy outcomes (pathological response and survival outcome), and safety profiles. Indirect comparisons between agents were conducted through network meta-analysis employing the mirror principle, utilizing both frequentist and Bayesian methodologies. FINDINGS Thirty-one trials (14,974 patients) were analyzed. Anti-PD-1 demonstrated superior pathological complete response (relative risk [RR]: 1.65, 95% confidence interval [CI]: 1.18-2.29, p = 0.003), major pathological response (RR: 1.43, 95% CI: 1.04-1.96, p = 0.026), and disease-free survival (hazard ratio [HR] = 0.82, 95% CI: 0.71-0.96, p = 0.0106) versus anti-PD-L1. Safety profiles were comparable overall, though anti-PD-1 correlated with higher grade 3-5 immune-related adverse events (irAEs). Frequentist and Bayesian analyses yielded consistent results. CONCLUSIONS Perioperative anti-PD-1 therapy shows enhanced efficacy but increased severe irAEs compared to anti-PD-L1, supporting agent-specific considerations in clinical practice. Further tumor-specific evaluations and mature data are warranted. FUNDING This work is supported in part by the CAMS Innovation Fund for Medical Sciences (2024-I2M-ZD-004) and so on.
Collapse
Affiliation(s)
- Chaoqi Zhang
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Peng Wu
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Dongyu Li
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Junhan Zhou
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Chuqi Lin
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Xuanyu Gu
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Dexin Shang
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Ruijie Ma
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Jingjing Liu
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Guochao Zhang
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Pan Wang
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Yun Che
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Qingpeng Zeng
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Jilin Peng
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Bohui Zhao
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Nan Sun
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
23
|
Jinushi K, Saito T, Kurose K, Suzuki S, Kojima T, Takahara T, Makino T, Ogawa T, Nishikawa H, Kakimi K, Iida S, Nakajima J, Doki Y, Oka M, Ueda R, Wada H. Phase I study on neoadjuvant combination immunotherapy with mogamulizumab and nivolumab for solid tumors. J Immunother Cancer 2025; 13:e010634. [PMID: 40180420 PMCID: PMC11966984 DOI: 10.1136/jitc-2024-010634] [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: 10/07/2024] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Effector regulatory T cells expressing C-C chemokine receptor 4 (CCR4) suppress antitumor immune responses. We conducted a phase I clinical trial to evaluate the safety and efficacy of preoperative combination therapy with mogamulizumab (an anti-CCR4 antibody) and nivolumab (an anti-programmed death-1 antibody) in patients with solid tumors. METHODS Patients with operable solid tumors were enrolled in a 3+3 design, with preoperative nivolumab (3.0 mg/kg) administered intravenously every 2 weeks three times and mogamulizumab at 0.1 mg/kg (cohort 1), 0.3 mg/kg (cohort 2), or 1.0 mg/kg (cohort 3) every week four times. The primary endpoints were safety and the effects of depleting Forkhead box P3+ (FoxP3+) T cells in the tumor. RESULTS 16 patients were enrolled between June 2016 and April 2020, including those with renal (n=7), lung (n=5), esophageal (n=3), and oral (n=1) cancers. Grade 3-4 treatment-related adverse events were observed in 6 of 16 patients, with lymphopenia (25%) and maculopapular rash (13%) being the most frequent. Grade 5 interstitial pneumonia was observed in one patient; however, the cause of death was disease progression. There were three partial responses (PRs) (one lung and two esophageal cancers) among clinical responses and one complete response (one lung cancer) and nine PRs (five kidney, two lung, and two esophageal cancers) among pathological responses. CCR4+FoxP3+ T cells were depleted in the tumors of all patients and increases in lymphocytes in tumor tissue according to the tumor immune microenvironment classification were observed in 50% of the patients, which correlated with a better prognosis. CONCLUSIONS The preoperative combination of mogamulizumab and nivolumab was safely managed, exerted antitumor effects, and may be an effective option in the preoperative setting. TRIAL REGISTRATION NUMBER The present study was registered with ClinicalTrials.gov as NCT02946671 (registration date 2016-10-05).
Collapse
Affiliation(s)
- Koichi Jinushi
- Department of Gastroenterological Surgery, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka Prefecture, Japan
- Department of Clinical Research in Tumor Immunology, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka Prefecture, Japan
| | - Takuro Saito
- Department of Gastroenterological Surgery, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka Prefecture, Japan
- Department of Clinical Research in Tumor Immunology, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka Prefecture, Japan
| | - Koji Kurose
- Department of Respiratory Medicine, Kawasaki Medical School, Kurashiki, Japan
| | - Susumu Suzuki
- Research Creation Support Center, Aichi Medical University, Nagakute, Aichi, Japan
- Tumor Immunology, Aichi Medical University, Nagakute, Aichi, Japan
| | - Takashi Kojima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center-Hospital East, Kashiwa, Chiba, Japan
| | - Taishi Takahara
- Department of Surgical Pathology, Aichi Medical University Hospital, Nagakute, Aichi Prefecture, Japan
| | - Tomoki Makino
- Department of Gastroenterological Surgery, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka Prefecture, Japan
| | - Tetsuya Ogawa
- Department of Otorhinolaryngology-Head and Neck Surgery, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, National Cancer Center Japan, Chuo, Tokyo, Japan
- Department of Immunology, Nagoya University Graduate School of Medicine Faculty of Medicine, Nagoya, Aichi Prefecture, Japan
- Division of Cancer Immune Multicellular System Regulation, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, Kyoto, Japan
| | - Kazuhiro Kakimi
- Department of Immunology, Kindai University Faculty of Medicine Graduate School of Medical Sciences, sayama, Osaka, Japan
- Department of Immunotherapeutics, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences and Medical School, Nagoya, Aichi Prefecture, Japan
| | - Jun Nakajima
- Department of Thoracic Surgery, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka Prefecture, Japan
| | - Mikio Oka
- Department of Immuno-Oncology, Kawasaki Medical School, Kurashiki, Japan
| | - Ryuzo Ueda
- Department of Immunology, Nagoya University Graduate School of Medicine Faculty of Medicine, Nagoya, Aichi Prefecture, Japan
| | - Hisashi Wada
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
24
|
De Silva P, Saad MA, Swain JWR, Mai Z, Kidd MD, Choe JJ, Camargo AP, Anand S, Chandrasekhara V, Pogue BW, Wang KK, Spring BQ, Maytin EV, Hasan T. Photodynamic priming with red light triggers adaptive immune responses in a pancreatic cancer mouse model. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2025; 265:113126. [PMID: 40007355 PMCID: PMC11895200 DOI: 10.1016/j.jphotobiol.2025.113126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025]
Abstract
The poor response of pancreatic ductal adenocarcinoma (PDAC) to treatment, including immunotherapy, is attributed to its tumor microenvironment (TME). An ongoing challenge is the desmoplastic and immunosuppressed TME that evades immune surveillance. Here, we investigate transient modulation of the TME to overcome immunosuppression using a light-activated process, termed photodynamic priming (PDP). As a first step, this study captures the temporal dynamics of variations in immune infiltrates and subsequent immune responses in the TME, spleen, and blood of the KPC mouse model of PDAC post-PDP. In response to PDP, there were transient increases in tumor infiltrating lymphocytes (TIL) in tumors. The TIL population post-PDP includes an enrichment of CD8+ T cells, accompanied by temporal increases in PD-1, CTLA-4, and TIM-3 immune checkpoints on both CD8+ T and CD4+ T cells. Significant increases in CD11C+MHC-11+ dendritic cells and proliferating lymphocytes are observed in the spleen within several hours post-tumor PDP, suggesting initiation of adaptive immune responses. These observations are followed by an expansion of CD44+CD62-CD8+ effector memory T cells in the blood over several days as evidence of a systemic immune response. Post-PDP TME alterations also included the reduced formation of blood (CD31+) and lymphatic (Lyve-1+) vessels as well as decreases in PD-L1 and collagen content. Collectively, these data suggest that PDP helps to mitigate immunosuppressive mechanisms and promote enhanced tumor permeability. The temporal dynamics of the processes elucidated here pave the way to develop strategies in future work for combined PDP-immunotherapy utilizing the immune checkpoint expression dynamics for precision therapy.
