1
|
Yuan D, Gao Y, Xia L, Liu H, Wu X, Ding X, Huang Y, Deng C, Li J, Dai W, Liu J, Ma J. Discovery of novel biphenyl compounds bearing hydroxamic acid moiety as the first PD-L1/class I HDACs dual inhibitors. J Enzyme Inhib Med Chem 2025; 40:2461190. [PMID: 39912413 PMCID: PMC11803765 DOI: 10.1080/14756366.2025.2461190] [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: 11/29/2024] [Revised: 01/06/2025] [Accepted: 01/27/2025] [Indexed: 02/07/2025] Open
Abstract
Herein, we firstly reported a series of biphenyl compounds bearing hydroxamic acid moiety as PD-L1/class I HDACs dual inhibitors. Among them, compound 14 displayed the strongest inhibitory activity in vitro against HDAC2 and HDAC3 with IC50 values of 27.98 nM and 14.47 nM, and had an IC50 value of 88.10 nM for PD-1/PD-L1 interaction. Importantly, 14 could upregulate the expression of PD-L1 and CXCL10 in a PD-L1 low-expression cancer cell line (MCF-7), highlighting the potential to enhance efficacy by recruiting T-cell infiltration into TME and improving the response of PD-1/PD-L1 inhibitor associated with PD-L1 low-expression. Besides, we identified another compound, 22, which possessed the strongest inhibitory activity against PD-1/PD-L1 interaction with an IC50 value of 12.47 nM, and effectively inhibited the proliferation of three cancer cell lines. Our results suggest that compounds 14 and 22 can be served as lead compounds of PD-L1/class I HDACs dual inhibitors for further optimisation.
Collapse
Affiliation(s)
- Dandan Yuan
- School of Medicine, Huaqiao University, Quanzhou, China
| | - Yali Gao
- Pharmacy Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Lin Xia
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Han Liu
- School of Medicine, Huaqiao University, Quanzhou, China
| | - Xingye Wu
- School of Medicine, Huaqiao University, Quanzhou, China
| | - Xueyan Ding
- School of Medicine, Huaqiao University, Quanzhou, China
| | - Yudan Huang
- School of Medicine, Huaqiao University, Quanzhou, China
| | | | - Jin Li
- School of Medicine, Huaqiao University, Quanzhou, China
| | - Wenqi Dai
- School of Medicine, Huaqiao University, Quanzhou, China
| | - Jieqing Liu
- School of Medicine, Huaqiao University, Quanzhou, China
| | - Junjie Ma
- School of Medicine, Huaqiao University, Quanzhou, China
| |
Collapse
|
2
|
Zhang Z, Zhao Q, Xu Q, Deng Q, Hua A, Wang X, Yang X, Li Z. A mitochondria-interfering nanocomplex cooperates with photodynamic therapy to boost antitumor immunity. Biomaterials 2025; 317:123094. [PMID: 39799701 DOI: 10.1016/j.biomaterials.2025.123094] [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: 11/07/2024] [Revised: 01/05/2025] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
Immunotherapeutics against triple-negative breast cancer (TNBC) hold great promise. In this work, we provide a combination therapy for simultaneous increasing tumor immunogenicity and down-regulating programmed cell death ligand 1 (PD-L1) to boost antitumor immunity in TNBC. We prepare bis (diethyldithiocarbamate)-copper/indocyanine green nanoparticles (CuET/ICG NPs) simply in aqueous with one-pot method. CuET/ICG NPs interfere mitochondria, reduce oxygen consumption, and alleviate tumor hypoxia to potentiate photodynamic therapy (PDT) for amplifying immunogenic cell death (ICD). Meanwhile, mitochondria dysfunction leads to energy stress and activates AMPK pathway. As a result, CuET/ICG NPs downregulates membrane PD-L1 (mPD-L1) on both 4T1 cancer cells and cancer stem cells (CSCs) through AMP-activated protein kinase (AMPK)-mediated pathway in hypoxia. Cooperatively, the combinational therapy activates antitumor immunity and triggers long lasting immune memory response to resist tumor re-challenge. Our study represents an attempt that conquers tumor immunosuppressive microenvironment with simple biomedical materials and multimodality treatments.
Collapse
Affiliation(s)
- Zhijie Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Qingfu Zhao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Qingqing Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Qingyuan Deng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Ao Hua
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Xing Wang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, PR China.
| | - Zifu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, PR China.
| |
Collapse
|
3
|
Aden D, Zaheer S, Sureka N, Trisal M, Chaurasia JK, Zaheer S. Exploring immune checkpoint inhibitors: Focus on PD-1/PD-L1 axis and beyond. Pathol Res Pract 2025; 269:155864. [PMID: 40068282 DOI: 10.1016/j.prp.2025.155864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025]
Abstract
Immunotherapy emerges as a promising approach, marked by recent substantial progress in elucidating how the host immune response impacts tumor development and its sensitivity to various treatments. Immune checkpoint inhibitors have revolutionized cancer therapy by unleashing the power of the immune system to recognize and eradicate tumor cells. Among these, inhibitors targeting the programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have garnered significant attention due to their remarkable clinical efficacy across various malignancies. This review delves into the mechanisms of action, clinical applications, and emerging therapeutic strategies surrounding PD-1/PD-L1 blockade. We explore the intricate interactions between PD-1/PD-L1 and other immune checkpoints, shedding light on combinatorial approaches to enhance treatment outcomes and overcome resistance mechanisms. Furthermore, we discuss the expanding landscape of immune checkpoint inhibitors beyond PD-1/PD-L1, including novel targets such as CTLA-4, LAG-3, TIM-3, and TIGIT. Through a comprehensive analysis of preclinical and clinical studies, we highlight the promise and challenges of immune checkpoint blockade in cancer immunotherapy, paving the way for future advancements in the field.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India.
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Monal Trisal
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| |
Collapse
|
4
|
Jiang Q, Chen Z, Jiang J, Chen Q, Lan H, Zhu J, Mao W. The role of cGAS-STING in remodeling the tumor immune microenvironment induced by radiotherapy. Crit Rev Oncol Hematol 2025; 209:104658. [PMID: 39956501 DOI: 10.1016/j.critrevonc.2025.104658] [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: 11/04/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/18/2025] Open
Abstract
The activation of the cGAS-STING pathway occurs when tumor cell DNA is damaged by ionizing radiation. Once triggered, this pathway reshapes the tumor immune microenvironment by promoting the maturation, activation, polarization, and immune-killing capacity of immune cells, as well as by inducing the release of interferons and the expression of immune-related genes. In addition, the gut microbiota and various mechanisms of programmed cell death interact with the cGAS-STING pathway, further influencing its function in remodeling the immune microenvironment after radiotherapy. Therefore, investigating the mechanisms of the cGAS-STING pathway in reshaping the tumor immune microenvironment post-radiotherapy can not only optimize the efficacy of combined radiotherapy and immunotherapy but also provide new research directions and potential targets for cancer treatment.
Collapse
Affiliation(s)
- Qingyu Jiang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Zhejiang Chinese Medical University, Hangzhou 310053, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Zhiheng Chen
- Department of Oncology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing 31400, China
| | - Jin Jiang
- Department of Oncology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing 31400, China
| | - Qianping Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Huiyin Lan
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Ji Zhu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China.
| | - Wei Mao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China.
| |
Collapse
|
5
|
Zhang R, Tan Y, Xu K, Huang N, Wang J, Liu M, Wang L. Cuproplasia and cuproptosis in hepatocellular carcinoma: mechanisms, relationship and potential role in tumor microenvironment and treatment. Cancer Cell Int 2025; 25:137. [PMID: 40205387 PMCID: PMC11983883 DOI: 10.1186/s12935-025-03683-4] [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: 05/19/2024] [Accepted: 02/08/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the main phenotype of liver cancer with a poor prognosis. Copper is vital in liver function, and HCC cells rely on it for growth and metastasis, leading to cuproplasia. Excessive copper can induce cell death, termed cuproptosis. Tumor microenvironment (TME) is pivotal in HCC, especially in immunotherapy, and copper is closely related to the TME pathogenesis. However, how these two mechanisms contribute to the TME is intriguing. MAIN BODY We conducted the latest progress literature on cuproplasia and cuproptosis in HCC, and summarized their specific roles in TME and treatment strategies. The mechanisms of cuproplasia and cuproptosis and their relationship and role in TME have been deeply summarized. Cuproplasia fosters TME formation, angiogenesis, and metastasis, whereas cuproptosis may alleviate mitochondrial dysfunction and hypoxic conditions in the TME. Inhibiting cuproplasia and enhancing cuproptosis in HCC are essential for achieving therapeutic efficacy in HCC. CONCLUSION An in-depth analysis of cuproplasia and cuproptosis mechanisms within the TME of HCC unveils their opposing nature and their impact on copper regulation. Grasping the equilibrium between these two factors is crucial for a deeper understanding of HCC mechanisms to shed light on novel directions in treating HCC.
Collapse
Affiliation(s)
- Ruoyu Zhang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli Area, Chaoyang District, Beijing, 100021, China
| | - Yunfei Tan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Unit III, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ke Xu
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli Area, Chaoyang District, Beijing, 100021, China
| | - Ning Huang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli Area, Chaoyang District, Beijing, 100021, China
| | - Jian Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Mei Liu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, P.O. Box 2258, 100021, Beijing, People's Republic of China.
| | - Liming Wang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli Area, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
6
|
Wahnou H, El Kebbaj R, Hba S, Ouadghiri Z, El Faqer O, Pinon A, Liagre B, Limami Y, Duval RE. Neutrophils and Neutrophil-Based Drug Delivery Systems in Anti-Cancer Therapy. Cancers (Basel) 2025; 17:1232. [PMID: 40227814 PMCID: PMC11988188 DOI: 10.3390/cancers17071232] [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: 03/08/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/15/2025] Open
Abstract
Neutrophils, the most abundant white blood cells, play a dual role in cancer progression. While they can promote tumor growth, metastasis, and immune suppression, they also exhibit anti-tumorigenic properties by attacking cancer cells and enhancing immune responses. This review explores the complex interplay between neutrophils and the tumor microenvironment (TME), highlighting their ability to switch between pro- and anti-tumor phenotypes based on external stimuli. Pro-tumorigenic neutrophils facilitate tumor growth through mechanisms such as neutrophil extracellular traps (NETs), secretion of pro-inflammatory cytokines, and immune evasion strategies. They contribute to angiogenesis, tumor invasion, and metastasis by releasing vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMPs). Conversely, anti-tumor neutrophils enhance cytotoxicity by generating reactive oxygen species (ROS), promoting antibody-dependent cell-mediated cytotoxicity (ADCC), and activating other immune cells such as cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells. Recent advances in neutrophil-based drug delivery systems have harnessed their tumor-homing capabilities to improve targeted therapy. Neutrophil-mimicking nanoparticles and membrane-coated drug carriers offer enhanced drug accumulation in tumors, reduced systemic toxicity, and improved therapeutic outcomes. Additionally, strategies to modulate neutrophil activity, such as inhibiting their immunosuppressive functions or reprogramming them towards an anti-tumor phenotype, are emerging as promising approaches in cancer immunotherapy. Understanding neutrophil plasticity and their interactions with the TME provides new avenues for therapeutic interventions. Targeting neutrophil-mediated mechanisms could enhance existing cancer treatments and lead to the development of novel immunotherapies, ultimately improving patient survival and clinical outcomes.
Collapse
Affiliation(s)
- Hicham Wahnou
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
| | - Riad El Kebbaj
- Sciences and Engineering of Biomedicals, Biophysics and Health Laboratory, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco;
| | - Soufyane Hba
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (A.P.); (B.L.)
| | - Zaynab Ouadghiri
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
| | - Othman El Faqer
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
| | - Aline Pinon
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (A.P.); (B.L.)
| | - Bertrand Liagre
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (A.P.); (B.L.)
| | - Youness Limami
- Sciences and Engineering of Biomedicals, Biophysics and Health Laboratory, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco;
| | | |
Collapse
|
7
|
Xi X, Guo S, Gu Y, Wang X, Wang Q. Challenges and opportunities in single-domain antibody-based tumor immunotherapy. Biochim Biophys Acta Rev Cancer 2025; 1880:189284. [PMID: 39947441 DOI: 10.1016/j.bbcan.2025.189284] [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: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025]
Abstract
Single-domain antibodies (sdAbs) have emerged as a promising tool in tumor immunotherapy, garnering significant attention in recent years due to their unique structure and superior properties. Unlike traditional antibodies, sdAbs exhibit several advantages, including small molecular weight, high stability, strong affinity, and high specificity. These characteristics enable sdAbs to effectively target and eliminate tumor cells within the complex tumor microenvironment. Moreover, their structural advantages enhance tissue penetration and reduce immunogenicity, thereby increasing their potential for clinical application. The potential applications of sdAbs include novel immune checkpoint inhibitors, bispecific antibody drugs, innovative immune cell therapies, antibody-drug conjugate therapies, and tumor molecular imaging diagnostics. Despite the promising prospects, several challenges of sdAb-based tumor immunotherapy still require further investigation. This review aims to summarize the status of sdAb-based immunotherapy, identify the challenges encountered, and evaluate the clinical research and application potential of sdAbs in this field.
Collapse
Affiliation(s)
- Xiaozhi Xi
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan 250022, People's Republic of China.; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, People's Republic of China.; Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 266003 Qingdao, People's Republic of China
| | - Shasha Guo
- Shandong Women's University, 250355 Jinan, People's Republic of China
| | - Yuchao Gu
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xuekai Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan 250022, People's Republic of China.; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, People's Republic of China
| | - Qiang Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan 250022, People's Republic of China.; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, People's Republic of China.; Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 266003 Qingdao, People's Republic of China.
| |
Collapse
|
8
|
Zhang A, Fan L, Liu Q, Zuo X, Zhu J. Immunological Effects of Proton Radiotherapy: New Opportunities and Challenges in Cancer Therapy. CANCER INNOVATION 2025; 4:e70003. [PMID: 40061827 PMCID: PMC11885950 DOI: 10.1002/cai2.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/23/2024] [Accepted: 01/16/2025] [Indexed: 03/14/2025]
Abstract
Radiation therapy can be categorised by particle type into photon, proton and heavy ion therapies. Proton radiotherapy is highlighted due to its unique physical properties, such as the Bragg peak and minimal exit dose, which offer superior dose distribution. This makes proton radiotherapy especially advantageous for treating tumours near vital organs with complex structures, such as gliomas near the brain, nasopharyngeal carcinoma near the brainstem and mediastinal tumours near the heart. Proton irradiation can induce distant effects through immunogenicity within the target area. The reduced low-dose zone outside the target provides better lymphatic system protection and immune benefits. Additionally, combining proton radiotherapy with immunotherapy may offer further biological advantages. These features make proton radiotherapy a promising option in cancer treatment. This article may aid in the understanding of proton radiotherapy and its immune effects and lead to new effective options for tumour treatment.
