1
|
Xu JX, Su YX, Chen YY, Huang YY, Chen ZS, Peng YC, Qi LN. Immune infiltration landscape and potential drug-targeted implications for hepatocellular carcinoma with 'progression/hyper-progression' recurrence. Ann Med 2025; 57:2456113. [PMID: 39865865 PMCID: PMC11774162 DOI: 10.1080/07853890.2025.2456113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 12/20/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND AND AIMS Hepatocellular carcinoma (HCC) recurrence was previously characterized into four types, and patients with progression/hyper-progression recurrence (type III-IV) have an extremely poor prognosis. However, the immune background of resectable HCC, particularly in patients who experience recurrence, remains underexplored. Therefore, this study aimed to describe the immune landscape of resectable HCC, especially postoperative type III-IV recurrent HCC, and explore potential immune-targeted anti-relapse strategies for treated populations. METHODS The differences in gene expression in patients with recurrent HCC (type I-II (solitary or multi-intrahepatic oligo recurrence) vs. type III-IV) were investigated using bulk sequencing. Multiple immune infiltration methods (single-sample gene set enrichment analysis (GSEA), Microenvironment Cell Populations-counter and ESTIMATE) were used, and patients were divided into four groups to identify four distinct immune subtypes: immune-enrichment/matrix-poor (IE1), immune-enrichment/matrix-rich (IE2), immune intermediate/matrix-rich (ITM) and immune desert/matrix-poor (ID). Co-expression and protein interaction analyses were used to identify characteristic genes in ITM closely associated with type III-IV recurrence, which was matched with drug targets for Huaier granules (HG) and lenvatinib. Virtual docking was used to identify potential therapeutic targets, and the results were verified using single-nuclei RNA sequencing and histological analysis. RESULTS ITM was closely related to type III-IV recurrence and exhibited immunotherapy potential. The potential efficacy of inhibiting CCNA2, VEGFA, CXCL8, PLK2, TIMP1, ITGB2, ALDOA, ANXA5 and CSK in ITM reversal was determined. Molecular docking demonstrated that the proteins of these genes could bind to HG or lenvatinib. The immunohistochemical findings demonstrated differential VEGFA (p < .01) and PLK2 (p < .001) expression in ITM type and ID in type III-IV recurrent HCC. CONCLUSIONS Three primary immunotypes of resectable HCC (IE2, ITM and ID) were identified, and HG and lenvatinib could potentially overcome immune checkpoint blockade (ICB) resistance in ITM patients with HCC, particularly those classified as type III-IV.
Collapse
Affiliation(s)
- Jing-Xuan Xu
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumour, Ministry of Education, Nanning, China
| | - Yue-Xiang Su
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumour, Ministry of Education, Nanning, China
| | - Yuan-Yuan Chen
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yi-Yue Huang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumour, Ministry of Education, Nanning, China
| | - Zu-Shun Chen
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yu-Chong Peng
- Department of General Surgery, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Lu-Nan Qi
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumour, Ministry of Education, Nanning, China
- Guangxi Liver Cancer Diagnosis and Treatment Engineering and Technology Research Center, Nanning, China
| |
Collapse
|
2
|
Bick F, Blanchetot C, Lambrecht BN, Schuijs MJ. Targeting γc family cytokines with biologics: current status and future prospects. MAbs 2025; 17:2468312. [PMID: 39967341 PMCID: PMC11845063 DOI: 10.1080/19420862.2025.2468312] [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: 11/12/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
Over the recent decades the market potential of biologics has substantially expanded, and many of the top-selling drugs worldwide are now monoclonal antibodies or antibody-like molecules. The common gamma chain (γc) cytokines, Interleukin (IL-)2, IL-4, IL-7, IL-9, IL-15, and IL-21, play pivotal roles in regulating immune responses, from innate to adaptive immunity. Dysregulation of cell signaling by these cytokines is strongly associated with a range of immunological disorders, which includes cancer as well as autoimmune and inflammatory diseases. Given the essential role of γc cytokines in maintaining immune homeostasis, the development of therapeutic interventions targeting these molecules poses unique challenges. Here, we provide an overview of current biologics targeting either single or multiple γc cytokines or their respective receptor subunits across a spectrum of diseases, primarily focusing on antibodies, antibody-like constructs, and antibody-cytokine fusions. We summarize therapeutic biologics currently in clinical trials, highlighting how they may offer advantages over existing therapies and standard of care, and discuss recent advances in this field. Finally, we explore future directions and the potential of novel therapeutic intervention strategies targeting this cytokine family.