Collapse
Affiliation(s)
- Pushpamali De Silva
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | - Mohammad A Saad
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | - Joseph W R Swain
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | - Zhiming Mai
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | - Madeline D Kidd
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA; Harvard University, Cambridge, MA, USA
| | - Joanna J Choe
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA; Boston College, Chestnut Hill, MA, USA
| | - Assiris P Camargo
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA; Bunker Hill Community College, Boston, MA, USA
| | - Sanjay Anand
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Brian W Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Kenneth K Wang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Bryan Q Spring
- Department of Physics, College of Science, Northeastern University, Boston, USA
| | - Edward V Maytin
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA; Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
25
|
Hua Q, Li Z, Weng Y, Wu Y, Zheng L. Myeloid cells: key players in tumor microenvironments. Front Med 2025; 19:265-296. [PMID: 40048137 DOI: 10.1007/s11684-025-1124-8] [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: 08/14/2024] [Accepted: 12/16/2024] [Indexed: 05/04/2025]
Abstract
Cancer is the result of evolving crosstalk between neoplastic cell and its immune microenvironment. In recent years, immune therapeutics targeting T lymphocytes, such as immune checkpoint blockade (ICB) and CAR-T, have made significant progress in cancer treatment and validated targeting immune cells as a promising approach to fight human cancers. However, responsiveness to the current immune therapeutic agents is limited to only a small proportion of solid cancer patients. As major components of most solid tumors, myeloid cells played critical roles in regulating the initiation and sustentation of adaptive immunity, thus determining tumor progression as well as therapeutic responses. In this review, we discuss emerging data on the diverse functions of myeloid cells in tumor progression through their direct effects or interactions with other immune cells. We explain how different metabolic reprogramming impacts the characteristics and functions of tumor myeloid cells, and discuss recent progress in revealing different mechanisms-chemotaxis, proliferation, survival, and alternative sources-involved in the infiltration and accumulation of myeloid cells within tumors. Further understanding of the function and regulation of myeloid cells is important for the development of novel strategies for therapeutic exploitation in cancer.
Collapse
Affiliation(s)
- Qiaomin Hua
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zhixiong Li
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yulan Weng
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yan Wu
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Limin Zheng
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
26
|
Anders MM, Mattos AZ, Debes JD, Beltran O, Coste P, Marín JI, Chagas AL, Menéndez J, Estupiñan EC, Ferrer JD, Mattos AA, Piñero F. Latin American expert opinion letter on the feasibility of systemic therapies in combination with locoregional therapies for hepatocellular carcinoma. Ann Hepatol 2025; 30:101905. [PMID: 40122521 DOI: 10.1016/j.aohep.2025.101905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/26/2024] [Accepted: 01/10/2025] [Indexed: 03/25/2025]
Abstract
Recent advances in the systemic treatment of advanced hepatocellular carcinoma (HCC) with immunotherapy have once again reignited discussion over the role of combined therapy in earlier stages. This year, different international meetings have presented recent results from clinical trials on adjuvant therapy alone (IMBrave-050) and combined with transarterial chemoembolization (EMERALD-1 and LEAP-12). Increased enthusiasm for the use of adjuvant and neoadjuvant therapy for liver transplantation, surgery, and local-regional treatment of HCC has been shown. However, the initial results from these trials should be interpreted cautiously as we wait for final analyses and effects on overall survival. In this position paper from the special interest group from the Latin American Association for the Study of Liver Diseases (ALEH), we underline the caveats of the applicability of these potential treatments in our region, explore points of agreement, and highlight areas of uncertainty. Moreover, we underscore the role of hepatologists in the clinical decision-making process and management of these patients.
Collapse
Affiliation(s)
| | - Angelo Z Mattos
- Graduate Program in Medicine: Hepatology. Federal University of Health Sciences of Porto Alegre, Brazil
| | - José D Debes
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | | - Pablo Coste
- Programa Nacional de Trasplante Hepático, Hospital R.A. Calderón Guardia, Costa Rica
| | | | - Aline Lopes Chagas
- Division of Clinical Gastroenterology and Hepatology, Hospital das Clínicas, Department of Gastroenterology, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Josemaría Menéndez
- Programa Nacional de Trasplante Hepático, Hospital Militar, Montevideo, Uruguay
| | - Enrique Carrera Estupiñan
- Hospital Eugenio Espejo, Departamento de Gastroenterología. Universidad San Francisco de Quito, Ecuador
| | | | - Angelo A Mattos
- Graduate Program in Medicine: Hepatology. Federal University of Health Sciences of Porto Alegre, Brazil
| | - Federico Piñero
- Hospital Universitario Austral, Austral University, School of Medicine, Buenos Aires, Argentina
| |
Collapse
|
27
|
Zhou L, Lian G, Zhou T, Cai Z, Yang S, Li W, Cheng L, Ye Y, He M, Lu J, Deng Q, Huang B, Zhou X, Lu D, Zhi F, Cui J. Palmitoylation of GPX4 via the targetable ZDHHC8 determines ferroptosis sensitivity and antitumor immunity. NATURE CANCER 2025:10.1038/s43018-025-00937-y. [PMID: 40108413 DOI: 10.1038/s43018-025-00937-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 02/27/2025] [Indexed: 03/22/2025]
Abstract
Ferroptosis is closely linked with various pathophysiological processes, including aging, neurodegeneration, ischemia-reperfusion injury, viral infection and, notably, cancer progression; however, its post-translational regulatory mechanisms remain incompletely understood. Here we revealed a crucial role of S-palmitoylation in regulating ferroptosis through glutathione peroxidase 4 (GPX4), a pivotal enzyme that mitigates lipid peroxidation. We identified that zinc finger DHHC-domain containing protein 8 (zDHHC8), an S-acyltransferase that is highly expressed in multiple tumors, palmitoylates GPX4 at Cys75. Through small-molecule drug screening, we identified PF-670462, a zDHHC8-specific inhibitor that promotes the degradation of zDHHC8, consequently attenuating GPX4 palmitoylation and enhancing ferroptosis sensitivity. PF-670462 inhibition of zDHHC8 facilitates the CD8+ cytotoxic T cell-induced ferroptosis of tumor cells, thereby improving the efficacy of cancer immunotherapy in a B16-F10 xenograft model. Our findings reveal the prominent role of the zDHHC8-GPX4 axis in regulating ferroptosis and highlight the potential application of zDHHC8 inhibitors in anticancer therapy.
Collapse
Affiliation(s)
- Liang Zhou
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences of Sun Yat-sen University, Guangzhou, China
| | - Guangyu Lian
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences of Sun Yat-sen University, Guangzhou, China
| | - Tao Zhou
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences of Sun Yat-sen University, Guangzhou, China
| | - Zhe Cai
- Guangzhou Institute of Pediatrics, Department of Allergy, Immunology and Rheumatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Shuai Yang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences of Sun Yat-sen University, Guangzhou, China
| | - Weining Li
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences of Sun Yat-sen University, Guangzhou, China
| | - Lilin Cheng
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences of Sun Yat-sen University, Guangzhou, China
| | - Ying Ye
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingfeng He
- Department of Oncology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianru Lu
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences of Sun Yat-sen University, Guangzhou, China
| | - Qifeng Deng
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences of Sun Yat-sen University, Guangzhou, China
| | - Bihui Huang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xiaoqian Zhou
- Department of Gastrointestinal Surgery, The First People's Hospital of Gui Yang, Gui Yang, China
| | - Desheng Lu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Cancer Research Center, Department of Pharmacology, Shenzhen University Medical School, Shenzhen, China
| | - Feng Zhi
- Department of Neurosurgery, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
28
|
Smeenk MM, van Diessen JN, Boellaard TN, Hartemink KJ, de Vries JF, van der Noort V, Badrising SK, Owers EC, Monkhorst K, van den Heuvel MM, Theelen WS. Tremelimumab plus Durvalumab prior to Chemoradiotherapy in Unresectable, Locally Advanced Non-Small Cell Lung Cancer: The Induction Trial. Clin Cancer Res 2025; 31:1037-1046. [PMID: 39821070 PMCID: PMC11911803 DOI: 10.1158/1078-0432.ccr-24-3476] [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: 10/22/2024] [Revised: 12/05/2024] [Accepted: 01/14/2025] [Indexed: 01/19/2025]
Abstract