Collapse
Affiliation(s)
- Anhang Zhang
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Department of Radiation Oncology Physics and TechnologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Shandong Provincial Key Medical and Health Laboratory of Pediatric Cancer Precision Radiotherapy (Shandong Cancer Hospital)JinanShandongChina
| | - Liyuan Fan
- Department of Radiation OncologyQilu Hospital of Shandong UniversityJinanShandongChina
| | - Qi Liu
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Department of Radiation Oncology Physics and TechnologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Shandong Provincial Key Medical and Health Laboratory of Pediatric Cancer Precision Radiotherapy (Shandong Cancer Hospital)JinanShandongChina
| | - Xiaoxin Zuo
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Department of Radiation Oncology Physics and TechnologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Shandong Provincial Key Medical and Health Laboratory of Pediatric Cancer Precision Radiotherapy (Shandong Cancer Hospital)JinanShandongChina
| | - Jian Zhu
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Department of Radiation Oncology Physics and TechnologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Shandong Provincial Key Medical and Health Laboratory of Pediatric Cancer Precision Radiotherapy (Shandong Cancer Hospital)JinanShandongChina
| |
Collapse
|
9
|
Hou R, Wu X, Wang C, Fan H, Zhang Y, Wu H, Wang H, Ding J, Jiang H, Xu J. Tumor‑associated neutrophils: Critical regulators in cancer progression and therapeutic resistance (Review). Int J Oncol 2025; 66:28. [PMID: 40017131 PMCID: PMC11900975 DOI: 10.3892/ijo.2025.5734] [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: 11/21/2024] [Accepted: 02/11/2025] [Indexed: 03/01/2025] Open
Abstract
Cancer is the second leading cause of death among humans worldwide. Despite remarkable improvements in cancer therapies, drug resistance remains a significant challenge. The tumor microenvironment (TME) is intimately associated with therapeutic resistance. Tumor‑associated neutrophils (TANs) are a crucial component of the TME, which, along with other immune cells, play a role in tumorigenesis, development and metastasis. In the current review, the roles of TANs in the TME, as well as the mechanisms of neutrophil‑mediated resistance to cancer therapy, including immunotherapy, chemotherapy, radiotherapy and targeted therapy, were summarized. Furthermore, strategies for neutrophil therapy were discussed and TANs were explored as potential targets for cancer treatment. In conclusion, the need to explore the precise roles, recruitment pathways and mechanisms of action of TANs was highlighted for the purpose of developing therapies that precisely target TANs and reverse drug resistance.
Collapse
Affiliation(s)
- Rui Hou
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Xi Wu
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Cenzhu Wang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Hanfang Fan
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Yuhan Zhang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Hanchi Wu
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Huiyu Wang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Junli Ding
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Huning Jiang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| | - Junying Xu
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Nanjing 214023, P.R. China
| |
Collapse
|
10
|
Li S, Yu Y, Xu Y, Zhou Y, Huang J, Jia J. Clinicopathological characteristics and the relationship of PD-L1 status, tumor mutation burden, and microsatellite instability in patients with esophageal carcinoma. BMC Cancer 2025; 25:576. [PMID: 40165109 PMCID: PMC11956183 DOI: 10.1186/s12885-025-13938-y] [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: 12/26/2024] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Despite significant advancements in the field of immunotherapy for esophageal cancer in recent years, only a minority of patients respond to these treatments, and effective predictive biomarkers remain elusive. Biomarkers such as programmed cell death 1 ligand 1 (PD-L1), tumor mutational burden (TMB), and microsatellite instability (MSI) are pivotal in guiding immune checkpoint inhibitor therapies. This study aimed to explore the correlation between the three biomarkers in patients with esophageal carcinoma. METHODS We collected one hundred esophageal squamous cell carcinoma (ESCC) tumor samples from patients who have been undergoing radical resection of esophageal carcinoma. Each tissue sample was divided into two parts for next-generation sequencing (NGS) and immunohistochemical staining. Mutations were identified using the NGS database, and TMB was calculated. Multiplex PCR targeting five loci (NR21, NR24, NR27, BAT25, and BAT26) was used to evaluate MSI. PD-L1 expression was determined through immunohistochemical analysis. RESULTS Among the 100 ESCC patients, 54% (54/100) exhibited positive PD-L1 expression, 57% (57/100) demonstrated high TMB (TMB-H), and only 1% (1/100) had high MSI (MSI-H). Within the subset of TMB-H cases, 32 showed positive PD-L1 expression, with a single case displaying high expression of all three biomarkers, and 21 cases displaying low expression of all three biomarkers. There was no statistical association between PD-L1 expression levels and TMB. Further analysis showed a significant correlation between TNM staging and PD-L1 expression levels in ESCC tissues, with higher positive rates of PD-L1 expression observed in advanced stages. Similarly, a significant relationship was observed between TMB and lymph node metastasis. CONCLUSIONS Based on our preliminary results, TMB and PD-L1 can serve as potential early screening clinical biomarkers and molecular targets for immune treatment in ESCC. However, there is no apparent statistical association between TMB and PD-L1 expression levels. Furthermore, PD-L1 and TMB may independently influence the efficacy of immunotherapy, highlighting the inadequacy of single-marker detection in effectively predicting treatment outcomes.
Collapse
Affiliation(s)
- Suyao Li
- The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei, China
| | - Yongling Yu
- The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei, China
| | - Yirong Xu
- Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Yue Zhou
- Wuxi People's Hospital, Wuxi, Jiangsu, China
| | - Junxing Huang
- The Affilitated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China.
| | - Jinghao Jia
- The Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei, China.
| |
Collapse
|
11
|
Rahimi A, Baghernejadan Z, Hazrati A, Malekpour K, Samimi LN, Najafi A, Falak R, Khorramdelazad H. Combination therapy with immune checkpoint inhibitors in colorectal cancer: Challenges, resistance mechanisms, and the role of microbiota. Biomed Pharmacother 2025; 186:118014. [PMID: 40157004 DOI: 10.1016/j.biopha.2025.118014] [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: 01/08/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Colorectal cancer (CRC) is still one of the leading causes of cancer deaths worldwide. Even though there has been progress in cancer immunotherapy, the results of applying immune checkpoint inhibitors (ICIs) have been unsatisfactory, especially in microsatellite stable (MSS) CRC. Single-agent ICIs that target programmed cell death-1 (PD-1)/ PD-L1, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), T cell Ig- and mucin-domain-containing molecule-3 (TIM-3), and lymphocyte activation gene (LAG)-3 have emerged as having specific benefits. However, many primary and secondary resistance mechanisms are available in the tumor microenvironment (TME) that prevent it from happening. Combination strategies, such as the use of anti-PD-1 and anti-CTLA-4, can be effective in overcoming these resistance pathways, but toxicities remain a significant concern. Moreover, ICIs have been integrated with various treatment modalities, including chemotherapy, radiotherapy, antibiotics, virotherapy, polyadenosine diphosphate-ribose polymerase (PARP) inhibitors, and heat shock protein 90 (HSP90) inhibitors. The outcomes observed in both preclinical and clinical settings have been encouraging. Interestingly, manipulating gut microbiota via fecal microbiota transplantation (FMT) has been identified as a new strategy to increase the efficacy of immunotherapy in CRC patients. Therefore, integrating ICIs with other treatment approaches holds promise in enhancing the prognosis of CRC patients. This review focuses on the unmet need for new biomarkers to select patients for combination therapies and the ongoing work to overcome resistance and immune checkpoint blockade.
Collapse
Affiliation(s)
- Ali Rahimi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Baghernejadan
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Alireza Najafi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
12
|
Mielcarska S, Kula A, Dawidowicz M, Waniczek D, Świętochowska E. Prognostic Significance of B7H3 Expression in Solid Tumors: A Systematic Review and Meta-Analysis. Int J Mol Sci 2025; 26:3044. [PMID: 40243697 PMCID: PMC11988431 DOI: 10.3390/ijms26073044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/07/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
B7H3 (CD276), an immunoregulatory molecule known for its role in immune evasion by transmitting inhibitory signals to T lymphocytes, has garnered significant attention in recent years as a promising target for cancer immunotherapy. This interest is largely due to its high expression in various types of solid tumors, coupled with low protein levels in normal tissues. However, studies examining the impact of B7H3 on survival outcomes have shown inconsistent results, leaving its prognostic significance not fully clarified. Therefore, this meta-analysis aimed to assess the relationship between B7H3 expression and various prognostic parameters in patients with solid malignancies. PubMed, Web of Science (WOS), Cochrane, SCOPUS, and Embase databases were searched for eligible articles published until November 2024. Statistical analysis was performed using R studio (version 4.3.2). The analysis included a total of 51 eligible studies comprising 11,135 patients. Results showed that overexpression of B7H3 is a negative predictor for all examined survival outcomes: OS (HR = 1.71, 95% CI = 1.44-2.03, p < 0.0001), DFS (HR = 2.02, 95% CI = 1.49-2.73, p < 0.0001), PFS (HR = 2.10, 95% CI = 1.44-3.06, p < 0.0001), RFS (HR = 1.66, 95% CI = 1.11-2.48, p = 0.01), and DSS (HR = 1.70, 95% CI = 1.24-2.32, p < 0.01). Despite the high heterogeneity observed across the studies, the sensitivity analysis confirmed the robustness of these results. This research suggests that B7H3 may serve as an effective biomarker for prognosis in solid tumors.
Collapse
Affiliation(s)
- Sylwia Mielcarska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-800 Zabrze, Poland
| | - Agnieszka Kula
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-514 Katowice, Poland; (A.K.); (M.D.); (D.W.)
| | - Miriam Dawidowicz
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-514 Katowice, Poland; (A.K.); (M.D.); (D.W.)
| | - Dariusz Waniczek
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-514 Katowice, Poland; (A.K.); (M.D.); (D.W.)
| | - Elżbieta Świętochowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-800 Zabrze, Poland
| |
Collapse
|
13
|
Xiong Y, Cheng L, Zhou YJ, Ge WH, Qian M, Yang H. Diagnosis and treatment of lung cancer: A molecular perspective. World J Clin Oncol 2025; 16:100361. [PMID: 40130062 PMCID: PMC11866089 DOI: 10.5306/wjco.v16.i3.100361] [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: 08/15/2024] [Revised: 12/23/2024] [Accepted: 01/09/2025] [Indexed: 01/21/2025] Open
Abstract
This editorial comments on the review by Da Silva et al, published in the World Journal of Clinical Oncology which focuses on the molecular perspectives of lung cancer. With the rapid development of molecular technology, new diagnostic methods are constantly emerging, including liquid biopsy, the identification of gene mutations, and the monitoring biomarkers, thus providing precise information with which to identify the occurrence and development of lung cancer. Biomarkers, such as circulating tumor cells, circulating tumor DNA, and circulating RNA can provide helpful information for clinical application. Common types of genetic mutations and immune checkpoints include epidermal growth factor receptor, anaplastic lymphoma kinase, c-ROS proto-oncogene 1, programmed death-1 and cytotoxic T-lymphocyte-associated protein. According to specific biomarkers, targeted therapy and immunotherapy can improve survival outcomes based on the types of gene mutation and immune checkpoints. The application of molecular approaches can facilitate our ability to control the progression of disease and select appropriate therapeutic strategies for patients with lung cancer.
Collapse
Affiliation(s)
- Yuan Xiong
- Department of Pharmacy, Nanjing Drum Tower Hospital, Nanjing 210008, Jiangsu Province, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu Province, China
| | - Long Cheng
- Department of Pharmacy, Nanjing Drum Tower Hospital, Nanjing 210008, Jiangsu Province, China
- Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing 210008, Jiangsu Province, China
| | - Yu-Jie Zhou
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Nanjing 210008, Jiangsu Province, China
| | - Wei-Hong Ge
- Department of Pharmacy, Nanjing Drum Tower Hospital, Nanjing 210008, Jiangsu Province, China
| | - Ming Qian
- Department of Pharmacy, Nanjing Drum Tower Hospital, Nanjing 210008, Jiangsu Province, China
| | - Hui Yang
- Department of Pharmacy, Nanjing Drum Tower Hospital, Nanjing 210008, Jiangsu Province, China
| |
Collapse
|
14
|
Guo R, Rao PG, Liao BZ, Luo X, Yang WW, Lei XH, Ye JM. Melatonin suppresses PD-L1 expression and exerts antitumor activity in hepatocellular carcinoma. Sci Rep 2025; 15:8451. [PMID: 40069331 PMCID: PMC11897332 DOI: 10.1038/s41598-025-93486-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 03/07/2025] [Indexed: 03/14/2025] Open
Abstract
Melatonin, also known as the pineal hormone, is secreted by the pineal gland and primarily regulates circadian rhythms. Additionally, it possesses immunomodulatory properties and anticancer effects. However, its specific mechanism in hepatocellular carcinoma (HCC) remains unclear, particularly regarding its effect on HCC-mediated immune escape through PD-L1 expression.In this study, in vitro experiments were conducted using Huh7 and HepG2 HCC cells. Melatonin treatment was applied to both cell types to observe changes in malignant phenotypes. Additionally, melatonin-pretreated Huh7 or HepG2 cells were co-cultured with T cells to simulate the tumor microenvironment. The results showed that melatonin inhibited cancer cell proliferation, migration, and invasion, as well as reduced PD-L1 expression in cancer cells, exhibiting similar anti-cancer effects in the co-culture system. In vivo experiments involved establishing ascitic HCC mouse models using H22 cells, followed by subcutaneous tumor models in Balb/c nude and Balb/c wild-type mice. Melatonin inhibited tumor growth and suppressed PD-L1 expression in cancer tissues in both subcutaneous tumor models, and it increased T lymphocyte activity in the spleen of Balb/c wild-type mice. Overall, the in vitro and in vivo experiments demonstrated that melatonin has dual anti-cancer effects in HCC: direct intrinsic anti-cancer activity and enhancement of anti-tumor immunity by reducing PD-L1 expression thereby inhibiting cancer immune escape. Furthermore, a decrease in the expression of the upstream molecule HIF-1α of PD-L1 and an increase in the expression levels of JNK, P38, and their phosphorylated forms were detected. Thus, the mechanism by which melatonin reduces PD-L1 may involve the downregulation of HIF-1α expression or the activation of the MAPK-JNK and MAPK-P38 pathways. This provides new insights and strategies for HCC treatment.
Collapse
Affiliation(s)
- Rui Guo
- Suzhou Medical College of Soochow University, Suzhou, China.
- Department of Anesthesiology, First Affiliated Hospital of Gannan Medical University, No.23 Qingnian Road, Ganzhou City, Jiangxi Province, China.
| | - Pan-Guo Rao
- Gannan Medical University, Ganzhou, Jiangxi, China
| | | | - Xin Luo
- Gannan Medical University, Ganzhou, Jiangxi, China
| | - Wen-Wen Yang
- Gannan Medical University, Ganzhou, Jiangxi, China
| | | | - Jun-Ming Ye
- Suzhou Medical College of Soochow University, Suzhou, China.
