1
|
Shi Y, Liu X, Liu A, Fang J, Meng Q, Ding C, Ai B, Gu Y, Zhang C, Zhou C, Wang Y, Shui Y, Yu S, Zhang D, Liu J, Zhang H, Zhou Q, Gao X, Chen M, Zhao J, Zhong W, Xu Y, Wang M. Programmed death-ligand 1 tumor proportion score in predicting the safety and efficacy of PD-1/PD-L1 antibody-based therapy in patients with advanced non-small cell lung cancer: A retrospective, multicenter, observational study. Chin Med J (Engl) 2025:00029330-990000000-01563. [PMID: 40413619 DOI: 10.1097/cm9.0000000000003620] [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: 06/13/2024] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND This study aimed to investigate the safety and efficacy of programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) antibody-based therapy in treating patients with advanced non-small cell lung cancer (NSCLC) according to different PD-L1 expression statuses in a real-world setting. METHODS This retrospective, multicenter, observational study enrolled adult patients who received PD-1/PD-L1 antibody-based therapy in China and met the following criteria: (1) had pathologically confirmed, unresectable stage III-IV NSCLC; (2) had a baseline PD-L1 tumor proportion score (TPS); and (3) had confirmed efficacy evaluation results after PD-1/PD-L1 treatment. Logistic regression, Kaplan-Meier analysis, and Cox regression were used to assess the progression-free survival (PFS), overall survival (OS), and immune-related adverse events (irAEs) as appropriate. RESULTS A total of 409 patients, 65.0% (n = 266) with a positive PD-L1 TPS (≥1%) and 32.8% (n = 134) with PD-L1 TPS ≥50%, were included in this study. Cox regression confirmed that patients with a PD-L1 TPS ≥1% had significantly improved PFS (hazard ratio [HR] 0.747, 95% confidence interval [CI] 0.573-0.975, P = 0.032). A total of 160 (39.1%) patients experienced 206 irAEs, and 27 (6.6%) patients experienced 31 grade 3-5 irAEs. The organs most frequently associated with irAEs were the skin (52/409, 12.7%), thyroid (40/409, 9.8%), and lung (34/409, 8.3%). Multivariate logistic regression revealed that a PD-L1 TPS ≥1% (odds ratio [OR] 1.713, 95% CI 1.054-2.784, P = 0.030) was an independent risk factor for irAEs. Other risk factors for irAEs included pretreatment absolute lymphocyte count >2.5 × 109/L (OR 3.772, 95% CI 1.377-10.329, P = 0.010) and pretreatment absolute eosinophil count >0.2 × 109/L (OR 2.006, 95% CI 1.219-3.302, P = 0.006). Moreover, patients who developed irAEs demonstrated improved PFS (13.7 months vs. 8.4 months, P <0.001) and OS (28.0 months vs. 18.0 months, P = 0.007) compared with patients without irAEs. CONCLUSIONS A positive PD-L1 TPS (≥1%) was associated with improved PFS and an increased risk of irAEs in a real-world setting. The onset of irAEs was associated with improved PFS and OS in patients with advanced NSCLC receiving PD-1/PD-L1-based therapy.
Collapse
Affiliation(s)
- Yuequan Shi
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Xiaoyan Liu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Anwen Liu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jian Fang
- Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Qingwei Meng
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
| | - Cuimin Ding
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050033, China
| | - Bin Ai
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Yangchun Gu
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing 100191, China
| | - Cuiying Zhang
- Department of Medical Oncology, Cancer Center, People's Hospital, Hohhot, Inner Mongolia Autonomous Region 750306, China
| | - Chengzhi Zhou
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Yan Wang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Yongjie Shui
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Siyuan Yu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Dongming Zhang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Jia Liu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Haoran Zhang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Qing Zhou
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Xiaoxing Gao
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Minjiang Chen
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Jing Zhao
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Wei Zhong
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Yan Xu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Mengzhao Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
2
|
Sakai T, Udagawa H, Izumi H, Umemura S, Zenke Y, Matsumoto S, Yoh K, Tomoyuki N, Tokiko N, Taki T, Sakamoto N, Sakashita S, Kojima M, Tsuboi M, Goto K, Ishii G. Clinicopathological Characterization of Squamous Cell Lung Carcinoma Adjacent to Emphysema. Pathol Int 2025. [PMID: 40396431 DOI: 10.1111/pin.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 02/25/2025] [Accepted: 05/06/2025] [Indexed: 05/22/2025]
Abstract
The study investigated the clinicopathological features and characteristic immune tumor microenvironment (TME) of lung squamous cell carcinoma (SqCC) adjacent to emphysematous lesions. 184 consecutive patients with peripheral-type SqCC who had undergone complete surgical resection were enrolled. The clinicopathological differences between emphysema-adjacent SqCC (EA-SqCC) and non-emphysema-adjacent SqCC (non-EA-SqCC) were examined. The immune TME, including tumor-infiltrating lymphocytes (TILs) and PD-L1 expression, was also analyzed. EA-SqCC was detected in 132 (71.7%) of the 184 patients. Patients with EA-SqCC had shorter recurrence-free survival (RFS) [median 58.2 months vs. not Reached (NR); hazard ratio (HR) 0.47; 95% CI 0.25-0.81, p < 0.01] and tended to have shorter overall survival (NR vs. NR; HR 0.47; 95% CI 0.27-1.03, p = 0.07) compared to patients with non-EA-SqCC. Evaluation of TILs in the cancer stroma showed the number of Foxp3+ TILs in the EA-SqCC group was significantly higher than that in the non-EA-SqCC group (median number 58 vs. 43, p < 0.01). However, there were no significant differences in the number of CD8 + T cells and the PD-L1 expression between the two groups. Immunosuppressive microenvironment is a characteristic feature of EA-SqCC, which may contribute to the poor prognosis of this disease.
Collapse
Affiliation(s)
- Tetsuya Sakai
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hibiki Udagawa
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hiroki Izumi
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shigeki Umemura
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshitaka Zenke
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shingo Matsumoto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kiyotaka Yoh
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Naito Tomoyuki
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Nakai Tokiko
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tetsuro Taki
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Naoya Sakamoto
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shingo Sakashita
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Motohiro Kojima
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masahiro Tsuboi
- Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Koichi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Genichiro Ishii
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
- Division of Innovative Pathology and Laboratory Medicine, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| |
Collapse
|
3
|
Yang P, Wang X, Yang J, Yan B, Sheng H, Li Y, Yang Y, Wang J. AI-Driven Multiscale Study on the Mechanism of Polygonati Rhizoma in Regulating Immune Function in STAD. ACS OMEGA 2025; 10:19770-19796. [PMID: 40415801 PMCID: PMC12096195 DOI: 10.1021/acsomega.5c00981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 04/26/2025] [Accepted: 04/30/2025] [Indexed: 05/27/2025]
Abstract
Polygonati Rhizoma, a traditional Chinese medicine, has demonstrated immunomodulatory and anticancer properties, yet its precise mechanisms in stomach adenocarcinoma (STAD) remain underexplored. This study aims to uncover the multitarget mechanisms of Polygonati Rhizoma in regulating the tumor immune microenvironment in STAD using artificial intelligence (AI)-driven network pharmacology, bioinformatics, and single-cell RNA sequencing, offering new insights into its immunotherapeutic potential. This study harnessed the power of AI to unravel the molecular mechanisms underlying Polygonati Rhizoma's effects. AI-driven methodologies screened 38 putative constituents, retaining 8 based on ADME criteria. Machine Learning algorithms predicted potential targets, which were cross-referenced with 5,569 immune-related genes from GeneCards, revealing 52 immune-associated targets. Differential expression analysis of the STAD data set identified 18 overlapping DEGs with prognostic significance and immune cell infiltration correlations. Key targets (AKT1, TP53, PTGS2 and VEGFA) emerged as central nodes in the network, with AI-assisted molecular docking confirming strong binding affinities, particularly between diosgenin and these core proteins. Molecular dynamics simulations further validated these interactions. Single-cell RNA sequencing revealed distinct target-gene expression patterns across malignant, stromal, and immune cell subsets in digestive-system tumors. In vitro, Polygonati Rhizoma extract significantly inhibited HGC-27 cell viability and increased intracellular ROS levels. These findings underscore the critical role of AI in integrating multiscale analyses, unveiling a multitarget immunomodulatory and antitumor mechanism for Polygonati Rhizoma in STAD, and providing a foundation for future preclinical and clinical studies.
Collapse
Affiliation(s)
- Peizheng Yang
- School
of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui230038, China
- Anhui
Provincial Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei230038, China
| | - Xiangyu Wang
- School
of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui230038, China
- Anhui
Provincial Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei230038, China
| | - Jianhua Yang
- Anhui
Provincial Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei230038, China
- School
of Medical Informatics Engineering, Anhui University of Chinese
Medicine, Hefei, Anhui230038, China
| | - Biaobiao Yan
- School
of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui230038, China
- Anhui
Provincial Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei230038, China
| | - Haiyang Sheng
- Global
Biometrics and Data Sciences, Bristol Myers
Squibb, Lawrenceville, New Jersey10154, United States
| | - Yan Li
- Key
Laboratory of Industrial Ecology and Environmental Engineering (MOE),
Department of Materials Sciences and Chemical Engineering, Dalian University of Technology, Dalian, Liaoning116023, China
| | - Yinfeng Yang
- Anhui
Provincial Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei230038, China
- School
of Medical Informatics Engineering, Anhui University of Chinese
Medicine, Hefei, Anhui230038, China
| | - Jinghui Wang
- School
of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui230038, China
- Anhui
Provincial Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei230038, China
| |
Collapse
|
4
|
Keshavarz Sadegh R, Saleki K, Rezaei N. Immune checkpoint inhibitor (ICI) therapy in central nervous system cancers: State-of-the-art and future outlook. Int Immunopharmacol 2025; 159:114837. [PMID: 40394797 DOI: 10.1016/j.intimp.2025.114837] [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: 01/11/2025] [Revised: 04/28/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Invasive central nervous system (CNS) cancers are an area where the development of breakthrough therapies is urgently needed. For instance, conditions such as glioblastoma multiforme (GBM) are associated with poor clinical prognosis, with the majority of trials offering no improvement to marginally enhanced survival. Unleashing the potential of targeting the immune system in CNS cancers has gained attention in recent years. Inhibition of immune checkpoints such as CTLA-4, PD-1/PD-L1, TIM-3, and LAG-3 has been attempted in recent trials. While potentially offering a notable edge over other immunotherapies, multi-organ adverse events have been found with the administration of immune checkpoint inhibitors (ICIs). The present review captures the state-of-the-art evidence on ICI treatments in different CNS cancers. Also, we discuss the value of combinational therapies involving ICIs as well as next-generation therapeutics such as bispecific antibodies targeting PD-1/LAG-3/TIM-3 and CRISPR-Cas9-edited PD-1-knock-out checkpoint-resistant CAR T-cells.
Collapse
Affiliation(s)
- Roghaye Keshavarz Sadegh
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; USERN MUBabol Office, Universal Scientific Education and Research Network (USERN), Babol, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Yang Y, Zhu L, Xu Y, Liang L, Liu L, Chen X, Li H, Liu H. The progress and prospects of targeting the adenosine pathway in cancer immunotherapy. Biomark Res 2025; 13:75. [PMID: 40390144 PMCID: PMC12090549 DOI: 10.1186/s40364-025-00784-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/26/2025] [Indexed: 05/21/2025] Open
Abstract
Despite the notable success of cancer immunotherapy, its effectiveness is often limited in a significant proportion of patients, highlighting the need to explore alternative tumor immune evasion mechanisms. Adenosine, a key metabolite accumulating in hypoxic tumor regions, has emerged as a promising target in oncology. Inhibiting the adenosinergic pathway not only inhibits tumor progression but also holds potential to enhance immunotherapy outcomes. Multiple therapeutic strategies targeting this pathway are being explored, ranging from preclinical studies to clinical trials. This review examines the complex interactions between adenosine, its receptors, and the tumor microenvironment, proposing strategies to target the adenosinergic axis to boost anti-tumor immunity. It also evaluates early clinical data on pharmacological inhibitors of the adenosinergic pathway and discusses future directions for improving clinical responses.
Collapse
Affiliation(s)
- Yuying Yang
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Lin Zhu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yantao Xu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Long Liang
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Li Liu
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Xiang Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Hui Li
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Hong Liu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
6
|
Yang A, Zhou M, Gao Y, Zhang Y. Mechanisms of CD8 + T cell exhaustion and its clinical significance in prognosis of anti-tumor therapies: A review. Int Immunopharmacol 2025; 159:114843. [PMID: 40394796 DOI: 10.1016/j.intimp.2025.114843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/22/2025]
Abstract
In recent years, immunotherapy has gradually become one of the main strategies for cancer treatment, with immune checkpoint inhibitors (ICIs) offering new possibilities for tumor therapy. However, some cancer patients exhibit low responses and resistance to ICIs treatment. T cell exhaustion, a process associated with tumor progression, refers to a subset of T cells that progressively lose effector functions and exhibit increased expression of inhibitory receptors. These exhausted T cells are considered key players in the therapeutic efficacy of immune checkpoint inhibitors. Therefore, understanding the impact of T cell exhaustion on tumor immunotherapy and the underlying mechanisms is critical for improving clinical treatment outcomes. Several elegant studies have provided insights into the prognostic value of exhausted T cells in cancers. In this review, we highlight the process of exhausted T cells and its predictive value in various cancers, as well as the relevant mechanisms behind it, providing new insights into the immunotherapy of cancer.
Collapse
Affiliation(s)
- Anrui Yang
- Department of Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Meng Zhou
- Department of Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Yixuan Gao
- Department of Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Ying Zhang
- Department of Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
7
|
Chao CJ, Zhang E, Trinh DN, Udofa E, Lin H, Silvers C, Huo J, He S, Zheng J, Cai X, Bao Q, Zhang L, Phan P, Elgendy SM, Shi X, Burdette JE, Lee SSY, Gao Y, Zhang P, Zhao Z. Integrating antigen capturing nanoparticles and type 1 conventional dendritic cell therapy for in situ cancer immunization. Nat Commun 2025; 16:4578. [PMID: 40379691 DOI: 10.1038/s41467-025-59840-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 05/02/2025] [Indexed: 05/19/2025] Open
Abstract
Eliciting a robust immune response against tumors is often hampered by the inadequate presence of effective antigen presenting cells and their suboptimal ability to present antigens within the immunosuppressive tumor microenvironment. Here, we report a cascade antigen relay strategy integrating antigen capturing nanoparticles (AC-NPs) and migratory type 1 conventional dendritic cells (cDC1s), named Antigen Capturing nanoparticle Transformed Dendritic Cell therapy (ACT-DC), to facilitate in situ immunization. AC-NPs are engineered to capture antigens directly from the tumor and facilitate their delivery to adoptively transferred migratory cDC1s, enhancing antigen presentation to the lymph nodes and reshaping the tumor microenvironment. Our findings suggest that ACT-DC improves in situ antigen collection, triggers a robust systemic immune response without the need for exogenous antigens, and transforms the tumor environment into a more "immune-hot" state. In multiple tumor models including colon cancer, melanoma, and glioma, ACT-DC in combination with immune checkpoint inhibitors eliminates primary tumors in 50-100% of treated mice and effectively rejects two separate tumor rechallenges. Collectively, ACT-DC could provide a broadly effective approach for in situ cancer immunization and tumor microenvironment modulation.
Collapse
Affiliation(s)
- Chih-Jia Chao
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Endong Zhang
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Duong N Trinh
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Edidiong Udofa
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Hanchen Lin
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Caylee Silvers
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jiawei Huo
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Shan He
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Jingtian Zheng
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Xiaoying Cai
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Qing Bao
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Luyu Zhang
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Philana Phan
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Sara M Elgendy
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Xiangqian Shi
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Joanna E Burdette
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
- University of Illinois Cancer Center, Chicago, IL, USA
| | - Steve Seung-Young Lee
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
- University of Illinois Cancer Center, Chicago, IL, USA
| | - Yu Gao
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
- University of Illinois Cancer Center, Chicago, IL, USA
| | - Peng Zhang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA.