PURPOSE The phase I induction trial (NCT04287894) assessed the feasibility and safety of induction immunotherapy (IIT) prior to concurrent chemoradiotherapy (cCRT) in patients with locally advanced non-small cell lung cancer (NSCLC). PATIENTS AND METHODS Patients with unresectable stage II/III NSCLC were eligible for inclusion. Patients received either one cycle of tremelimumab (75 mg) with two cycles of durvalumab (1,500 mg) in cohort I, one cycle of tremelimumab (300 mg) with two cycles of durvalumab in cohort II, or one cycle of tremelimumab (300 mg) with one cycle of durvalumab in cohort III. After IIT, a comprehensive radiological and pathological restaging was performed followed by cCRT. The combined primary endpoint was the feasibility and safety of IIT-cCRT. RESULTS Fifteen of 17 included patients were treated per protocol. IIT-cCRT was completed in 13 of the 15 patients within the predefined feasibility criteria. Grade ≥3 immune-related adverse events occurred in seven of the 15 patients, of which six were treated in the high-dose tremelimumab cohorts, thereby violating the safety criteria in cohorts II and III. The low-dose tremelimumab cohort (I) complied with safety criteria. Eleven patients had multilevel N2 or N3 disease at baseline; eight of these patients were downstaged to either N0/N1 or single-level N2 after IIT. Multiparametric MRI accurately identified nodal downstaging in all seven patients. CONCLUSIONS Induction with high-dose tremelimumab plus durvalumab prior to cCRT in unresectable locally advanced NSCLC was associated with unacceptable toxicity, although IIT resulted in clinically relevant nodal downstaging in eight of the 11 patients with baseline multilevel N2 or N3 disease. Multiparametric MRI showed potential for evaluating treatment response.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/therapy
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/mortality
- Female
- Male
- Middle Aged
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Lung Neoplasms/drug therapy
- Lung Neoplasms/mortality
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Aged
- Chemoradiotherapy/methods
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Adult
- Neoplasm Staging
- Treatment Outcome
Collapse
Affiliation(s)
- Michiel M. Smeenk
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Judi N.A. van Diessen
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Thierry N. Boellaard
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Koen J. Hartemink
- Department of Surgery, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Thoracic Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jeltje F. de Vries
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Sushil K. Badrising
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Emilia C. Owers
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Kim Monkhorst
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | | |
Collapse
|
29
|
Ning L, Li X, Xu Y, Si Y, Zhao H, Ren Q. Multi-Omics Analysis Revealed That TAOK1 Can Be Used as a Prognostic Marker and Target in a Variety of Tumors, Especially in Cervical Cancer. Onco Targets Ther 2025; 18:335-353. [PMID: 40109409 PMCID: PMC11920640 DOI: 10.2147/ott.s506582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Background Thousand and One Kinase 1 (TAOK1), a member of the MAPK kinase family, plays a crucial role in processes like microtubule dynamics, DNA damage response, and neurodevelopment. While TAOK1 is linked to tumorigenesis, its oncogenic role across cancers remains unclear. This study aims to explore the relationship between TAOK1 expression, prognosis, and immune function in various cancers. Methods We analyzed TAOK1 expression in multiple cancers using TCGA, GEO, CCLE, and other bioinformatics databases. The correlation between TAOK1 expression and immune cell infiltration was assessed with the ESTIMATE algorithm. We also examined associations with tumor stemness, DNA methylation, gene copy number alterations, and drug sensitivity. The oncogenic role of TAOK1 was further evaluated in vitro with SiHa and A2780 cells and in vivo with TAOK1 overexpression in SiHa cells. Results TAOK1 is a key prognostic biomarker in various cancers and its high expression is associated with poor prognosis. It showed a significant negative correlation with immune cell infiltration and immune checkpoints. GSEA identified its involvement in key tumour pathways, highlighting the therapeutic potential of inhibiting the TAOK1 gene. The high expression of TAOK1 is associated with DNA methylation and gene copy number variation, and in addition its upstream regulator, EP300, is closely associated with TAOK1 expression. In vitro cellular experiments demonstrated that inhibition of TAOK1 reduced the proliferation of SiHa and A2780 cells, whereas overexpression of TAOK1 in SiHa cells promoted growth. These findings were further validated in vivo by nude mouse tumourigenicity assay and human tissue immunohistochemistry. Conclusion TAOK1 serves as a promising prognostic biomarker and potential therapeutic target, especially for cervical cancer. These results support its clinical potential in cancer prognosis and treatment strategies.
Collapse
Affiliation(s)
- Li Ning
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, People's Republic of China
| | - Xiu Li
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, People's Republic of China
| | - Yating Xu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, People's Republic of China
| | - Yu Si
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, People's Republic of China
| | - Hongting Zhao
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, People's Republic of China
| | - Qingling Ren
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
- The Chinese Clinical Medicine Innovation Center of Obstetrics, Gynecology, and Reproduction in Jiangsu Province, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
30
|
Liu J, Li P, Zhang Y, Zheng L. Transcriptome combined with single-cell sequencing explored prognostic markers associated with T cell exhaustion characteristics in head and neck squamous carcinoma. Sci Rep 2025; 15:8209. [PMID: 40065044 PMCID: PMC11893791 DOI: 10.1038/s41598-025-91299-z] [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: 09/04/2024] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSC) ranks among the most prevalent cancers worldwide, characterized by significant heterogeneity and a complex immune microenvironment. T cell exhaustion is pivotal in the pathogenesis of HNSC, where depleted T cells exhibit reduced proliferative capacity and diminished effector function, facilitating tumor immune escape and subsequent disease progression. A thorough understanding of the primary mechanisms driving T cell depletion within the tumor microenvironment is essential for enhancing the efficacy of immunotherapeutic approaches in HNSC, with profound implications for patient outcomes. In this study, a single-cell atlas of HNSC was constructed, enabling an in-depth analysis of T cell heterogeneity. The differentiation trajectory of T cells, transitioning from normal tissue to HNSC, was characterized, revealing a predisposition toward depletion in the C2 T cell subgroup. A subsequent cross-analysis of significantly upregulated differentially expressed genes in the C2 T cell subset identified five characteristics pertinent to T cell C2, which informed the development of a clinical prognostic model. Additionally, maximum half inhibitory concentration (IC50) values for various pharmacological agents were calculated, leading to the identification of eleven drugs relevant to the risk model, providing an intriguing starting point for further work in personalized cancer treatment. However, certain limitations of this study must be acknowledged. While T cell heterogeneity and differentiation trajectories were mapped, the interrelationships among these subpopulations remain poorly understood. Further research is required to elucidate the specific biological processes and molecular evolutionary mechanisms involved. The insights from this study provide a valuable foundation for future investigations into the molecular mechanisms and immune landscape associated with the progression from normal tissue to malignant squamous cell carcinoma.
Collapse
Affiliation(s)
- Jie Liu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Penghui Li
- Department of Gastrointestinal surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Yuanyuan Zhang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Lian Zheng
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- The First Affiliated Hospital of Zhengzhou University, NO.1 Jianshedong Road, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
31
|
Uher O, Hadrava Vanova K, Labitt R, Petrlakova K, Ye J, Wang H, Masarik M, Jakubek M, Zenka J, Zhuang Z, Pacak K. Neoadjuvant intratumoral MBT(A) immunotherapy prevents distant metastases and recurrence in murine models. Cancer Lett 2025; 612:217464. [PMID: 39809356 DOI: 10.1016/j.canlet.2025.217464] [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: 09/17/2024] [Revised: 12/15/2024] [Accepted: 01/11/2025] [Indexed: 01/16/2025]
Abstract
Neoadjuvant immunotherapy represents a pioneering approach in the preoperative treatment of cancer, providing new strategies for tumor reduction and improved patient outcomes by modulating the immune response. This study investigated neoadjuvant immunotherapy using intratumoral administration of mannan-BAM, Toll-like receptor ligands, and anti-CD40 antibody (MBTA therapy) followed by surgery in murine models of MTT pheochromocytoma, B16-F10 melanoma, and 4T1 and E0771.lmb mammary carcinomas. In the MTT pheochromocytoma model, it was found that neoadjuvant MBTA therapy followed by surgery could prevent the development of distant metastases in 100% of treated animals, compared to a 60% mortality rate in the control group due to metastatic disease after surgery. These outcomes were achieved even in tumors three times larger than those in the control group. In the aggressive 4T1 model, neoadjuvant MBTA therapy resulted in slower tumor progression and a significant prolongation of survival. In the B16-F10 and E0771.lmb models, neoadjuvant MBTA therapy also protected animals from metastases development and tumor recurrence upon rechallenge with tumor cells after surgery. Transcriptomic analysis revealed enhanced effector immune cell infiltration, cytotoxicity, and antigen presentation in retransplanted tumors from MBTA-treated mice, indicating robust immune memory. Notably, the exclusion of the anti-CD40 antibody from the neoadjuvant MBTA therapy (MBT therapy) yielded comparable outcomes in protection against metastases development. These findings advocate for further investigation of intratumoral neoadjuvant MBTA therapy for immunologically "cold" tumors, including those at high risk of metastases or recurrence.