Collapse
Affiliation(s)
- Fabian Bick
- Argenx BV, Zwijnaarde, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | | | - Bart N. Lambrecht
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Martijn J. Schuijs
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
3
|
Reader CS, Liao W, Potter-Landua BJ, Veyssier CS, Seal CJ, Brewis N, Morrow M. The tetravalent, bispecific properties of FS118, an anti-LAG-3/PD-L1 antibody, mediate LAG-3 shedding from CD4 + and CD8 + tumor-infiltrating lymphocytes. Anticancer Drugs 2025; 36:447-458. [PMID: 39960386 PMCID: PMC12061380 DOI: 10.1097/cad.0000000000001705] [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: 11/26/2024] [Accepted: 12/06/2024] [Indexed: 03/01/2025]
Abstract
Tumor-infiltrating lymphocytes (TILs) often have upregulated expression of immune checkpoint receptors, such as programmed cell death 1 (PD-1) and lymphocyte-activation gene 3 (LAG-3). Patients treated with antibodies targeting PD-1 or its ligand (PD-L1) can develop resistance or relapse, with LAG-3 upregulation on T cells being one possible mechanism. FS118 is a tetravalent, bispecific antibody comprising a full-length IgG 1 anti-PD-L1 antibody with bivalent LAG-3-binding capability in the fragment crystallizable region. Here we demonstrate how the structure of FS118 is important for its function. We generated variants of FS118 and tested their ability to mediate LAG-3 shedding using staphylococcal enterotoxin B assays, antigen recall assays, and soluble LAG-3 ELISAs. Mediated by metalloproteases ADAM10 and ADAM17, FS118 induced shedding of LAG-3 from the surface of both CD4 + and CD8 + T cells. We also determined the effect of surrogate antibodies on immune cell LAG-3 expression and proliferation in syngeneic mouse models. In vivo , the bivalent LAG-3 binding sites of a mouse surrogate of FS118 and their location in the fragment crystallizable region were important for eliciting maximal reduction in LAG-3 levels on the surface of TILs, as variants with a single LAG-3 binding site in the fragment crystallizable region, or with reversed orientation of the LAG-3 and PD-L1 binding sites, were less efficient at inducing shedding. We also show that PD-L1, not PD-1, binding drives the LAG-3 reduction on TILs. We hypothesize that the LAG-3 bivalency in the fragment crystallizable region of FS118 allows LAG-3 clustering, which optimizes cleavage by ADAM10/ADAM17 and thus shedding.
Collapse
|
4
|
Sultan MH, Zhan Q, Wang Y, Xia Y, Jia X. Precision oncolytic viral therapy in colorectal cancer: Genetic targeting and immune modulation for personalized treatment (Review). Int J Mol Med 2025; 56:104. [PMID: 40342021 PMCID: PMC12081034 DOI: 10.3892/ijmm.2025.5545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/09/2025] [Indexed: 05/11/2025] Open
Abstract
Colorectal cancer (CRC) is a leading health issue and treatments to eradicate it, such as conventional chemotherapy, are non‑selective and come with a number of complications. The present review focuses on the relatively new area of precision oncolytic viral therapy (OVT), with genetic targeting and immune modifications that offer a new future for CRC treatment. In the present review, an overview of the selection factors that are considered optimal for an oncolytic virus, mechanisms of oncolysis and immunomodulation applied to the OVT, as well as new strategies to improve the efficacy of this method are described. Additionally, cause‑and‑effect relationships are examined for OVT efficacy, mediated by the tumor microenvironment, and directions for genetic manipulation of viral specificity are explored. The possibility of synergy between OVT and immune checkpoint inhibitors and other treatment approaches are demonstrated. Incorporating the details of the present review, biomarker‑guided combination therapies in precision OVT for individualized CRC care, significant issues and future trends in this required area of medicine are highlighted. Increasingly, OVT is leaving the experimental stage and may become routine practice; it provides a new perspective on overcoming CRC and highlights the importance of further research and clinical work.