- University of Illinois Cancer Center, Chicago, IL, USA.
| |
Collapse
|
8
|
Gao YQ, Tan YJ, Fang JY. Roles of the gut microbiota in immune-related adverse events: mechanisms and therapeutic intervention. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01026-w. [PMID: 40369317 DOI: 10.1038/s41571-025-01026-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2025] [Indexed: 05/16/2025]
Abstract
Immune checkpoint inhibitors (ICIs) constitute a major breakthrough in the field of cancer therapy; their use has resulted in improved outcomes across various tumour types. However, ICIs can cause a diverse range of immune-related adverse events (irAEs) that present a considerable challenge to the efficacy and safety of these treatments. The gut microbiota has been demonstrated to have a crucial role in modulating the tumour immune microenvironment and thus influences the effectiveness of ICIs. Accumulating evidence indicates that alterations in the composition and function of the gut microbiota are also associated with an increased risk of irAEs, particularly ICI-induced colitis. Indeed, these changes in the gut microbiota can contribute to the pathogenesis of irAEs. In this Review, we first summarize the current clinical challenges posed by irAEs. We then focus on reported correlations between alterations in the gut microbiota and irAEs, especially ICI-induced colitis, and postulate mechanisms by which these microbial changes influence the occurrence of irAEs. Finally, we highlight the potential value of gut microbial changes as biomarkers for predicting irAEs and discuss gut microbial interventions that might serve as new strategies for the management of irAEs, including faecal microbiota transplantation, probiotic, prebiotic and/or postbiotic supplements, and dietary modulations.
Collapse
Affiliation(s)
- Ya-Qi Gao
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong-Jie Tan
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
Zhang J, Song Z, Zhang Y, Zhang C, Xue Q, Zhang G, Tan F. Recent advances in biomarkers for predicting the efficacy of immunotherapy in non-small cell lung cancer. Front Immunol 2025; 16:1554871. [PMID: 40406096 PMCID: PMC12095235 DOI: 10.3389/fimmu.2025.1554871] [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: 01/03/2025] [Accepted: 04/18/2025] [Indexed: 05/26/2025] Open
Abstract
Lung cancer continues to be the primary cause of cancer-related deaths globally, with non-small cell lung cancer (NSCLC) accounting for approximately 85% of all instances. Recently, immune checkpoint inhibitors (ICIs) have transformed the treatment approach for NSCLC, however, only a subset of patients experiences significant benefits. Therefore, identifying reliable biomarkers to forecast the efficacy of ICIs is crucial for ensuring the safety and effectiveness of treatments, becoming a major focus of current research efforts. This review highlights the recent advances in predictive biomarkers for the efficacy of ICIs in the treatment of NSCLC, including PD-L1 expression, tertiary lymphoid structures (TLS), tumor-infiltrating lymphocytes (TILs), tumor genomic alterations, transcriptional signatures, circulating biomarkers, and the microbiome. Furthermore, it underscores the pivotal roles of liquid biopsy, sequencing technologies, and digital pathology in biomarker discovery. Special attention is given to the predictive value of TLS, circulating biomarkers, and transcriptional signatures. The review concludes that the integration of multiple biomarkers holds promise for achieving more accurate efficacy predictions and optimizing personalized immunotherapy strategies. By providing a comprehensive overview of the current progress, this review offers valuable insights into biomarker-based precision medicine for NSCLC and outlines future research directions.
Collapse
Affiliation(s)
- Jiacheng Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zehao Song
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuanjie Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chentong Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guochao Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
10
|
Nykaza I, Murciano-Goroff YR, Desilets A, Harada G, Postow MA, Callahan MK, Lee CH, Rudin CM, Kelsen DP, Stadler ZK, Wibmer AG, Hechtman JF, Drilon A, Friedman CF. Sarcoid-like reactions in patients treated with checkpoint inhibitors for advanced solid tumors. Oncologist 2025; 30:oyaf017. [PMID: 40349135 PMCID: PMC12065934 DOI: 10.1093/oncolo/oyaf017] [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: 06/17/2024] [Accepted: 12/23/2024] [Indexed: 05/14/2025] Open
Abstract
IMPORTANCE While new intrathoracic adenopathy in a patient with cancer can represent progression of disease, the differential diagnosis is broad. Sarcoid-like reactions (SLR) remain an underreported source of lymphadenopathy in patients treated with immune checkpoint inhibitors (ICI), with limited reports in patients with cancers other than melanoma. OBJECTIVE To characterize SLRs among patients treated with ICI for advanced solid tumors. METHODS Data were collected on the clinical, pathologic, and radiographic presentation of patients treated with ICI who developed clinical or imaging findings suggestive of an SLR, including the presence of hilar or mediastinal lymphadenopathy, cutaneous/subcutaneous involvement, and/or worsening of existing sarcoidosis on ICI. RESULTS Twelve patients were identified as having experienced an SLR. While 6 patients had melanoma, SLRs were also observed among patients with lung, gynecologic, and genitourinary cancers, including high-grade serous ovarian carcinoma, and an angiomyolipoma. Median time from initiation of ICI to diagnosis of an SLR was 3.4 months (range: 1.8-9.1). All but one patient (92%) were deemed to have had a radiographic response to ICI. CONCLUSIONS AND RELEVANCE Clinicians should maintain the awareness of the possibility of SLRs in patients receiving ICI, particularly in patients whose scans show evidence of "mixed" response, with decreases in certain lesions coupled with new/increasing intrathoracic lymphadenopathy and/or other systemic signs of sarcoid.
Collapse
Affiliation(s)
- Ian Nykaza
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Yonina R Murciano-Goroff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States
| | - Antoine Desilets
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Guilherme Harada
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Michael A Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States
| | - Margaret K Callahan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States
| | - Chung-Han Lee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States
| | - David Paul Kelsen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Andreas G Wibmer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Jaclyn F Hechtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States
| | - Claire F Friedman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States
| |
Collapse
|
11
|
Zhang ZF, Zhang Y, Chen YW, Cao GS, Zheng XD, Sun R, Peng H, Tian ZG, Sun HY. CD200R blockade enhances anti-tumor immunity by unleashing NK and CD8 + T cells in tumor. Acta Pharmacol Sin 2025:10.1038/s41401-025-01556-0. [PMID: 40329005 DOI: 10.1038/s41401-025-01556-0] [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: 11/09/2024] [Accepted: 03/30/2025] [Indexed: 05/08/2025]
Abstract
Immune checkpoint inhibitors have revolutionized cancer therapy, but a large proportion of patients do not respond well to current checkpoint immunotherapies. CD200R (also known as OX2R) is a transmembrane glycoprotein of the immunoglobulin superfamily that is mainly expressed on myeloid and lymphoid-derived immunocompetent cells such as myeloid cells, natural killer (NK), and CD8+ T cells. In this study, we investigated the therapeutic potential and cellular mechanisms of targeting CD200R in tumor immunotherapy. We established 4 subcutaneous tumor mouse models using MC38 (colon cancer), MCA205 (fibrosarcoma), LLC (lung cancer), and EO771 (mammary cancer) cell lines. We found that CD200R was highly expressed on tumor-infiltrating NK and CD8+ T cells with exhausted phenotypes in the four subcutaneous tumor mouse models. Either genetic ablation or antibody blockade of CD200R retarded tumor growth and prolonged the survival of tumor-bearing mice by preventing or reversing exhaustion of both NK cells and CD8+ T cells. The combined therapy of CD200R antibody with anti-PD-1/anti-PD-L1 synergistically inhibited tumor growth. By depletion of NK or/and CD8+ T cells, we demonstrated that both cell types contributed to the anti-tumor efficacy of CD200R blockade in tumor-bearing mice. Further, the blockade of human CD200R significantly enhanced human NK cell function and inhibited human tumor growth in PBMC-reconstituted xenograft mice. Our results demonstrate that CD200R is a potential immune checkpoint molecule that can suppress the tumoricidal activities of NK and CD8+ T cells, and could thus be exploited as a therapeutic target in the future.
Collapse
Affiliation(s)
- Zheng-Feng Zhang
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Yu Zhang
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ya-Wen Chen
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Guo-Shuai Cao
- Hefei TG ImmunoPharma Corporation Limited, Hefei, 230027, China
| | - Xiao-Dong Zheng
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Rui Sun
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Hui Peng
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Zhi-Gang Tian
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Hao-Yu Sun
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
12
|
Joly F, Castel H, Compter A, Nicola C, Duivon M, Lange M. Neuropsychological and central neurologic effects of cancer immunotherapy: the start of a new challenge. J Clin Exp Neuropsychol 2025:1-20. [PMID: 40323211 DOI: 10.1080/13803395.2025.2498713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 04/22/2025] [Indexed: 05/15/2025]
Abstract
INTRODUCTION Cognitive difficulties are frequently reported after cancer treatments, such as chemotherapy or hormone therapy, and have a negative impact on patients' quality of life. Recently, some studies have shown that new cancer treatments, such as immunotherapy agents, can induce cognitive changes. METHOD This review presents the central neurological immune adverse events of immunotherapy treatments including Immune Checkpoint Inhibitors (ICI) and Chimeric Antigen Receptor (CAR) T-cell therapy. The physiopathological mechanisms and risk factors are developed and clinical studies on immunotherapy agents and cognition (among adult patients, using validated questionnaires and/or cognitive tests), psychological factors and quality of life were presented. RESULTS Neurological toxicities are frequently observed with CAR-T cell therapies at acute stage, such as the immune effector cell-associated neurotoxicity syndrome (ICANS), inducing cognitive disorders such as disorientation and aphasia. However, few studies have accurately assessed the impact of immunotherapy on cognition. The methodology of these studies is heterogeneous and they mainly included nonspecific self-report questionnaires of cognitive complaints. Variable results have been obtained concerning the cognitive impact of ICI and CAR-T cell several months following immunotherapy: overall, while some studies reported cognitive difficulties (mainly processing speed and executive functions), the majority has not. Although anxiety and depression are frequently reported in patients treated with ICI or CAR-T cells, these symptoms tend to decrease after the start of immunotherapy. The current neurobiological investigations are too fragmentary to explain neurological symptoms and potential cognitive alteration, but neuroinflammation, vascular inflammation, brain blood barrier disruption, and immune cell brain infiltration would constitute common mechanisms relayed by CAR-T and to a lesser degree, ICI. CONCLUSIONS Acute neurological toxicities following CAR-T cell therapies are a major issue. Further studies are needed to better assess cognitive difficulties after the initiation of immunotherapy, in particular ICI, to better understand the physiopathology, including imaging studies, and risk factors.
Collapse
Affiliation(s)
- Florence Joly
- ANTICIPE U1086 INSERM-UCN, Equipe Labellisée Ligue Contre le Cancer, Centre François Baclesse, Normandie Université UNICAEN, Caen, France
- Services Unit PLATON, Cancer and Cognition Platform, University of Caen Normandy, Caen, France
- Clinical Research Department, Centre François Baclesse, Caen, France
- Medical oncology department, CHU de Caen, Caen, France
| | - Hélène Castel
- Services Unit PLATON, Cancer and Cognition Platform, University of Caen Normandy, Caen, France
- UNIROUEN, INSERM, U1245, Cancer and Brain Genomics, Normandie University, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Annette Compter
- Department of Neuro-Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Celeste Nicola
- UNIROUEN, INSERM, U1245, Cancer and Brain Genomics, Normandie University, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Mylène Duivon
- ANTICIPE U1086 INSERM-UCN, Equipe Labellisée Ligue Contre le Cancer, Centre François Baclesse, Normandie Université UNICAEN, Caen, France
- Services Unit PLATON, Cancer and Cognition Platform, University of Caen Normandy, Caen, France
| | - Marie Lange
- ANTICIPE U1086 INSERM-UCN, Equipe Labellisée Ligue Contre le Cancer, Centre François Baclesse, Normandie Université UNICAEN, Caen, France
- Services Unit PLATON, Cancer and Cognition Platform, University of Caen Normandy, Caen, France
- Clinical Research Department, Centre François Baclesse, Caen, France
| |
Collapse
|
13
|
Abudula M, Astuti Y, Raymant M, Sharma V, Schmid MC, Mielgo A. Macrophages suppress CD8 + T cell cytotoxic function in triple negative breast cancer via VISTA. Br J Cancer 2025:10.1038/s41416-025-03013-5. [PMID: 40316725 DOI: 10.1038/s41416-025-03013-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 03/13/2025] [Accepted: 03/31/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Immunotherapy targeting negative immune checkpoint regulators to enhance the anti-tumour immune response holds promise in the treatment of TNBC. V-domain Ig suppressor of T-cell activation (VISTA) is an immune checkpoint molecule, known to be upregulated and involved in modulating tumour immunity in TNBC. However, how VISTA affects immune response and its therapeutic potential in TNBC remains unclear. METHOD Here, we examined VISTA expression and cellular distribution in TNBC patients' samples and pre-clinical TNBC mouse model. Functional assays were performed to assess the impact of VISTA blockade on macrophage phenotypes, CD8 + T cell infiltration and activation, and overall anti-tumour immune response. RESULTS In this study we show that VISTA expression levels are increased in TNBC patients' samples and pre-clinical mouse models compared to non-involved breast tissue and VISTA is mainly expressed on tumour infiltrating macrophages and neutrophils. Blocking VISTA reverts macrophages immunosuppressive phenotypes, increases CD8 + T cell infiltration and activation, and enhances an anti-tumour immune response. Mechanistically, we show that neutralising VISTA on macrophages enhances their immune-stimulatory functions and inhibits the suppressive effect of macrophages on CD8 + T cells activation. CONCLUSION These findings provide the rationale for the development of anti-VISTA targeting strategies in the treatment of TNBC.
Collapse
Affiliation(s)
- Maidinaimu Abudula
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Yuliana Astuti
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Meirion Raymant
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Vijay Sharma
- School of Medicine and Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
- Department of Cellular Pathology, Liverpool Clinical Laboratories, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Michael C Schmid
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Ainhoa Mielgo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK.
| |
Collapse
|
14
|
Nakai S, Karayama M, Inoue Y, Yasui H, Hozumi H, Suzuki Y, Furuhashi K, Fujisawa T, Enomoto N, Funayama S, Ichikawa S, Goshima S, Inui N, Suda T. Utility of serum Krebs von den Lungen-6 and surfactant protein-D levels for the diagnosis of immune checkpoint inhibitor-induced immune-related pneumonitis. Respir Investig 2025; 63:259-264. [PMID: 39970788 DOI: 10.1016/j.resinv.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/24/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Immune-related pneumonitis (irP) is a severe immune-related adverse event that can occur after treatment with immune checkpoint inhibitors (ICIs); accurate monitoring and early diagnosis are crucial. Serum Krebs von den Lungen-6 (KL-6) and surfactant protein-D (SP-D) are used to diagnose and monitor the disease activity of various types of interstitial lung disease. This study evaluated the usefulness of KL-6 and SP-D for diagnosing irP in cancer patients receiving ICIs. PATIENTS AND METHODS This retrospective observational study included cancer patients treated with more than two cycles of ICIs from September 2014 to October 2023. Receiver operating characteristic (ROC) analysis was used to assess the diagnostic utility of KL-6 and SP-D. RESULTS Of the 724 cancer patients, 631 were included in the analysis, and 64 developed irP. The ROC analysis showed that KL-6 and SP-D had areas under the curve of 0.803 and 0.845, respectively. Serum KL-6 ≥ 500 U/mL had a sensitivity of 65.6% and a specificity of 83.4% for the diagnosis of irP, while SP-D ≥ 110 ng/mL had a sensitivity of 66.7% and a specificity of 88.6%. Combining both KL-6 ≥ 500 U/mL and SP-D ≥ 110 ng/mL resulted in a specificity of 96.6%, with a sensitivity of 46.7%. Combining either KL-6 ≥ 500 U/mL or SP-D ≥ 110 ng/mL resulted in a sensitivity of 85.0%, with a specificity of 75.5%. SP-D levels were significantly associated with irP severity, while KL-6 levels were not. CONCLUSIONS Serum KL-6 and SP-D are useful for diagnosing irP in cancer patients receiving ICIs.
Collapse
Affiliation(s)
- Shogo Nakai
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan
| | - Masato Karayama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan; Department of Chemotherapy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan.
| | - Yusuke Inoue
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan
| | - Hideki Yasui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan
| | - Hironao Hozumi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan
| | - Yuzo Suzuki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan
| | - Kazuki Furuhashi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan
| | - Tomoyuki Fujisawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan
| | - Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan
| | - Satoshi Funayama
- Department of Radiology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan
| | - Shintaro Ichikawa
- Department of Radiology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan
| | - Satoshi Goshima
- Department of Radiology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan; Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan
| |
Collapse
|
15
|
Wang Q, Chen Z, Lu X, Lin H, Feng H, Weng N, Chen L, Liu M, Long L, Huang L, Deng Y, Zheng K, Zheng X, Li Y, Cai T, Zheng J, Yang W. Methionine Metabolism Dictates PCSK9 Expression and Antitumor Potency of PD-1 Blockade in MSS Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2501623. [PMID: 40125618 PMCID: PMC12097065 DOI: 10.1002/advs.202501623] [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] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Indexed: 03/25/2025]
Abstract
Nutrient metabolisms are vitally interrelated to cancer progression and immunotherapy. However, the mechanisms by which nutrient metabolisms interact to remodel immune surveillance within the tumor microenvironment remain largely unexplored. Here it is demonstrated that methionine restriction inhibits the expression of proprotein convertase subtilisin/kexin type 9 (PCSK9), a key regulator of cholesterol homeostasis and a potential target for cancer immunotherapy, in colorectal cancer (CRC) but not in the liver. Mechanistically, methionine is catabolized to S-adenosylmethionine (SAM), promoting mRNA transcription of PCSK9 through increased DNA methyltransferase 1 (DNMT1)-mediated DNA methylation and suppression of sirtuin 6 (SIRT6) expression. Furthermore, both PCSK9 inhibition and dietary methionine restriction (DMR) potentiate PD-1 blockade therapy and foster the infiltration of CD8+ T cells in Colon 26 tumor-bearing mice-a proficient mismatch repair (pMMR)/microsatellite stable (MSS) CRC model that exhibits limited response to anti-PD-1 therapy. Moreover, combining 5-fluorouracil (5-FU) chemotherapy with PCSK9 inhibition and PD-1 blockade further augments therapeutic efficacy for MSS CRC. The findings establish a mechanistic link between amino acid metabolism and cholesterol metabolism within the tumor microenvironment where tumor cells sense methionine to regulate PCSK9 expression, highlighting promising combination therapeutic strategies that may greatly benefit MSS CRC patients.