Collapse
MESH Headings
- Animals
- Female
- Neoadjuvant Therapy/methods
- Mice
- Immunotherapy/methods
- Mice, Inbred C57BL
- Neoplasm Recurrence, Local/prevention & control
- Neoplasm Recurrence, Local/immunology
- Cell Line, Tumor
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Melanoma, Experimental/pathology
- Melanoma, Experimental/secondary
- CD40 Antigens/immunology
- CD40 Antigens/antagonists & inhibitors
- Mice, Inbred BALB C
- Disease Models, Animal
- Neoplasm Metastasis
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/therapy
Collapse
Affiliation(s)
- Ondrej Uher
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Katerina Hadrava Vanova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Rachael Labitt
- Research Animal Management Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Katerina Petrlakova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Juan Ye
- Neuro-Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Herui Wang
- Neuro-Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Michal Masarik
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; BIOCEV (Biotechnology and Biomedicine Center in Vestec), First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Milan Jakubek
- BIOCEV (Biotechnology and Biomedicine Center in Vestec), First Faculty of Medicine, Charles University, Vestec, Czech Republic; Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jan Zenka
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice, Czech Republic
| | - Zhengping Zhuang
- Neuro-Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA; AKESO, Prague 5, Czech Republic.
| |
Collapse
|
32
|
Zhou S, Qin Y, Lei A, Li Y, Yang P, Liu H, Sun Y, Zhang J, Deng C, Chen Y. Neoadjuvant and Adjuvant Immunotherapy in the Treatment of Oral Squamous Cell Carcinoma. J Biochem Mol Toxicol 2025; 39:e70199. [PMID: 40034087 DOI: 10.1002/jbt.70199] [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: 11/28/2024] [Revised: 01/22/2025] [Accepted: 02/20/2025] [Indexed: 03/05/2025]
Abstract
Oral squamous cell carcinoma (OSCC) is experiencing a progressive increase in global incidence. Regrettably, this entity is typically discovered at an advanced stage in the majority of patients, which indicates increased therapeutic challenges and a poorer prognosis. Programmed cell death protein 1 (PD-1) appears to have a significant role in immunotherapy and monoclonal antibodies targeting this molecule have been utilized as a therapeutic intervention. A decade of research indicates that neoadjuvant immunotherapy has garnered greater interest than adjuvant immunotherapy in OSCC. This may be due to neoadjuvant immunotherapy serving as a preventive and adjunctive treatment. Enhanced outcomes may be achieved by optimizing the cancer microenvironment before surgery. Of note, neoadjuvant immunotherapy has been introduced preoperatively for untreated OSCC.
Collapse
Affiliation(s)
- Songlin Zhou
- Anhui Engineering Research Center for Oral Materials and Application, Wannan Medical College, Wuhu, China
- College of Oral Medicine, Wannan Medical College, Wuhu, China
| | - Yutao Qin
- Anhui Engineering Research Center for Oral Materials and Application, Wannan Medical College, Wuhu, China
- College of Oral Medicine, Wannan Medical College, Wuhu, China
| | - Anwen Lei
- Anhui Engineering Research Center for Oral Materials and Application, Wannan Medical College, Wuhu, China
- Xuancheng City People's Hospital, Xuancheng, China
| | - Yue Li
- Anhui Engineering Research Center for Oral Materials and Application, Wannan Medical College, Wuhu, China
- College of Oral Medicine, Wannan Medical College, Wuhu, China
| | - Peiru Yang
- Anhui Engineering Research Center for Oral Materials and Application, Wannan Medical College, Wuhu, China
- College of Oral Medicine, Wannan Medical College, Wuhu, China
| | - Hai Liu
- Anhui Engineering Research Center for Oral Materials and Application, Wannan Medical College, Wuhu, China
- College of Oral Medicine, Wannan Medical College, Wuhu, China
| | - Yi Sun
- Anhui Engineering Research Center for Oral Materials and Application, Wannan Medical College, Wuhu, China
- College of Oral Medicine, Wannan Medical College, Wuhu, China
| | - Jue Zhang
- Anhui Engineering Research Center for Oral Materials and Application, Wannan Medical College, Wuhu, China
- College of Oral Medicine, Wannan Medical College, Wuhu, China
| | - Chao Deng
- Anhui Engineering Research Center for Oral Materials and Application, Wannan Medical College, Wuhu, China
- College of Oral Medicine, Wannan Medical College, Wuhu, China
| | - Yu Chen
- Anhui Engineering Research Center for Oral Materials and Application, Wannan Medical College, Wuhu, China
- College of Oral Medicine, Wannan Medical College, Wuhu, China
| |
Collapse
|
33
|
Ruan L, Wang L. Adoptive cell therapy against tumor immune evasion: mechanisms, innovations, and future directions. Front Oncol 2025; 15:1530541. [PMID: 40094019 PMCID: PMC11906336 DOI: 10.3389/fonc.2025.1530541] [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: 11/19/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025] Open
Abstract
Tumors employ a range of strategies to evade detection and eradication by the host's immune system. These include downregulating antigen expression, altering antigen presentation processes, and inhibiting immune checkpoint pathways. etc. Adoptive Cell Therapy (ACT) represents a strategy that boosts anti-tumor immunity. This is achieved by amplifying or genetically engineering immune cells, which are either sourced from the patient or a donor, in a laboratory setting. Subsequently, these cells are reintroduced into the patient to bolster their immune response against cancer. ACT has successfully restored anti-tumor immune responses by amplifying the activity of T cells from patients or donors. This review focuses on the mechanisms underlying tumor escape, including alterations in tumor cell antigens, the immunosuppressive tumor microenvironment (TME), and modulation of immune checkpoint pathways. It further explores how ACT can avddress these factors to enhance therapeutic efficacy. Additionally, the review discusses the application of gene-editing technologies (such as CRISPR) in ACT, highlighting their potential to strengthen the anti-tumor capabilities of T cells. Looking forward, the personalized design of ACT, combined with immune checkpoint inhibitors and targeted therapies, is expected to significantly improve treatment outcomes, positioning this approach as a key strategy in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Liqin Ruan
- Department of Hepatobiliary Surgery, JiuJiang City Key Laboratory of Cell Therapy, JiuJiang No.1 People's Hospital, Jiujiang, Jiangxi, China
| | - Lu Wang
- Department of Oncology, JiuJiang City Key Laboratory of Cell Therapy, JiuJiang No.1 People's Hospital, Jiujiang, Jiangxi, China
| |
Collapse
|
34
|
Wang X, He S, Gong X, Lei S, Zhang Q, Xiong J, Liu Y. Neutrophils in colorectal cancer: mechanisms, prognostic value, and therapeutic implications. Front Immunol 2025; 16:1538635. [PMID: 40092983 PMCID: PMC11906667 DOI: 10.3389/fimmu.2025.1538635] [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: 12/03/2024] [Accepted: 02/04/2025] [Indexed: 03/19/2025] Open
Abstract
Neutrophils, the most abundant myeloid cells in human peripheral blood, serve as the first defense line against infection and are also significantly involved in the initiation and progression of cancer. In colorectal cancer (CRC), neutrophils exhibit a dual function by promoting tumor events and exerting antitumor activity, which is related to the heterogeneity of neutrophils. The neutrophil extracellular traps (NETs), gut microbiota, and various cells within the tumor microenvironment (TME) are involved in shaping the heterogeneous function of neutrophils. This article provides an updated overview of the complex functions and underlying mechanisms of neutrophils in CRC and their pivotal role in guiding prognosis assessment and therapeutic strategies, aiming to offer novel insights into neutrophil-associated treatment approaches for CRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yang Liu
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Zhou B, Chen M, Hao Z, Li L, Zhang Y, Fang B, Shao M, Ren G, Wang K, Liu H, Zhu J, Zhang X, Yuan S, Sitou I, Zhao J, Huang J, Yu Z, Qiu F. Zinc-copper bimetallic nanoplatforms trigger photothermal-amplified cuproptosis and cGAS-STING activation for enhancing triple-negative breast cancer immunotherapy. J Nanobiotechnology 2025; 23:137. [PMID: 39994712 PMCID: PMC11849371 DOI: 10.1186/s12951-025-03186-4] [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: 11/25/2024] [Accepted: 02/01/2025] [Indexed: 02/26/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by high rates of metastasis and recurrence, along with a low sensitivity to immunotherapy, resulting in a paucity of effective therapeutic strategies. Herein, we have developed polydopamine-coated zinc-copper bimetallic nanoplatforms (Cu-ZnO2@PDA nanoplatforms, abbreviated CZP NPs) that can efficiently induce photothermal amplified cuproptosis and cGAS-STING signaling pathway activation, thereby reversing the immunosuppressive tumor microenvironment of TNBC, upregulating PD-L1 expression, and boosting the efficacy of anti-programmed death-ligand 1 antibody (αPD-L1)-based immunotherapy. Within the acidic tumor microenvironment (TME), CZP NPs spontaneously release copper and zinc ions and hydrogen peroxide, generating highly oxidative hydroxyl radicals and downregulating iron-sulfur cluster proteins. These actions lead to the disruption of mitochondrial integrity, the release of mitochondrial DNA (mtDNA) and irreversible cuproptosis. The further synergy between mtDNA and zinc ions potentiates the activation of the cGAS-STING signaling pathway, triggering a robust antitumor immune response and sensitizing TNBC to αPD-L1 therapy. Additionally, using an 808 nm near-infrared laser for photothermal therapy significantly augments these effects, resulting in a cascade amplification of therapeutic efficacy against TNBC. The strategic combination of CZP NPs with αPD-L1 markedly bolsters antitumor immunity and suppresses tumor growth. Collectively, our findings present a promising synergistic strategy for TNBC treatment by linking cuproptosis, cGAS-STING activation, photothermal therapy, and immunotherapy.