Collapse
Affiliation(s)
- Muhammad Haris Sultan
- College of Life Sciences and Medicine, Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
- Center for Translational Medicine and Precision Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, P.R. China
| | - Qi Zhan
- College of Life Sciences and Medicine, Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Yigang Wang
- College of Life Sciences and Medicine, Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Yulong Xia
- Center for Translational Medicine and Precision Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiaoyuan Jia
- College of Life Sciences and Medicine, Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| |
Collapse
|
5
|
Lee J, McClure S, Weichselbaum RR, Mimee M. Designing live bacterial therapeutics for cancer. Adv Drug Deliv Rev 2025; 221:115579. [PMID: 40228606 PMCID: PMC12067981 DOI: 10.1016/j.addr.2025.115579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Humans are home to a diverse community of bacteria, many of which form symbiotic relationships with their host. Notably, tumors can also harbor their own unique bacterial populations that can influence tumor growth and progression. These bacteria, which selectively colonize hypoxic and acidic tumor microenvironments, present a novel therapeutic strategy to combat cancer. Advancements in synthetic biology enable us to safely and efficiently program therapeutic drug production in bacteria, further enhancing their potential. This review provides a comprehensive guide to utilizing bacteria for cancer treatment. We discuss key considerations for selecting bacterial strains, emphasizing their colonization efficiency, the delicate balance between safety and anti-tumor efficacy, and the availability of tools for genetic engineering. We also delve into strategies for precise spatiotemporal control of drug delivery to minimize adverse effects and maximize therapeutic impact, exploring recent examples of engineered bacteria designed to combat tumors. Finally, we address the underlying challenges and future prospects of bacterial cancer therapy. This review underscores the versatility of bacterial therapies and outlines strategies to fully harness their potential in the fight against cancer.
Collapse
Affiliation(s)
- Jaehyun Lee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA
| | - Sandra McClure
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago 60637, USA; The Ludwig Center for Metastasis Research, University of Chicago, Chicago 60637, USA
| | - Mark Mimee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA; Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
6
|
Sun L, Liu Y, Sun Q, Wang G, Du B, Liu B, Gao T, Zhao P, Yang Y, Rong R. Polysaccharides from traditional Chinese medicine and their nano-formulated delivery systems for cancer immunotherapy. Carbohydr Polym 2025; 357:123416. [PMID: 40158963 DOI: 10.1016/j.carbpol.2025.123416] [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: 11/25/2024] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 04/02/2025]
Abstract
Cancer immunotherapy has evolved into a new generation strategy in the field of anti-tumor treatment. Polysaccharides derived from Traditional Chinese Medicine (TCM) are gaining recognition as powerful immunomodulators in cancer therapy, noted for their multi-target and multi-pathway actions. Owing to their beneficial properties such as water solubility, biocompatibility, and chemical structure modifiability, TCM polysaccharides can also serve as carriers for hydrophobic drugs in the development of innovative drug delivery systems, enhancing synergistic antitumor effects. In this article, we summarize the diverse mechanisms of immunoregulation by TCM polysaccharides in tumor therapy. The applications of these polysaccharides as both active ingredients and drug carriers within nanodelivery systems for cancer immunotherapy are also introduced. Additionally, extensive research on TCM polysaccharides in clinical settings has been collected. Furthermore, discussions are presented on the development prospects and challenges faced by these polysaccharides in the field of tumor immunotherapy. Our goal is to improve researchers' comprehension of TCM polysaccharides in cancer immunotherapy, providing promising strategies to optimize cancer treatment and benefit diverse patient populations.