Collapse
Affiliation(s)
- Qi‐Long Wang
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Zijie Chen
- Guangdong Provincial Key Laboratory of Molecular Oncologic PathologyDepartment of PathologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Xiaofei Lu
- Guangdong Provincial Key Laboratory of Molecular Oncologic PathologyDepartment of PathologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Huizhen Lin
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Guangdong Provincial Key Laboratory of Molecular Oncologic PathologyDepartment of PathologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Huolun Feng
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Nuozhou Weng
- Department of General SurgeryZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Liwen Chen
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Guangdong Provincial Key Laboratory of Molecular Oncologic PathologyDepartment of PathologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Mengnan Liu
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Guangdong Provincial Key Laboratory of Molecular Oncologic PathologyDepartment of PathologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Li Long
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Guangdong Provincial Key Laboratory of Molecular Oncologic PathologyDepartment of PathologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Lingjun Huang
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Yongmei Deng
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Kehong Zheng
- Department of General SurgeryZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Xiaojun Zheng
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Yong Li
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Ting Cai
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Jiabin Zheng
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Wei Yang
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Guangdong Provincial Key Laboratory of Molecular Oncologic PathologyDepartment of PathologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| |
Collapse
|
16
|
A N, Zou F, Chen M, Liu M, Zhang H, Cheng S, Liu Y. FAM72 family members serves as prognostic biomarker in liver hepatocellular carcinoma. Pathol Res Pract 2025; 269:155893. [PMID: 40081285 DOI: 10.1016/j.prp.2025.155893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/14/2025] [Accepted: 03/02/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Liver hepatocellular carcinoma (LIHC) is a common cancer with poor prognosis. The FAM72 gene family enhances neuronal self-renewal, potentially increasing tumor formation, but its functional and predictive relevance in LIHC remains unclear. We sought to investigate the function of the FAM72 gene family in LIHC in the present study. METHODS We acquired TCGA-LIHC expression and phenotypic data as well as extensive clinicopathologic information from the UCSC Xena Database (https://xenabrowser.net/datapages/) database. We analyzed the association between FAM72 gene family expression in LIHC and patient prognosis and immune infiltration; Genomic and functional enrichment analysis for FAM72 genes was analyzed. Finally, Western blot method, quantitative real-time polymerase chain reaction and CCK8 detection and cell invasion experiments were used to verify the effect of FAM72A expression on LIHC. RESULTS The expression of FAM72 gene family is different between LIHC and normal liver tissues. The expression of FAM72 gene family increased with increasing grading of LIHC tissues. The expression of FAM72 gene family was significantly reduced in LIHC stage IV. LIHC tissues expressed significantly more FAM72 genes than did normal tissues at the T stage (p < 0.001,). which has a good value in the diagnosis of LIHC (AUC greater than 0.85), and was strongly linked with the tumor stage in LIHC. Based on Cox analysis of univariate data, the FAM72 gene family was associated with poor overall survival (OS) in patients with LIHC. Analysis of multifactorial Cox data revealed an independent relationship between FAM72 expression and OS. Increased FAM72 gene expression is associated with poor survival rates and immune cell infiltration. Methylation levels were associated with the prognosis of patients with LIHC. Ultimately, our findings revealed that FAM72A is abundantly expressed in LIHC cells, facilitating proliferation and metastasis. CONCLUSION These findings indicate that the FAM72 gene family is a potential molecular marker for poor prognosis in LIHC, providing additional insights into the development of therapeutic approaches and prognostic markers.
Collapse
Affiliation(s)
- Naer A
- Thyroid and Breast Surgery Department, Shenzhen Bao'an District Songgang People's Hospital, No.2 Shajiang Road, Shenzhen, Guangdong, China
| | - Feilong Zou
- Thyroid and Breast Surgery Department, Shenzhen Bao'an District Songgang People's Hospital, No.2 Shajiang Road, Shenzhen, Guangdong, China
| | - Meiyan Chen
- Thyroid and Breast Surgery Department, Shenzhen Bao'an District Songgang People's Hospital, No.2 Shajiang Road, Shenzhen, Guangdong, China
| | - Meiling Liu
- Thyroid and Breast Surgery Department, Shenzhen Bao'an District Songgang People's Hospital, No.2 Shajiang Road, Shenzhen, Guangdong, China
| | - Huishan Zhang
- Thyroid and Breast Surgery Department, Shenzhen Bao'an District Songgang People's Hospital, No.2 Shajiang Road, Shenzhen, Guangdong, China
| | - Shaohua Cheng
- Thyroid and Breast Surgery Department, Shenzhen Bao'an District Songgang People's Hospital, No.2 Shajiang Road, Shenzhen, Guangdong, China.
| | - Yunhong Liu
- Thyroid and Breast Surgery Department, Shenzhen Bao'an District Songgang People's Hospital, No.2 Shajiang Road, Shenzhen, Guangdong, China.
| |
Collapse
|
17
|
Saeidpour Masouleh S, Nasiri K, Ostovar Ravari A, Saligheh Rad M, Kiani K, Sharifi Sultani A, Nejati ST, Nabi Afjadi M. Advances and challenges in CAR-T cell therapy for head and neck squamous cell carcinoma. Biomark Res 2025; 13:69. [PMID: 40312353 PMCID: PMC12044960 DOI: 10.1186/s40364-025-00783-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) remains among the most aggressive malignancies with limited treatment options, especially in recurrent and metastatic cases. Despite advances in surgery, radiotherapy, chemotherapy, and immune checkpoint inhibitors, survival rates remain suboptimal due to tumor heterogeneity, immune evasion, and treatment resistance. In recent years, Chimeric Antigen Receptor (CAR) T-cell therapy has revolutionized hematologic cancer treatment by genetically modifying T cells to target tumor-specific antigens like CD19, CD70, BCMA, EGFR, and HER2, leading to high remission rates. Its success is attributed to precise antigen recognition, sustained immune response, and long-term immunological memory, though challenges like cytokine release syndrome and antigen loss remain. Notably, its translation to solid tumors, including HNSCC, faces significant challenges, such as tumor microenvironment (TME)-induced immunosuppression, antigen heterogeneity, and limited CAR T-cell infiltration. To address these barriers, several tumor-associated antigens (TAAs), including EGFR, HER2 (ErbB2), B7-H3, CD44v6, CD70, CD98, and MUC1, have been identified as potential CAR T-cell targets in HNSCC. Moreover, innovative approaches, such as dual-targeted CAR T-cells, armored CARs, and CRISPR-engineered modifications, aim to enhance efficacy and overcome resistance. Notably, combination therapies integrating CAR T-cells with immune checkpoint inhibitors (e.g., PD-1/CTLA-4 blockade) and TGF-β-resistant CAR T designs are being explored to improve therapeutic outcomes. This review aimed to elucidate the current landscape of CAR T-cell therapy in HNSCC, by exploring its mechanisms, targeted antigens, challenges, emerging strategies, and future therapeutic potential.
Collapse
Affiliation(s)
| | - Kamyar Nasiri
- Faculty of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Ava Ostovar Ravari
- Faculty of Dentistry, Haybusak University of Medical Sciences, Yerevan, Armenia
| | - Mona Saligheh Rad
- Faculty of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Kiarash Kiani
- Faculty of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | | | | | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
18
|
Bai J, Gao Y, Zhang G. The treatment of breast cancer in the era of precision medicine. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0510. [PMID: 40269562 PMCID: PMC12032834 DOI: 10.20892/j.issn.2095-3941.2024.0510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/05/2025] [Indexed: 04/25/2025] Open
Abstract
The management of breast cancer, one of the most common and heterogeneous malignancies, has transformed with the advent of precision medicine. This review explores current developments in genetic profiling, molecular diagnostics, and targeted therapies that have revolutionized breast cancer treatment. Key innovations, such as cyclin-dependent kinases 4/6 (CDK4/6) inhibitors, antibody-drug conjugates (ADCs), and immune checkpoint inhibitors (ICIs), have improved outcomes for hormone receptor-positive (HR+), HER2-positive (HER2+), and triple-negative breast cancer (TNBC) subtypes remarkably. Additionally, emerging treatments, such as PI3K inhibitors, poly (ADP-ribose) polymerase (PARP) inhibitors, and mRNA-based therapies, offer new avenues for targeting specific genetic mutations and improving treatment response, particularly in difficult-to-treat breast cancer subtypes. The integration of liquid biopsy technologies provides a non-invasive approach for real-time monitoring of tumor evolution and treatment response, thus enabling dynamic adjustments to therapy. Molecular imaging and artificial intelligence (AI) are increasingly crucial in enhancing diagnostic precision, personalizing treatment plans, and predicting therapeutic outcomes. As precision medicine continues to evolve, it has the potential to significantly improve survival rates, decrease recurrence, and enhance quality of life for patients with breast cancer. By combining cutting-edge diagnostics, personalized therapies, and emerging treatments, precision medicine can transform breast cancer care by offering more effective, individualized, and less invasive treatment options.
Collapse
Affiliation(s)
- Jingwen Bai
- The Breast Center of Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University & Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Yiyang Gao
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, School of Medicine, Xiamen University, Xiamen 361100, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361100, China
| | - Guojun Zhang
- The Breast Center of Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University & Peking University Cancer Hospital Yunnan, Kunming 650118, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, School of Medicine, Xiamen University, Xiamen 361100, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361100, China
| |
Collapse
|
19
|
Fan Z, Yi Z, Li S, He J. Parabacteroides distasonis promotes CXCL9 secretion of tumor-associated macrophages and enhances CD8 +T cell activity to trigger anti-tumor immunity against anti-PD-1 treatment in non-small cell lung cancer mice. BMC Biotechnol 2025; 25:30. [PMID: 40241108 PMCID: PMC12004837 DOI: 10.1186/s12896-025-00963-9] [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: 09/26/2024] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Parabacteroides distasonis (P. distasonis) could regulate inflammatory markers, promote intestinal barrier integrity, and block tumor formation in colon. However, the regulatory effect of P. distasonis on non-small cell lung cancer (NSCLC) remains unknown. This study aimed to investigate the regulatory effect of P. distasonis on NSCLC and its impact on tumor immunity. METHODS We first established a mouse model of Lewis lung cancer, and administered P. distasonis and intrabitoneal injection of anti-mouse PD-1 monoclonal antibody to assess the impact of P. distasonis on tumor immunity, and mouse intestinal barrier. Then, we explored the effect of P. distasonis on CD8+T cells and CXCL9 secretion mediated by tumor-associated macrophages (TAM). We used the TLR1/2 complex inhibitor CPT22 to evaluate its effect on macrophage activation. Finally, we explored the effect of P. distasonis on CD8+T cells and CXCL9 secreted by TAM in vivo. RESULTS In vivo, P. distasonis enhanced anti-tumor effects of anti-PD-1 in NSCLC mice, improved intestinal barrier integrity, recruited macrophages, and promoted M1 polarization. In vitro, CD86 and iNOS levels in BMDM were elevated and CD206 and Arg1 levels were suppressed in membrane fraction of P. distasonis (PdMb) group in comparison to Control group. With additional CPT22 pre-treatment, the levels of CD86 and iNOS in BMDM were reduced, and the levels of CD206 and Arg1 were increased. Compared to PBS group, P. distasonis group exhibited higher proportion of CD8+T cells in tumor tissues, along with increased positive proportion of GZMB and IFN-γ in CD8+T cells. Additionally, in comparison to Control group, PdMb group showed an elevated proportion of GZMB+T and IFN-γ+T cells within CD8+T cells, and secretion of IFN-γ, TNF-α, perforin, and GZMB in CD8+T cell supernatant increased. Moreover, the proportion of CXCL9+F4/80+ macrophages in tumor tissues was higher in P. distasonis group compared to PBS group. In comparison to Control group, CXCL9 protein level in BMDM and CXCL9 secretion level in BMDM supernatant were increased in PdMb group. Finally, P. distasonis enhanced CD8+T cell activity by secreting CXCL9 from macrophages in vivo. CONCLUSIONS P. distasonis promoted CXCL9 secretion of TAM and enhanced CD8+T cell activity to trigger anti-tumor immunity against anti-PD-1 treatment in NSCLC mice.
Collapse
Affiliation(s)
- Zhijun Fan
- Department of Cardiothoracic Surgery, The People's Hospital of Liuyang, Changsha, China
| | - Zheng Yi
- Department of Cardiothoracic Surgery, The People's Hospital of Liuyang, Changsha, China
| | - Sheng Li
- Department of Gastrointestinal Surgery, The Central Hospital of Shaoyang, Shaoyang, China
| | - Junjun He
- Department of Gastrointestinal Surgery, The Central Hospital of Shaoyang, Shaoyang, China.
| |
Collapse
|
20
|
Sakaguchi H, Nanjo S, Sato S, Kotani H, Nishiyama A, Yamashita K, Ohtsubo K, Suzuki C, Shimojima M, Yano S, Takeuchi S. Effectiveness of Additional Immunosuppressive Drugs for Corticosteroid-refractory Immune Checkpoint Inhibitor-induced Myocarditis: Two Case Reports. Intern Med 2025; 64:1205-1210. [PMID: 39231667 PMCID: PMC12097839 DOI: 10.2169/internalmedicine.4162-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) improve the outcomes of several types of cancer. However, they are also associated with various immune-related adverse events including myocarditis. ICI-induced myocarditis is a rare, potentially life-threatening adverse event. We herein report two cases of corticosteroid-refractory ICI-induced myocarditis. In both cases, additional immunosuppressive therapies, such as intravenous immunoglobulin and tacrolimus, successfully resolved myocarditis. Given the corticosteroid-refractory nature of these cases, we suggest that prompt addition of other immunosuppressive drugs to corticosteroid therapy should be considered in the treatment of ICI-induced myocarditis.