- Gannan Medical University, Ganzhou, Jiangxi, China.
| |
Collapse
|
15
|
Han X, Guan J, Guo L, Jiao Q, Wang K, Hou F, Liu S, Yang S, Huang C, Cong W, Wang H. A CT-based interpretable deep learning signature for predicting PD-L1 expression in bladder cancer: a two-center study. Cancer Imaging 2025; 25:27. [PMID: 40065444 PMCID: PMC11892212 DOI: 10.1186/s40644-025-00849-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND To construct and assess a deep learning (DL) signature that employs computed tomography imaging to predict the expression status of programmed cell death ligand 1 in patients with bladder cancer (BCa). METHODS This retrospective study included 190 patients from two hospitals who underwent surgical removal of BCa (training set/external validation set, 127/63). We used convolutional neural network and radiomics machine learning technology to generate prediction models. We then compared the performance of the DL signature with the radiomics machine learning signature and selected the optimal signature to build a nomogram with the clinical model. Finally, the internal forecasting process of the DL signature was explained using Shapley additive explanation technology. RESULTS On the external validation set, the DL signature had an area under the curve of 0.857 (95% confidence interval: 0.745-0.932), and demonstrated superior prediction performance in comparison with the other models. SHAP expression images revealed that the prediction of PD-L1 expression status is mainly influenced by the tumor edge region, particularly the area close to the bladder wall. CONCLUSIONS The DL signature performed well in comparison with other models and proved to be a valuable, dependable, and interpretable tool for predicting programmed cell death ligand 1 expression status in patients with BCa.
Collapse
Affiliation(s)
- Xiaomeng Han
- Department of Radiology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong, 266003, China
| | - Jing Guan
- Department of Radiology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei, 050000, China
| | - Li Guo
- College of Computer Science and Technology, Qingdao University, Qingdao, Shandong, 266071, China
| | - Qiyan Jiao
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Kexin Wang
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Feng Hou
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, China
| | - Shunli Liu
- Department of Radiology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong, 266003, China
| | - Shifeng Yang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250022, China
| | - Chencui Huang
- Department of Research Collaboration, R&d Center, Beijing Deepwise & League of PHD Technology Co., Ltd, Beijing, 100080, China
| | - Wenbin Cong
- Department of Radiology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong, 266003, China.
| | - Hexiang Wang
- Department of Radiology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong, 266003, China.
| |
Collapse
|
16
|
Fan Y, Ji X, Yuan K, Wu Q, Lou M. HDAC1 Mediates Immunosuppression of the Tumor Microenvironment in Non-Small Cell Lung Cancer. J Inflamm Res 2025; 18:3333-3347. [PMID: 40078575 PMCID: PMC11900795 DOI: 10.2147/jir.s509316] [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: 11/29/2024] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Background Studies have demonstrated that histone deacetylase 1 (HDAC1) enables cancer cells to evade killing mediated by cytotoxic T lymphocytes. However, there are no studies on the immunological aspects of HDAC1 in non-small cell lung cancer (NSCLC). Methods In this research, we used the Cancer Genome Atlas (TCGA) public database combined with tissue microarray (TMA) to investigate HDAC1 expression and prognosis in NSCLC. According to the median value of HDAC1 expression in the TCGA dataset, samples of patients with NSCLC were classified into high- and low-expression cohorts. Subsequently, the biological characteristics of HDAC1 in high- and low-expression cohorts in terms of signaling pathways, immune cell infiltration, immune cell function, and genomic stability were investigated to better understand the effect of HDAC1 in the tumor microenvironment (TME) of NSCLC. Additionally, we selected tissue samples with HDAC1 overexpression in TMA2 and performed immunohistochemical staining of CD8+ T cells to observe the distribution of CD8+ T cells in the tumor. Results The findings revealed that overexpression of HDAC1 in NSCLC was associated with poor prognosis. Analysis of signaling pathway enrichment indicated that HDAC1 downregulated immune-related signaling pathways in NSCLC. Immune cell infiltration, immune cell function, and genomic stability analyses suggested that the TME alteration mediated by HDAC1 in the high-expression cohort was consistent with the "immune desert" phenotype. Furthermore, CD8+ T immunohistochemical staining experiments of tissue samples with HDAC1 overexpression in NSCLC revealed few CD8+ T cells distributed in the tumor parenchyma and interstitium. Conclusion Conclusively, our findings from several biological analyses revealed that HDAC1 is overexpressed in NSCLC and induces TME immunosuppression.
Collapse
Affiliation(s)
- Yongfei Fan
- Department of Thoracic Surgery, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
- Heart and Lung Disease Laboratory, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
| | - Xiang Ji
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, People’s Republic of China
| | - Kai Yuan
- Department of Thoracic Surgery, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
- Heart and Lung Disease Laboratory, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
| | - Qiyong Wu
- Department of Thoracic Surgery, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
- Heart and Lung Disease Laboratory, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
| | - Ming Lou
- Department of Thoracic Surgery, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
- Heart and Lung Disease Laboratory, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
| |
Collapse
|
17
|
Wu Y, Yan Y, Guo Y, Niu M, Zhou B, Zhang J, Zhou P, Chu Q, Mei Q, Yi M, Wu K. Anti-TGF-β/PD-L1 bispecific antibody synergizes with radiotherapy to enhance antitumor immunity and mitigate radiation-induced pulmonary fibrosis. J Hematol Oncol 2025; 18:24. [PMID: 40038780 DOI: 10.1186/s13045-025-01678-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Despite the success of immune checkpoint inhibitors (ICIs) in multiple malignant tumors, a significant proportion of patients remain unresponsive to treatment. Radiotherapy (RT) elicits immunogenic antitumor responses but concurrently activates several immune evasion mechanisms. Our earlier research demonstrated the efficacy of YM101, an anti-TGF-β/PD-L1 bispecific antibody, in stroma-rich tumors. Nevertheless, YM101 has demonstrated reduced effectiveness in non-inflamed tumors characterized by poor immune cell infiltration. This study investigated the potential synergy between RT and YM101 in overcoming immunotherapy resistance and mitigating RT-induced pulmonary fibrosis. METHODS The antitumor activity and survival outcomes of RT plus YM101 treatment in vivo were explored in several non-inflamed murine tumor models. Furthermore, the inhibition of pulmonary metastases was assessed in a pulmonary metastasis model. The impact of RT on dendritic cell (DC) maturation was quantified by flow cytometry, whereas cytokine and chemokine secretions were measured by ELISA. To comprehensively characterize changes in the tumor microenvironment, we utilized a combination of methods, including flow cytometry, IHC staining, multiplex inmunofluorecence and RNA sequencing. Additionally, we evaluated the impact of YM101 on RT-induced pulmonary fibrosis. RESULTS RT plus YM101 significantly inhibited tumor growth, prolonged survival and inhibited pulmonary metastases compared with monotherapies in non-inflamed tumors with poor immune infiltration. RT promoted DC maturation in a dose-dependent manner and increased the secretions of multiple proinflammatory cytokines. Mechanistically, RT plus YM101 simultaneously increased the infiltration and activation of intratumoral DCs and tumor-infiltrating lymphocytes and reshaped the tumor microenvironment landscape. Notably, YM101 attenuated both RT-induced peritumoral fibrosis and pulmonary fibrosis. CONCLUSIONS Our findings suggest that RT combined with YM101 enhances antitumor immunity and overcomes resistance in non-inflamed tumors in preclinical models, while simultaneously showing potential in mitigating RT-induced fibrosis. This combination therapy demonstrates promise in overcoming ICI resistance, while potentially sparing normal pulmonary tissue, thereby providing a strong rationale for further clinical investigations.
Collapse
Affiliation(s)
- Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Yuheng Yan
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Yarong Guo
- Cancer Center, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Binghan Zhou
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Jing Zhang
- Wuhan YZY Biopharma Co., Ltd., Biolake, C2-1, No.666 Gaoxin Road, Wuhan, 430075, China
| | - Pengfei Zhou
- Wuhan YZY Biopharma Co., Ltd., Biolake, C2-1, No.666 Gaoxin Road, Wuhan, 430075, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Qi Mei
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.
- Cancer Center, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
| | - Ming Yi
- Department of Breast Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, 310000, China.
| | - Kongming Wu
- Cancer Center, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
- Cancer Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430032, China.
| |
Collapse
|
18
|
Xu Y, Zhang J, Pan D, Yan J, Chen C, Wang L, Wang X, Yang M, Xu Y. Development of Novel Peptide-Based Radiotracers for Detecting FGL1 Expression in Tumors. Mol Pharm 2025; 22:1605-1614. [PMID: 39893698 DOI: 10.1021/acs.molpharmaceut.4c01293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
A novel immune checkpoint, FGL1, is a potentially viable target for tumor immunotherapy. The development of FGL1-targeted PET probes could provide significant insights into the immune system's status and the evaluation of treatment efficacy. A ClusPro 2.0 server was used to analyze the interaction between FGL1 and LAG3, and the candidate peptides were identified by using the Rosetta peptide derivate protocol. Three candidate peptides targeting FGL1, named FGLP21, FGLP22, and FGLP23, with a simulated affinity of -9.56, -8.55, and -8.71 kcal/mol, respectively, were identified. The peptides were readily conjugated with p-NCS-benzyl-NODA-GA, and the resulting compounds were successfully labeled with 68Ga in approximately 70% yields and radiochemical purity greater than 95%. In vitro competitive cell-binding assay demonstrated that all probes bound to FGL1 with IC50 ranging from 100 nM to 160 nM. Among the probes, PET imaging revealed that 68Ga-NODA-FGLP21 exhibited the best tumor imaging performance in mice bearing FGL1 positive Huh7 tumor. At 60 min p.i., the tumor uptake of 68Ga-NODA-FGLP21 was significantly higher than those of 68Ga-NODA-FGLP22 and 68Ga-NODA-FGLP23, respectively (2.51 ± 0.11% ID/g vs 1.00 ± 0.16% ID/g and 1.49 ± 0.05% ID/g). Simultaneously, the tumor-to-muscle uptake ratios of the former were also higher than those of the latter, respectively (19.40 ± 2.30 vs 9.65 ± 0.62 and 12.45 ± 0.72). In the presence of unlabeled FGLP21, the uptake of 68Ga-NODA-FGLP21 in Huh7 xenograft decreased to 0.81 ± 0.09% ID/g at 60 min p.i., which is similar to that observed in the FGL1 negative U87 MG tumor (0.46 ± 0.03% ID/g). The results were consistent with the immunohistochemical analysis and ex vivo autoradiography. No significant radioactivity was accumulated in normal organs, except for kidneys. In summary, a preclinical study confirmed that the tracer 68Ga-NODA-FGLP21 has the potential to specifically detect FGL1 expression in tumors with good contrast to the background.
Collapse
Affiliation(s)
- Yue Xu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jinyuan Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Donghui Pan
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Junjie Yan
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Chongyang Chen
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Lizhen Wang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Xinyu Wang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Min Yang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Yuping Xu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| |
Collapse
|
19
|
Andour L, Hagenaars SC, Gregus B, Tőkes AM, Karancsi Z, Tollenaar RAEM, Kroep JR, Kulka J, Mesker WE. The prognostic value of the tumor-stroma ratio compared to tumor-infiltrating lymphocytes in triple-negative breast cancer: a review. Virchows Arch 2025; 486:427-444. [PMID: 39904885 PMCID: PMC11950021 DOI: 10.1007/s00428-025-04039-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/17/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
Previous literature extensively explored biomarkers to personalize treatment for breast cancer patients. The clinical need is especially high in patients with triple-negative breast cancer (TNBC) due to its aggressive nature and limited treatment modalities. This review aims to evaluate the value of tumor-infiltrating lymphocytes (TILs) and tumor-stroma ratio (TSR) as prognostic biomarkers in TNBC patients and assess their clinical potential. A literature search was conducted in PubMed, Embase, Emcare, Web of Science, and Cochrane Library. Papers comparing survival outcomes of TNBC patients with low/high or negative/positive TSR and immune cells were included. The most frequently mentioned subgroups of TILs were selected and reported in this review. Data from 43 articles on TILs and eight articles on TSR were included. Among TNBC patients, high CD8 expression was generally associated with better survival. Notable, the poor survival outcomes were related to high intra-tumoral PD-L1 expression, whereas high stromal PD-L1 expression more often was correlated with favorable outcomes. For the TSR, a high amount of stroma in the primary tumor of TNBC patients was consistently associated with worse survival. This review highlights that a high number of CD8-positive T-cells is a promising prognostic factor for TNBC patients. PD-L1 expression analyzed for intra-tumoral and stromal expression separately reports strong but contrasting information. Finally, the TSR shows potential to be an important prognostic marker, especially for TNBC patients. Utilizing both biomarkers, either on itself or combined, could enhance clinical decision-making and personalization of treatment.
Collapse
Affiliation(s)
- Layla Andour
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Sophie C Hagenaars
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Barbara Gregus
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Anna Mária Tőkes
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Zsófia Karancsi
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Rob A E M Tollenaar
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Janina Kulka
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Wilma E Mesker
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
20
|
Bae SU, Lee HW, Park JY, Seo I, Cho JM, Kim JY, Lee JY, Lee YJ, Baek SK, Kim NK, Byun SJ, Kim S. Neoadjuvant chemoradiotherapy up-regulates PD-L1 in radioresistant colorectal cancer. Clin Transl Radiat Oncol 2025; 51:100906. [PMID: 39811542 PMCID: PMC11732604 DOI: 10.1016/j.ctro.2024.100906] [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: 07/18/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
Background Combining radiotherapy (RT) with immune checkpoint inhibitors (ICIs) is a promising strategy that can enhance the therapeutic efficacy of ICIs. However, little is known about RT-induced changes in the expression of immune checkpoints, such as PD-L1, and their clinical implications in colorectal cancer (CRC). This study aimed to investigate the association between responsiveness to RT and changes in PD-L1 expression in human CRC tissue and cell lines. Methods Tissue specimens from preoperative biopsy via sigmoidoscopy and surgical resection were obtained from 24 patients with locally advanced rectal cancer (LARC) who underwent neoadjuvant chemoradiation therapy (CRT) between August 2016 and December 2017. Immunohistochemistry for PD-L1 in formalin-fixed paraffin-embedded tissue was performed from the endoscopic biopsy and surgical specimens. RNA sequencing was performed using 11 pairs of human LARC tissues before and after irradiation. After exposing human CRC cells to radiation, we investigated changes in the expression levels of PD-L1 and its regulatory signaling pathways. Results Patients were classified by tumor regression grade into responders (grade 2; 9 patients, 37.5 %) and non-responders (grades 3, 4, or 5; 15 patients, 62.5 %). In the non-responder group, 13 patients had low PD-L1 expression, but neoadjuvant CRT increased PD-L1 expression in 7 patients (53.9 %) (McNemar's test, p=0.034). CRT up-regulated PD-L1 in non-responder LARC tissues. Similarly, radiation increased PD-L1 in radioresistant DLD-1 cells more than in radiosensitive HCT116 cells, also affecting PD-L1-regulating genes and immune checkpoints in CRC cells. Conventional fractionated radiation treatment further increased PD-L1 in DLD-1 cells compared to HCT116 cells. Conclusions This study demonstrated that radiation induces an increase in PD-L1 expression, which is more pronounced in radioresistant CRC, proving the theoretical framework for a combined treatment strategy with a PD-L1 blockade for locally advanced rectal cancer.