Collapse
Affiliation(s)
- Linlin Sun
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yuting Liu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Qihui Sun
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Guimei Wang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Baoxiang Du
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Bodong Liu
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Tian Gao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Pan Zhao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yong Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China; Collaborative Innovation Center for Antiviral Traditional Chinese Medicine in Shandong Province, Jinan 250355, PR China; Shandong Antiviral Engineering Research Center of Traditional Chinese Medicine, Jinan 250355, PR China.
| | - Rong Rong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| |
Collapse
|
7
|
Zhang Y, Yan Y, Liu J, Xia H, Zhou J, Cui Y, Huang X, Chang J, Zhang W, Chen W, Zhang Q, Wang S, Wang Y, Chen B. An Endoplasmic Reticulum Stress-Specific Nanoinducer Selectively Evokes Type-II Immunogenic Cell Death for Pyroptotic Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2501953. [PMID: 40434207 DOI: 10.1002/adma.202501953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 05/15/2025] [Indexed: 05/29/2025]
Abstract
Specific induction of endoplasmic reticulum (ER) stress-initiated type-II immunogenic cell death (ICD) shows great potential in boosting tumor immunogenicity and anti-tumor immunotherapy. However, it remains challenging to selectively provoke type-II ICD, due to the lack of highly efficient ER targeting strategy. Here, a pH/Cathepsin-Activatable Nanoplatform (PCAN) is reported to specifically photo-induce ER stress (PCANER) and type-II ICD for cancer immunotherapy. PCANER integrates the long-circulating properties of nanomedicines with pH/cathepsin B dual-gated design, exhibiting excellent ER targeting with a colocalization efficacy of 83% in cancer tissues. Through directly intensifying glucose-regulated protein 78 and calreticulin exposure, PCANER augments type-II ICD and pyroptotic cancer cell death with high immune priming to cascade-amplify the cancer-immunity cycle, while the mild type-I ICD induced by lysosome stress (PCANLy) exhibits negligible antitumor efficacy. By leveraging the spatiotemporal subcellular organelle targeting of PCAN technology, this study achieves precise tuning of the type of ICD and cellular pyroptosis-based cancer therapy. This study offers new insights into the design of organelle level-targeted nanomedicines, paving the way for dissecting and modulating the cell death mechanism to boost cancer immunotherapy.
Collapse
Affiliation(s)
- Yimeng Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yue Yan
- Department of Central Laboratory, Peking University First Hospital, Beijing, 100034, China
| | - Jianxiong Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Heming Xia
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jiayi Zhou
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yi Cui
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xinyu Huang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jian Chang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Weiwei Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Wei Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Chemical Biology Center, Peking University, Beijing, China
- Ningbo Institute of Marine Medicine, Peking University, China
| | - Binlong Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| |
Collapse
|
8
|
Wang Y, Jin RU, Xu J, Lin DC, Sun Z, Xu Y, Li QK, Zhang H. Harnessing technologies to unravel gastric cancer heterogeneity. Trends Cancer 2025:S2405-8033(25)00107-4. [PMID: 40425443 DOI: 10.1016/j.trecan.2025.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 05/29/2025]
Abstract
Gastric cancer arises from complex carcinogenic factor interactions, with limited treatment options due to the lack of targetable driver gene mutations and significant tumor heterogeneity. Recent studies have provided promising novel approaches to improve our understanding of gastric cancer heterogeneity through integrated characterization, combining genomics with emerging technologies. Delineating the molecular changes and targeting specific molecular subtypes will enhance the efficacy of gastric cancer treatment and improve clinical outcomes. This review provides a comprehensive overview of current technologies used in gastric cancer research, highlighting key discoveries and treatment strategies driven by these innovations. Finally, we discuss the emerging technology-guided directions and potential breakthroughs that could enhance the understanding of gastric cancer tumor heterogeneity, ultimately improving clinical outcomes.