Collapse
Affiliation(s)
| | - Shigeki Nanjo
- Department of Respiratory Medicine, Kanazawa University Hospital, Japan
| | - Shigeki Sato
- Department of Medical Oncology, Kanazawa University Hospital, Japan
| | - Hiroshi Kotani
- Department of Medical Oncology, Kanazawa University Hospital, Japan
| | | | - Kaname Yamashita
- Department of Medical Oncology, Kanazawa University Hospital, Japan
| | - Koushiro Ohtsubo
- Department of Medical Oncology, Kanazawa University Hospital, Japan
| | - Chiaki Suzuki
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Japan
| | - Masaya Shimojima
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Japan
| | - Seiji Yano
- Department of Respiratory Medicine, Kanazawa University Hospital, Japan
| | - Shinji Takeuchi
- Department of Medical Oncology, Kanazawa University Hospital, Japan
| |
Collapse
|
21
|
Ding C, Zhang Y, Xia T, Li J, Yao W, Zhang Q, Han Z, Wang J, Cao Z, Hu J, Wei L. Perioperative the BTLA inhibitor (tifcemalimab) combined with toripalimab and chemotherapy for resectable locally advanced thoracic esophageal squamous cell carcinoma trial (BT-NICE trial): a prospective, single-arm, exploratory study. Front Immunol 2025; 16:1542877. [PMID: 40276504 PMCID: PMC12018478 DOI: 10.3389/fimmu.2025.1542877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/18/2025] [Indexed: 04/26/2025] Open
Abstract
Background The treatment of cancer has brought about a paradigm shift with the introduction of immune checkpoint blockade (ICB) therapy, which is mostly dependent on inhibiting PD-1/PD-L1 and CTLA-4. However, recent studies have shown limited efficacy of this treatment in esophageal squamous cell carcinoma (ESCC). Preliminary studies have found that tifcemalimab (the world's first anti-BTLA blocking monoclonal antibody) combined with toripalimab (PD-1) and chemotherapy has shown favorable safety and efficacy in several solid cancers. This study aimed to evaluate the safety and efficacy of neoadjuvant tifcemalimab combined with toripalimab and chemotherapy following esophagectomy for resectable ESCC, and the association of adjuvant immunotherapy with improved survival outcomes. Methods Patients with pathologically confirmed cT1b-3N1-3M0 or cT2-3N0M0 thoracic ESCC were treated with neoadjuvant tifcemalimab (200mg, iv, d1) in combination with toripalimab (240mg, iv, d1) and chemotherapy (paclitaxel 135-175 mg/m2, d1 + cisplatin 75 mg/m2, d1) every 3 weeks for 2 cycles. Patients undergoing esophagectomy with pathological complete response (pCR) were administered up to 15 cycles of adjuvant tifcemalimab (200 mg) and toripalimab (240 mg), whereas patients without pCR received tifcemalimab in combination with toripalimab and adjuvant chemotherapy for 2 cycles, followed by tifcemalimab in combination with toripalimab immunotherapy up to 13 cycles. The patient with incomplete resection was decided to receive radiotherapy after a multidisciplinary consultation. The primary endpoint of this study was the pCR rate. The secondary endpoints include major pathological response rate (MPR), objective response rate (ORR), disease control rate (DCR), adverse events, R0 resection rate, event-free survival (EFS), and overall survival (OS). Discussion The Ethics Committee of Henan Provincial People's Hospital has approved the protocol (No 2024-132-03). This study is the world's first prospective clinical trial to evaluate the safety and efficacy of the BTLA inhibitor in combination with PD-1 and chemotherapy as neoadjuvant/adjuvant therapy for locally advanced thoracic ESCC. We predicted that perioperative combination immunotherapy as a potentially preferred and effective treatment strategy may lead to better survival outcomes.
Collapse
Affiliation(s)
- Chengzhi Ding
- Department of Thoracic Surgery, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Yahao Zhang
- Department of Thoracic Surgery, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Tian Xia
- Department of Thoracic Surgery, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Jiwei Li
- Department of Thoracic Surgery, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Wenjian Yao
- Department of Thoracic Surgery, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Quan Zhang
- Department of Thoracic Surgery, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Zhijun Han
- Department of Thoracic Surgery, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Jianjun Wang
- Department of Thoracic Surgery, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Zhikun Cao
- Department of Thoracic Surgery, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Jinlong Hu
- Department of Oncology, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Li Wei
- Department of Thoracic Surgery, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital, Zhengzhou, China
| |
Collapse
|
22
|
Kulkarni AD, Mukarrama T, Barlow BR, Kim J. Recent advances in non-invasive in vivo tracking of cell-based cancer immunotherapies. Biomater Sci 2025; 13:1939-1959. [PMID: 40099377 PMCID: PMC11980607 DOI: 10.1039/d4bm01677g] [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] [Indexed: 03/19/2025]
Abstract
Immunotherapy has been at the forefront of cancer treatment research in recent years due to an increased understanding of the immune system's role in cancer and the substantial benefits it has demonstrated compared to conventional treatment methods. In particular, immune cell-based approaches utilizing T cells, natural killer (NK) cells, macrophages, and more have shown great potential as cancer treatments. While these treatments hold promise, there are still numerous issues that limit their clinical translation, including a lack of understanding of their mechanisms and inconsistent responses to treatment. Traditionally, tissue or blood samples are collected as a means of monitoring treatment progression. However, these in vitro diagnostics are invasive and provide limited information about the real-time status of the treatment or its long-term effectiveness. To address these limitations, novel non-invasive imaging modalities have been developed. These include optical imaging, X-ray computed tomography (CT), magnetic resonance imaging (MRI), positron emission tomography (PET) and single-photon emission computed tomography (SPECT), and photoacoustic (PA) imaging. This review focuses on methods for tracking cell-based cancer immunotherapies using these in vivo imaging modalities, thereby enhancing real-time monitoring of their therapeutic effect and predictions of their long-term efficacy.
Collapse
Affiliation(s)
- Anika D Kulkarni
- Department of Biomedical Engineering, University of California, Davis, Davis, 95616, USA.
| | - Tasneem Mukarrama
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, 95817, USA
| | - Brendan R Barlow
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, 95817, USA
| | - Jinhwan Kim
- Department of Biomedical Engineering, University of California, Davis, Davis, 95616, USA.
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, 95817, USA
| |
Collapse
|
23
|
Jiang Q, Braun DA, Clauser KR, Ramesh V, Shirole NH, Duke-Cohan JE, Nabilsi N, Kramer NJ, Forman C, Lippincott IE, Klaeger S, Phulphagar KM, Chea V, Kim N, Vanasse AP, Saad E, Parsons T, Carr-Reynolds M, Carulli I, Pinjusic K, Jiang Y, Li R, Syamala S, Rachimi S, Verzani EK, Stevens JD, Lane WJ, Camp SY, Meli K, Pappalardi MB, Herbert ZT, Qiu X, Cejas P, Long HW, Shukla SA, Van Allen EM, Choueiri TK, Churchman LS, Abelin JG, Gurer C, MacBeath G, Childs RW, Carr SA, Keskin DB, Wu CJ, Kaelin WG. HIF regulates multiple translated endogenous retroviruses: Implications for cancer immunotherapy. Cell 2025; 188:1807-1827.e34. [PMID: 40023154 PMCID: PMC11988688 DOI: 10.1016/j.cell.2025.01.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 11/14/2024] [Accepted: 01/31/2025] [Indexed: 03/04/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC), despite having a low mutational burden, is considered immunogenic because it occasionally undergoes spontaneous regressions and often responds to immunotherapies. The signature lesion in ccRCC is inactivation of the VHL tumor suppressor gene and consequent upregulation of the HIF transcription factor. An earlier case report described a ccRCC patient who was cured by an allogeneic stem cell transplant and later found to have donor-derived T cells that recognized a ccRCC-specific peptide encoded by a HIF-responsive endogenous retrovirus (ERV), ERVE-4. We report that ERVE-4 is one of many ERVs that are induced by HIF, translated into HLA-bound peptides in ccRCCs, and capable of generating antigen-specific T cell responses. Moreover, ERV expression can be induced in non-ccRCC tumors with clinical-grade HIF stabilizers. These findings have implications for leveraging ERVs for cancer immunotherapy.
Collapse
Affiliation(s)
- Qinqin Jiang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - David A Braun
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Yale Center of Cellular and Molecular Oncology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Karl R Clauser
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Vijyendra Ramesh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Nitin H Shirole
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Joseph E Duke-Cohan
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Nicholas J Kramer
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Cleo Forman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Isabelle E Lippincott
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Susan Klaeger
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Kshiti M Phulphagar
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Vipheaviny Chea
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Nawoo Kim
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Allison P Vanasse
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Eddy Saad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | | | | | - Isabel Carulli
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Katarina Pinjusic
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Yijia Jiang
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Rong Li
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sudeepa Syamala
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Suzanna Rachimi
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Eva K Verzani
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Jonathan D Stevens
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02215, USA
| | - William J Lane
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02215, USA
| | - Sabrina Y Camp
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Kevin Meli
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | | | - Zachary T Herbert
- Molecular Biology Core Facilities, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Xintao Qiu
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Paloma Cejas
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Henry W Long
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sachet A Shukla
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Toni K Choueiri
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA
| | - L Stirling Churchman
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Jennifer G Abelin
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | | | | | - Richard W Childs
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Steven A Carr
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA.
| | - Derin B Keskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Computer Science, Metropolitan College, Boston University, Boston, MA 02215, USA; Section for Bioinformatics, Department of Health Technology, Technical University of Denmark 2800 Lyngby, Denmark.
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA.
| | - William G Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
24
|
Xue J, Liu C, Shao J, Wang L, Han Y, Wang J, Wang J. Predictive Value of Neutrophil-to-Lymphocyte Ratio for Immune Checkpoint Inhibitor-Related Myocarditis Among Patients Treated for Non-Small-Cell Lung Cancer. CANCER INNOVATION 2025; 4:e163. [PMID: 39981496 PMCID: PMC11840423 DOI: 10.1002/cai2.163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 09/10/2024] [Accepted: 11/05/2024] [Indexed: 02/22/2025]
Abstract
Background The predictive value of the neutrophil-to-lymphocyte ratio (NLR) for immune checkpoint inhibitors (ICIs) in various tumors remains uncertain despite its use in forecasting the effectiveness of immunotherapy. The purpose of our research was to determine the prognostic significance of NLR for immune checkpoint inhibitor-related myocarditis in non-small-cell lung cancer (NSCLC) patients. Methods We enrolled and monitored patients with NSCLC who received ICI therapy at the Fifth Medical Center of Chinese PLA General Hospital between January 1, 2018, and February 20, 2021. NLR was determined before and soon after each cycle of ICIs. All participants in this study were periodically examined for troponin and brain natriuretic peptide (BNP), and an electrocardiogram (ECG) and echocardiography were done. Cox's proportional hazards regression model and receiver operating characteristic (ROC) were used to assess the predictive value for ICI-related myocarditis. Results A total of 146 patients received ICI treatment and completed a follow-up. Of these, 17 patients (11.64%) developed ICI-related myocarditis that met the diagnostic criteria. The initial cycle revealed that the NLR was a reliable predictor of potential myocarditis related to ICIs, with an area under the curve (AUC) of 0.833 and a 95% confidence interval (CI) of 0.721-0.945. Following the initial round of ICI treatment, an NLR elevation (NLR ≥ 3.25) appeared to be the most significant standalone indicator of ICI-related myocarditis (HR: 11.094; 95% CI: 3.186-38.631; p < 0.001). Conclusions Our study confirmed that NLR elevation in the early phase after ICI treatment of NSCLC is a reliable predictive factor of ICI-related myocarditis. Regular and frequent cardiac monitoring may help to avoid the occurrence of severe and fatal cases.
Collapse
Affiliation(s)
- Jian Xue
- Senior Department of CardiologyThe Sixth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Chuanbin Liu
- Western Medical Branch of Chinese PLA General HospitalBeijingChina
- Department of EmergencyThe First Medical Center of Chinese PLA General HospitalBeijingChina
| | - Jun Shao
- Department of General SurgeryThe First Medical Center of Chinese PLA General HospitalBeijingChina
| | - Li Wang
- Western Medical Branch of Chinese PLA General HospitalBeijingChina
| | - Yating Han
- Western Medical Branch of Chinese PLA General HospitalBeijingChina
| | - Jing Wang
- Department of General SurgeryThe First Medical Center of Chinese PLA General HospitalBeijingChina
| | - Jinda Wang
- Senior Department of CardiologyThe Sixth Medical Center of Chinese PLA General HospitalBeijingChina
| |
Collapse
|
25
|
Wirth TC, Saborowski A, Kuehnle E, Fischer M, Bültmann E, von Kaisenberg C, Merten R. Chemo- and Radiotherapy of Gastrointestinal Tumors during Pregnancy. Visc Med 2025; 41:64-73. [PMID: 40201111 PMCID: PMC11975343 DOI: 10.1159/000540428] [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: 03/24/2024] [Accepted: 07/16/2024] [Indexed: 04/10/2025] Open
Abstract
Background Gastrointestinal cancers account for approximately one-fourth of all cancer cases and one-third of all tumor-related deaths worldwide. For the most frequent gastrointestinal tumor entities including colorectal, gastric, esophageal, and liver cancer, the incidence is expected to increase by more than 50% until 2040. While most gastrointestinal cancers are diagnosed beyond the age of fertility and predominantly in men, the increasing incidence of gastrointestinal malignancies in patients below the age of fifty suggests a growing importance in women of childbearing age. While localized cancers in pregnant women can either be monitored or treated surgically, more advanced stages might require radio- or chemotherapy to control tumor growth until delivery. Under these circumstances, critical decisions have to be made to preserve maternal health on the one side and minimize harm to the infant on the other side. Summary Here we summarize data from case reports, meta-analyses, and registries of women undergoing radio- or chemotherapy during pregnancy and provide guidance for therapeutic decision-making in pregnant women suffering from gastrointestinal cancers. Key Message After the first trimester, most chemotherapeutic regimens can be safely administered to pregnant patients with gastrointestinal cancers. With appropriate safety measures, both radiotherapy and radiochemotherapy can be applied to pregnant patients with rectal cancers.
Collapse
Affiliation(s)
- Thomas Christian Wirth
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Anna Saborowski
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Elna Kuehnle
- Department of Obstetrics and Gynaecology, Hannover Medical School, Hannover, Germany
| | - Mirko Fischer
- Department of Radiation Oncology, Hannover Medical School, Hannover, Germany
| | - Eva Bültmann
- Institute of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
| | | | - Roland Merten
- Department of Radiation Oncology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
26
|
Braghieri L, Gharaibeh A, Nkashama L, Abushouk A, Abushawer O, Mehdizadeh‐Shrifi A, Honnekeri B, Calabrese C, Menon V, Funchain P, Collier P, Sadler D, Moudgil R. Long-term cardiovascular outcomes of immune checkpoint inhibitor-related myocarditis: A large single-centre analysis. ESC Heart Fail 2025; 12:1237-1245. [PMID: 39482568 PMCID: PMC11911570 DOI: 10.1002/ehf2.15131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/11/2024] [Accepted: 10/03/2024] [Indexed: 11/03/2024] Open
Abstract
AIMS Immune checkpoint inhibitors (ICI) are the cornerstone of modern oncology; however, side effects such as ICI-related myocarditis (irM) can be fatal. Recently, Bonaca proposed criteria for irM; however, it is unknown if they correlate well with cardiovascular (CV) ICI-related adverse events. Additionally, whether incident irM portends worse long-term CV outcomes remains unclear. We aimed to determine the incidence of long-term CV comorbidities and CV mortality among irM patients. PATIENTS AND METHODS The ICI-related adverse event (irAE) registry was queried to identify irM patients by using Bonaca criteria. Random controls were selected after excluding patients with other concomitant irAEs. Patients' demographics, comorbidities and myocarditis presenting features were gathered. Outcomes included 2-year freedom from CV comorbidities (composite of atrial fibrillation, stroke, myocardial infarction and heart failure) and freedom from CV death. IrM was treated as a time-varying covariate. RESULTS Seventy-six patients developed irM at a median of 167 days (mean age 69, 63.2% male, 47% lung cancer). Majority of patients had new wall motion abnormalities or EKG changes on presentation. Mean LVEF was 43%, median peak TnT was 0.81, and median NTproBNP was 2057 at irM onset. Two-year freedom from CV comorbidities (67% vs 86.8%, P < 0.001) and death (93.4% vs 99.3%, P = 0.003) was lower among irM patients. Incident irM was an independent predictor of CV death (HR 8.28, P = 0.048), but not CV comorbidities (HR 2.21, P = 0.080). CONCLUSIONS This is the largest case-control study on irM highlighting worse long-term CV outcomes. Future studies are needed to establish appropriate therapeutic strategies and efficient screening strategies for irM survivors.