Collapse
Affiliation(s)
- Sung Uk Bae
- Department of Surgery, School of Medicine, Keimyung University and Dongsan Hospital, Daegu, Republic of Korea
- Department of Medicine, The Graduate School, Yonsei University, Seoul, Republic of Korea
- Institute of Medical Science & Institute for Cancer Research, Keimyung University, Daegu, Republic of Korea
| | - Hye Won Lee
- Institute of Medical Science & Institute for Cancer Research, Keimyung University, Daegu, Republic of Korea
- Department of Pathology, School of Medicine, Keimyung University and Dongsan Hospital, Daegu, Republic of Korea
| | - Jee Young Park
- Department of Surgery, School of Medicine, Keimyung University and Dongsan Hospital, Daegu, Republic of Korea
- Department of Pathology, School of Medicine, Keimyung University and Dongsan Hospital, Daegu, Republic of Korea
- Department of Radiation Oncology, School of Medicine, Keimyung University and Dongsan Hospital, Daegu, Republic of Korea
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Incheol Seo
- Department of Immunology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Jae-Min Cho
- Department of Surgery, School of Medicine, Keimyung University and Dongsan Hospital, Daegu, Republic of Korea
- Department of Pathology, School of Medicine, Keimyung University and Dongsan Hospital, Daegu, Republic of Korea
| | - Jin Young Kim
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, Keimyung University and Dongsan Hospital, Daegu, Republic of Korea
| | - Ju Yup Lee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, Keimyung University and Dongsan Hospital, Daegu, Republic of Korea
| | - Yoo Jin Lee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, Keimyung University and Dongsan Hospital, Daegu, Republic of Korea
| | - Seong Kyu Baek
- Department of Surgery, School of Medicine, Keimyung University and Dongsan Hospital, Daegu, Republic of Korea
| | - Nam Kyu Kim
- Division of Colorectal Surgery, Department of Surgery, Severance Hospital, Colorectal Cancer Clinic, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang Jun Byun
- Institute of Medical Science & Institute for Cancer Research, Keimyung University, Daegu, Republic of Korea
- Department of Radiation Oncology, School of Medicine, Keimyung University and Dongsan Hospital, Daegu, Republic of Korea
| | - Shin Kim
- Institute of Medical Science & Institute for Cancer Research, Keimyung University, Daegu, Republic of Korea
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| |
Collapse
|
21
|
Jing D, Zhang J, Li Z, Yan W, Guo Y. Nanomotors activating both cGAS-STING pathway and immune checkpoint blockade for tumor therapy and bioimaging. Talanta 2025; 284:127258. [PMID: 39586211 DOI: 10.1016/j.talanta.2024.127258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 11/27/2024]
Abstract
Cellular innate immune response is closely related to cGAS-STING pathway and PD-1/PD-L1 immune checkpoint blockade. The lack of tissue penetration of STING agonists and nanomedicines in conventional approaches reduces their immunotherapeutic efficacy. At the same time, because the cGAS-STING signaling pathway is silent in many breast cancer cells, it cannot play its role. To address these challenges, here, we developed a silica nanomotor based on bubble propulsion. Its hollow structure was packed with the photosensitizer Ce6 molecule. Under 808 nm laser irradiation, Ce6 produced 1O2, which lead to intracellular DNA damage and further activated the cGAS-STING pathway, stimulating the maturation of DC cells, and enhancing the tumor infiltration of CD8+ T cells. The nanomotor had an asymmetrical structure. One side of the nanomotor was modified with Pt nanoparticle. This asymmetric modification can catalyze H2O2 in the environment, producing an asymmetric concentration of O2, which realized the bubble driving nanomotor movement and enhances penetration into breast cancer cells of nanomotor. The other side of the nanomotor was modified the LXL-1 aptamer, triphenylphosphine and peptide CLP002. Peptide CLP002 specifically bound residues of PD-L1 interaction with PD-1, blocked the mutual binding between PD-1 and PD-L1, and further improved the immune response ability of tumor infiltrating T cells. In this study, we developed a multi-pronged immunotherapy strategy of intelligent target finding, breaking through the physiological barrier through kinetic energy, accurately intervening the target and bioimaging, providing a new idea for breast cancer cells targeted therapy.
Collapse
Affiliation(s)
- Dan Jing
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Ji Zhang
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Ziyi Li
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Wennan Yan
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Yingshu Guo
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China.
| |
Collapse
|
22
|
Li X, Wang Y, Wang Y, Xie H, Gong R, Wu X, Chen J, Sun C, Gu Y. Anti-tumor activity of an αPD-L1-PE38 immunotoxin delivered by engineered Nissle 1917. Int J Biol Macromol 2025; 295:139537. [PMID: 39788238 DOI: 10.1016/j.ijbiomac.2025.139537] [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: 09/20/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
Although immune checkpoint inhibitors specifically targeting the PD-1/PD-L1 axis have exhibited remarkable clinical success, they are not uniformly effective across all patient cohorts. Immunotoxins, a novel class of cancer therapeutics, offering a promising alternative. PD-L1, which is also present in certain normal tissues, limits its suitability as an ideal target for immunotoxins. The probiotic strain of E. coli Nissle 1917 (EcN) could target and colonize to solid tumors, which positions it as a promising candidate for tumor tissue-specific delivery of anti-tumor proteins. In this study, we constructed a PD-L1-targeted immunotoxin, designated as αPD-L1-PE38, by fusing an anti-PD-L1 nanobody and a clinically validated PE38 toxin. This immunotoxin exhibited potent cytotoxic activity against tumor cells while showed slightly cytotoxic activity against normal cells. To effectively deliver the αPD-L1-PE38 to tumor tissues, we engineered the EcN strain to release the immunotoxin induced by L-arabinose. Upon induction, the immunotoxin was efficiently secreted, and exhibited robust anti-tumor activity mainly by inducing cell apoptosis both in vitro and in vivo. Furthermore, we enhanced the immunotoxin's affinity for PD-L1 by optimizing the linker between the nanobody and PE38 toxin. The engineered EcN expressing the optimized immunotoxin, achieved superior anti-tumor activity. Collectively, our study suggests that the delivery of immunotoxins through live bacteria to improve safety and efficacy is a promising option in cancer therapeutics.
Collapse
Affiliation(s)
- Xinrui Li
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yangui Wang
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yanqing Wang
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Huilin Xie
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Ruxin Gong
- School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xiao Wu
- Department of Respiratory and Critical Care Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao 266042, China
| | - Jin Chen
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Changning Sun
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Yuchao Gu
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| |
Collapse
|
23
|
Jin S, Li T, Liu L, Gao T, Zhang T, Yuan D, Di J, Guo Z, Luo Z, Yuan H, Liu J. V-domain immunoglobulin suppressor of T-cell activation and programmed death receptor 1 dual checkpoint blockade enhances antitumour immunity and survival in glioblastoma. Br J Pharmacol 2025; 182:1306-1323. [PMID: 39626657 DOI: 10.1111/bph.17404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/30/2024] [Accepted: 10/19/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE The current therapy cannot meet the needs of glioblastoma (GBM). V-domain immunoglobulin suppressor of T-cell activation (VISTA) is significantly up-regulated in GBM patients; however, its therapeutic potential in GBM is still unclear. EXPERIMENTAL APPROACH Flow cytometry was used to detect the expression of VISTA and the co-expression pattern of VISTA and programmed death receptor 1 (PD-1) on brain infiltrating lymphocytes of GBM mice. Monoclonal antibody therapy was used to evaluate the therapeutic effect of α-VISTA monotherapy and α-VISTA combined with α-PD-1 on GBM mice. Transcriptome analysis, flow cytometry, and immunofluorescence were used to detect changes of immune microenvironment in mouse brain tumours. Immunofluorescence and TCGA data analysis were used to further validate the combined treatment strategy on patient data. KEY RESULTS Compared with normal mice, the frequency of VISTA expression and co-expression of VISTA and PD-1 on tumour-infiltrating lymphocytes (TILs) in tumour-bearing mice was increased. Anti-VISTA monotherapy significantly up-regulated multiple immune stimulation-related pathways and moderately prolonged mouse survival time. Blocking the immune checkpoint VISTA and PD-1 significantly prolonged the survival time of mice and cured about 80% of the mice; CD8+ T cells played an important role in this process. In addition, we found that the expression of VISTA and PD-1 was significantly up-regulated in GBM patients by immunofluorescence, and patients with high expression of VISTA and PD-1 were associated with poor overall survival. This combination of blocking the immune checkpoint VISTA and PD-1 may achieve clinical transformation in GBM.
Collapse
Affiliation(s)
- Shasha Jin
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Tao Li
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Liu Liu
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ting Gao
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Tingting Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Dingyi Yuan
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jianwen Di
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhanying Guo
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhijie Luo
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Haoliang Yuan
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jun Liu
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
24
|
Liang BG, Zheng YM, Shen HY, Yang GH, Xu WX, Tan CJ, Ke AW, Qin WZ. Cordycepin mediates pyroptosis in HCC through the upregulation of TXNIP and synergizes with anti-PD-L1 immunotherapy. Hepatol Commun 2025; 9:e0633. [PMID: 40008893 PMCID: PMC11868431 DOI: 10.1097/hc9.0000000000000633] [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: 07/16/2024] [Accepted: 11/14/2024] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors are effective treatments for HCC; however, their therapeutic efficacy is often limited by the development of drug resistance. Therefore, investigating new combination therapeutics involving immune checkpoint inhibitors is critical to improving patient prognosis. In this study, we investigated the therapeutic effect of cordycepin (COR) in HCC and its synergistic effect with anti-programmed cell death ligand 1 (anti-PD-L1) immunotherapy. METHODS We selected 2 HCC cell lines to investigate the effects of COR on HCC growth using in vivo and in vitro experiments. We performed RNA sequencing of the MHCC97H cell line treated with or without COR to understand the underlying mechanism and identify the key regulatory genes. Through in vivo and in vitro experiments on gene knockdown cells, we identified thioredoxin-interacting protein as a key molecule involved in the role of COR. Next, we used mouse subcutaneous and orthotopic tumor models to evaluate the therapeutic effects of COR, atezolizumab (a programmed death-ligand 1 [PD-L1] inhibitor), or their combination. Multiple immunofluorescence staining revealed that the combination of atezolizumab and COR therapy greatly increased the number of tumor-infiltrating CD8+ T cells and PD-L1 expression in HCC compared to monotherapy. RESULTS Our study revealed that COR significantly inhibited HCC growth both in vitro and in vivo. Mechanistically, we showed that COR induces endoplasmic reticulum stress, which upregulates thioredoxin-interacting protein expression and leads to HCC cell pyroptosis. In addition, the combination treatment with COR and PD-L1 inhibitors profoundly inhibited HCC. CONCLUSIONS Overall, our study successfully established a combined therapeutic strategy using COR and PD-L1 inhibitors. This strategy has significant synergistic effects on cancer cells, highlighting its importance in cancer therapy.
Collapse
Affiliation(s)
- Bu-Gang Liang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
- Department of Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, PR China
| | - Yi-Min Zheng
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
- Department of Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, PR China
| | - Hong-Ye Shen
- Department of Endoscopy Center, Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai Collaborative Innovation Center of Endoscopy, Shanghai, China
| | - Guo-Huan Yang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
- Department of Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, PR China
| | - Wen-Xin Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
- Department of Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, PR China
| | - Chang-Jun Tan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
- Department of Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, PR China
| | - Ai-Wu Ke
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
- Department of Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, PR China
| | - Wen-Zheng Qin
- Department of Endoscopy Center, Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai Collaborative Innovation Center of Endoscopy, Shanghai, China
| |
Collapse
|
25
|
Zhao X, Wang X, Liu S, Cheng P, Chen J, Liu J. Severe thyroiditis induced by sintilimab monotherapy in a patient with non-small cell lung cancer: a case report and literature review. Front Immunol 2025; 16:1548452. [PMID: 40070833 PMCID: PMC11893825 DOI: 10.3389/fimmu.2025.1548452] [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: 12/19/2024] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Thyroid dysfunction is a common immune-related adverse event (irAE) associated with immune checkpoint inhibitors (ICIs) that target PD-1, PD-L1, and CTLA-4. Nevertheless, the incidence of severe cases, defined as grade 3 or higher, remains rare. This report presents a detailed case study of severe thyroiditis in a patient with non-small cell lung cancer (NSCLC) who developed grade 3 thyroiditis following a single cycle of sintilimab monotherapy. The clinical presentation in this patient was remarkable for its early onset, occurring one week after the initiation of sintilimab therapy, and for its severe manifestations. During hospitalization, a prompt and accurate differential diagnosis was performed. Sintilimab treatment was discontinued, and the patient was promptly started on high-dose glucocorticoids, with a tapering schedule implemented as the condition improved or reached Common Terminology Criteria for Adverse Events (CTCAE) grade 1 or lower. The patient subsequently developed overt hypothyroidism, necessitating the initiation of thyroxine replacement therapy. Furthermore, we provide a comprehensive review of the mechanisms and risk factors associated with thyroid dysfunction immune-related adverse events (TD-irAEs). It is imperative for clinicians to meticulously monitor the clinical symptoms exhibited by patients. For those presenting with symptoms, prompt diagnosis and appropriate symptomatic management are essential. Additionally, regular thyroid function testing is recommended for high-risk patients, and we advocate for the assessment of baseline levels of thyroid peroxidase antibodies (TPOAb) and thyroglobulin antibodies (TGAb) prior to initiating ICI treatment.
Collapse
Affiliation(s)
- Xiaolin Zhao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Xiaoyu Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Surui Liu
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Pian Cheng
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Jinjuan Chen
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Jie Liu
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
26
|
Yang M, Wang X, Peng M, Wang F, Hou S, Xing R, Chen A. Nanomaterials Enhanced Sonodynamic Therapy for Multiple Tumor Treatment. NANO-MICRO LETTERS 2025; 17:157. [PMID: 39992547 PMCID: PMC11850698 DOI: 10.1007/s40820-025-01666-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/08/2025] [Indexed: 02/25/2025]
Abstract
Sonodynamic therapy (SDT) as an emerging modality for malignant tumors mainly involves in sonosensitizers and low-intensity ultrasound (US), which can safely penetrate the tissue without significant attenuation. SDT not only has the advantages including high precision, non-invasiveness, and minimal side effects, but also overcomes the limitation of low penetration of light to deep tumors. The cytotoxic reactive oxygen species can be produced by the utilization of sonosensitizers combined with US and kill tumor cells. However, the underlying mechanism of SDT has not been elucidated, and its unsatisfactory efficiency retards its further clinical application. Herein, we shed light on the main mechanisms of SDT and the types of sonosensitizers, including organic sonosensitizers and inorganic sonosensitizers. Due to the development of nanotechnology, many novel nanoplatforms are utilized in this arisen field to solve the barriers of sonosensitizers and enable continuous innovation. This review also highlights the potential advantages of nanosonosensitizers and focus on the enhanced efficiency of SDT based on nanosonosensitizers with monotherapy or synergistic therapy for deep tumors that are difficult to reach by traditional treatment, especially orthotopic cancers.