Collapse
Affiliation(s)
- Bangyi Zhou
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Mengyao Chen
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Zhixing Hao
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Lili Li
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Yixin Zhang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Baoru Fang
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, P. R. China
| | - Miner Shao
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Guohong Ren
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Ke Wang
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, P. R. China
| | - Huiying Liu
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Jingxuan Zhu
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Xinyi Zhang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Shuyan Yuan
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - I Sitou
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Jing Zhao
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
| | - Jian Huang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
| | - Zhangsen Yu
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, P. R. China.
| | - Fuming Qiu
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
| |
Collapse
|
36
|
Cheng R, Li S, Ma X, Zhuang W, Lei Y, He J, Liang C, Nie W, Xie HY. Intratumoral antigen-presenting cell activation by a nanovesicle for the concurrent tertiary lymphoid structure de novo neogenesis. SCIENCE ADVANCES 2025; 11:eadr1299. [PMID: 39970209 PMCID: PMC11837995 DOI: 10.1126/sciadv.adr1299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025]
Abstract
Tertiary lymphoid structures (TLSs) usually lead to significantly improved clinical benefits in immunotherapy but are rarely observed within native tumors. The current approaches are difficult in effectively inducing TLS formation, let alone fully exploiting its anticancer efficacy. Here, a biomimetic nanovesicle (ADU-S@M1) is constructed to target tumors and then to produce abundant activated antigen-presenting cells (APCs) in situ by polarizing the tumor-associated macrophages toward M1 phenotype and promoting dendritic cell maturation. These activated APCs effectively initiate the TLS de novo neogenesis by acting as lymphoid tissue inducer cells that secrete lymphotoxin α and tumor necrosis factor α while normalizing the intratumoral vasculatures. In addition, they induce robust in situ adaptive immune responses by presenting the antigens released from the M1 cell-destroyed tumors and transporting them to the nearby TLS. Therefore, the development of tumors in mice, especially immune-cold tumors, was efficiently prevented, providing a promising strategy for promoting cancer immunotherapy.
Collapse
Affiliation(s)
- Ran Cheng
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P.R. China
| | - Sucheng Li
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P.R. China
| | - Xianbin Ma
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P.R. China
| | - Wanru Zhuang
- Chemical Biology Center, Peking University, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Beijing 100191, P.R. China
| | - Yao Lei
- Chemical Biology Center, Peking University, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Beijing 100191, P.R. China
| | - Jiaqi He
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P.R. China
| | - Chao Liang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P.R. China
| | - Weidong Nie
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P.R. China
| | - Hai-Yan Xie
- Chemical Biology Center, Peking University, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Beijing 100191, P.R. China
| |
Collapse
|
37
|
Wang SY, Wang KJ. Reevaluating Calculus bovis: Modulating the liver cancer immune microenvironment via the Wnt/β-catenin pathway. World J Gastroenterol 2025; 31:99750. [PMID: 39958448 PMCID: PMC11752708 DOI: 10.3748/wjg.v31.i6.99750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/05/2024] [Accepted: 12/10/2024] [Indexed: 01/10/2025] Open
Abstract
In this article, we comment on the work published by Huang et al, which explores the mechanisms by which Calculus bovis (CB) modulates the liver cancer immune microenvironment via the Wnt/β-catenin signalling pathway. The study demonstrates that active components in CB effectively inhibit the activation of the Wnt/β-catenin pathway, significantly reducing the polarization of M2 tumor-associated macrophages. Both in vivo and in vitro experiments have validated the anti-tumour effects of CB, revealing its complex mechanisms of action through the modulation of immune cell functions within the tumour microenvironment. This article highlights CB's therapeutic potential in liver cancer treatment and calls for further investigations into its mechanisms and clinical applications to develop safer, more effective options for patients. The study also revealed that key components of CB, such as bilirubin and bile acids, inhibit tumour cell proliferation and promote apoptosis through multiple pathways. Future research should explore the mechanisms of action of CB and its potential integration with existing treatments to improve the therapeutic outcomes of liver cancer patients. With multidisciplinary collaboration and advanced research, CB could become a key component of comprehensive liver cancer treatment, offering new hope for patients.
Collapse
Affiliation(s)
- Shi-Yue Wang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Kai-Juan Wang
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| |
Collapse
|
38
|
Barcellini L, Nardin S, Sacco G, Ferrante M, Rossi G, Barletta G, Bennicelli E, Dellepiane C, Tagliamento M, Ramella Pollone B, Lucente L, Coco S, Marconi S, Santamaria S, Pariscenti GL, Genova C. Immune Checkpoint Inhibitors and Targeted Therapies in Early-Stage Non-Small-Cell Lung Cancer: State-of-the-Art and Future Perspectives. Cancers (Basel) 2025; 17:652. [PMID: 40002247 PMCID: PMC11853691 DOI: 10.3390/cancers17040652] [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: 01/13/2025] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Approximately 25-30% of non-small-cell lung cancer (NSCLC) patients are diagnosed when the disease is still resectable, although the risk of recurrence is significant. Recently, approaches based on targeted agents or immune checkpoint inhibitors (ICIs) have modified the management of such patients. However, some questions remain unanswered. Objectives: Our aim is to assess the current evidence on approaches involving targeted agents and ICIs in resectable NSCLC, to provide an up-to-date overview of the subject, and to identify areas of current debate, Methods: We analyzed randomized trials on ICIs and targeted therapies in early-stage NSCLC, published or presented at international oncology meetings throughout the last 5 years. Results: Osimertinib and alectinib have shown robust results in the adjuvant setting for molecularly identified patient subgroups, while ICIs have achieved robust data in the neoadjuvant/perioperative setting, with less consistent data on the pure adjuvant approach. Circulating tumor DNA levels may offer a possible biomarker for therapeutic decisions, albeit more prospective data are needed. Conclusions: Targeted agents and ICIs are revolutionizing early-stage NSCLC, similarly to what was observed in advanced disease. Prospective studies designed to compare neoadjuvant, adjuvant, and perioperative approaches and to assess the role of circulating biomarkers are warranted.
Collapse
Affiliation(s)
- Lucrezia Barcellini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genoa, 16126 Genoa, Italy; (L.B.); (G.S.); (M.F.); (M.T.); (B.R.P.); (L.L.)
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (G.B.); (S.S.)
| | - Simone Nardin
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genoa, 16126 Genoa, Italy; (L.B.); (G.S.); (M.F.); (M.T.); (B.R.P.); (L.L.)
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (G.B.); (S.S.)
| | - Gianluca Sacco
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genoa, 16126 Genoa, Italy; (L.B.); (G.S.); (M.F.); (M.T.); (B.R.P.); (L.L.)
- U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (G.R.); (E.B.); (C.D.); (S.C.); (S.M.)
| | - Michele Ferrante
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genoa, 16126 Genoa, Italy; (L.B.); (G.S.); (M.F.); (M.T.); (B.R.P.); (L.L.)
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (G.B.); (S.S.)
| | - Giovanni Rossi
- U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (G.R.); (E.B.); (C.D.); (S.C.); (S.M.)
| | - Giulia Barletta
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (G.B.); (S.S.)
| | - Elisa Bennicelli
- U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (G.R.); (E.B.); (C.D.); (S.C.); (S.M.)
| | - Chiara Dellepiane
- U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (G.R.); (E.B.); (C.D.); (S.C.); (S.M.)
| | - Marco Tagliamento
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genoa, 16126 Genoa, Italy; (L.B.); (G.S.); (M.F.); (M.T.); (B.R.P.); (L.L.)
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (G.B.); (S.S.)
| | - Beatrice Ramella Pollone
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genoa, 16126 Genoa, Italy; (L.B.); (G.S.); (M.F.); (M.T.); (B.R.P.); (L.L.)
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (G.B.); (S.S.)
| | - Luca Lucente
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genoa, 16126 Genoa, Italy; (L.B.); (G.S.); (M.F.); (M.T.); (B.R.P.); (L.L.)
- U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (G.R.); (E.B.); (C.D.); (S.C.); (S.M.)
| | - Simona Coco
- U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (G.R.); (E.B.); (C.D.); (S.C.); (S.M.)
| | - Silvia Marconi
- U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (G.R.); (E.B.); (C.D.); (S.C.); (S.M.)
| | - Sara Santamaria
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (G.B.); (S.S.)
| | - Gian Luca Pariscenti
- Department of Thoracic Surgery, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy;
| | - Carlo Genova
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genoa, 16126 Genoa, Italy; (L.B.); (G.S.); (M.F.); (M.T.); (B.R.P.); (L.L.)