Collapse
Affiliation(s)
- Yuefan Wang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Ramon U Jin
- Division of Oncology and Gastroenterology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Joanne Xu
- College of Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Ding Chiao Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Zhenyu Sun
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yuanwei Xu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Qing K Li
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Hui Zhang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
9
|
Zhong X, Han J, Li H, Shen X, Yu B, Chen T, Li H, Li J, Pang J, Qian L, Wu W, Tong X, Ding B. Glycosylated protein-related microenvironmental features in breast cancer are associated with patient prognosis. Mamm Genome 2025:10.1007/s00335-025-10137-9. [PMID: 40411577 DOI: 10.1007/s00335-025-10137-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Accepted: 05/10/2025] [Indexed: 05/26/2025]
Abstract
The tumor microenvironment (TME) and aberrant glycosylation have been suggested to play key roles in cancer. This study integrated differentially expressed genes (DEGs) and weighted gene coexpression network analysis (WGCNA) to identify tumor microenvironment-related genes and construct a TME-risk prognostic signature (TMERS) through LASSO Cox regression. After batch effect removal, 44 TME-prognosis-related genes (TMEPGs) were identified and classified into three molecular subtypes via K-means clustering. The finalized 22-gene TMERS model demonstrated robust prognostic predictive capacity in GEO datasets. The results revealed distinct immune profiles and prognostic stratifications among genetic subtypes and risk groups, confirming that the TMERS is an independent prognostic indicator for breast cancer (BRCA). Glycosyltransferase genes (GTs) have potential therapeutic relevance through immune regulation, with TMEPG member killer cell lectin like receptor B1 (KLRB1) significantly correlated with BRCA prognosis. Cellular experiments demonstrated that KLRB1 overexpression suppressed BRCA cell proliferation and migration. This work establishes a novel prognostic model for BRCA while highlighting KLRB1 as a potential biomarker, providing new insights into TME-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoxiao Zhong
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
| | - Jiaxuan Han
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Huan Li
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Xiangyu Shen
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Bowen Yu
- Department of Gastrointestinal Surgery, Third XiangYa Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Ting Chen
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Haobing Li
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421200, Hunan, China
| | - Jun Li
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Jin Pang
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Liyuan Qian
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Wei Wu
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Xiaoliang Tong
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, 410000, China.
| | - Boni Ding
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
| |
Collapse
|
10
|
Wang M, Liu H, Huang J, Cai T, Xu ZP, Zhang L. Advancing cancer gene therapy: the emerging role of nanoparticle delivery systems. J Nanobiotechnology 2025; 23:362. [PMID: 40394591 PMCID: PMC12090605 DOI: 10.1186/s12951-025-03433-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 05/01/2025] [Indexed: 05/22/2025] Open
Abstract
Gene therapy holds immense potential due to its ability to precisely target oncogenes, making it a promising strategy for cancer treatment. Advances in genetic science and bioinformatics have expanded the applications of gene delivery technologies beyond detection and diagnosis to potential therapeutic interventions. However, traditional gene therapy faces significant challenges, including limited therapeutic efficacy and the rapid degradation of genetic materials in vivo. To address these limitations, multifunctional nanoparticles have been engineered to encapsulate and protect genetic materials, enhancing their stability and therapeutic effectiveness. Nanoparticles are being extensively explored for their ability to deliver various genetic payloads-including plasmid DNA, messenger RNA, and small interfering RNA-directly to cancer cells. This review highlights key gene modulation strategies such as RNA interference, gene editing systems, and chimeric antigen receptor (CAR) technologies, alongside a diverse array of nanoscale delivery systems composed of polymers, lipids, and inorganic materials. These nanoparticle-based delivery platforms aim to improve targeted transport of genetic material into cancer cells, ultimately enhancing the efficacy of cancer therapies.
Collapse
Affiliation(s)
- Maoze Wang
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, 315040, China
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518107, China
| | - Huina Liu
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, 315040, China
| | - Jinling Huang
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518107, China
- School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Ting Cai
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, 315040, China.
| | - Zhi Ping Xu
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, 315040, China.
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518107, China.
- School of Medicine, Hangzhou City University, Hangzhou, 310015, China.
| | - Lingxiao Zhang
- Interdisciplinary Nanoscience Center (INANO), Aarhus University, Aarhus C, DK-8000, Denmark.
| |
Collapse
|
11
|
Dey NS, Dey S, Brown N, Senarathne S, Campos Reis L, Sengupta R, Lindoso JA, James SR, Gilbert L, Boucher D, Chatterjee M, Goto H, Ranasinghe S, Kaye PM. IL-32-producing CD8+ memory T cells define immunoregulatory niches in human cutaneous leishmaniasis. J Clin Invest 2025; 135:e182040. [PMID: 40371647 PMCID: PMC12077899 DOI: 10.1172/jci182040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 03/12/2025] [Indexed: 05/16/2025] Open
Abstract
Human cutaneous leishmaniasis (CL) is characterized by chronic skin pathology. Experimental and clinical data suggest that immune checkpoints (ICs) play a crucial role in disease outcome, but the cellular and molecular niches that facilitate IC molecule expression during leishmaniasis are ill defined. In Sri Lankan patients with CL, indoleamine 2,3-dioxygenase 1 (IDO1) and programmed death-ligand 1 (PD-L1) were enriched in skin lesions, and reduced PD-L1 expression early after treatment initiation was predictive of a cure rate following antimonial therapy. Here, we used spatial cell interaction mapping to identify IL-32-expressing CD8+ memory T cells and Tregs as key components of the IDO1/PD-L1 niche in Sri Lankan patients with CL and in patients with distinct forms of dermal leishmaniasis in Brazil and India. Furthermore, the abundance of IL-32+ cells and IL-32+CD8+ T cells at treatment initiation was negatively correlated with the rate of cure in Sri Lankan patients. This study provides insights into the spatial mechanisms underpinning IC expression during CL and offers a strategy for identifying additional biomarkers of treatment response.