Collapse
Affiliation(s)
- Mengyao Yang
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Xin Wang
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Mengke Peng
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Fei Wang
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Senlin Hou
- The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People's Republic of China.
| | - Ruirui Xing
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, People's Republic of China.
| | - Aibing Chen
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China.
| |
Collapse
|
27
|
Li J, Jiang L, Ma Q, Zhang Z, Zheng S, Qiu J, Pang Y, Wang J. Evodiamine inhibits programmed cell death ligand 1 expression via the PI3K/AKT signaling pathway to regulate antitumor immunity in melanoma. Sci Rep 2025; 15:6649. [PMID: 39994441 PMCID: PMC11850830 DOI: 10.1038/s41598-025-91137-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
Malignant melanoma, a rare and aggressive skin cancer, arises from the transformation of cutaneous melanocytes and is associated with a poor prognosis. Evodiamine (EVO), a bioactive compound derived from traditional Chinese herbal medicine, has demonstrated significant inhibitory effects on various tumor cells. In this study, we aimed to investigate the potential of EVO in regulating melanoma immunity and elucidate its underlying mechanisms. Experimental results revealed that the IC50 value of EVO in B16-F10 cells for 24, 48, and 72 h were 11.73, 5.083, and 4.604 µM, respectively. EVO inhibited the proliferation, invasion, and metastasis of B16-F10 cells by more than 50%, while promoting apoptosis of higher concentration of EVO. EVO also significantly suppressed tumor growth by more than 80% and reduced spleen damage in tumor-bearing mice. Treatment with EVO led to a marked increase in T-cell subsets in the spleen, bone marrow, and tumors, with immunohistochemical (IHC) staining in particular showing about 50% higher. Furthermore, EVO inhibited the expression of programmed cell death ligand 1 (PD-L1) and the PI3K/AKT signaling pathway-related proteins in both B16-F10 cells and tumors. These findings suggest that EVO exerts antitumor effects by enhancing the tumor immune microenvironment and indicates its potential as a therapeutic agent for melanoma.
Collapse
Affiliation(s)
- Jiamin Li
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Li Jiang
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Qianlong Ma
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Zhenglong Zhang
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Shengping Zheng
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Jing Qiu
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China
| | - Yunqing Pang
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China.
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China.
| | - Jing Wang
- Department of Periodontology, School/Hospital of Stomatology, Lanzhou University, 199 Donggang Western Rd, Lanzhou, 730000, Gansu, People's Republic of China.
- Clinical Research Center for Oral Diseases, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
28
|
Zhao SQ, Chen MJ, Chen F, Gao ZF, Li XP, Hu LY, Cheng HY, Xuan JY, Fei JG, Song ZW. ENTPD8 overexpression enhances anti-PD-L1 therapy in hepatocellular carcinoma via miR-214-5p inhibition. iScience 2025; 28:111819. [PMID: 39995876 PMCID: PMC11849663 DOI: 10.1016/j.isci.2025.111819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/20/2024] [Accepted: 01/13/2025] [Indexed: 02/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths globally, with poor prognosis due to late diagnosis and limited treatment options. In this study, we evaluated the expression of ectonucleoside triphosphate diphosphohydrolase 8 (ENTPD8) in HCC tissues and its clinical significance. Immunohistochemistry, The Cancer Genome Atlas (TCGA) data, and single-cell expression analysis revealed reduced ENTPD8 levels in liver cancer compared to adjacent tissues, with ENTPD8 primarily expressed in tumor cells within the tumor tissue. In vitro assays demonstrated that ENTPD8 inhibits HCC cell proliferation, invasion, and migration. Mechanistically, ENTPD8 regulates programmed death-ligand 1 (PD-L1) expression through miR-214-5p modulation. In vivo, ENTPD8 overexpression combined with anti-PD-L1 treatment enhanced therapeutic efficacy in HCC mouse models. These findings suggest that ENTPD8 may serve as a prognostic marker and therapeutic target for HCC, offering potential strategies for improving treatment outcomes.
Collapse
Affiliation(s)
- Si-qi Zhao
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Min-jie Chen
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Fei Chen
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhao-feng Gao
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Xiao-ping Li
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Ling-yu Hu
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Hai-ying Cheng
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jin-yan Xuan
- Department of General Practice, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jian-guo Fei
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Zheng-wei Song
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
29
|
Delbari P, Ahmadvand MH, Mirjani MS, Hajikarimloo B, Hamidi Rad R, Kargar-Soleimanabad S, Edalat M, Bahri A, Shahir Eftekhar M, Mohammadzadeh I, Habibi MA. The safety and efficacy of anti-PD-1/PD-L1 monoclonal antibodies for lung cancer brain metastasis: a systematic review and meta-analysis on brain metastasis. Neurosurg Rev 2025; 48:253. [PMID: 39969599 DOI: 10.1007/s10143-025-03418-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
Individuals with non-small cell lung cancer (NSCLC) who develop brain metastases face a poor prognosis and limited treatment options. Programmed cell death protein 1 (PD-1) inhibitors, such as pembrolizumab and nivolumab, have emerged as a promising immunotherapy for treating lung cancer with brain metastases. This systematic review and meta-analysis according to Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guideline and evaluate the safety and efficacy of anti-PD-1/PD-L1 monoclonal antibodies in treating lung cancer patients with brain metastases. A comprehensive literature search was conducted to identify relevant studies up to 27 August 2023. Data on overall survival (OS), progression-free survival, radiological response rates, and adverse events were extracted. All statistical analysis was performed using STATA v.17. Our literature search yielded 39 eligible studies involving 15,428 patients. The overall response rate for PD-1 inhibitors was substantial, with pooled rates of 39% for overall response, 7% for complete response (CR), 27% for partial response (PR), and 31% for stable disease (SD). The pooled 6-month OS rate was 77%, and the 1-year OS rate was 61%. Subgroup analyses revealed higher PD-L1 expression levels and the use of platinum-based chemotherapy alongside immunotherapy were associated with improved outcomes. PD-1/PD-L1 inhibitors have demonstrated promising efficacy and safety in treating lung cancer patients with brain metastases, as evidenced by significant improvements in OS, PFS, and response rates. Incorporating PD-1/PD-L1 inhibitors into the treatment regimen, particularly for patients with high PD-L1 expression, has the potential to improve clinical outcomes in this patient population.
Collapse
Affiliation(s)
- Pouria Delbari
- Faculty of Medicine, Tehran University of Medical Science, Tehran, Iran
| | | | | | - Bardia Hajikarimloo
- Department of Neurological Surgery, University of Virginia, Charlottesville, USA
| | - Romina Hamidi Rad
- Department of medicine, Tehran Medical Branch, Islamic Azad University, Tehran, Iran
| | | | - Mehrshad Edalat
- Department of medicine, Tehran Medical Branch, Islamic Azad University, Tehran, Iran
| | - Amirmohammad Bahri
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Shahir Eftekhar
- Department of Surgery, School of Medicine, Shahid Beheshti Hospital, Qom University of Medical Sciences, Qom, Iran
| | - Ibrahim Mohammadzadeh
- Skull Base Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Habibi
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
30
|
Zhu Y, Zhang X, Jin J, Wang X, Liu Y, Gao J, Hang D, Fang L, Zhang H, Liu H. Engineered oncolytic virus coated with anti-PD-1 and alendronate for ameliorating intratumoral T cell hypofunction. Exp Hematol Oncol 2025; 14:16. [PMID: 39955603 PMCID: PMC11829442 DOI: 10.1186/s40164-025-00611-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/07/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Glioblastoma is a highly aggressive and devastating primary brain tumor that is resistant to conventional therapies. Oncolytic viruses represent a promising therapeutic approach for glioblastoma by selectively lysing tumor cells and eliciting an anti-tumor immune response. However, the clinical efficacy of oncolytic viruses is often hindered by challenges such as short persistence, host antiviral immune responses, and T cell dysfunction. METHODS We have developed a novel therapeutic strategy by "dressing" oncolytic viruses with anti-PD-1 antibodies and alendronate (PD-1/Al@OV) to prevent premature clearance of the oncolytic viruses and enhance T cell function, thereby improving immunotherapy outcomes against glioma. RESULTS We found that in the high reactive oxygen species environment of the tumor, PD-1/Al@OV disassembled to release oncolytic viruses, anti-PD-1, and alendronate. The released anti-PD-1 blocked the PD-1/PD-L1 pathway, activating T cells; the alendronate eliminated tumor-associated macrophages, increasing the concentration of oncolytic viruses; and the oncolytic viruses directly lysed cancer cells, enhancing intratumoral T cell infiltration. CONCLUSION This approach effectively improved the immunosuppressive microenvironment of glioblastoma and achieved a robust anti-tumor effect. Consequently, this study presents a novel strategy for immune combination therapy and the improvement of the glioblastoma immune microenvironment, thereby offering new prospects for the clinical application of oncolytic viruses.
Collapse
Affiliation(s)
- Yufu Zhu
- Institute of Nervous System Diseases, Xuzhou Medical University, No.84 Huaihai West Road, Xuzhou, 221002, China.
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, No.99 Huaihai West Road, Xuzhou, 221002, China.
| | - Xuefeng Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, No.84 Huaihai West Road, Xuzhou, 221002, China
| | - Jiaqi Jin
- Institute of Nervous System Diseases, Xuzhou Medical University, No.84 Huaihai West Road, Xuzhou, 221002, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, No.99 Huaihai West Road, Xuzhou, 221002, China
| | - Xiaoqian Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, No.84 Huaihai West Road, Xuzhou, 221002, China
| | - Yang Liu
- Department of Neurosurgery, The First Hospital of China Medical University, No. 155, Nanjing Bei Street, Shenyang, 110001, China
| | - Jian Gao
- Institute of Nervous System Diseases, Xuzhou Medical University, No.84 Huaihai West Road, Xuzhou, 221002, China
| | - Diancheng Hang
- Institute of Nervous System Diseases, Xuzhou Medical University, No.84 Huaihai West Road, Xuzhou, 221002, China
| | - Lin Fang
- Cancer Institute, Xuzhou Medical University, No. 209, Tongshan Road, Xuzhou, 221004, China.
| | - Hengzhu Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, No.84 Huaihai West Road, Xuzhou, 221002, China.
- Department of Neurosurgery, The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou University, No. 98, Nantong West Road, Yangzhou, 225009, China.
| | - Hongmei Liu
- Institute of Nervous System Diseases, Xuzhou Medical University, No.84 Huaihai West Road, Xuzhou, 221002, China.
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088, Xueyuan Avenue, Shenzhen, 518055, China.
| |
Collapse
|
31
|
Wu Y, Liu Y, Wu H, Tong M, Du L, Ren S, Che Y. Advances in Ultrasound-Targeted Microbubble Destruction (UTMD) for Breast Cancer Therapy. Int J Nanomedicine 2025; 20:1425-1442. [PMID: 39925678 PMCID: PMC11804227 DOI: 10.2147/ijn.s504363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 01/08/2025] [Indexed: 02/11/2025] Open
Abstract
Breast cancer is one of the most common types of cancer in women worldwide and is a leading cause of cancer deaths among women. As a result, various treatments have been developed to combat this disease. Breast cancer treatment varies based on its stage and type of pathology. Among the therapeutic options, ultrasound has been employed to assist in the treatment of breast cancer, including radiation therapy, chemotherapy, targeted immunotherapy, hormonal therapy, and, more recently, radiofrequency ablation for early-stage and inoperable patients. One notable advancement is ultrasound-targeted microbubble destruction (UTMD), which is gradually becoming a highly effective and non-invasive anti-tumor modality. This technique can enhance chemical, genetic, immune, and anti-vascular therapies through its physical and biological effects. Specifically, UTMD improves drug transfer efficiency and destroys tumor neovascularization while reducing toxic side effects on the body during tumor treatment. Given these developments, the application of ultrasound-assisted therapy to breast cancer has gained significant attention from research scholars. In this review, we will discuss the development of various therapeutic modalities for breast cancer and, importantly, highlight the application of ultrasound microbubble-targeted disruption techniques in breast cancer treatment.
Collapse
Affiliation(s)
- Yunfeng Wu
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Yuxi Liu
- Department of Ultrasound, Shandong Second Medical University Affiliated Hospital, Shan Dong, Weifang, People’s Republic of China
| | - Han Wu
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Mengying Tong
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Linyao Du
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Shuangsong Ren
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Ying Che
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| |
Collapse
|
32
|
Yu B, Shao S, Ma W. Frontiers in pancreatic cancer on biomarkers, microenvironment, and immunotherapy. Cancer Lett 2025; 610:217350. [PMID: 39581219 DOI: 10.1016/j.canlet.2024.217350] [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: 09/20/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Pancreatic cancer remains one of the most challenging malignancies to treat due to its late-stage diagnosis, aggressive progression, and high resistance to existing therapies. This review examines the latest advancements in early detection, and therapeutic strategies, with a focus on emerging biomarkers, tumor microenvironment (TME) modulation, and the integration of artificial intelligence (AI) in data analysis. We highlight promising biomarkers, including microRNAs (miRNAs) and circulating tumor DNA (ctDNA), that offer enhanced sensitivity and specificity for early-stage diagnosis when combined with multi-omics panels. A detailed analysis of the TME reveals how components such as cancer-associated fibroblasts (CAFs), immune cells, and the extracellular matrix (ECM) contribute to therapy resistance by creating immunosuppressive barriers. We also discuss therapeutic interventions that target these TME components, aiming to improve drug delivery and overcome immune evasion. Furthermore, AI-driven analyses are explored for their potential to interpret complex multi-omics data, enabling personalized treatment strategies and real-time monitoring of treatment response. We conclude by identifying key areas for future research, including the clinical validation of biomarkers, regulatory frameworks for AI applications, and equitable access to innovative therapies. This comprehensive approach underscores the need for integrated, personalized strategies to improve outcomes in pancreatic cancer.
Collapse
Affiliation(s)
- Baofa Yu
- Taimei Baofa Cancer Hospital, Dongping, Shandong, 271500, China; Jinan Baofa Cancer Hospital, Jinan, Shandong, 250000, China; Beijing Baofa Cancer Hospital, Beijing, 100010, China; Immune Oncology Systems, Inc, San Diego, CA, 92102, USA.
| | - Shengwen Shao
- Institute of Microbiology and Immunology, Huzhou University School of Medicine, Huzhou, Zhejiang, 313000, China.
| | - Wenxue Ma
- Department of Medicine, Sanford Stem Cell Institute, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
33
|
Hu H, Wang Q, Yu D, Tao X, Guo M, Tian S, Zhang Q, Xu M, Geng X, Zhang H, Xu H, Li L, Xie S, Chen K, Zhu W, Li X, Xu H, Li B, Zhang W, Liu S. Berberine Derivative B68 Promotes Tumor Immune Clearance by Dual-Targeting BMI1 for Senescence Induction and CSN5 for PD-L1 Degradation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413122. [PMID: 39721027 PMCID: PMC11831439 DOI: 10.1002/advs.202413122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/05/2024] [Indexed: 12/28/2024]
Abstract
Promoting tumor cell senescence arrests the cell cycle of tumor cells and activates the immune system to eliminate these senescent cells, thereby suppressing tumor growth. Nevertheless, PD-L1 positive senescent tumor cells resist immune clearance and possess the ability to secret various cytokines and inflammatory factors that stimulate the growth of tumor cells. Consequently, drugs capable of both triggering senescence in tumor cells and concurrently diminishing the expression of PD-L1 to counteract immune evasion are urgently needed. Here, a berberine derivative B68 is developed, which specifically induces tumor cell senescence by targeting BMI1. B68 also involves the degradation of PD-L1 by targeting CSN5, thereby disrupting the immunosuppressive PD-1/PD-L1 interaction and enabling rapid clearance of senescent tumor cells. This approach simultaneously inhibits tumor progression and activates T cell immunity, as evidenced by the robust antitumor response following B68-induced immunization of senescent cancer cells. Moreover, the synergistic effect of B68 with anti-CTLA4 therapy further enhances antitumor immunity, and its ability to induce senescence in cancer cells triggers a strong protective response by dendritic and CD8+ T cells. These findings provide a scientific basis for developing a new tumor treatment strategy based on senescence induction and lay the foundation for further preclinical research.