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (G.B.); (S.S.)
| |
Collapse
|
39
|
Li B, Zhang C, Xu X, Shen Q, Luo S, Hu J. Manipulating the cGAS-STING Axis: advancing innovative strategies for osteosarcoma therapeutics. Front Immunol 2025; 16:1539396. [PMID: 39991153 PMCID: PMC11842356 DOI: 10.3389/fimmu.2025.1539396] [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: 12/04/2024] [Accepted: 01/15/2025] [Indexed: 02/25/2025] Open
Abstract
This paper explored the novel approach of targeting the cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) synthase-stimulator of interferon genes (cGAS-STING) pathway for the treatment of osteosarcoma (OS). Osteosarcoma is a common malignancy in adolescents. Most patients die from lung metastasis. It reviewed the epidemiology and pathological characteristics of OS, highlighting its highly malignant nature and tendency for pulmonary metastasis, underscoring the importance of identifying new therapeutic targets. The cGAS-STING pathway was closely associated with the malignant biological behaviors of OS cells, suggesting that targeting this pathway could be a promising therapeutic strategy. Currently, research on the role of the cGAS-STING pathway in OS treatment has been limited, and the underlying mechanisms remain unclear. Therefore, further investigation into the mechanisms of the cGAS-STING pathway in OS and the exploration of therapeutic strategies based on this pathway are of great significance for developing more effective treatments for OS. This paper offered a fresh perspective on the treatment of OS, providing hope for new therapeutic options for OS patients by targeting the cGAS-STING pathway.
Collapse
Affiliation(s)
- BingBing Li
- Department of Pediatrics, Shaoxing Central Hospital, The Central Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Cheng Zhang
- Department of Pediatrics, Shaoxing Central Hospital, The Central Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - XiaoJuan Xu
- Department of Pediatrics, Shaoxing Central Hospital, The Central Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - QiQin Shen
- Department of Orthopedics, Shaoxing Central Hospital, The Central Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - ShuNan Luo
- Department of Surgery, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - JunFeng Hu
- Department of Pain, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| |
Collapse
|
40
|
Zhang X, Tang D, Xiao H, Li B, Shang K, Zhao D. Activating the cGAS-STING Pathway by Manganese-Based Nanoparticles Combined with Platinum-Based Nanoparticles for Enhanced Ovarian Cancer Immunotherapy. ACS NANO 2025; 19:4346-4365. [PMID: 39846241 DOI: 10.1021/acsnano.4c12237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Recent research has demonstrated that activating the cGAS-STING pathway can enhance interferon production and the activation of T cells. A manganese complex, called TPA-Mn, was developed in this context. The reactive oxygen species (ROS)-sensitive nanoparticles (NPMn) loaded with TPA-Mn are developed. NPMn activates the cGAS-STING pathway via cGAS activation (i.e., 1.6-fold enhancement of P-STING), which in turn increases the secretion of pro-inflammatory cytokines (e.g., TNF-α, IL-6, and IL-2). This promotes dendritic cell maturation, enhances the infiltration of cytotoxic T lymphocytes, and reduces the percentage of immunosuppressive regulatory T cells. In addition, it is crucial to emphasize that cisplatin-induced DNA damage can potentially trigger activation of the cGAS-STING pathway. NPMn, in combination with low-dose NPPt, a carrier of a Cis(IV) prodrug capable of causing DNA damage, augments the cGAS-STING pathway activation and significantly activates the tumor immune microenvironment (TIME). Furthermore, combined with anti-PD-1 antibody, NPPt+NPMn shows synergistic efficacy in both ovarian cancer peritoneal metastases and recurrence models. It not only effectively eliminates tumors but also induces a strong immune memory response, providing a promising strategy for the clinical management of ovarian cancer. This work offers a design of manganese-based nanoparticles for immunotherapy.
Collapse
Affiliation(s)
- Xiangling Zhang
- Department of Gynecology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Dongsheng Tang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Bin Li
- Department of Gynecology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Kun Shang
- Department of Nuclear Medicine, Peking University People's Hospital, Beijing 100044, P. R. China
| | - Dan Zhao
- Department of Gynecology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| |
Collapse
|
41
|
Li Y, Cao Y, Wang X, Li C, Zhao L, Li H. Effects of perioperative treatment of resectable adenocarcinoma of esophagogastric junction by immunotherapy (Adebrelimab) combined with chemotherapy (XELOX): protocol for a single-center, open-labeled study (AEGIS trial, neoadjuvant immunochemotherapy). BMC Cancer 2025; 25:198. [PMID: 39905364 PMCID: PMC11792203 DOI: 10.1186/s12885-025-13589-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND For resectable adenocarcinoma of the esophagogastric junction (AEG), current treatment exploration primarily focuses on perioperative chemotherapy regimens combined with PD-1/PD-L1 inhibitors, but the long-term survival benefits of still require further investigation, and the use of upfront immunotherapy is typically restricted to patients with metastatic MSI-H (M1 MSI-H) disease due to their potential responsiveness to immunological agents. Adebrelimab, as a novel PD-L1 antibody, has not yet been proven for its efficacy and safety in adenocarcinoma of the esophagogastric junction. METHODS The AEGIS study is a prospective, open-labeled, single-arm, phase II clinical trial. A total of 26 patients with AEG will be enrolled. The primary endpoint is the pathologic complete response (pCR) rate after perioperative neoadjuvant immunochemotherapy. Secondary outcomes of the study include the objective response rate (ORR), R0 resection rate, major pathological response (MPR) rate, and pCR rate in combined positive score(CPS) ≥ 5 and MSI-H populations, event-free survival (EFS), and overall survival (OS). The exploratory outcomes are the biomarkers related to therapeutic efficacy, such as PD-L1 expression, microsatellite instability (MSI), tumor mutational burden(TMB), Epstein-Barr virus(EBV) infection, and circulating tumor DNA(ctDNA). DISCUSSION This trail aims to verify the efficacy and safety of the perioperative treatment regimen of anti-PD-L1 (Adebrelimab) combined with chemotherapy (capecitabine plus oxaliplatin, XELOX) for patients with resectable AEG. Considering the differences in chemotherapy regimen tolerance between Asian and Western populations, this study intends to evaluate the suitability of Adebrelimab combined with XELOX chemotherapy for the Asian population. TRIAL REGISTRATION ClinicalTrials.gov: NCT06482788. The trial was prospectively registered on 22 May 2024, https://clinicaltrials.gov/study/NCT06482788 .
Collapse
Affiliation(s)
- Yingyi Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuqin Cao
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xipeng Wang
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chengqiang Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Liqin Zhao
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hecheng Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
42
|
Lim BJW, Liu M, Wang L, Kong SLY, Yin T, Yan C, Xiang K, Cao C, Wu H, Mihai A, Tay FPL, Wang E, Jiang Q, Ma Z, Tan L, Chia RN, Qin D, Pan CC, Wang XF, Li QJ. Neoadjuvant anti-4-1BB confers protection against spontaneous metastasis through low-affinity intratumor CD8 + T cells in triple-negative breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.29.635356. [PMID: 39975187 PMCID: PMC11838326 DOI: 10.1101/2025.01.29.635356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Neoadjuvant immunotherapy seeks to harness the primary tumor as a source of relevant tumor antigens to enhance systemic anti-tumor immunity through improved immunological surveillance. Despite having revolutionized the treatment of patients with high-risk early-stage triple-negative breast cancer (TNBC), a significant portion of patients remain unresponsive and succumb to metastatic recurrence post-treatment. Here, we found that optimally scheduled neoadjuvant administration of anti-4-1BB monotherapy was able to counteract metastases and prolong survival following surgical resection. Phenotypic and transcriptional profiling revealed enhanced 4-1BB expression on tumor-infiltrating intermediate (T int ), relative to progenitor (T prog ) and terminally exhausted (T term ) T cells. Furthermore, T int was enriched in low-affinity T cells. Treatment with anti-4-1BB drove clonal expansion of T int , with reduced expression of tissue-retention marker CD103 in T prog . This was accompanied by increased TCR clonotype sharing between paired tumors and pre-metastatic lungs. Further interrogation of sorted intratumor T cells confirmed enhanced T cell egress into circulation following anti-4-1BB treatment. In addition, gene signature extracted from anti-4-1BB treated T int was consistently associated with improved clinical outcomes in BRCA patients. Combinatorial neoadjuvant anti-4-1BB and ablation of tumor-derived CXCL16 resulted in enhanced therapeutic effect. These findings illustrate the intratumor changes underpinning the efficacy of neoadjuvant anti-4-1BB, highlighting the reciprocity between local tissue-retention and distant immunologic fortification, suggesting treatment can reverse the siphoning of intratumor T cells to primary tumor, enabling redistribution to distant tissues and subsequent protection against metastases.