Collapse
Affiliation(s)
- Nidhi S. Dey
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Shoumit Dey
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Naj Brown
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Sujai Senarathne
- Department of Parasitology, Faculty of Medical Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | - Luiza Campos Reis
- Department of Preventive Medicine, Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ritika Sengupta
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| | - Jose A.L. Lindoso
- Secretaria de Saúde do Estado de São Paulo, Instituto de Infectologia Emílio Ribas, São Paulo, Brazil
- University of São Paulo, Faculty of Medicine, Department of Infectious and Parasitic Diseases, São Paulo, Brazil
| | - Sally R. James
- Technology Facility, Department of Biology, University of York, York, United Kingdom
| | - Lesley Gilbert
- Technology Facility, Department of Biology, University of York, York, United Kingdom
| | - Dave Boucher
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Mitali Chatterjee
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| | - Hiro Goto
- Department of Preventive Medicine, Instituto de Medicina Tropical de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Shalindra Ranasinghe
- Department of Parasitology, Faculty of Medical Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | - Paul M. Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| |
Collapse
|
12
|
Huang R, Wang Y, Teng H, Xu M, He K, Shen Y, Guo G, Feng X, Li T, Zhou B, Bajenoff M, Lawrence T, Liang Y, Lu L, Zhang L. Tyrosinase in melanoma inhibits anti-tumor activity of PD-1 deficient T cells. BMC Biol 2025; 23:135. [PMID: 40375241 PMCID: PMC12083179 DOI: 10.1186/s12915-025-02237-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Melanoma is one of the most commonly diagnosed malignancies and serves as a model for studying immunotherapy. The B16 melanoma model, resembling human cold tumors that lack T cell infiltration and show minimal response to PD-1 blockade, is widely used for studying melanoma and its resistance to immunotherapy. Therefore, understanding the molecular basis that prevents T cell-mediated anti-tumor activity in B16 melanoma is of great significance. RESULTS In this study, we generated tyrosinase knockout B16 melanoma cells using CRISPR/Cas9 and discovered that tyrosinase in melanoma significantly inhibits the anti-tumor activity of T cells. Tyrosinase deficiency leads to a 3.80-fold increase in T-cell infiltration and enhances T-cell activation within the tumor. Single-cell RNA sequencing reveals an altered cold tumor immunophenotype in tyrosinase-deficient B16 melanoma. In wild-type mice, T cells in tyrosinase-deficient tumors express elevated levels of PD-1 and Foxp3. However, strikingly, in PD-1 deficient mice, the loss of tyrosinase in B16 melanoma unleashes the anti-tumor activity of PD-1 deficient T cells. This enhanced anti-tumor activity is explained by significantly increased tumor T cell infiltration accompanied by reduced frequencies of regulatory T cells in PD-1 knockout mice. CONCLUSIONS These findings suggest that targeting tyrosinase could convert cold tumors into an immune-responsive state in vivo using murine models. Inhibiting tyrosinase could enhance the effectiveness of PD-1 blockade, offering a new approach for melanoma patients who fail in current PD-1 inhibitor treatment.