Collapse
Affiliation(s)
- Hongmei Hu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Qun Wang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Dianping Yu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Xiaoyu Tao
- State Key Laboratory of Chemical Biology, Drug Discovery and Design CenterShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- University of Chinese Academy of SciencesBeijing100049China
| | - Mengmeng Guo
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Saisai Tian
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghai200433China
| | - Qing Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Mengting Xu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Xiangxin Geng
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Hongwei Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Hanchi Xu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Linyang Li
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Shize Xie
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghai200433China
| | - Kaixian Chen
- University of Chinese Academy of SciencesBeijing100049China
- State Key Laboratory of Drug ResearchDrug Discovery and Design CenterShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Weiliang Zhu
- University of Chinese Academy of SciencesBeijing100049China
- State Key Laboratory of Drug ResearchDrug Discovery and Design CenterShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Xu‐Wen Li
- State Key Laboratory of Chemical Biology, Drug Discovery and Design CenterShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hanchen Xu
- Institute of Digestive DiseasesLonghua HospitalShanghai University of Traditional Chinese MedicineShanghai200032China
| | - Bo Li
- University of Chinese Academy of SciencesBeijing100049China
- State Key Laboratory of Drug ResearchDrug Discovery and Design CenterShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Weidong Zhang
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghai200433China
- Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100193China
| | - Sanhong Liu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| |
Collapse
|
34
|
Li J, Luo Y, Fu Q, Tang S, Zhang P, Frazer IH, Liu X, Wang T, Ni G. Caerin 1.1/1.9-mediated antitumor immunity depends on IFNAR-Stat1 signalling of tumour infiltrating macrophage by autocrine IFNα and is enhanced by CD47 blockade. Sci Rep 2025; 15:3789. [PMID: 39885296 PMCID: PMC11782643 DOI: 10.1038/s41598-025-87687-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 01/21/2025] [Indexed: 02/01/2025] Open
Abstract
Previously, we demonstrated that natural host-defence peptide caerin 1.1/caerin 1.9 (F1/F3) increases the efficacy of anti-PD-1 and therapeutic vaccine, in a HPV16 + TC-1 tumour model, but the anti-tumor mechanism of F1/F3 is still unclear. In this study, we explored the impact of F1/F3 on the tumor microenvironment in a transplanted B16 melanoma model, and further investigated the mechanism of action of F1/F3 using monoclonal antibodies to deplete relevant cells, gene knockout mice and flow cytometry. We show that F1/F3 is able to inhibit the growth of melanoma B16 tumour cells both in vitro and in vivo. Depletion of macrophages, blockade of IFNα receptor, and Stat1 inhibition each abolishes F1/F3-mediated antitumor responses. Subsequent analysis reveals that F1/F3 increases the tumour infiltration of inflammatory macrophages, upregulates the level of IFNα receptor, and promotes the secretion of IFNα by macrophages. Interestingly, F1/F3 upregulates CD47 level on tumour cells; and blocking CD47 increases F1/F3-mediated antitumor responses. Furthermore, F1/F3 intratumor injection, CD47 blockade, and therapeutic vaccination significantly increases the survival time of B16 tumour-bearing mice. These results indicate that F1/F3 may be effective to improve the efficacy of ICB and therapeutic vaccine-based immunotherapy for human epithelial cancers and warrants consideration for clinical trials.
Collapse
Affiliation(s)
- Junjie Li
- Key Laboratory of Cancer Immunotherapy of Guangdong Tertiary Education, Guangdong CAR-T Treatment Related Adverse Reaction Key Laboratory, The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China
- Zhongao Biomedical Technology (Guangdong) Co., Ltd, Zhongshan, 528403, Guangdong, China
| | - Yuandong Luo
- Medical School of Guizhou University, Guiyang, 550000, Guizhou, China
| | - Quanlan Fu
- Medical School of Guizhou University, Guiyang, 550000, Guizhou, China
| | - Shuxian Tang
- Cancer Research Institute, Foshan First People's Hospital, Foshan, 528000, Guangdong, China
| | - Pingping Zhang
- Cancer Research Institute, Foshan First People's Hospital, Foshan, 528000, Guangdong, China
| | - Ian H Frazer
- Diamantia Institute, Translational Research Institute, University of Queensland, Woolloongabba, Brisbane, QLD, 4002, Australia
| | - Xiaosong Liu
- Key Laboratory of Cancer Immunotherapy of Guangdong Tertiary Education, Guangdong CAR-T Treatment Related Adverse Reaction Key Laboratory, The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China
- Cancer Research Institute, Foshan First People's Hospital, Foshan, 528000, Guangdong, China
| | - Tianfang Wang
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia.
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia.
| | - Guoying Ni
- Key Laboratory of Cancer Immunotherapy of Guangdong Tertiary Education, Guangdong CAR-T Treatment Related Adverse Reaction Key Laboratory, The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China.
- Cancer Research Institute, Foshan First People's Hospital, Foshan, 528000, Guangdong, China.
| |
Collapse
|
35
|
Huang Q, Ren H, Bao Z, Jiang Y, Luo Y, Yao L. The impact of neoadjuvant immunotherapy on the clinical efficacy of stage IIIA-N2 non-small cell lung cancer patients. Int Immunopharmacol 2025; 146:113809. [PMID: 39708486 DOI: 10.1016/j.intimp.2024.113809] [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: 08/06/2024] [Revised: 11/24/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024]
Abstract
OBJECTIVE To explore the impact of neoadjuvant immunotherapy on the clinical efficacy of stage IIIA-N2 non-small cell lung cancer (NSCLC) patients. METHODS The retrospective study was conducted on 120 patients with stage IIIA-N2 NSCLC admitted to our hospital during April 2020 to April 2022. The control group received local chemotherapy, while the combination group received neoadjuvant immunotherapy on the basis of chemotherapy. The treatment efficacy, immune function, PD-1 and PD-L1 (SP142) expression levels, and changes in inflammatory factors were compared between the two groups. Kaplan Meier survival curve was used to analyze the overall survival rate. RESULTS Total effective rate in the control group was 53.33 % (15.00 % recovery + 16.66 % significant effect + 21.66 % effective), and the combined group had a higher total effective rate of 85.00 % (41.66 % recovery + 23.33 % significant effective + 20.00 % effective) (P < 0.001). After intervention, the combination group had largely increased immune indicators levels, including CD3+, CD4+ and CD4+/CD8+ (P < 0.001), but reduced levels of CD8+, PD-1 and PD-L1 (SP142) than the control group (P < 0.001). After intervention, the levels of inflammatory factors in the combination group were also reduced than the control group (P < 0.001). Compared to the control group with an adverse reaction rate of 31.66 % (8.33 % gastrointestinal reaction + 11.66 % hair loss + 6.66 % proteinuria + 5.00 % diarrhea), the combined group had much lower adverse reaction rate of 11.66 % (1.66 % gastrointestinal reaction + 5.00 % hair loss + 3.33 % proteinuria + 1.66 % diarrhea) (P < 0.05). After 24 months of follow-up, the overall survival rate was 58.33 % (35/60) and 40.00 % (24/60) in the combination group and the control group, respectively. The Kaplan Meier survival curve analysis showed a statistically significant difference in overall survival rate between the two groups (P < 0.05). CONCLUSION Neoadjuvant immunotherapy had a more significant therapeutic effect on stage IIIA-N2 NSCLC patients by reducing immunosuppressive and inflammatory factors, improving immune function, and reducing the occurrence of adverse reactions.
Collapse
Affiliation(s)
- Qin Huang
- Department of Oncology, The Second People's Hospital of Banan District, Chongqing 400054, China.
| | - Hongbo Ren
- Department of Oncology, The Second People's Hospital of Banan District, Chongqing 400054, China
| | - Zhonghui Bao
- Department of Oncology, The Second People's Hospital of Banan District, Chongqing 400054, China
| | - Yi Jiang
- Department of Oncology, The Second People's Hospital of Banan District, Chongqing 400054, China
| | - Yan Luo
- Department of Oncology, The Second People's Hospital of Banan District, Chongqing 400054, China
| | - Lan Yao
- Department of Oncology, The Second People's Hospital of Banan District, Chongqing 400054, China
| |
Collapse
|
36
|
Lin H, Gao Y, Zhu L, Guo Y, Zhang L, Xie J, Yang D, Liu J, Dong Q, Zhu Z. Rational Design of Single‐Atom Nanozymes for Combination Cancer Immunotherapy. ADVANCED FUNCTIONAL MATERIALS 2025. [DOI: 10.1002/adfm.202416563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Indexed: 02/03/2025]
Abstract
AbstractRemodeling of the tumor immune microenvironment and enhancement of antitumor immune responses are necessary to overcome immunotherapy resistance in tumors. However, tumor heterogeneity and complexity of immune evasion mechanisms pose significant therapeutic challenges. Nanozymes exhibit enzyme‐like characteristics and unique nanomaterial properties, showing potential for tumor therapy. However, design of effective nanozymes remains complex, inefficient, and functionally limited. Therefore, in this study, a novel strategy combining rationally designed single‐atom nanozymes (SAzymes) with immune checkpoint blockade (ICB) therapy is established. Molybdenum SAzymes supported on graphitic carbon nitride (Mo SAs) are constructed using 25 transition metal candidates from the 4th to 6th periods based on high‐throughput calculations and optimal piezoelectric‐enhanced multienzyme‐like activities. Upon activation by ultrasound, Mo SAs exerted potent therapeutic effects against ICB‐resistant tumors and remodeled the tumor immune microenvironment by inducing tumor immunogenic cell death, alleviating tumor hypoxia, and modulating chemokine expression in tumors. Combination of Mo SAs with anti‐programmed death protein‐1 antibodies further enhanced their antitumor efficacy, highlighting their potential to treat ICB‐resistant tumors.
Collapse
Affiliation(s)
- Hanchao Lin
- Key Laboratory of Whole‐Period Monitoring and Precise Intervention of Digestive Cancer Shanghai Municipal Health Commission Minhang Hospital Fudan University 170 Xingsong Road Shanghai 201199 China
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute Fudan University 12 Middle Urumuqi Road Shanghai 200040 China
| | - Yonghui Gao
- College of Materials Science and Engineering Qingdao University of Science and Technology 53 Zhengzhou Road Qingdao Shandong 266042 China
| | - Le Zhu
- Key Laboratory of Whole‐Period Monitoring and Precise Intervention of Digestive Cancer Shanghai Municipal Health Commission Minhang Hospital Fudan University 170 Xingsong Road Shanghai 201199 China
| | - Yu Guo
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute Fudan University 12 Middle Urumuqi Road Shanghai 200040 China
| | - Lumin Zhang
- Key Laboratory of Whole‐Period Monitoring and Precise Intervention of Digestive Cancer Shanghai Municipal Health Commission Minhang Hospital Fudan University 170 Xingsong Road Shanghai 201199 China
| | - Jiali Xie
- College of Materials Science and Engineering Qingdao University of Science and Technology 53 Zhengzhou Road Qingdao Shandong 266042 China
| | - Dongqin Yang
- Department of Laboratory Medicine Huashan Hospital Fudan University 12 Middle Urumqi Road Shanghai 200040 China
| | - Jing Liu
- College of Materials Science and Engineering Qingdao University of Science and Technology 53 Zhengzhou Road Qingdao Shandong 266042 China
| | - Qiongzhu Dong
- Key Laboratory of Whole‐Period Monitoring and Precise Intervention of Digestive Cancer Shanghai Municipal Health Commission Minhang Hospital Fudan University 170 Xingsong Road Shanghai 201199 China
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute Fudan University 12 Middle Urumuqi Road Shanghai 200040 China
| | - Zhiling Zhu
- College of Materials Science and Engineering Qingdao University of Science and Technology 53 Zhengzhou Road Qingdao Shandong 266042 China
- Key Laboratory of Optic‐electric Sensing and Analytical Chemistry for Life Science MOE; Shandong Key Laboratory of Biochemical Analysis Qingdao University of Science and Technology 53 Zhengzhou Road Qingdao Shandong 266042 China
| |
Collapse
|
37
|
Al-Matouq J, Al-Ghafli H, Alibrahim NN, Alsaffar N, Radwan Z, Ali MD. Unveiling the Interplay Between the Human Microbiome and Gastric Cancer: A Review of the Complex Relationships and Therapeutic Avenues. Cancers (Basel) 2025; 17:226. [PMID: 39858007 PMCID: PMC11763844 DOI: 10.3390/cancers17020226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/23/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
The human microbiota plays a crucial role in maintaining overall health and well-being. The gut microbiota has been implicated in developing and progressing various diseases, including cancer. This review highlights the related mechanisms and the compositions that influence cancer pathogenesis with a highlight on gastric cancer. We provide a comprehensive overview of the mechanisms by which the microbiome influences cancer development, progression, and response to treatment, with a focus on identifying potential biomarkers for early detection, prevention strategies, and novel therapeutic interventions that leverage microbiome modulation. This comprehensive review can guide future research and clinical practices in understanding and harnessing the microbiome to optimize gastric cancer therapies.
Collapse
Affiliation(s)
- Jenan Al-Matouq
- Department of Medical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia; (H.A.-G.); (N.N.A.); (N.A.); (Z.R.)
| | - Hawra Al-Ghafli
- Department of Medical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia; (H.A.-G.); (N.N.A.); (N.A.); (Z.R.)
| | - Noura N. Alibrahim
- Department of Medical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia; (H.A.-G.); (N.N.A.); (N.A.); (Z.R.)
| | - Nida Alsaffar
- Department of Medical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia; (H.A.-G.); (N.N.A.); (N.A.); (Z.R.)
| | - Zaheda Radwan
- Department of Medical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia; (H.A.-G.); (N.N.A.); (N.A.); (Z.R.)
| | - Mohammad Daud Ali
- Department of Pharmacy, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia;
| |
Collapse
|
38
|
Peng H, Chao Z, Wang Z, Hao X, Xi Z, Ma S, Guo X, Zhang J, Zhou Q, Qu G, Gao Y, Luo J, Wang Z, Wang J, Li L. Biomechanics in the tumor microenvironment: from biological functions to potential clinical applications. Exp Hematol Oncol 2025; 14:4. [PMID: 39799341 PMCID: PMC11724500 DOI: 10.1186/s40164-024-00591-7] [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: 09/16/2024] [Accepted: 12/10/2024] [Indexed: 01/15/2025] Open
Abstract
Immune checkpoint therapies have spearheaded drug innovation over the last decade, propelling cancer treatments toward a new era of precision therapies. Nonetheless, the challenges of low response rates and prevalent drug resistance underscore the imperative for a deeper understanding of the tumor microenvironment (TME) and the pursuit of novel targets. Recent findings have revealed the profound impacts of biomechanical forces within the tumor microenvironment on immune surveillance and tumor progression in both murine models and clinical settings. Furthermore, the pharmacological or genetic manipulation of mechanical checkpoints, such as PIEZO1, DDR1, YAP/TAZ, and TRPV4, has shown remarkable potential in immune activation and eradication of tumors. In this review, we delved into the underlying biomechanical mechanisms and the resulting intricate biological meaning in the TME, focusing mainly on the extracellular matrix, the stiffness of cancer cells, and immune synapses. We also summarized the methodologies employed for biomechanical research and the potential clinical translation derived from current evidence. This comprehensive review of biomechanics will enhance the understanding of the functional role of biomechanical forces and provide basic knowledge for the discovery of novel therapeutic targets.