Collapse
Affiliation(s)
- Lorenzo Braghieri
- Department of Internal MedicineCleveland Clinic FoundationClevelandOhioUSA
| | - Ahmad Gharaibeh
- Department of Internal MedicineCleveland Clinic FoundationClevelandOhioUSA
| | - Lubika Nkashama
- Department of Internal MedicineWashU/Barnes‐Jewish HospitalSt. LouisMissouriUSA
| | | | - Osama Abushawer
- Department of Internal MedicineCleveland Clinic FoundationClevelandOhioUSA
| | | | - Bianca Honnekeri
- Department of Internal MedicineCleveland Clinic FoundationClevelandOhioUSA
| | - Cassandra Calabrese
- Department of Rheumatologic and Immunologic DiseaseCleveland Clinic FoundationClevelandOhioUSA
| | - Venu Menon
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic InstituteCleveland Clinic FoundationClevelandOhioUSA
| | - Pauline Funchain
- Department of Hematology & OncologyTaussig Cancer Center, Cleveland ClinicClevelandOhioUSA
| | - Patrick Collier
- Department of Cardiovascular Medicine, Division of Cardiac Imaging, Heart, Vascular and Thoracic InstituteCleveland Clinic FoundationClevelandOhioUSA
| | - Diego Sadler
- Department of Cardiovascular Medicine, Division of Cardiac Imaging, Heart, Vascular and Thoracic InstituteCleveland Clinic FoundationWestonFloridaUSA
| | - Rohit Moudgil
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic InstituteCleveland Clinic FoundationClevelandOhioUSA
| |
Collapse
|
27
|
Wu CS, Li HP, Hsieh CH, Lin YT, Yi-Feng Chang I, Chung AK, Huang Y, Ueng SH, Hsiao YC, Chien KY, Luo JD, Chen CH, Liao WC, Hung JL, Yuan SN, OuYang CN, Chiang WF, Chien CY, Chuang HC, Chu LJ, Liu H, Yang CY, Robles AI, Rodriguez H, Lin HH, Yang HY, Hsueh C, Chang KP, Yu JS, Chang YS. Integrated multi-omics analyses of oral squamous cell carcinoma reveal precision patient stratification and personalized treatment strategies. Cancer Lett 2025; 614:217482. [PMID: 39842500 DOI: 10.1016/j.canlet.2025.217482] [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: 12/09/2024] [Revised: 01/10/2025] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
Oral cavity squamous cell carcinoma (OSCC), a leading subtype of head and neck cancer, exhibits high global incidence and mortality rates. Despite advancements in surgery and radiochemotherapy, approximately one-third of patients experience relapse. To improve current targeted and immunotherapy strategies for recurrent OSCC, we conducted multi-omics analyses on pretreatment OSCC samples (cohorts 1 and 2, n = 137) and identified A3A and EGFR, both at the RNA and protein levels, as inversely expressed markers for patient stratification and response prediction. Survival analysis demonstrated that elevated A3A or PD-L1 expression levels correlated to improved responses to anti-PD-1 therapy in patients (cohort 3a, n = 50, IHC). In contrast, high RRAS expression (cohort 4, n = 252, qRT-PCR) was significantly associated with OSCC recurrence. Cell-based experiments revealed that RRAS was involved in radiotherapy and cisplatin resistance through the EGFR/RRAS/AKT/ERK signaling pathway. In OSCC patient-derived xenograft (PDX) mouse models, treatments with cisplatin and cetuximab (anti-EGFR) effectively reduced tumor size in EGFR-high-derived (#34) but not A3A-high-derived (#22) PDX tumors. Our study demonstrated that A3A-high tumors were immune-hot and responsive to anti-PD-1 therapy, whereas EGFR-high tumors exhibited chr.7p11.2 gains and DNA repair alterations. Additionally, RRAS-high tumors were associated with OSCC recurrence via AKT and ERK phosphorylation and demonstrate improved clinical outcomes with cetuximab therapy (cohort 3b, n = 49, IHC). This study emphasizes the significance of A3A and EGFR expression levels in OSCC patient stratification and precision therapy, suggesting the use of anti-PD-1 or anti-EGFR treatments, respectively based on these biomarkers. Furthermore, RRAS emerges as a novel prognostic marker for local recurrence.
Collapse
Affiliation(s)
- Chi-Sheng Wu
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan; Department of Otolaryngology-Head & Neck Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City, 33305, Taiwan
| | - Hsin-Pai Li
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan; Department of Microbiology and Immunology, Chang Gung University, Taoyuan City, 33302, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, at Linkou, Taoyuan City, 33305, Taiwan.
| | - Chia-Hsun Hsieh
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, at Linkou, Taoyuan City, 33305, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, New Taipei Municipal City TuCheng Hospital, New Taipei City, Taiwan; College of Medicine, Chang Gung University, Taoyuan City, 33302, Taiwan
| | - Yu-Tsun Lin
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan
| | - Ian Yi-Feng Chang
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City, 33305, Taiwan; Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Taoyuan City, 33305, Taiwan
| | - An-Ko Chung
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan
| | - Yenlin Huang
- Department of Pathology, Chang Gung Memorial Hospital at Linkou, Taoyuan City, 33305, Taiwan; School of Medicine, National Tsing-Hua University, Hsinchu, 300044, Taiwan; Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, at Linkou, Taoyuan City, 33305, Taiwan
| | - Shir-Hwa Ueng
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan; Department of Pathology, Chang Gung Memorial Hospital at Linkou, Taoyuan City, 33305, Taiwan
| | - Yung-Chin Hsiao
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan; Department of Otolaryngology-Head & Neck Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City, 33305, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Kun-Yi Chien
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan; Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan City, 33302, Taiwan
| | - Ji-Dung Luo
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan; Bioinformatics Resource Center, The Rockefeller University, 1230 York Avenue, New York City, NY, USA, 10065
| | - Chia-Hua Chen
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan; Department of Anatomy, School of Medicine, Chang Gung University, Taoyuan City, 33302, Taiwan
| | - Wei-Chao Liao
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan; Department of Nephrology, Chang Gung Memorial Hospital at Linkou, Taoyuan City, 33305, Taiwan
| | - Jui-Lung Hung
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan
| | - Sheng-Ning Yuan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan
| | - Chun-Nan OuYang
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan
| | - Wei-Fan Chiang
- Department of Dentistry, Chi-Mei Medical Center, Liouying, Taiwan; School of Dentistry, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Chih-Yen Chien
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hui-Ching Chuang
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Lichieh Julie Chu
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan; Liver Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan City, 33305, Taiwan
| | - Hsuan Liu
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan; Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan City, 33302, Taiwan; Division of Colon and Rectal Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City, 33305, Taiwan
| | - Chia-Yu Yang
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan; Department of Otolaryngology-Head & Neck Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City, 33305, Taiwan; Department of Microbiology and Immunology, Chang Gung University, Taoyuan City, 33302, Taiwan
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Rockville, MD, 20850, USA
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Rockville, MD, 20850, USA
| | - Hsi-Hsien Lin
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan City, 33302, Taiwan; Department of Pathology, Chang Gung Memorial Hospital at Linkou, Taoyuan City, 33305, Taiwan; Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital at Keelung, Keelung, 20401, Taiwan
| | - Huang-Yu Yang
- College of Medicine, Chang Gung University, Taoyuan City, 33302, Taiwan; Department of Nephrology, Chang Gung Memorial Hospital at Linkou, Taoyuan City, 33305, Taiwan
| | - Chuen Hsueh
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan; Department of Pathology, Chang Gung Memorial Hospital at Linkou, Taoyuan City, 33305, Taiwan
| | - Kai-Ping Chang
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan; Department of Otolaryngology-Head & Neck Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City, 33305, Taiwan; College of Medicine, Chang Gung University, Taoyuan City, 33302, Taiwan.
| | - Jau-Song Yu
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan; Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan City, 33302, Taiwan; Liver Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan City, 33305, Taiwan.
| | - Yu-Sun Chang
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 33302, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| |
Collapse
|
28
|
Patalakh I, Wandersee A, Schlüter J, Erdmann M, Hackstein H, Cunningham S. Influence of the Immune Checkpoint Inhibitors on the Hemostatic Potential of Blood Plasma. Transfus Med Hemother 2025; 52:120-131. [PMID: 40201622 PMCID: PMC11975347 DOI: 10.1159/000535926] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/18/2023] [Indexed: 04/10/2025] Open
Abstract
Introduction Immune checkpoint inhibitors (ICIs) have revolutionized classical treatment approaches of various cancer entities, but are also associated with a number of side effects. One of these may be life-threatening clotting disorders with the risk of thrombotic or hemorrhagic complications, the mechanisms of which are still poorly understood. In the present study, we analyzed the direct effects of pembrolizumab, nivolumab, and ipilimumab on platelet aggregation as well as plasma coagulation followed by fibrinolysis in an ex vivo model. Methods Microplate spectrometry was used to analyze aggregation, coagulation, and fibrinolysis in platelet-free (PFP) and platelet-rich (PRP) healthy donor plasma samples treated with pembrolizumab, nivolumab, ipilimumab, and appropriate isotype controls. Aggregation was induced by TRAP-6. Clotting of PFP and PRP followed by lysis was initiated with a tissue factor in a mixture of phosphatidylserine:phosphatidylcholine and the addition of t-PA. Among other parameters, the area under the curve (AUC) was used to compare the effect of ICIs on aggregation, coagulation, and fibrinolysis. Results Upon direct contact with platelets, pembrolizumab stimulated platelet aggregation in PRP, while nivolumab and ipilimumab promoted disaggregation with corresponding changes in the AUC. Pembrolizumab and nivolumab, both PD-1 receptor inhibitors, had no effect on the plasma coagulation cascade. Ipilimumab, a CTLA-4 receptor inhibitor, significantly increased the rate of PRP clotting. When clotting was followed by lysis, all ICIs were found to prolong the growth of the PRP-derived fibrin clot and delay its elimination. This was manifested by an increase in AUC relative to control PRP. Conclusion This study characterizes the potential impact of pembrolizumab, nivolumab, and ipilimumab on hemostasis. Nivolumab and ipilimumab are able to reduce aggregation and increase the procoagulant properties of platelets, which can cause side effects associated with hemostatic imbalance leading to thrombosis or bleeding. The observed ICI-specific effects may contribute to our understanding of the mechanisms by which ICI affects platelets and suggest how, in a clinical setting, to reduce coagulation disorders during ICI treatment in the future.
Collapse
Affiliation(s)
- Irina Patalakh
- Department of Transfusion Medicine and Hemostaseology, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
- Department of Chemistry and Biochemistry of Enzymes, Palladin Institute of Biochemistry of the National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Alexandra Wandersee
- Department of Transfusion Medicine and Hemostaseology, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| | - Julian Schlüter
- Department of Transfusion Medicine and Hemostaseology, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| | - Michael Erdmann
- Department of Dermatology, Uniklinikum Erlangen, Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Holger Hackstein
- Department of Transfusion Medicine and Hemostaseology, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| | - Sarah Cunningham
- Department of Transfusion Medicine and Hemostaseology, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
29
|
Zhang Y, Cao Z, Jia H, Feng Y, Sun X, Wu H, Xu B, Wei Z. Immune checkpoint inhibitor induces cardiac injury by impairing efferocytosis of macrophages via MerTK cleavage. Int Immunopharmacol 2025; 150:114263. [PMID: 39938164 DOI: 10.1016/j.intimp.2025.114263] [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: 08/01/2024] [Revised: 02/01/2025] [Accepted: 02/06/2025] [Indexed: 02/14/2025]
Abstract
Cancer immunotherapy is a well-established therapeutic approach for various types of cancer. However, its clinical utility is usually limited by cardiovascular adverse events. Immune Checkpoint Inhibitors (ICIs) can induce diverse forms of cardiotoxicity, with myocarditis being the most fatal complication. The underlying mechanism of its occurrence remains elusive. Therefore, this study aims to elucidate the impact of programmed death-1 (PD-1) inhibitor on myocarditis development in mice. Myeloid-epithelial-reproductive tyrosine kinase (MerTK) receptors, located on the surface of macrophages, play a pivotal role in phagocytic regulation. We established a mouse model of autoimmune myocarditis by injecting 6-week-old normal male BALB/c mice with PD-1 inhibitor and cardiac troponin I peptide fragments, which resulted in elevated levels of serum soluble MerTK (SolMer) and reduced numbers of MerTK-CD68 double-positive macrophages, accompanied by cardiac injury in mice. In vitro, PD-1 inhibitor promotes a disintegrin and metalloproteinase17 (ADAM17)-mediated shed of the MerTK, forming SolMer, through MKK3/P38 MAPK pathway, leading to downregulation of MerTK expression on the macrophage surface. This results in the inhibition of efferocytosis and impairment of tissue repair function, ultimately contributing to myocarditis development. TAPI-0 inhibited the activity of ADAM17, while SB203580 inhibited the phosphorylation of P38 MAPK. Both inhibitors effectively restored the inhibition of efferocytosis induced by the PD-1 inhibitor. In vitro, when the PD-1 receptor on the surface of RAW264.7 macrophages was knocked down and then stimulated with a PD-1 inhibitor, no further significant alterations in the pathway were elicited. In conclusion, the PD-1 inhibitor induces the shedding of MerTK in macrophages by binding to the PD-1 receptor on the surface of macrophages and activating the MKK3/P38 MAPK/ADAM17 pathway, leading to impaired efferocytosis. Elucidation of this molecular mechanism holds promise for improved prognosis and therapeutic strategies in cancer patients.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, 358 Zhongshan Road 210008 Nanjing, China
| | - Zhenzhu Cao
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, 358 Zhongshan Road 210008 Nanjing, China
| | - Huihui Jia
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, 358 Zhongshan Road 210008 Nanjing, China
| | - Yuting Feng
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008 China
| | - Xuan Sun
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008 China
| | - Han Wu
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008 China.
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, 358 Zhongshan Road 210008 Nanjing, China; Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008 China.
| | - Zhonghai Wei
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, 358 Zhongshan Road 210008 Nanjing, China; Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008 China.
| |
Collapse
|
30
|
Chen S, Liu R, Duan S, Zhang B, Wang Y, Li X, Zhao Y, Li Z, Zhou Q, Zhang R, Zhang L, Xu X, Jang R, Zhang J, Li Y, Cai X, Zhang L. Ultrasound-guided percutaneous radiofrequency ablation combined with anti-PD-1 for the treatment of prostate cancer: an experimental study. Front Oncol 2025; 15:1527763. [PMID: 40196732 PMCID: PMC11973081 DOI: 10.3389/fonc.2025.1527763] [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: 11/13/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Background This study seeks to investigate the potential synergistic effects of combining ultrasound-guided percutaneous radiofrequency ablation with anti-PD-1 therapy on prostate cancer, utilizing animal models. Methods A mouse model of prostate cancer was established by subcutaneous injection of 1 × 106 Myc-Cap cells on the right side of FVB mice. When the volume of the tumors reached about 400mm3, the mice were randomly divided into four groups and received corresponding intervention treatments. Among them, Group 1 was the blank control group, Group 2 was the simple anti-PD-1 treatment group, Group 3 was the simple radiofrequency ablation group, and Group 4 is the group that received percutaneous radiofrequency ablation combined with anti-PD-1 therapy under ultrasound guidance. The growth of the tumors was observed in mice after treatment in each group, tumor tissues were collected, and the immune status of the mice was analyzed through flow cytometry, immunohistochemistry, immunofluorescence, and other methods. Results Compared with other treatment groups, ultrasound-guided percutaneous radiofrequency ablation combined with anti-PD-1 therapy significantly reduced the weight and volume of the tumors, demonstrating more effective tumor suppression. At the same time, combination therapy can promote the aggregation of T-cells within the tumor and increase the proportion of cytotoxic T-cells, increase the proportion of M1 macrophages and iNOS expression, and decrease the proportion of M2 macrophages and Arg expression in the local area of the tumors. Conclusion Local ablation can improve the therapeutic effect of PD-1 monoclonal antibody. Our preliminary results suggest that ultrasound-guided percutaneous radiofrequency ablation, in combination with anti-PD-1 treatment, produces synergistic effects. These effects may be driven by changes in immune cell populations within the tumor's immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Si Chen
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ruiqing Liu
- Department of Interventional Therapy, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
- Henan Provincial International Joint Laboratory of Ultrasonic Nanotechnology and Artificial Intelligence in Precision Theragnostic Systems, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Shaobo Duan
- Department of Ultrasound, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Henan University People’s Hospital, Zhengzhou, Henan, China
- Henan Provincial International Joint Laboratory of Ultrasonic Nanotechnology and Artificial Intelligence in Precision Theragnostic Systems, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Beibei Zhang
- Henan Provincial International Joint Laboratory of Ultrasonic Nanotechnology and Artificial Intelligence in Precision Theragnostic Systems, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Yuzhou Wang
- Henan Provincial International Joint Laboratory of Ultrasonic Nanotechnology and Artificial Intelligence in Precision Theragnostic Systems, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Xiaoxiao Li
- Henan University People’s Hospital, Henan Provincial People’s Hospital, Henan University, Zhengzhou, Henan, China
| | - Yingying Zhao
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Zesheng Li
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Qi Zhou
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Rui Zhang
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Linlin Zhang
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoxia Xu
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ru Jang
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Juan Zhang
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yaqiong Li
- Henan Provincial International Joint Laboratory of Ultrasonic Nanotechnology and Artificial Intelligence in Precision Theragnostic Systems, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Xiguo Cai
- Henan Provincial Clinical Research Center for Rehabilitation Medicine, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Lianzhong Zhang
- Department of Ultrasound, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Henan University People’s Hospital, Zhengzhou, Henan, China
- Henan Provincial International Joint Laboratory of Ultrasonic Nanotechnology and Artificial Intelligence in Precision Theragnostic Systems, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
- Henan Provincial Clinical Research Center for Rehabilitation Medicine, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| |
Collapse
|
31
|
Kazim M, Ganguly A, Malespini SM, Thang L, Patel NL, Kim C, Kalen JD, Difilippantonio S, Yoo E. Granzyme-targeting quenched activity-based probes for assessing tumor response to immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.13.643086. [PMID: 40161750 PMCID: PMC11952571 DOI: 10.1101/2025.03.13.643086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Molecular imaging of immune activation holds tremendous potential for the development of novel immunotherapy. In particular, chemical probes capable of detecting immune responses before changes in tumor size occur can guide early therapeutic strategies. Here, we present quenched activity-based probes targeting granzymes as a biomarker of antitumor immunity. Through optimization of peptide recognition element and functional chemical warhead, we have developed an optical imaging probe Cy5-IEPCyaPhP-QSY21, which rapidly reacts with GzmB at substoichiometric concentrations and enables efficient, selective labeling of the active enzyme in a complex proteome. With high specificity and minimal background signal, this probe produces GzmB-induced near-infrared fluorescence signals in the tumors of living mice shortly after injection. Both in vivo and ex vivo fluorescence signals correlate with GzmB expression and activity, and the population of CD8+ cells in tumor tissues. Moreover, it demonstrates the potential to track tumor response to immunotherapy. Thus, this study offers a chemical tool for assessing immune-mediated anticancer activity using noninvasive optical imaging.