Collapse
|
43
|
Shi J, Gao H, Wu Y, Luo C, Yang G, Luo Q, Jia B, Han C, Liu Z, Wang F. Nuclear imaging of PD-L1 expression promotes the synergistic antitumor efficacy of targeted radionuclide therapy and immune checkpoint blockade. Eur J Nucl Med Mol Imaging 2025; 52:955-969. [PMID: 39472367 DOI: 10.1007/s00259-024-06962-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: 06/17/2024] [Accepted: 10/18/2024] [Indexed: 01/23/2025]
Abstract
PURPOSE In order to maximize synergistic effect of targeted radionuclide therapy (TRT) and immune checkpoint blockade (ICB) as well as reduce the toxicity, we pioneered a strategy guided by PD-L1-targeted nuclear medicine imaging for the combination of TRT and ICB towards precision cancer therapy. METHODS As a novel targeted radiotherapeutic agent, 177Lu-AB-3PRGD2 targeting integrin αvβ3 was developed to achieve sustained antitumor effect by introducing an albumin binder (AB) into the structure of 3PRGD2. The 177Lu-AB-3PRGD2 TRT as well as different types of combination therapies of 177Lu-AB-3PRGD2 TRT and anti-PD-L1 ICB were performed in animal models. The changes of PD-L1 expression in tumors after TRT were evaluated in vitro and in vivo by PD-L1-specific SPECT/CT imaging of 99mTc-MY1523. RESULTS 177Lu-AB-3PRGD2 showed improved tumor uptake and prolonged tumor retention, leading to significantly enhanced tumor growth suppression. Moreover, 177Lu-AB-3PRGD2 TRT remodeled the tumor immune microenvironment by upregulating PD-L1 expression and increasing tumor-infiltrating CD8+ T cells, facilitating immunotherapy. We found that the anti-PD-L1 treatment was more effective during the upregulation of tumor PD-L1 expression, and the time window could be determined by 99mTc-MY1523 SPECT/CT. CONCLUSION We developed a novel and long-acting radiotherapeutic agent 177Lu-AB-3PRGD2, and pioneered a strategy guided by PD-L1-targeted nuclear medicine imaging for the combination of TRT and ICB towards precision cancer therapy, optimizing the therapeutic efficacy and reducing the cost and potential toxicity risks. This strategy could also be adapted for clinical practice, combining conventional radiotherapy or chemotherapy with ICB to enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Jiyun Shi
- Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, International Cancer Institute, Peking University, Beijing, 100191, China
- Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hannan Gao
- Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, International Cancer Institute, Peking University, Beijing, 100191, China
- Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yue Wu
- Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, International Cancer Institute, Peking University, Beijing, 100191, China
| | - Chuangwei Luo
- Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, International Cancer Institute, Peking University, Beijing, 100191, China
| | - Guangjie Yang
- Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, International Cancer Institute, Peking University, Beijing, 100191, China
| | - Qi Luo
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Bing Jia
- Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, International Cancer Institute, Peking University, Beijing, 100191, China
| | - Chuanhui Han
- Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, International Cancer Institute, Peking University, Beijing, 100191, China
| | - Zhaofei Liu
- Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, International Cancer Institute, Peking University, Beijing, 100191, China
| | - Fan Wang
- Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, International Cancer Institute, Peking University, Beijing, 100191, China.
- Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- Guangzhou National Laboratory, Guangzhou, 510005, China.
| |
Collapse
|
44
|
Zhao M, Schoenfeld JD, Egloff AM, Hanna GJ, Haddad RI, Adkins DR, Uppaluri R. T cell dynamics with neoadjuvant immunotherapy in head and neck cancer. Nat Rev Clin Oncol 2025; 22:83-94. [PMID: 39658611 DOI: 10.1038/s41571-024-00969-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2024] [Indexed: 12/12/2024]
Abstract
Immune-checkpoint inhibitors (ICIs) are being tested as neoadjuvant therapies in various solid tumours, including in patients with head and neck squamous cell carcinoma (HNSCC), with promising results. Key findings thus far include that this approach is well-tolerated with favourable clinical outcomes including promising pathological response rates in initial studies. Pathological responses are likely to be increased by combining other agents with anti-PD-(L)1 antibodies. Comparisons of baseline biopsy samples with post-treatment surgical specimens have enabled correlative studies utilizing multiomic and immunogenomic methods. Data from these studies suggest that pretreatment intratumoural tissue-resident memory CD8+ T cells are key drivers of tumour regression and give rise to both local and systemic antitumour immune responses. Analyses of systemic responses have defined a PD-1+KLRG1- circulating CD8+ T cell subpopulation that is highly predictive of response, and revealed the interrelationships between intratumoural clones and circulating CD8+ T cells. Lastly, interrogation of T cell populations within lymph nodes is beginning to delineate the immune crosstalk between the primary tumour and tumour-draining lymph nodes and how this relationship might be disrupted with tumour infiltration of the latter. In this Review, we examine data from trials testing neoadjuvant ICIs in patients with HNSCC, focusing on human papillomavirus-unrelated disease, and highlight correlative immunogenomic findings from these trials.
Collapse
Affiliation(s)
- Maryann Zhao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jonathan D Schoenfeld
- Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Boston, MA, USA
| | - Ann Marie Egloff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Glenn J Hanna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Robert I Haddad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Douglas R Adkins
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine/Medical Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ravindra Uppaluri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
45
|
Xie Z, Shao J, Shen Z, Ye Z, Okada Y, Okuzaki D, Okada N, Tachibana M. HDAC1-3 inhibition triggers NEDD4-mediated CCR2 downregulation and attenuates immunosuppression in myeloid-derived suppressor cells. Cancer Immunol Immunother 2025; 74:81. [PMID: 39891718 PMCID: PMC11787094 DOI: 10.1007/s00262-024-03931-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/21/2024] [Indexed: 02/03/2025]
Abstract
Myeloid-derived suppressor cells (MDSCs) play a critical role in cancer progression and resistance, thus representing promising targets for immunotherapy. Despite the established role of histone deacetylases (HDACs) in epigenetic regulation of cell fate and function, their specific impact on MDSCs remains elusive. We sought to investigate the effects and underlying mechanisms of HDAC on MDSCs using various HDAC inhibitors. Our results indicate that HDAC1-3 inhibitors reduce CCR2 expression, a chemokine receptor that mediates the migration of monocytic (M-)MDSCs to tumors and attenuated the immunosuppressive activity of MDSCs. In an orthotropic hepatocellular carcinoma (HCC) murine model, HDAC1-3 inhibitors reduced the infiltration of M-MDSCs, increased the number of natural killer cells in tumors, and suppressed tumor growth. Our results also suggest that HDAC1-3 inhibitors potentiate the antitumor effects of anti-programmed cell death protein 1 antibodies. ATAC-seq and RNA-seq analyses revealed 115 genes epigenetically upregulated by HDAC1-3 inhibitors, primarily linked to transcriptional regulation and ubiquitination. We further elucidated that HDAC1-3 inhibitors facilitate CCR2 protein degradation through ubiquitination-mediated by NEDD4 E3 ligase. Our findings reveal a novel mechanism of action of HDAC1-3 inhibitors in MDSCs and suggest a potential synergistic immunotherapy strategy for clinical benefit in HCC.
Collapse
Affiliation(s)
- Zhiqi Xie
- Wuyi First People's Hospital, Affiliated Hospital, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Jinjin Shao
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, 310053, China
| | - Zeren Shen
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Zhichao Ye
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, 310053, China
| | - Yoshiaki Okada
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Daisuke Okuzaki
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Frontier Research Center, Osaka University, Osaka, 565-0871, Japan
| | - Naoki Okada
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Masashi Tachibana
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan.
- Laboratory for Context-Dependent Cell Immunology, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan.
| |
Collapse
|
46
|
Huang Q, Xu L, Ye L. Functional subsets of tumor-specific CD8 + T cells in draining lymph nodes and tumor microenvironment. Curr Opin Immunol 2025; 92:102506. [PMID: 39591663 DOI: 10.1016/j.coi.2024.102506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 10/21/2024] [Accepted: 10/26/2024] [Indexed: 11/28/2024]
Abstract
Accumulating evidence demonstrates that tumor-specific CD8+ T cells in tumor-draining lymph nodes (TdLNs) act as an upstream reservoir of exhausted subsets within tumor microenvironment (TME). This reservoir primarily consists of progenitor exhausted CD8+ T (TPEX) cells and newly defined tumor-specific memory subsets (TTSM). We propose that these two subsets work together to mediate the antitumor effects of PD-1/PD-L1 immune checkpoint blockade (ICB) in a spatiotemporal manner. Although PD-1/PD-L1 ICB monotherapy drives the proliferation and further differentiation of these subsets, it does not alter the programmed differentiation trajectory from TTSM cells to TPEX cells, ultimately leading to the development of terminally exhausted CD8+ T cells. This phenomenon may partly explaining the frequent relapse in patients following initial ICB therapy. In this review, we focus on the phenotypic and functional heterogeneity of tumor-specific CD8+ T cells in both TdLNs and the TME and discuss the implications of these studies for ICB. Our insights aim to illuminate new strategies for advancing tumor immunotherapies.