Collapse
Affiliation(s)
- Hao Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
- The Second Clinical School, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Zheng Chao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Zefeng Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaodong Hao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Zirui Xi
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
- The Second Clinical School, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Sheng Ma
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Xiangdong Guo
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Junbiao Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Qiang Zhou
- Department of Urology, Qinghai University Affiliated Hospital, Qinghai University Medical College, Xining, 810001, Qinghai, China
| | - Guanyu Qu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
- The Second Clinical School, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Yuan Gao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
- The Second Clinical School, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Jing Luo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhihua Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China.
- Taikang Tongji (Wuhan) Hospital, 420060, Wuhan, China.
| | - Jing Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China.
| | - Le Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China.
| |
Collapse
|
39
|
Tian JZ, Zhang L, Lin FY, He RJ, Tian WR, Yan L, Huang GX, Ai JW, Pei B, Li DS. The efficacy and safety of PD-1 inhibitors combined with chemotherapy treatment for advanced esophageal cancer: a network meta-analysis. Front Med (Lausanne) 2025; 11:1515263. [PMID: 39867925 PMCID: PMC11759289 DOI: 10.3389/fmed.2024.1515263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/27/2024] [Indexed: 01/28/2025] Open
Abstract
Objective This study systematically evaluated the efficacy of programmed death 1 (PD-1) inhibitors combined with chemotherapy for advanced esophageal cancer (EC). Methods PubMed, Embase, Web of Science, Scopus, and Cochrane Library were searched to identify related randomized controlled trials (RCTs). Results Seven RCTs involving 4,363 participants were included. The results of the direct comparison showed that, compared with chemotherapy alone, PD-1 inhibitors combined with chemotherapy significantly improved overall survival (OS) (HR = 0.69, 95%CI = 0.63-0.74), progression-free survival (PFS) (HR = 0.63, 95%CI = 0.58-0.67), objective response rate (ORR) (RR = 1.41, 95%CI = 1.28-1.57), but were associated with a slight increase in treatment-related adverse events (AEs) (RR = 1.08, 95%CI = 1.03-1.14). The results of the network meta-analysis showed that toripalimab, sintilimab or camrelizumab, and nivolumab combined with chemotherapy were the best in OS, PFS, and ORR, respectively, with camrelizumab showing the lowest incidence of AEs. Conclusion These results suggest that PD-1 inhibitors combined with chemotherapy provide superior clinical benefits over chemotherapy alone, albeit with a moderate increase in AEs. However, further verification through multi-center, high-quality RCTs with larger sample sizes is needed to confirm these findings. Systematic review registration https//wwwcrdyorkacuk/prospero/display_recordphp?ID=CRD42024627485
Collapse
Affiliation(s)
- Jian-Zhou Tian
- Evidence-Based Medicine Center, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Li Zhang
- Department of Central Sterile Supply, Fujian Medical University Union Hospital, Fuzhou, China
| | - Fu-Yong Lin
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ren-Jiao He
- Department Three of Orthopedics/Plastic Surgery, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Wen-Rong Tian
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Liu Yan
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Guo-Xin Huang
- Evidence-Based Medicine Center, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Jin-Wei Ai
- Evidence-Based Medicine Center, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department Three of Orthopedics/Plastic Surgery, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Bin Pei
- Evidence-Based Medicine Center, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
- Department Three of Orthopedics/Plastic Surgery, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - De-Sheng Li
- Department Three of Orthopedics/Plastic Surgery, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| |
Collapse
|
40
|
Liu Y, Tan H, Dai J, Lin J, Zhao K, Hu H, Zhong C. Targeting macrophages in cancer immunotherapy: Frontiers and challenges. J Adv Res 2025:S2090-1232(24)00622-2. [PMID: 39778768 DOI: 10.1016/j.jare.2024.12.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Cancer immunotherapy has emerged as a groundbreaking approach in cancer treatment, primarily realized through the manipulation of immune cells, notably T cell adoption and immune checkpoint blockade. Nevertheless, the manipulation of T cells encounters formidable hurdles. Macrophages, serving as the pivotal link between innate and adaptive immunity, play crucial roles in phagocytosis, cytokine secretion, and antigen presentation. Consequently, macrophage-targeted therapies have garnered significant attention. AIM OF REVIEW We aim to provide the most cutting-edge insights and future perspectives for macrophage-targeted therapies, fostering the development of novel and effective cancer treatments. KEY SCIENTIFIC CONCEPTS OF REVIEW To date, the forefront strategies for macrophage targeting encompass: altering their plasticity, harnessing CAR-macrophages, and targeting phagocytosis checkpoints. Macrophages are characterized by their remarkable diversity and plasticity, offering a unique therapeutic target. In this context, we critically analyze the innovative strategies aimed at transforming macrophages from their M2 (tumor-promoting) to M1 (tumor-suppressing) phenotype. Furthermore, we delve into the design principles, developmental progress, and advantages of CAR-macrophages. Additionally, we illuminate the challenges encountered in targeting phagocytosis checkpoints on macrophages and propose potential strategies to overcome these obstacles.
Collapse
Affiliation(s)
- Yu'e Liu
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China; Department of Pediatric Hematology-Oncology, Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
| | - Huabing Tan
- Department of Infectious Diseases, Hepatology Institute, Renmin Hospital, Hubei University of Medicine, Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, Hubei Province 442000, China; General internal medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430048, China
| | - Jingyuan Dai
- School of Computer Science and Information Systems, Northwest Missouri State University, Maryville, MO 64468, USA
| | - Jianghua Lin
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Kaijun Zhao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China.
| | - Haibo Hu
- Department of Cardiothoracic Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China.
| | - Chunlong Zhong
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China.
| |
Collapse
|
41
|
Zheng Y, Wang J, Zhao G, Zhang Z, Shao Y, Lu B, Zhang Y, Chen R, Sun L, Xie X, Ding J, Zheng J, Chai D. Targeting Siglec-E facilitates tumor vaccine-induced antitumor immunity in renal carcinoma. J Immunother Cancer 2025; 13:e010521. [PMID: 39755580 PMCID: PMC11749828 DOI: 10.1136/jitc-2024-010521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 11/28/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Siglec-E is an immune checkpoint inhibitory molecule. Expression of Siglec-E on the immune cells has been shown to promote tumor regression. This study aimed to develop an adenovirus (Ad) vaccine targeting Siglec-E and carbonic anhydrase IX (CAIX) (Ad-Siglec-E/CAIX) and to evaluate its potential antitumor effects in several preclinical renal cancer models. METHODS Ad vaccines encoding Siglec-E or CAIX were developed and evaluated for their therapeutic potential in mouse subcutaneous, lung metastatic, and orthotopic tumor models. The expression of Ad-Siglec-E/CAIX was confirmed via PCR and flow cytometry. Immune responses induced by Ad-Siglec-E/CAIX were assessed in vitro and in vivo using flow cytometry, immunohistochemistry, ELISA, histological analysis, cell proliferation, enzyme-linked immunosorbent spot, cytotoxic T lymphocytes (CTL) killing, and cell depletion assays. RESULTS Ad-Siglec-E/CAIX vaccine induced the increase of tumor-infiltrated immune cells, and significantly suppressed the subcutaneous tumor growth of renal carcinoma. Immunization with Ad-Siglec-E/CAIX promoted the induction and maturation of CD11c+ dendritic cells and their subsets, which in turn enhanced tumor-specific CD8+ T cell immune responses, as evidenced by increased CD8+ T cell proliferation and CTL activity. Importantly, the deletion of CD8+ T cells in vivo abolished the antitumor effect of the Ad-Siglec-E/CAIX vaccine, highlighting the essential role of functional CD8+ T cell responses. The potent therapeutic efficacy of the Ad-Siglec-E/CAIX vaccine was also observed in lung metastasis and orthotopic tumor models through tumor-specific CD8+ T cell immune responses. CONCLUSIONS Our results indicate that targeting Siglec-E enhances the therapeutic efficacy of Ad-CAIX against renal carcinoma, providing a promising therapeutic option for solid tumors.
Collapse
Affiliation(s)
- Yanyan Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiawei Wang
- Department of Oncology, Ninghe District Hospital of Tianjin, Tianjin, China
| | - Guangya Zhao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zichun Zhang
- Department of Urology, The Yancheng Clinical College of Xuzhou Medical University, Yancheng City No 1 People's Hospital, Yancheng, Jiangsu, China
| | - Yingxiang Shao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Bowen Lu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuchen Zhang
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Renjin Chen
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Li Sun
- Department of Oncology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaohui Xie
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Jiage Ding
- Department of Oncology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
42
|
Chang L, Liu J, Zhu J, Guo S, Wang Y, Zhou Z, Wei X. Advancing precision medicine: the transformative role of artificial intelligence in immunogenomics, radiomics, and pathomics for biomarker discovery and immunotherapy optimization. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0376. [PMID: 39749734 PMCID: PMC11795263 DOI: 10.20892/j.issn.2095-3941.2024.0376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/27/2024] [Indexed: 01/04/2025] Open
Abstract
Artificial intelligence (AI) is significantly advancing precision medicine, particularly in the fields of immunogenomics, radiomics, and pathomics. In immunogenomics, AI can process vast amounts of genomic and multi-omic data to identify biomarkers associated with immunotherapy responses and disease prognosis, thus providing strong support for personalized treatments. In radiomics, AI can analyze high-dimensional features from computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography/computed tomography (PET/CT) images to discover imaging biomarkers associated with tumor heterogeneity, treatment response, and disease progression, thereby enabling non-invasive, real-time assessments for personalized therapy. Pathomics leverages AI for deep analysis of digital pathology images, and can uncover subtle changes in tissue microenvironments, cellular characteristics, and morphological features, and offer unique insights into immunotherapy response prediction and biomarker discovery. These AI-driven technologies not only enhance the speed, accuracy, and robustness of biomarker discovery but also significantly improve the precision, personalization, and effectiveness of clinical treatments, and are driving a shift from empirical to precision medicine. Despite challenges such as data quality, model interpretability, integration of multi-modal data, and privacy protection, the ongoing advancements in AI, coupled with interdisciplinary collaboration, are poised to further enhance AI's roles in biomarker discovery and immunotherapy response prediction. These improvements are expected to lead to more accurate, personalized treatment strategies and ultimately better patient outcomes, marking a significant step forward in the evolution of precision medicine.
Collapse
Affiliation(s)
- Luchen Chang
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jiamei Liu
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jialin Zhu
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Shuyue Guo
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yao Wang
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zhiwei Zhou
- Departments of Biochemistry and Radiation Oncology, UT Southwestern Medical Center, Dallas 75390, USA
| | - Xi Wei
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
43
|
Zhang S, Gong L, Sun Y, Zhang F, Gao W. An ultra-long-acting L-asparaginase synergizes with an immune checkpoint inhibitor in starvation-immunotherapy of metastatic solid tumors. Biomaterials 2025; 312:122740. [PMID: 39096839 DOI: 10.1016/j.biomaterials.2024.122740] [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: 12/31/2023] [Revised: 07/05/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024]
Abstract
Metastasis stands as the primary contributor to mortality associated with tumors. Chemotherapy and immunotherapy are frequently utilized in the management of metastatic solid tumors. Nevertheless, these therapeutic modalities are linked to serious adverse effects and limited effectiveness in preventing metastasis. Here, we report a novel therapeutic strategy named starvation-immunotherapy, wherein an immune checkpoint inhibitor is combined with an ultra-long-acting L-asparaginase that is a fusion protein comprising L-asparaginase (ASNase) and an elastin-like polypeptide (ELP), termed ASNase-ELP. ASNase-ELP's thermosensitivity enables it to generate an in-situ depot following an intratumoral injection, yielding increased dose tolerance, improved pharmacokinetics, sustained release, optimized biodistribution, and augmented tumor retention compared to free ASNase. As a result, in murine models of oral cancer, melanoma, and cervical cancer, the antitumor efficacy of ASNase-ELP by selectively and sustainably depleting L-asparagine essential for tumor cell survival was substantially superior to that of ASNase or Cisplatin, a first-line anti-solid tumor medicine, without any observable adverse effects. Furthermore, the combination of ASNase-ELP and an immune checkpoint inhibitor was more effective than either therapy alone in impeding melanoma metastasis. Overall, the synergistic strategy of starvation-immunotherapy holds excellent promise in reshaping the therapeutic landscape of refractory metastatic tumors and offering a new alternative for next-generation oncology treatments.
Collapse
Affiliation(s)
- Sanke Zhang
- Biomedical Engineering Department, Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China; Peking University International Cancer Institute, Beijing, 100191, China; Peking University-Yunnan Baiyao International Medical Research Center, Beijing, 100191, China
| | - Like Gong
- Biomedical Engineering Department, Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China; Peking University International Cancer Institute, Beijing, 100191, China; Peking University-Yunnan Baiyao International Medical Research Center, Beijing, 100191, China
| | - Yuanzi Sun
- Biomedical Engineering Department, Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China; Peking University International Cancer Institute, Beijing, 100191, China; Peking University-Yunnan Baiyao International Medical Research Center, Beijing, 100191, China
| | - Fan Zhang
- Biomedical Engineering Department, Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China; Peking University International Cancer Institute, Beijing, 100191, China; Peking University-Yunnan Baiyao International Medical Research Center, Beijing, 100191, China
| | - Weiping Gao
- Biomedical Engineering Department, Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China; Peking University International Cancer Institute, Beijing, 100191, China; Peking University-Yunnan Baiyao International Medical Research Center, Beijing, 100191, China; Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China.
| |
Collapse
|
44
|
Chen Y, Guo Y, Cheng W, Fan J, Li J, Song J, Yang X, Wang K, Huang J. Sequentially Activated Smart DNA Nanospheres for Photoimmunotherapy and Immune Checkpoint Blockade. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410632. [PMID: 39588591 PMCID: PMC11744728 DOI: 10.1002/advs.202410632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/09/2024] [Indexed: 11/27/2024]
Abstract
Due to the inherent immunosuppression and immune evasion of cancer cells, combining photoimmunotherapy with immune checkpoint blockade leverages phototherapy and immune enhancement, overcoming mutual limitations and demonstrating significant anticancer potential. The main challenges include nonspecific accumulation of agents, uncontrolled activation, and drug carrier safety. Smart DNA nanospheres (NS) is developed with targeted delivery and controllable release of photosensitizers and immune agents to achieve effective synergistic therapy and minimize side effects. The multifunctional NS incorporate a targeting module for programming aptamers, a response module for programming i-motif and DNA/RNA hybrid sequences, and a therapeutic module for packaging photosensitizers and PD-L1 siRNA. NS navigate to the tumor site and are sequentially activated by intracellular acid and enzymes to release photosensitizers and programmed death ligand 1 (PD-L1) small interfering RNA (siRNA)a. Besides tumor killing and immune promotion, activated NS downregulate PD-L1 expression, alleviating immune tolerance and evasion, thus enhancing the immune response. These results indicate that NS significantly enhance antitumor immune responses, synergistically improve antitumor efficacy, and reduce systemic toxicity. This study broadens the application of DNA nanomaterials in precision drug delivery and tumor therapy.