Collapse
Affiliation(s)
- Muhammad Kazim
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, 21702, United States
| | - Arghya Ganguly
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, 21702, United States
| | - Sebastian M. Malespini
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, 21702, United States
| | - Lai Thang
- Animal Research Technical Support, Laboratory of Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, 21702, United States
| | - Nimit L. Patel
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, 21702, United States
| | - Caleb Kim
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, 21702, United States
| | - Joseph D. Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, 21702, United States
| | - Simone Difilippantonio
- Animal Research Technical Support, Laboratory of Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, 21702, United States
| | - Euna Yoo
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, 21702, United States
| |
Collapse
|
32
|
Matheson BE, Jaremko JL, Dowhanik A, Gill J, Gallant C, Walker J, Armani N, Leslie WD, Kolinsky M, Boyd SK, Ye C. Assessing the effects of immune checkpoint inhibitors on bone utilizing machine learning-assisted opportunistic quantitative computed tomography. J Bone Miner Res 2025; 40:396-403. [PMID: 39849845 PMCID: PMC11909731 DOI: 10.1093/jbmr/zjaf009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/10/2024] [Accepted: 01/14/2025] [Indexed: 01/25/2025]
Abstract
Immune checkpoint inhibitors (ICIs) are widely used in cancer treatment, yet their impact on bone health remains unclear. This study aimed to perform a retrospective cohort study utilizing routine CT scans from patients with melanoma to perform opportunistic QCT analysis to investigate the effects of ICI treatment on skeletal health, including volumetric BMD (vBMD) measurements and osteoarthritis (OA) parameters. A previously established machine learning-assisted opportunistic QCT pipeline was used to estimate lumbar spine vBMD from baseline and 12-mo follow-up CT scans in patients with melanoma treated with ICI therapy and those not treated with ICI therapy. Facet joint OA, osteophyte formation, and endplate sclerosis were also graded. Independent and paired t tests were used to determine any differences in vBMD and OA parameters between ICI users and non-ICI users. Multivariable linear regression models were used to control for confounding variables. Non-ICI users had a significant decrease in vBMD of -6.96 mg/cm3 from baseline to follow-up, whereas the ICI users had no significant change. There was a significant difference in change in vBMD from baseline to follow-up between the 2 groups, with the non-ICI users experiencing a 11.22 mg/cm3 larger decrease in vBMD. After adjusting for baseline age, sex, baseline vBMD, and change in OA parameters, this difference remained significant at -13.04 mg/cm3. Among the ICI users, those who had a decline in vBMD had a lower baseline vBMD compared with those who had increased vBMD. Neither group showed a significant change in OA parameters over the follow-up period, nor a difference in change between ICI and non-ICI users, even after adjusting for sex, age, and baseline OA parameters. While the effects of ICI treatment on vBMD may vary based on baseline bone health, ICIs do not significantly impact OA parameters in the short term.
Collapse
Affiliation(s)
- Bryn E Matheson
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Jacob L Jaremko
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Alexandra Dowhanik
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Jasmine Gill
- Division of Dermatology, Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Cassandra Gallant
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - John Walker
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Nathan Armani
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - William D Leslie
- Departments of Internal Medicine and Radiology, University of Manitoba, Winnipeg, MB R3A 1R9, Canada
| | - Michael Kolinsky
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Steven K Boyd
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Department of Radiology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Carrie Ye
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2G3, Canada
| |
Collapse
|
33
|
Hu Q, Shi Y, Wang H, Bing L, Xu Z. Post-translational modifications of immune checkpoints: unlocking new potentials in cancer immunotherapy. Exp Hematol Oncol 2025; 14:37. [PMID: 40087690 PMCID: PMC11907956 DOI: 10.1186/s40164-025-00627-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/27/2025] [Indexed: 03/17/2025] Open
Abstract
Immunotherapy targeting immune checkpoints has gained traction across various cancer types in clinical settings due to its notable advantages. Despite this, the overall response rates among patients remain modest, alongside issues of drug resistance and adverse effects. Hence, there is a pressing need to enhance immune checkpoint blockade (ICB) therapies. Post-translational modifications (PTMs) are crucial for protein functionality. Recent research emphasizes their pivotal role in immune checkpoint regulation, directly impacting the expression and function of these key proteins. This review delves into the influence of significant PTMs-ubiquitination, phosphorylation, and glycosylation-on immune checkpoint signaling. By targeting these modifications, novel immunotherapeutic strategies have emerged, paving the way for advancements in optimizing immune checkpoint blockade therapies in the future.
Collapse
Affiliation(s)
- Qiongjie Hu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang Province, China
- The Third Affiliated Hospital of Zhejiang, Chinese Meical University, Hangzhou, 310013, China
| | - Yueli Shi
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang Province, China
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China
| | - Huang Wang
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liuwen Bing
- The Third Affiliated Hospital of Zhejiang, Chinese Meical University, Hangzhou, 310013, China.
| | - Zhiyong Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang Province, China.
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China.
| |
Collapse
|
34
|
Carr AS, Vonberg FW, Koay S, Young K, Shaw H, Olsson-Brown A, Willis M. Neurological complications of immune checkpoint inhibitors: a practical guide. Pract Neurol 2025; 25:116-126. [PMID: 39592208 DOI: 10.1136/pn-2024-004327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2024] [Indexed: 11/28/2024]
Abstract
Immune checkpoint inhibition unleashes the power of the immune system against tumour cells. Immune checkpoint inhibitors (ICIs) block the inhibitory effects of cytotoxic T-lymphocyte associated protein 4 (CTLA-4), programmed death protein 1 (PD-1), programmed death ligand 1 (PD-L1) and lymphocyte activation gene 3 (LAG-3) molecules on T-cells, and so enhance physiological cytotoxic effects. ICIs can significantly improve survival from cancers, including those previously associated with poor treatment response, such as metastatic melanoma. However, on-target off-tumour effects of ICIs result in immune-related adverse events. These toxicities are common and require new multidisciplinary expertise to manage. ICI neurotoxicity is relatively rare but ominous due to its severity, heterogenous manifestations and potential for long-term disability. Neurotoxic syndromes are novel and often present precipitously. Here, we describe ICI mechanisms of action, their impact on cancer outcomes and their frequency of immune-related adverse events. We focus particularly on neurotoxicity. We discuss the current appreciation of neurotoxic syndromes, management strategies and outcomes based on clinical expertise and consensus, multi-specialty guidance. The use of immunotherapy is expanding exponentially across multiple cancer types and so too will our approach to these cases.
Collapse
Affiliation(s)
- Aisling S Carr
- Centre for Neuromuscular Diseases, National Hospital for Neurology and Neurosurgery, London, UK
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Frederick William Vonberg
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Shiwen Koay
- Centre for Neuromuscular Diseases, National Hospital for Neurology and Neurosurgery, London, UK
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Kate Young
- Renal and Melanoma Unit, Royal Marsden Hospital Chelsea, London, London, UK
| | - Heather Shaw
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, London, UK
| | - Anna Olsson-Brown
- Sussex Cancer Centre, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
- Department of Clinical and Molecular Pharmacology, University of Liverpool, Liverpool, UK
| | - Mark Willis
- Department of Neurology, University Hospital of Wales, Cardiff, UK
| |
Collapse
|
35
|
Zang H, Liu T, Wang X, Cheng S, Zhu X, Huang C, Duan L, Zhao X, Guo F, Wang X, Zhang C, Yang F, Gu Y, Hu H, Gao S. PD-1 IR2 promotes tumor evasion via deregulating CD8 + T cell function. J Immunother Cancer 2025; 13:e010529. [PMID: 40050045 PMCID: PMC11887316 DOI: 10.1136/jitc-2024-010529] [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: 09/07/2024] [Accepted: 02/22/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND The programmed cell death 1 (PD-1) is an immune checkpoint that mediates immune evasion of tumors. Alternative splicing (AS) such as intron retention (IR) plays a crucial role in the immune-related gene processing and its function. However, it is not clear whether PDCD1 encoding PD-1 exists as an IR splicing isoform and what underlying function of such isoform plays in tumor evasion. METHODS An AS isoform of human PDCD1, characterized by the second IR and named PD-1IR2, was identified by reverse transcription-PCR (RT-PCR) and Sanger sequencing. The expression profile of PD1IR2 was assessed by quantitative RT-PCR and flow cytometry, while its function was evaluated through immune cell proliferation, cytokine interleukin 2 secretion, and tumor cell killing assays. PDCD1IR2 CKI mice which specifically conditional knock-in PDCD1IR2 in T cells and humanized peripheral blood mononuclear cells (PBMC)-NOG (NOD.Cg-PrkdcscidIL2rgtm1Sug/JicCrl) mice were utilized to further confirm the physiological function of PD-1IR2 in vivo. RESULTS PD-1IR2 is expressed in a variety of human leukemia cell lines and tumor-infiltrating lymphocytes. PD-1IR2 expression is induced on T cell activation and regulated by the RNA-binding protein hnRNPLL. PD-1IR2 negatively regulates the immune function of CD8+ T cells, indicated by inhibiting T cell proliferation, cytokine production, and tumor cell killing in vitro. PD-1IR2+ CD8+ T cells show impaired antitumor function, which consequently promote tumor evasion in a conditional knock-in mouse model and a PBMC-engrafted humanized NOG mouse model. PD-1IR2 mice exhibit resistance to anti-PD-L1 therapy compared with wild-type mice. CONCLUSIONS PD-1IR2 is a potential immune checkpoint that may mediate potential resistance to immune checkpoint therapy.
Collapse
Affiliation(s)
- Haojing Zang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, Shanxi, China
| | - Tongfeng Liu
- School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
- Medical College, Guizhou University, Guiyang, Guizhou, China
| | - Xiaodong Wang
- School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Shuwen Cheng
- School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
- Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Xiaofeng Zhu
- School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
- Medical College, Guizhou University, Guiyang, Guizhou, China
| | - Chang Huang
- School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
- Medical College, Guizhou University, Guiyang, Guizhou, China
| | - Liqiang Duan
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, Shanxi, China
- School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
| | - Xujie Zhao
- School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
| | - Fang Guo
- School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
- The Key Laboratory of Medical Molecular Cell Biology of Shanxi Province, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, Shanxi, China
| | - Xuetong Wang
- School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Chang Zhang
- School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
- Department of oncology, The Key Laboratory of Advanced Interdisciplinary Studies, First Affiliated Hospital of Guangzhou Medical University State Key Laboratory of Respiratory Disease, Guangzhou, Guangdong, China
| | - Facai Yang
- School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
| | - Yinmin Gu
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Hongbo Hu
- Center for Immunology and Hematology, Department of Biotherapy and Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shan Gao
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, China
- School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
36
|
Zhang L, Kao G, Zhao Y, Zhang Z, Kim HS, Shi X, Cheng Q, Hou T, Lenz HJ, Zhang Y. Genetically reprogrammed exosomes for immunotherapy of acute myeloid leukemia. Mol Ther 2025; 33:1091-1104. [PMID: 39815621 PMCID: PMC11897778 DOI: 10.1016/j.ymthe.2025.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 11/24/2024] [Accepted: 01/10/2025] [Indexed: 01/18/2025] Open
Abstract
Current treatments for acute myeloid leukemia (AML) remain challenging and are characterized by poor clinical outcomes. Exosomes, cell-derived membranous vesicles, have been emerging as a new modality of therapy. Here, we designed and generated genetically reprogrammed exosomes with surface-displayed antibodies and immunoregulatory proteins, namely programmed immune-engaging exosomes (PRIME Exos). By simultaneously targeting T cells and AML cells expressing C-type lectin-like molecule-1 (CLL-1), PRIME Exos can elicit tumor-specific immune responses and sustain cellular immunity against AML by modulating programmed death 1 (PD-1)- and CD27-mediated immune checkpoint pathways. In preclinical models of AML, PRIME Exos have shown promising efficacy and safety for suppressing leukemia expansion. This study developed a new exosome-based approach for AML immunotherapy.
Collapse
MESH Headings
- Exosomes/genetics
- Exosomes/metabolism
- Exosomes/immunology
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Humans
- Animals
- Immunotherapy/methods
- Mice
- Cell Line, Tumor
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Disease Models, Animal
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Lei Zhang
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Guoyun Kao
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Yuanteng Zhao
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Zeyu Zhang
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Hyo Sun Kim
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Xiaojing Shi
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Qinqin Cheng
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Tianling Hou
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Yong Zhang
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; Department of Chemistry, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA 90089, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA; Research Center for Liver Diseases, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
37
|
Tripathi S, Sharma Y, Kumar D. Unveiling the link between chronic inflammation and cancer. Metabol Open 2025; 25:100347. [PMID: 39876904 PMCID: PMC11772974 DOI: 10.1016/j.metop.2025.100347] [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: 11/10/2024] [Revised: 01/05/2025] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
The highly nuanced transition from an inflammatory process to tumorigenesis is of great scientific interest. While it is well known that environmental stimuli can cause inflammation, less is known about the oncogenic modifications that chronic inflammation in the tissue microenvironment can bring about, as well as how these modifications can set off pro-tumorigenic processes. It is clear that no matter where the environmental factors come from, maintaining an inflammatory microenvironment encourages carcinogenesis. In addition to encouraging angiogenesis and metastatic processes, sustaining the survival and proliferation of malignant transformed cells, and possibly altering the efficacy of therapeutic agents, inflammation can negatively regulate the antitumoral adaptive and innate immune responses. Because chronic inflammation has multiple pathways involved in tumorigenesis and metastasis, it has gained recognition as a marker of cancer and a desirable target for cancer therapy. Recent advances in our knowledge of the molecular mechanisms that drive cancer's progression demonstrate that inflammation promotes tumorigenesis and metastasis while suppressing anti-tumor immunity. In many solid tumor types, including breast, lung, and liver cancer, inflammation stimulates the activation of oncogenes and impairs the body's defenses against the tumor. Additionally, it alters the microenvironment of the tumor. As a tactical approach to cancer treatment, these findings have underscored the importance of targeting inflammatory pathways. This review highlights the role of inflammation in cancer development and metastasis, focusing on its impact on tumor progression, immune suppression, and therapy resistance. It examines current anti-inflammatory strategies, including NSAIDs, cytokine modulators, and STAT3 inhibitors, while addressing their potential and limitations. The review emphasizes the need for further research to unravel the complex mechanisms linking inflammation to cancer progression and identify molecular targets for specific cancer subtypes.
Collapse
Affiliation(s)
- Siddhant Tripathi
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Yashika Sharma
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Dileep Kumar
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| |
Collapse
|
38
|
Zhuang AB, Xi Z, Cheng YX, Zhang CH, Li WG. Current status and future perspectives of immunotherapy for abdominal liposarcoma: From basic research to clinical practice. Shijie Huaren Xiaohua Zazhi 2025; 33:81-88. [DOI: 10.11569/wcjd.v33.i2.81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/06/2024] [Accepted: 12/17/2024] [Indexed: 02/28/2025] Open
Abstract
Liposarcoma is a highly heterogeneous type of soft tissue sarcoma originating from adipose tissue, characterized by complex biological behavior and invasiveness. Traditional treatments have shown limited efficacy in high-grade and metastatic liposarcoma, with unsatisfactory patient outcomes. In recent years, the breakthroughs of immunotherapy in various solid tumors have sparked interest in its potential application to liposarcoma. This review systematically examines the progress in basic research and clinical practice of immunotherapy for liposarcoma, discussing the tumor immune microenvironment, mechanisms of immune evasion, the application of immune checkpoint inhibitors, combination therapy strategies, the challenges faced, as well as the future direction, with an aim to provide a theoretical basis for personalized treatment of liposarcoma, promote the development of novel immunotherapy strategies, and ultimately improve patient prognosis and quality of life.