Collapse
Affiliation(s)
- Qizhao Huang
- Institute of Immunological Innovation and Translation, Chongqing Medical University, Chongqing, China; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Lifan Xu
- Institute of Immunology, Third Military Medical University, Chongqing, China; National Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China.
| | - Lilin Ye
- Institute of Immunology, Third Military Medical University, Chongqing, China.
| |
Collapse
|
47
|
Salimi Asl A, Davari M, Ghorbani A, Seddighi N, Arabi K, Saburi E. Neoadjuvant immunotherapy and oncolytic virotherapy in HPV positive and HPV negative skin cancer: A comprehensive review. Int Immunopharmacol 2025; 146:113790. [PMID: 39673996 DOI: 10.1016/j.intimp.2024.113790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Skin cancer is the most common new cancer among Caucasians. This cancer has different types, of which non-melanoma skin cancer is the most common type. Various factors affect this disease, one of which is viral infections, including HPV. This virus plays an important role in skin cancer, especially cSCCs. There are various options for the treatment of skin cancer, and today special attention has been paid to treatments based on therapeutic goals, immunotherapy and combination therapy. In this study, we have investigated treatments based on immunotherapy and virotherapy and the effect of HPV virus on the effectiveness of these treatments in skin cancer. Treatments based on virotherapy are performed for a long time in combination with other common treatments such as radiotherapy and chemotherapy in order to have a greater effect and lower its side effects, which include: shortness of breath, tachycardia, lowering blood pressure in the patient. Also, the most important axis of immunotherapy is to focus on PD1-PDL1, despite abundant evidence on the importance of immunotherapy, many studies investigate the use of immunotherapy inhibitors in the adjuvant and neoadjuvant setting in various cancers. Also, previous findings show conflicting evidence of the effect of HPV status on the response to immunotherapy.
Collapse
Affiliation(s)
- Ali Salimi Asl
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Mohsen Davari
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Atousa Ghorbani
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Narjes Seddighi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Kimia Arabi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran.
| | - Ehsan Saburi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics and Molecular Medicine Department, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
48
|
Topalian SL, Pardoll DM. Neoadjuvant anti-PD-1-based immunotherapy: evolving a new standard of care. J Immunother Cancer 2025; 13:e010833. [PMID: 39855712 PMCID: PMC11759207 DOI: 10.1136/jitc-2024-010833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Neoadjuvant (presurgical) anti-programmed cell death protein-1 (PD-1)-based immunotherapy as a new approach to cancer treatment has been developing on an accelerated trajectory since the seminal clinical trial results from studies in lung cancer and melanoma were published in 2018. Groundbreaking regulatory approvals in triple-negative breast cancer, non-small cell lung cancer and melanoma will certainly be followed by additional approvals in other disease indications, as clinical and basic research are burgeoning globally in hundreds of clinical trials across dozens of cancer types. As this field is evolving, it is addressing gaps in our understanding of biological mechanisms underlying PD-1 pathway blockade and their synergy with other antineoplastic drugs, probing mechanisms of response and resistance to neoadjuvant immunotherapy, optimizing efficacious clinical strategies, and analyzing commonalities and differences across cancer types. Knowledge gained thus far provides a firm foundation from which to launch the next phase of translational research in this expanding arena of biomedical investigation.
Collapse
Affiliation(s)
- Suzanne L Topalian
- Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, Maryland, USA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, USA
| | - Drew M Pardoll
- The Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, Maryland, USA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, USA
- Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
49
|
Zhao L, Gui Y, Cai J, Deng X. Biometallic ions and derivatives: a new direction for cancer immunotherapy. Mol Cancer 2025; 24:17. [PMID: 39815289 PMCID: PMC11734411 DOI: 10.1186/s12943-025-02225-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: 09/26/2024] [Accepted: 01/01/2025] [Indexed: 01/18/2025] Open
Abstract
Biometallic ions play a crucial role in regulating the immune system. In recent years, cancer immunotherapy has become a breakthrough in cancer treatment, achieving good efficacy in a wide range of cancers with its specificity and durability advantages. However, existing therapies still face challenges, such as immune tolerance and immune escape. Biometallic ions (e.g. zinc, copper, magnesium, manganese, etc.) can assist in enhancing the efficacy of immunotherapy through the activation of immune cells, enhancement of tumor antigen presentation, and improvement of the tumor microenvironment. In addition, biometallic ions and derivatives can directly inhibit tumor cell progression and offer the possibility of effectively overcoming the limitations of current cancer immunotherapy by promoting immune responses and reducing immunosuppressive signals. This review explores the role and potential application prospects of biometallic ions in cancer immunotherapy, providing new ideas for future clinical application of metal ions as part of cancer immunotherapy and helping to guide the development of more effective and safe therapeutic regimens.
Collapse
Affiliation(s)
- Lin Zhao
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China
| | - Yajun Gui
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China
| | - Jing Cai
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China
| | - Xiangying Deng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China.
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China.
- Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
50
|
Yang Z, Chen Y, Miao Y, Yan H, Chen K, Xu Y, Su L, Zhang L, Yan Y, Chi H, Fu J, Wang L. Elucidating stearoyl metabolism and NCOA4-mediated ferroptosis in gastric cancer liver metastasis through multi-omics single-cell integrative mendelian analysis: advancing personalized immunotherapy strategies. Discov Oncol 2025; 16:46. [PMID: 39812999 PMCID: PMC11735723 DOI: 10.1007/s12672-025-01769-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The metabolism of stearoyl-GPE plays a key role in the liver metastasis of gastric cancer. This investigation delves into the mechanisms underlying the intricate tumor microenvironment (TME) heterogeneity triggered by stearoyl metabolism in gastric cancer with liver metastasis (LMGC), offering novel perspectives for LMGC. OBJECTIVE Utilizing Mendelian randomization, we determined that stearoyl metabolism significantly contributes to the progression of gastric cancer (GC). Following this, bulk transcriptome analyses and single-cell multiomics techniques to investigate the roles of stearoyl-GPE metabolism-related genes, particularly NCOA4, in regulating LMGC TME. RESULTS Our analysis highlights the crucial role of stearoyl metabolism in modulating the complex microenvironment of LMGC, particularly impacting monocyte cells. Through single-cell sequencing and spatial transcriptomics, we have identified key metabolic genes specific to stearoyl metabolism within the monocyte cell population, including NCOA4. Regarding the relationship between ferroptosis, stearoyl metabolism, and LMGC findings, it is plausible that stearoyl metabolism and LMGC pathways intersect with mechanisms involved in ferroptosis. Ferroptosis, characterized by iron-dependent lipid peroxidation, represents a regulated form of cell death. The activity of Stearoyl-CoA desaturase (SCD), a critical enzyme in stearoyl metabolism, has been associated with the modulation of lipid composition and susceptibility to ferroptosis. Furthermore, the LMGC is integral to cellular processes related to oxidative stress and lipid metabolism, both of which are significant factors in the context of ferroptosis. CONCLUSION This study enhances the understanding of the relationship between stearoyl metabolism and ferroptosis in promoting liver metastasis of gastric cancer and its role in the regulation of tumor heterogeneity. In addition, this study contributes to a deeper understanding of the dynamics of gastric cancer tumor microenvironment (TME) and provides a basis for the development of better interventions to combat cancer metastasis.
Collapse
Affiliation(s)
- Zhongqiu Yang
- Department of General Surgery, Dazhou Central Hospital, Dazhou, 635000, China
| | - Yuquan Chen
- School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, 3004, Australia
| | - Yaping Miao
- General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Haisheng Yan
- General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Kexin Chen
- General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yaoqin Xu
- General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Lanqian Su
- School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Lanyue Zhang
- School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yalan Yan
- School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Hao Chi
- School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- Western Institute of Digital-Intelligent Medicine, 401329, Chongqing, China.
| | - Jin Fu
- Department of Laboratory Medicine, Chonggang General Hospital, Chongqing, 400080, China.
| | - Lexin Wang
- Western Institute of Digital-Intelligent Medicine, 401329, Chongqing, China.
| |
Collapse
|