Collapse
Affiliation(s)
- Yu Chen
- State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Chemistry and Chemical EngineeringKey Laboratory for Bio‐Nanotechnology and Molecular Engineering of Hunan ProvinceHunan UniversityChangsha410082P. R. China
| | - Yu Guo
- State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Chemistry and Chemical EngineeringKey Laboratory for Bio‐Nanotechnology and Molecular Engineering of Hunan ProvinceHunan UniversityChangsha410082P. R. China
| | - Wen Cheng
- State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Chemistry and Chemical EngineeringKey Laboratory for Bio‐Nanotechnology and Molecular Engineering of Hunan ProvinceHunan UniversityChangsha410082P. R. China
| | - Jiahao Fan
- State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Chemistry and Chemical EngineeringKey Laboratory for Bio‐Nanotechnology and Molecular Engineering of Hunan ProvinceHunan UniversityChangsha410082P. R. China
| | - Jiacheng Li
- State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Chemistry and Chemical EngineeringKey Laboratory for Bio‐Nanotechnology and Molecular Engineering of Hunan ProvinceHunan UniversityChangsha410082P. R. China
| | - Jiajia Song
- State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Chemistry and Chemical EngineeringKey Laboratory for Bio‐Nanotechnology and Molecular Engineering of Hunan ProvinceHunan UniversityChangsha410082P. R. China
| | - Xiaohai Yang
- State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Chemistry and Chemical EngineeringKey Laboratory for Bio‐Nanotechnology and Molecular Engineering of Hunan ProvinceHunan UniversityChangsha410082P. R. China
| | - Kemin Wang
- State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Chemistry and Chemical EngineeringKey Laboratory for Bio‐Nanotechnology and Molecular Engineering of Hunan ProvinceHunan UniversityChangsha410082P. R. China
| | - Jin Huang
- State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Chemistry and Chemical EngineeringKey Laboratory for Bio‐Nanotechnology and Molecular Engineering of Hunan ProvinceHunan UniversityChangsha410082P. R. China
| |
Collapse
|
45
|
Arafat Hossain M. A comprehensive review of immune checkpoint inhibitors for cancer treatment. Int Immunopharmacol 2024; 143:113365. [PMID: 39447408 DOI: 10.1016/j.intimp.2024.113365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Immunology-based therapies are emerging as an effective cancer treatment, using the body's immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
46
|
Huo Y, Wang D, Yang S, Xu Y, Qin G, Zhao C, Lei Q, Zhao Q, Liu Y, Guo K, Ouyang S, Sun T, Wang H, Fan F, Han N, Liu H, Chen H, Miao L, Liu L, Duan Y, Lv W, Liu L, Zhang Z, Cang S, Wang L, Zhang Y. Optimal timing of anti-PD-1 antibody combined with chemotherapy administration in patients with NSCLC. J Immunother Cancer 2024; 12:e009627. [PMID: 39706602 PMCID: PMC11667274 DOI: 10.1136/jitc-2024-009627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 11/17/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Anti-programmed cell death 1 (PD-1) antibody combined with chemotherapy simultaneously is regarded as the standard treatment for patients with advanced non-small cell lung cancer (NSCLC) by current clinical guidelines. Different immune statuses induced by chemotherapy considerably affect the synergistic effects of the chemo-anti-PD-1 combination. Therefore, it is necessary to determine the optimal timing of combination treatment administration. METHODS The dynamic immune status induced by chemotherapy was observed in paired peripheral blood samples of patients with NSCLC using flow cytometry and RNA sequencing. Ex vivo studies and metastatic lung carcinoma mouse models were used to evaluate immune activity and explore the optimal combination timing. A multicenter prospective clinical study of 170 patients with advanced NSCLC was performed to assess clinical responses, and systemic immunity was assessed using omics approaches. RESULTS PD-1 expression on CD8+ T cells was downregulated on day 1 (D1) and D2, but recovered on D3 after chemotherapy administration, which is regulated by the calcium influx-P65 signaling pathway. Programmed cell death 1 ligand 1 expression in myeloid-derived suppressor cells was markedly reduced on D3. RNA sequencing analysis showed that T-cell function began to gradually recover on D3 rather than on D1. In addition, ex vivo and in vivo studies have shown that anti-PD-1 treatment on D3 after chemotherapy may enhance the antitumor response and considerably inhibit tumor growth. Finally, in clinical practice, a 3-day-delay sequential combination enhanced the objective response rate (ORR, 68%) and disease control rate (DCR, 98%) compared with the simultaneous combination (ORR=37%; DCR=81%), and prolonged progression-free survival to a greater extent than the simultaneous combination. The new T-cell receptor clones were effectively expanded, and CD8+ T-cell activity was similarly recovered. CONCLUSIONS A 3-day-delay sequential combination might increase antitumor responses and clinical benefits compared with the simultaneous combination.
Collapse
Affiliation(s)
- Yachang Huo
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dan Wang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuangning Yang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yujie Xu
- Department of Oncology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Guohui Qin
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chenhui Zhao
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qingyang Lei
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qitai Zhao
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yaqing Liu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Kaiyuan Guo
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Songyun Ouyang
- Department of Respiratory and Critical Care Sleep Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ting Sun
- Department of Respiratory and Critical Care Sleep Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongmin Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feifei Fan
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Na Han
- Department of Oncology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hong Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongjie Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lijun Miao
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuqing Duan
- Department of Tumor Immunotherapy, the Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, Hebei, China
| | - Wei Lv
- Department of Tumor Immunotherapy, the Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, Hebei, China
| | - Lihua Liu
- Department of Tumor Immunotherapy, the Fourth Hospital of Hebei Medical University and Hebei Cancer Institute, Shijiazhuang, Hebei, China
| | - Zhixin Zhang
- Department of Technology, Chengdu ExAb Biotechnology Ltd, Chengdu, Sichuan, China
| | - Shundong Cang
- Department of Oncology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Liping Wang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- School of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
47
|
Chen K, Li Y, Ni J, Yang X, Zhou Y, Pang Y, Ye R, Chen H, Yu S, Wang P, Zhu Z. Identification of a novel subtype of SPP1 + macrophages expressing SIRPα: implications for tumor immune evasion and treatment response prediction. Exp Hematol Oncol 2024; 13:119. [PMID: 39696410 DOI: 10.1186/s40164-024-00587-3] [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: 09/15/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND SPP1 + macrophages are among the major phagocytic cells, yet promoting tumor immune evasion and predicting unfavorable prognosis, in various cancer types. Meanwhile, the predictive value of the abundance of SPP1 + macrophages in patients receiving immunotherapy remains debatable, indicating the potential existence of subtypes of SPP1 + macrophages with diverse biological functions. METHODS The single cell RNA sequencing data of myeloid cells integrated from several cancers including esophageal squamous cell carcinoma was analyzed for characterizing the function and cellular interactions of SPP1 + macrophages expressing SIRPα. Multiplexed immunohistochemistry was used to quantify the quantity and spatial distribution of SPP1 + macrophages expressing SIRPα. Kaplan-Meier method was used for survival analysis. In vitro and in vivo studies investigating the function of SPP1 + macrophages were performed. RESULTS SPP1 + macrophages possessed a high phagocytic signature and could engulf more tumor cells in vitro and in vivo. SIRPα expression could represent the phagocytic activity of SPP1 + macrophages and delineated subsets of SPP1 + macrophages with different functions. SPP1 + SIRPα + macrophages showed close spatial distance to tumor cells and positively correlated with PD1 + CD8 + T cells. A high abundance of SPP1 + SIRPα + macrophages at baseline corresponded to patients' response to PD-1/PD-L1 inhibitors. CONCLUSION A novel subtype of SPP1 + macrophages expressing SIRPα was identified and their abundance predicted patients' response to PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Kun Chen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yida Li
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Jianjiao Ni
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Xi Yang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Yue Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Yechun Pang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Ruiting Ye
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Hongru Chen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Silai Yu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Peng Wang
- Department of Hepatic Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China.
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China.
- Institute of Thoracic Oncology, Fudan University, Shanghai, China.
| |
Collapse
|
48
|
Park HB, An EK, Kim SJ, Ryu D, Zhang W, Pack CG, Kim H, Kwak M, Im W, Ryu JH, Lee PCW, Jin JO. Anti-PD-L1 Antibody Fragment Linked to Tumor-Targeting Lipid Nanoparticle Can Eliminate Cancer and Its Metastasis via Photoimmunotherapy. ACS NANO 2024; 18:33366-33380. [PMID: 39603816 DOI: 10.1021/acsnano.4c08448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Effective cancer therapy aims to treat primary tumors and metastatic and recurrent cancer. Immune checkpoint blockade-mediated immunotherapy has shown promising effects against tumors; however, its efficacy in metastatic or recurrent cancer is limited. Here, based on the advantages of nanomedicine, lipid nanoparticles (LNPs) that can target tumors are synthesized for photothermal therapy (PTT) and immunotherapy to treat primary and metastatic recurrent cancer. These LNPs, termed piLNPs, are encapsulated with indocyanine green and incorporated with the antigen (Ag)-binding fragment of the anti-PD-L1 antibody for targeting tumors and immunotherapy. Intravenously injected piLNPs in 4T1 breast tumor-bearing BALB/c mice effectively target the 4T1 tumor and are suitable for performing PTT using a near-infrared laser. Moreover, lung metastatic 4T1 tumor growth is completely prevented in mice previously cured of the 4T1 breast tumor by piLNP treatment and rechallenged with lung 4T1 metastatic cancer. Blockage of the second challenged metastatic 4T1 breast cancer by piLNP is due to the activation of Ag-specific T cells. Cytotoxic T lymphocytes from piLNP-cured mice selectively attack 4T1 breast cancer cells. Therefore, piLNP can be used as a multifunctional breast cancer treatment composition that can target tumors, treat primary tumors, and prevent metastasis and recurrence.
Collapse
Affiliation(s)
- Hae-Bin Park
- Department of Microbiology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Eun-Koung An
- Department of Microbiology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - So-Jung Kim
- Department of Microbiology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Dayoung Ryu
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Wei Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Chan-Gi Pack
- Department of Biomedical Engineering, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Hyuncheol Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, South Korea
| | - Minseok Kwak
- Department of Chemistry and Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, South Korea
| | - Wonpil Im
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Ja-Hyoung Ryu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, South Korea
| | - Peter C W Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Jun-O Jin
- Department of Microbiology, Brain Korea 21 Project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| |
Collapse
|
49
|
Palmer T, Kessler MD, Shao XM, Balan A, Yarchoan M, Zaidi N, Lopez-Vidal TY, Saeed AM, Gore J, Azad NS, Jaffee EM, Favorov AV, Anagnostou V, Karchin R, Gaykalova DA, Fertig EJ, Danilova L. SpliceMutr Enables Pan-Cancer Analysis of Splicing-Derived Neoantigen Burden in Tumors. CANCER RESEARCH COMMUNICATIONS 2024; 4:3137-3150. [PMID: 39470352 PMCID: PMC11648103 DOI: 10.1158/2767-9764.crc-23-0309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 07/05/2024] [Accepted: 10/25/2024] [Indexed: 10/30/2024]
Abstract
SIGNIFICANCE SpliceMutr shows that splicing antigenicity changes in response to ICI therapies and that native modulation of the splicing machinery through mutations increases the contribution of splicing to the neoantigen load of some The Cancer Genome Atlas cancer subtypes. Future studies of the relationship between splicing antigenicity and immune checkpoint inhibitor response pan-cancer are essential to establish the interplay between antigen heterogeneity and immunotherapy regimen on patient response.
Collapse
Affiliation(s)
- Theron Palmer
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University, Baltimore, Maryland
| | - Michael D. Kessler
- Convergence Institute, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Xiaoshan M. Shao
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Archana Balan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Mark Yarchoan
- Convergence Institute, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Neeha Zaidi
- Convergence Institute, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Tamara Y. Lopez-Vidal
- Convergence Institute, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Biochemistry, Cellular and Molecular Biology (BCMB) Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ali M. Saeed
- Convergence Institute, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Jessica Gore
- Department of Otorhinolaryngology-Head and Neck Surgery, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Nilofer S. Azad
- Convergence Institute, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Elizabeth M. Jaffee
- Convergence Institute, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Alexander V. Favorov
- Convergence Institute, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Laboratory of Systems Biology and Computational Genetics, Vavilov Institute of General Genetics, Moscow, Russia
| | - Valsamo Anagnostou
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Rachel Karchin
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Daria A. Gaykalova
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Otorhinolaryngology-Head and Neck Surgery, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Elana J. Fertig
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, Maryland
| | - Ludmila Danilova
- Convergence Institute, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
50
|
Feng Y, Gao M, Xu X, Liu H, Lu K, Song Z, Yu J, Liu X, Han X, Li L, Qiu L, Qian Z, Zhou S, Zhang H, Wang X. Elevated serum magnesium levels prompt favourable outcomes in cancer patients treated with immune checkpoint blockers. Eur J Cancer 2024; 213:115069. [PMID: 39489925 DOI: 10.1016/j.ejca.2024.115069] [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: 09/23/2024] [Accepted: 10/10/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Magnesium deficiency influences the activation and cytotoxicity of immune cells. Nevertheless, whether serum magnesium levels influence the clinical outcomes of immune checkpoint blockers (ICBs) treatment still remains ambiguous. There is an urgent need for clinical research to elucidate the relationship between serum magnesium levels and the outcomes of ICB therapy. Such insights could offer new perspectives on immunotherapy for cancer. METHODS A multi-center retrospective study involving in pan-cancer patients treated with ICBs at three large cancer centers from August 2012 to May 2023 was conducted. The primary objective was to assess the correlation between serum magnesium levels and therapeutic response in patients receiving ICBs, and further evaluate the associations between serum magnesium levels and progression-free survival (PFS) and overall survival (OS). RESULTS A total of 1441 patients treated with ICBs, including 1042 with lung cancer, 270 with esophageal cancer, and 129 with Hodgkin lymphoma, were enrolled in this study. We found that patients with elevated serum magnesium levels exhibited a favourable response to ICBs treatment. The optimal cut-off point for serum magnesium level (0.79 mmol/L) was applied for stratifying patients into distinct groups. In the three tumor cohorts, patients in high magnesium level group (Mg2+ ≥ 0.79 mmol/L) had longer PFS and OS than those in low magnesium level group (Mg2+ < 0.79 mmol/L). Univariate and multivariate analyses confirmed that the serum Mg2+ level serves as an independent prognostic factor for cancer patients receiving ICBs therapy. CONCLUSION Our multi-center study demonstrated that among patients receiving ICBs therapy, those with elevated serum magnesium levels exhibit significantly better clinical outcomes than those with low serum magnesium levels. Further prospective validation studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Yingfang Feng
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Meng Gao
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China; The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010030, China
| | - Xiyue Xu
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Hengqi Liu
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Ke Lu
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China; Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Zheng Song
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Jingwei Yu
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Xia Liu
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Xue Han
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Lanfang Li
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Lihua Qiu
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Zhengzi Qian
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Shiyong Zhou
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Huilai Zhang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China.
| | - Xianhuo Wang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine / Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China.
| |
Collapse
|