Collapse
Affiliation(s)
- Ao-Bo Zhuang
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Zhe Xi
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Ying-Xue Cheng
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Chen-He Zhang
- School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Wen-Gang Li
- Department of Hepatobiliary and Pancreatic Surgery, Xiang'an Hospital of Xiamen University, Xiamen 361102, Fujian Province, China
- Cancer Research Center of Xiamen University, Xiamen 361005, Fujian Province, China
| |
Collapse
|
39
|
Shi R, Sun J, Zhou Z, Shi M, Wang X, Gao Z, Zhao T, Li M, Shu Y. Integration of multiple machine learning approaches develops a gene mutation-based classifier for accurate immunotherapy outcomes. NPJ Precis Oncol 2025; 9:54. [PMID: 40011681 DOI: 10.1038/s41698-025-00842-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 02/17/2025] [Indexed: 02/28/2025] Open
Abstract
In addition to traditional biomarkers like PD-(L)1 expression and tumor mutation burden (TMB), more reliable methods for predicting immune checkpoint blockade (ICB) response in cancer patients are urgently needed. This study utilized multiple machine learning approaches on nonsynonymous mutations to identify key mutations that are most significantly correlated to ICB response. We proposed a classifier, Gene mutation-based Predictive Signature (GPS), to categorize patients based on their predicted response and clinical outcomes post-ICB therapy. GPS outperformed conventional predictors when validated in independent cohorts. Multi-omics analysis and multiplex immunohistochemistry (mIHC) revealed insights into tumor immunogenicity, immune responses, and the tumor microenvironment (TME) in lung adenocarcinoma (LUAD) across different GPS groups. Finally, we validated distinct responses of different GPS samples to ICB in an ex-vivo tumor organoid-PBMC co-culture model. Overall, our findings highlight a simple, robust classifier for accurate ICB response prediction, which could reduce costs, shorten testing times, and facilitate clinical implementation.
Collapse
Affiliation(s)
- Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Sun
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meiqi Shi
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xin Wang
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Zhaojia Gao
- Department of Thoracic Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Tianyu Zhao
- Institute and Clinic for Occupational, Social and Environmental Medicine, LMU University Hospital Munich, Munich, Germany
| | - Minglun Li
- Department of Radiation Oncology, Lueneburg Municipal Hospital, Lueneburg, Germany
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
40
|
Du F, Wang G, Dai Q, Huang J, Li J, Liu C, Du K, Tian H, Deng Q, Xie L, Zhao X, Zhang Q, Yang L, Li Y, Wu Z, Zhang Z. Targeting novel regulated cell death: disulfidptosis in cancer immunotherapy with immune checkpoint inhibitors. Biomark Res 2025; 13:35. [PMID: 40012016 DOI: 10.1186/s40364-025-00748-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
The battle against cancer has evolved over centuries, from the early stages of surgical resection to contemporary treatments including chemotherapy, radiation, targeted therapies, and immunotherapies. Despite significant advances in cancer treatment over recent decades, these therapies remain limited by various challenges. Immune checkpoint inhibitors (ICIs), a cornerstone of tumor immunotherapy, have emerged as one of the most promising advancements in cancer treatment. Although ICIs, such as CTLA-4 and PD-1/PD-L1 inhibitors, have demonstrated clinical efficacy, their therapeutic impact remains suboptimal due to patient-specific variability and tumor immune resistance. Cell death is a fundamental process for maintaining tissue homeostasis and function. Recent research highlights that the combination of induced regulatory cell death (RCD) and ICIs can substantially enhance anti-tumor responses across multiple cancer types. In cells exhibiting high levels of recombinant solute carrier family 7 member 11 (SLC7A11) protein, glucose deprivation triggers a programmed cell death (PCD) pathway characterized by disulfide bond formation and REDOX (reduction-oxidation) reactions, termed "disulfidptosis." Studies suggest that disulfidptosis plays a critical role in the therapeutic efficacy of SLC7A11high cancers. Therefore, to investigate the potential synergy between disulfidptosis and ICIs, this study will explore the mechanisms of both processes in tumor progression, with the goal of enhancing the anti-tumor immune response of ICIs by targeting the intracellular disulfidptosis pathway.
Collapse
Affiliation(s)
- Fei Du
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Guojun Wang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qian Dai
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Jiang Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Junxin Li
- Department of pharmacy, Zigong Fourth People's Hospital, Zigong, 643000, China
| | - Congxing Liu
- Department of Pharmacy, Chengfei Hospital, Chengdu, 610000, China
| | - Ke Du
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pediatrics, Luzhou Maternal and Child Health Hospital, Luzhou Second People's Hospital, Luzhou, 646000, Sichuan, China
| | - Hua Tian
- School of Nursing, Chongqing College of Humanities, Science & Technology, Chongqing, 401520, China
| | - Qiwei Deng
- Heruida Pharmaceutical Co.,ltd, Haikou, Hainan, 570100, China
| | - Longxiang Xie
- The TCM Hospital of Longquanyi District, Chengdu, 610100, Sichuan, China
| | - Xin Zhao
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qimin Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Lan Yang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhigui Wu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhuo Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
41
|
Zhao WJ, Wang ML, Zhao YF, Zhao WP, Huang QH, Lu ZW, Jia F, Shi JJ, Liu BS, Han WH, Lu HW, Zhang BC, Wang ZX. Pan-cancer analysis reveals SMARCAL1 expression is associated with immune cell infiltration and poor prognosis in various cancers. Sci Rep 2025; 15:6591. [PMID: 39994264 PMCID: PMC11850860 DOI: 10.1038/s41598-025-88955-9] [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: 06/07/2024] [Accepted: 02/03/2025] [Indexed: 02/26/2025] Open
Abstract
Although immune checkpoint inhibition in particular has shown promise in cancer immunotherapy, it is not always efficient. Recent studies suggest that SMARCAL1 may play a role in tumor immune evasion, yet its pan-cancer role is unclear. We conducted a comprehensive analysis of SMARCAL1 using TCGA, GTEx, and CCLE databases, evaluating its expression, genetic alterations, epigenetic modifications, and their clinical correlations across 33 cancer types. Our findings indicate that SMARCAL1 is overexpressed in several cancers, such as Glioma, LUAD, KIRC, and LIHC, impacting prognosis. Elevated SMARCAL1 is linked to poor outcomes in Glioma, LUAD, and LIHC but correlates with better survival in KIRC. We also found significant associations between SMARCAL1 expression and DNA methylation in 13 cancers. Furthermore, SMARCAL1 expression correlates with immune infiltration, suggesting it as a potential therapeutic target in cancer immunotherapy. This study underscores the need for further research on SMARCAL1 to enhance immunotherapeutic strategies.
Collapse
Affiliation(s)
- Wu-Jie Zhao
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Meng-Lei Wang
- Department of Digestive Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Yun-Fang Zhao
- Jitang College of North China University of Science and Technology, Tangshan, 063000, Hebei, China
| | - Wen-Peng Zhao
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Qiong-Hui Huang
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, 350108, Fujian, China
| | - Zhen-Wei Lu
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, 350108, Fujian, China
| | - Fang Jia
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, Guangdong, China
| | - Jin-Jin Shi
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Bo-Sen Liu
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Wan-Hong Han
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Han-Wen Lu
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Bing-Chang Zhang
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China.
| | - Zhan-Xiang Wang
- Department of Neurosurgery and Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China.
| |
Collapse
|
42
|
Iemwimangsa N, Anantaya D, Oranratnachai S, Thamrongjirapat T, Lumjiaktase P, Teoh VH, Khiewngam K, Monnamo N, Sanvarinda P, Incharoen P, Charoenyingwattan A, Sensorn I, Dejthevaporn T, Sirachainan E, Chantratita W, Reungwetwattana T, Trachu N. Dynamic changes in immune repertoire profiles in patients with stage III unresectable non-small cell lung cancer during consolidation treatment with immunotherapy. BMC Cancer 2025; 25:333. [PMID: 39994571 PMCID: PMC11853222 DOI: 10.1186/s12885-025-13716-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND One-year of immune checkpoint inhibitor (ICI) treatment after concurrent chemoradiation (CCRT) in unresectable stage III non-small cell lung cancer (NSCLC) is a standard of care. The precise predictive biomarkers are under investigations either immunological markers or clinical characteristics. Here, we explored immune repertoire of T cell receptor β-chain (TCRβ) during ICI treatment. METHODS During August 2019 and September 2021, stage III NSCLC, post CCRT patients from Ramathibodi Hospital was enrolled. All patients were treated by durvalumab after CCRT. Blood samples were collected together with clinical data and tumor assessment every 3-4 months until disease progression or discontinuation of treatment due to adverse events. CDR3 region and TCRΒ polymorphisms was explored by RNA sequencing using Next-Generation Sequencing (NGS) TCR beta short-read assay. Bioinformatic analysis was performed to analyze clonal diversity, TCR convergence frequency and the Shannon diversity from each timepoint. Immune repertoire and clinical correlation were explored using Spearman's correlation and Pearson's correlation. RStudio software version 2021 build 372 was used for analyses. A significance level was at P < 0.05. RESULTS Forty-four blood samples from 12 patients were analyzed. Mean duration of durvalumab treatment was 284 days. After durvalumab treatment, increasing of TCR convergence frequency was found compared to baseline (R = 0.36). Interestingly, it was also significantly higher in non-progressive disease (non-PD) patients compared with progressive disease (PD) patients (P = 0.011). Furthermore, Shannon diversity was higher increasing in PD patients compared with non-PD patients. Taken together, our study found that increasing of TCR convergence with less T-cell diversity in non-PD patients probably demonstrated a T cell-specific clonal expansion response to durvalumab treatment in this population. CONCLUSIONS TCRβ repertoire is the potential biomarker for predicting durvalumab treatment response in post CCRT stage III NSCLC patients. However, a larger cohort with long-read assay should be explored.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/therapy
- Lung Neoplasms/pathology
- Lung Neoplasms/genetics
- Lung Neoplasms/drug therapy
- Male
- Female
- Middle Aged
- Aged
- Neoplasm Staging
- Immune Checkpoint Inhibitors/therapeutic use
- Antibodies, Monoclonal/therapeutic use
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Chemoradiotherapy
- Immunotherapy/methods
- High-Throughput Nucleotide Sequencing
- Biomarkers, Tumor
Collapse
Affiliation(s)
- Nareenart Iemwimangsa
- Center for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Dulyathat Anantaya
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Faculty of Medicine Ramathibodi Hospital, Ramathibodi Lung Cancer Consortium (RLC), Mahidol University, Bangkok, Thailand
| | - Songporn Oranratnachai
- Oncology Unit Sriphat Medical Center, Faculty of Medicine, Chiangmai University, Chiangmai, Thailand
| | - Thanaporn Thamrongjirapat
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Faculty of Medicine Ramathibodi Hospital, Ramathibodi Lung Cancer Consortium (RLC), Mahidol University, Bangkok, Thailand
| | - Putthapoom Lumjiaktase
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | | - Khantong Khiewngam
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Nanamon Monnamo
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Tungpayathai, Rajathewee, Bangkok, 10400, Thailand
| | | | - Pimpin Incharoen
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Angkana Charoenyingwattan
- Center for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Insee Sensorn
- Center for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Thitiya Dejthevaporn
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Faculty of Medicine Ramathibodi Hospital, Ramathibodi Lung Cancer Consortium (RLC), Mahidol University, Bangkok, Thailand
| | - Ekaphop Sirachainan
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Faculty of Medicine Ramathibodi Hospital, Ramathibodi Lung Cancer Consortium (RLC), Mahidol University, Bangkok, Thailand
| | - Wasun Chantratita
- Center for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Thanyanan Reungwetwattana
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Faculty of Medicine Ramathibodi Hospital, Ramathibodi Lung Cancer Consortium (RLC), Mahidol University, Bangkok, Thailand
| | - Narumol Trachu
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Tungpayathai, Rajathewee, Bangkok, 10400, Thailand.
| |
Collapse
|
43
|
Zhang H, Meng X, Wang Z, Zhou X, Liu Y, Li N. Predicting PD-L1 in Lung Adenocarcinoma Using 18F-FDG PET/CT Radiomic Features. Diagnostics (Basel) 2025; 15:543. [PMID: 40075791 PMCID: PMC11899397 DOI: 10.3390/diagnostics15050543] [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: 01/17/2025] [Revised: 02/15/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: This study aims to retrospectively analyze the clinical and imaging data of 101 patients with lung adenocarcinoma who underwent [18F]FDG PET/CT examination and were pathologically confirmed in the Department of Nuclear Medicine at Peking University Cancer Hospital. This study explores the predictive value and important features of [18F]FDG PET/CT radiomics for PD-L1 expression levels in lung adenocarcinoma patients, assisting in screening patients who may benefit from immunotherapy. Methods: 101 patients with histologically confirmed lung adenocarcinoma who received pre-treatment [18F] FDG PET/CT were included. Among them, 44 patients were determined to be PD-L1 positive and 57 patients were determined to be PD-L1 negative based on immunohistochemical assays. Clinical data, PET/CT radiomics parameters, conventional metabolic parameters, and observed CT characteristics were included in the modeling. Random Forest was used in feature denoising, while Forward Stepwise Regression and the Least Absolute Shrinkage and Selection Operator were used in feature selection. Models based on Tree, Discriminant, Logistic Regression, and Support Vector Machine were trained and evaluatedto explore the value of clinical data, PET/CT radiomics parameters, conventional metabolic parameters, and observed CT characteristics. Results: All models showed some predictive ability in distinguishing PD-L1 positive from PD-L1 negative samples. Among the multimodal imaging, clinical data were incorporated into the models, with clinical stage and gender selected by Forward Stepwise Regression, while clinical stage, smoking history, and gender were selected by LASSO. When incorporating clinical data and thin-section CT-derived images into the models, nodular type, spiculation, and CT Shape Flatness were selected by Forward Stepwise Regression, while nodular type and spiculation were selected by LASSO. When incorporating clinical data, PET/CT radiomics, observed CT characteristics, and conventional metabolic information. Forward Stepwise Regression selected TLGlean, MTV, nodule component, PET Shape Sphericity, while LASSO selected SULmax, MTV, nodular type, PET Shape Sphericity, and spiculation. Conclusions: The integration of clinical data, PET/CT radiomics, and conventional metabolic parameters effectively predicted PD-L1 expression, thereby assisting the selection of patients who would benefit from immunotherapy. Observed CT characteristics and conventional metabolic information play an important role in predicting PD-L1 expression levels.
Collapse
Affiliation(s)
- Huiyuan Zhang
- Department of Nuclear Medicine, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China;
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing Key Laboratory of Research, Investigation and Evaluation of Radiopharmaceuticals, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing 100142, China (Y.L.)
| | - Zhe Wang
- United Imaging Healthcare Group, Central Research Institute, Shanghai 201900, China
| | - Xin Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing Key Laboratory of Research, Investigation and Evaluation of Radiopharmaceuticals, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing 100142, China (Y.L.)
| | - Yang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing Key Laboratory of Research, Investigation and Evaluation of Radiopharmaceuticals, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing 100142, China (Y.L.)
| | - Nan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing Key Laboratory of Research, Investigation and Evaluation of Radiopharmaceuticals, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing 100142, China (Y.L.)
| |
Collapse
|
44
|
Niu X, Zhao W, Zhou X, Luo F, Xiao Y, Luo T, Sui X, Li W, Dong Q, Yang X, He Z, Shang W, Sun Y, Gao Y. Chidamide functions as a VISTA/PSGL-1 blocker for cancer immunotherapy. Cancer Immunol Immunother 2025; 74:104. [PMID: 39932560 PMCID: PMC11813839 DOI: 10.1007/s00262-025-03955-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: 10/08/2024] [Accepted: 01/22/2025] [Indexed: 02/14/2025]
Abstract
The response rates of PD-1/PD-L1 blockade in cancer immunotherapy are relatively low, necessitating the development of novel immune checkpoint inhibitors. Compared with other immune checkpoints, VISTA interacts with its ligand PSGL-1 only under acidic conditions in the tumor microenvironment to suppress the function of CD8+ T cells. On the other hand, drug repurposing offers advantages such as time efficiency and high safety. However, the development of VISTA/PSGL-1 inhibitor based on drug repurposing is still infancy. Here, by screening a library of marketed drugs, we identified Chidamide had a strong binding affinity toward VISTA (KD = 5 nM) and blocked VISTA/PSGL-1 under acidic conditions, thereby significantly enhancing the function of CD8+ T cells and inhibiting the tumor growth in immunocompetent murine CT26 tumor model. This study represents the first discovery of Chidamide as VISTA/PSGL-1 blocker for cancer immunotherapy.
Collapse
Affiliation(s)
- Xiaoshuang Niu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Wenshan Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiuman Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Feiyu Luo
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Youmei Xiao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Tao Luo
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Xinghua Sui
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Wanqiong Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Qingyu Dong
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Xin Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Zhuoying He
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Wenzhi Shang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Yixuan Sun
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Yanfeng Gao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
45
|
Delgado-Almenta V, Blaya-Cánovas JL, Calahorra J, López-Tejada A, Griñán-Lisón C, Granados-Principal S. Cancer Vaccines and Beyond: The Transformative Role of Nanotechnology in Immunotherapy. Pharmaceutics 2025; 17:216. [PMID: 40006583 PMCID: PMC11859086 DOI: 10.3390/pharmaceutics17020216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/20/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Cancer is one of the leading causes of morbidity and mortality globally, responsible for approximately 10 million deaths in 2022 and an estimated 21 million new cases in 2024. Traditional cancer treatments such as surgery, radiation therapy, and chemotherapy often present limitations in efficacy and side effects. However, immunotherapeutic vaccines have emerged as a promising approach, leveraging the body's immune system to target and eliminate cancer cells. This review examines the evolving landscape of cancer vaccines, differentiating between preventive and therapeutic strategies and highlighting the significance of tumor-specific antigens, including tumor-associated antigens (TAAs) and neoantigens. Recent advancements in vaccine technology, particularly through nanotechnology, have resulted in the development of nanovaccines, which enhance antigen stability, optimize delivery to immune cells, and promote robust immune responses. Notably, clinical data indicate that patients receiving immune checkpoint inhibitors can achieve overall survival rates of approximately 34.8 months compared to just 15.7 months for traditional therapies. Despite these advancements, challenges remain, such as the immunosuppressive tumor microenvironment and tumor heterogeneity. Emerging evidence suggests that combining nanovaccines with immunomodulators may enhance therapeutic efficacy by overcoming these obstacles. Continued research and interdisciplinary collaboration will be essential to fully exploit the promise of nanovaccines, ultimately leading to more effective and accessible treatments for cancer patients. The future of cancer immunotherapy appears increasingly hopeful as these innovative strategies pave the way for enhanced patient outcomes and an improved quality of life in oncology.
Collapse
Affiliation(s)
- Violeta Delgado-Almenta
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
| | - Jose L. Blaya-Cánovas
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Jesús Calahorra
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Araceli López-Tejada
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18011 Granada, Spain
| | - Carmen Griñán-Lisón
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18011 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), Centro de Investigación Biomédica (CIBM), University of Granada, 18016 Granada, Spain
| | - Sergio Granados-Principal
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18011 Granada, Spain
| |
Collapse
|
46
|
Gu Y, Zou X, Zhu J, Wu G. Efficacy and safety of camrelizumab combined with chemotherapy as second-line treatment for locally advanced, recurrent, or metastatic esophageal squamous cell carcinoma. World J Surg Oncol 2025; 23:38. [PMID: 39905538 PMCID: PMC11792583 DOI: 10.1186/s12957-025-03690-9] [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: 10/06/2024] [Accepted: 01/28/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND This study aims to evaluate the efficacy and safety of camrelizumab in combination with chemotherapy as a second-line treatment for patients with locally advanced, recurrent, or metastatic esophageal squamous cell carcinoma (ESCC). METHODS In this retrospective, single-center observational study, we collected medical records of patients with locally advanced, recurrent, or metastatic ESCC who received either camrelizumab combined with chemotherapy or chemotherapy alone as second-line treatment between July 1, 2019, and May 31, 2023. We evaluated short-term efficacy, including overall response rate (ORR) and disease control rate (DCR), as well as survival outcomes, including progression-free survival (PFS) and overall survival (OS). Safety was also assessed. Additionally, factors influencing OS in ESCC patients were analyzed. RESULTS A total of 60 patients with locally advanced, recurrent, or metastatic ESCC were included, with 30 receiving camrelizumab combined with chemotherapy and 30 receiving chemotherapy alone as second-line treatment. There were no statistically significant differences in ORR (33.33% vs. 13.33%) and DCR (73.33% vs. 56.67%) between the combination therapy and chemotherapy-alone groups (P > 0.05). However, the median PFS was significantly longer in the combination therapy group compared to the chemotherapy group (4.7 months vs. 3.4 months, P = 0.048). Additionally, the median OS was significantly improved in the combination therapy group compared to the chemotherapy group (11.7 months vs. 6.5 months, P = 0.003). Age and history of radical surgery were significantly associated with OS in patients receiving camrelizumab combined with chemotherapy as second-line treatment (P < 0.05). CONCLUSION Second-line treatment with camrelizumab combined with chemotherapy is well-tolerated and associated with favorable oncological outcomes in patients with locally advanced, recurrent, or metastatic ESCC. Furthermore, younger patients and those who have undergone radical surgery may derive greater benefit from camrelizumab combined with chemotherapy as a second-line treatment.
Collapse
Affiliation(s)
- Yinfang Gu
- Department of Oncology, Cancer Center, Meizhou People's Hospital (Huangtang Hospital, Meizhou Academy of Medical Sciences, 63 Huangtang Road, Meizhou, 514031, Guangdong, People's Republic of China.
- Guangdong Provincial Engineering and Technological Research Center for Clinical Molecular Diagnosis and Antibody Drugs, 63 Huangtang Road, Meizhou, 514031, Guangdong, People's Republic of China.
| | - Xiaofang Zou
- Department of Oncology, Cancer Center, Meizhou People's Hospital (Huangtang Hospital, Meizhou Academy of Medical Sciences, 63 Huangtang Road, Meizhou, 514031, Guangdong, People's Republic of China
- Guangdong Provincial Engineering and Technological Research Center for Clinical Molecular Diagnosis and Antibody Drugs, 63 Huangtang Road, Meizhou, 514031, Guangdong, People's Republic of China
| | - Junlin Zhu
- Department of Oncology, Cancer Center, Meizhou People's Hospital (Huangtang Hospital, Meizhou Academy of Medical Sciences, 63 Huangtang Road, Meizhou, 514031, Guangdong, People's Republic of China
- Guangdong Provincial Engineering and Technological Research Center for Clinical Molecular Diagnosis and Antibody Drugs, 63 Huangtang Road, Meizhou, 514031, Guangdong, People's Republic of China
| | - Guowu Wu
- Department of Oncology, Cancer Center, Meizhou People's Hospital (Huangtang Hospital, Meizhou Academy of Medical Sciences, 63 Huangtang Road, Meizhou, 514031, Guangdong, People's Republic of China.
- Guangdong Provincial Engineering and Technological Research Center for Clinical Molecular Diagnosis and Antibody Drugs, 63 Huangtang Road, Meizhou, 514031, Guangdong, People's Republic of China.
| |
Collapse
|
47
|
Jo A, Shoji T, Otsuka H, Abe M, Tatsuki S, Chiba Y, Sato S, Takatori E, Kaido Y, Nagasawa T, Kagabu M, Baba T. Treatment strategies for advanced and recurrent endometrial cancer using immune checkpoint inhibitors. Int J Clin Oncol 2025; 30:229-240. [PMID: 39812928 DOI: 10.1007/s10147-024-02689-8] [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: 08/02/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025]
Abstract
Doxorubicin + cisplatin and paclitaxel + carboplatin are standard chemotherapy regimens for endometrial cancer. The development of PD-1 and PDL-1 antibody drugs has led to the use of these agents for endometrial cancer in other countries. The KEYNOTE-775 trial for advanced or recurrent endometrial cancer demonstrated the benefits of pembrolizumab and lenvatinib combination therapy, and the results of this trial led to the approval of its coverage for recurrent cancer by the Japanese health insurance system. Currently, treatment with immune checkpoint inhibitors is transitioning from second-line to first-line therapy. In a global randomized phase III study, the drugs dostarlimab, durvalumab, and atezolizumab, which are not yet approved in Japan, showed better results in the study arms than in the control arm. Additionally, biomarkers have been developed for endometrial cancer, enabling gynecologists to pursue treatment options based on the biomarkers detected for better treatment outcomes. In this article, we review the clinical trials of immune checkpoint inhibitors for advanced or recurrent endometrial cancer.
Collapse
Affiliation(s)
- Ami Jo
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Tadahiro Shoji
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan.
| | - Haruka Otsuka
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Marina Abe
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Shunsuke Tatsuki
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Yohei Chiba
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Sho Sato
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Eriko Takatori
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Yoshitaka Kaido
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Takayuki Nagasawa
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Masahiro Kagabu
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Tsukasa Baba
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| |
Collapse
|
48
|
Münter D, de Faria FW, Richter M, Aranda-Pardos I, Hotfilder M, Walter C, Paga E, Inserte C, Albert TK, Roy R, Rahman S, Riedel NC, Müller V, Pascher A, Wiebe K, Schmid I, Vokuhl C, Winkler B, Jüttner E, Vieth S, Mücke U, Kluiver TA, Peng WC, Rossig C, Schlué J, Madadi-Sanjani O, Sandmann S, Hartmann W, A-Gonzalez N, Soehnlein O, Kerl K. Multiomic analysis uncovers a continuous spectrum of differentiation and Wnt-MDK-driven immune evasion in hepatoblastoma. J Hepatol 2025:S0168-8278(25)00068-6. [PMID: 39900120 DOI: 10.1016/j.jhep.2025.01.031] [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: 04/04/2024] [Revised: 12/04/2024] [Accepted: 01/27/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND & AIMS Hepatoblastoma is the most common pediatric cancer of the liver, with the majority of cases displaying activating mutations in the Wnt/β-catenin pathway. Understanding the complex milieu of the tumor microenvironment has resulted in promising new therapies for adult cancers, but similar approaches in pediatric cancers are still lacking. We aimed to provide a comprehensive analysis of the tumor microenvironment of hepatoblastoma, unveiling its spatial architecture and key signaling mechanisms. METHODS Single-cell/-nucleus RNA-sequencing (RNA-seq) (n = 15), spatial transcriptomics (n = 22), and multiplex immunofluorescence stainings (n = 7) of treated, untreated, and metastasized pediatric hepatoblastomas were performed. An RNA-seq validation cohort (n = 110) including hepatoblastoma, non-tumor and fetal liver samples and single-cell RNA-seq data of healthy immune cells were used for further analysis. Western blotting and RNA-seq of hepatoblastoma and macrophage cell lines were conducted for experimental validation. RESULTS Of four identified transcriptional tumor programs, "Developmental" and "Metabolic" reflected different hepatic differentiation stages, while "Cycling" was enriched in undifferentiated cells and relapsed samples, and "Intermediate" displayed high activity in samples from patients with poor outcomes. We discovered an increased ratio of anti-to pro-inflammatory immune cells and evidence of immune exclusion from tumor areas. Wnt-responsive upregulation of the immunomodulator midkine in hepatoblastoma cells was associated with a change in macrophage phenotype, which could be partially reversed through midkine inhibition. CONCLUSIONS Hepatoblastoma cells exist along a continuous spectrum of hepatic differentiation and inhabit an altered immune environment. Wnt signaling augments midkine expression, which appears to be involved in shaping the immune environment by modifying macrophages to enable immune evasion, thereby providing a potential therapeutic target. IMPACT AND IMPLICATIONS Despite hepatoblastoma being the most common pediatric liver cancer, there has been a critical knowledge gap in understanding how the tumor microenvironment and immune landscape contribute to disease progression. Our novel findings, revealing a continuous spectrum of tumor differentiation states and Wnt-MDK-driven immune evasion, are significant for pediatric oncology clinicians and researchers, improving our functional understanding of the immune environment of hepatoblastoma. The identification of midkine as a tumor-specific immunomodulator suggests a potential for developing new targeted therapies, though further mechanistic and practical validation would be needed to realize clinical translation of these findings.
Collapse
Affiliation(s)
- Daniel Münter
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Flavia W de Faria
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Mathis Richter
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | | | - Marc Hotfilder
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Carolin Walter
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Enya Paga
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Clara Inserte
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany; Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Thomas K Albert
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Rajanya Roy
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Shariyah Rahman
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Nicole C Riedel
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Volker Müller
- Department of Pediatric Surgery, University Hospital Münster, Münster, Germany
| | - Andreas Pascher
- Department of General, Visceral and Transplantation Surgery, University Hospital Münster, Münster, Germany
| | - Karsten Wiebe
- Department of Cardiothoracic Surgery, University Hospital Münster, Münster, Germany
| | - Irene Schmid
- Department of Pediatric Oncology and Hematology, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | - Beate Winkler
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eva Jüttner
- Department of Pathology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Simon Vieth
- Department of Pediatrics, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Urs Mücke
- Pediatric Oncology and Hematology, Medical School of Hanover, Hanover, Germany
| | - Thomas A Kluiver
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Weng Chuan Peng
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Claudia Rossig
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany; Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Jerome Schlué
- Institute for Pathology, Medical School of Hanover, Hanover, Germany
| | - Omid Madadi-Sanjani
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Sandmann
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Wolfgang Hartmann
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Münster, Germany
| | | | - Oliver Soehnlein
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Kornelius Kerl
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany.
| |
Collapse
|
49
|
Li J, Li Y, Tao L, Zhang C, Zuo Z. Diagnostic and Prognostic Value of Cardiac Magnetic Resonance for Cardiotoxicity Caused by Immune Checkpoint Inhibitors: A Systematic Review and Meta-Analysis. Rev Cardiovasc Med 2025; 26:25508. [PMID: 40026491 PMCID: PMC11868891 DOI: 10.31083/rcm25508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/31/2024] [Accepted: 11/11/2024] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND The complex process of cardiac magnetic resonance (CMR) and the uncertainty of each parameter in the diagnosis and prognosis of cardiotoxicity limit its promotion in the cardiac evaluation of patients treated with immune checkpoint inhibitors (ICI). METHODS A comprehensive search was conducted across PubMed, Web of Science, Embase, China National Knowledge Infrastructure (CNKI), and Cochrane databases for relevant articles published up until September 28, 2024. RESULTS After screening, 8 articles were included in this study. The analysis revealed that following ICI treatment, the left ventricular global longitudinal strain (GLS) increased significantly [weighted mean difference (WMD) 2.33; 95% confidence interval (CI) 1.26, 3.41; p < 0.01], while the global radial strain (GRS) decreased [WMD -4.73; 95% CI -6.74, -2.71; p < 0.01]. Additionally, T1 and T2 values increased [standardized mean difference (SMD) 1.14; 95% CI 0.59, 1.68; p < 0.01] and [SMD 1.11; 95% CI 0.64, 1.58; p < 0.01], respectively. An elevated T2 was associated with a higher occurrence of major adverse cardiovascular events (MACE), with a hazard ratio of 1.36 (95% CI 1.12, 1.64). CONCLUSIONS Our findings demonstrate that T1, T2, and GLS increase, while GRS decreases following ICI administration. By consolidating these critical metrics, we propose a streamlined, abbreviated (non-contrast) CMR protocol that can be completed within 15 minutes, thereby facilitating the integration of CMR in cardio-oncology. THE PROSPERO REGISTRATION CRD42023437238, https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023437238.
Collapse
Affiliation(s)
- Jialian Li
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Yanwei Li
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Li Tao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Chuan Zhang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Zhong Zuo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| |
Collapse
|
50
|
Ostroumov D, Benne N, Lozano Vigario F, Escalona-Rayo O, Dodz K, Sauer S, Suhl LL, Wedemeyer HH, Kühnel F, Slütter B, Wirth TC. Sequential STING and CD40 agonism drives massive expansion of tumor-specific T cells in liposomal peptide vaccines. Cell Mol Immunol 2025; 22:150-160. [PMID: 39741195 PMCID: PMC11782543 DOI: 10.1038/s41423-024-01249-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 11/12/2024] [Accepted: 11/27/2024] [Indexed: 01/02/2025] Open
Abstract
The clinical use of cancer vaccines is hampered by the low magnitude of induced T-cell responses and the need for repetitive antigen stimulation. Here, we demonstrate that liposomal formulations with incorporated STING agonists are optimally suited to deliver peptide antigens to dendritic cells in vivo and to activate dendritic cells in secondary lymphoid organs. One week after liposomal priming, systemic administration of peptides and a costimulatory agonistic CD40 antibody enables ultrarapid expansion of T cells, resulting in massive expansion of tumor-specific T cells in the peripheral blood two weeks after priming. In the MC-38 colon cancer model, this synthetic prime-boost regimen induces rapid regression and cure of large established subcutaneous cancers via the use of a single tumor-specific neoantigen. These experiments demonstrate the feasibility of liposome-based heterologous vaccination regimens to increase the therapeutic efficacy of peptide vaccines in the context of immunogenic adjuvants and costimulatory booster immunizations. Our results provide a rationale for the further development of modern liposomal peptide vaccines for cancer therapy.
Collapse
Affiliation(s)
- Dmitrij Ostroumov
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Naomi Benne
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Fernando Lozano Vigario
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Oscar Escalona-Rayo
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Ksenia Dodz
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Sarah Sauer
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Lena Luisa Suhl
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Hans Heiner Wedemeyer
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Florian Kühnel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Bram Slütter
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Thomas Christian Wirth
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|