Collapse
Affiliation(s)
- Rong Huang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China.
| | - Yingbin Wang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Haitao Teng
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Mengjun Xu
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Kexin He
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Yingzhuo Shen
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Guo Guo
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
- Centre d'immunologie de Marseille-Luminy, Aix-Marseille University, CNRS, INSERM, Marseille, France
| | - Xinyu Feng
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Tianhan Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Binhui Zhou
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
| | - Marc Bajenoff
- Centre d'immunologie de Marseille-Luminy, Aix-Marseille University, CNRS, INSERM, Marseille, France
| | - Toby Lawrence
- Centre d'immunologie de Marseille-Luminy, Aix-Marseille University, CNRS, INSERM, Marseille, France
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Yinming Liang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China.
- Center of Disease Model and Immunology, Hunan Academy of Chinese Medicine, Changsha, China.
| | - Liaoxun Lu
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China.
| | - Lichen Zhang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China.
- Center of Disease Model and Immunology, Hunan Academy of Chinese Medicine, Changsha, China.
| |
Collapse
|
13
|
Zhang F, Ramar S, Wang Y, Xu H, Zhang K, Awadasseid A, Rao G, Zhang W. Advances in cancer immunotherapy using small-molecular inhibitors targeting the PD-1/PD-L1 interaction. Bioorg Med Chem 2025; 127:118238. [PMID: 40367914 DOI: 10.1016/j.bmc.2025.118238] [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: 01/20/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
Cancer cells evade immune responses by interacting with PD-1 and its ligand, PD-L1. Although monoclonal antibodies targeting this pathway have revolutionized oncology, their high production costs, poor oral bioavailability, and limited tumor penetration remain significant challenges. Small-molecule inhibitors provide a promising alternative, offering advantages such as improved tumor penetration and cost-effectiveness. This review highlights advancements in small-molecule PD-1/PD-L1 inhibitors, focusing on their mechanisms, structural designs, and therapeutic potential. Key innovations, including biphenyl scaffolds, heterocyclic frameworks, enhance binding efficiency and immune activation. The article effectively integrates fundamental principles of drug chemistry with real-world clinical needs, offering a comprehensive approach to the design of PD-1/PD-L1 small-molecule inhibitors. It systematically classifies various molecular structures, analyzes relevant industrial cases, and incorporates the most recent research findings. By examining these aspects, it uncovers the underlying logic driving the design process and provides a fresh, innovative perspective on advancing the field of immune small-molecule inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Feng Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China
| | - Sivaramakarthikeyan Ramar
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China; Moganshan Institute ZJUT, Deqing 313202, China
| | - Yu Wang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China
| | - Haoran Xu
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China
| | - Koutian Zhang
- Zhejiang Qingzhenghong Technology Co., Ltd, Hangzhou 311121, China
| | - Annoor Awadasseid
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China; Zhejiang Qingzhenghong Technology Co., Ltd, Hangzhou 311121, China.
| | - Guowu Rao
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China.
| | - Wen Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China; Zhejiang Jieyuan Med-Tech Co., Ltd., Hangzhou 311113, China.
| |
Collapse
|
14
|
Zhang Y, Zhou J, Wang Y, Wu Y, Li Y, Wang B, Liu G, Gong Q, Luo K, Jing J. Stimuli-responsive polymer-dasatinib prodrug to reprogram cancer-associated fibroblasts for boosted immunotherapy. J Control Release 2025; 381:113606. [PMID: 40054628 DOI: 10.1016/j.jconrel.2025.113606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025]
Abstract
The barriers from cancer-associated fibroblasts (CAFs) have diminished the clinical efficacy of immunotherapy for triple-negative breast cancer (TNBC). The obstacles from CAFs often result in poor drug penetration, constrained cytotoxic T lymphocyte infiltration, and an immunosuppressive microenvironment. Herein, chondroitin sulfate (CS) was engineered to conjugate dasatinib (DAS), a tyrosine kinase inhibitor, via the cathepsin B (CTSB)-responsive GFLG linker to produce CS-GFLG-DAS (CGD), which could be employed to reverse the CAF phenotype and regulate the biosynthesis of extracellular matrix (ECM), thus enhancing the efficacy of immune checkpoint blockade (ICB) therapy. Upon reaching the tumor site, DAS released from CGD in response to overexpressed CTSB in the tumor microenvironment could transform CAFs into a quiescent state instead of killing them to prevent CAFs from producing abundant ECM, thereby promoting deep penetration of CGD to effectively kill tumor cells. In addition, ECM remodeling facilitated tumor infiltration of cytotoxic T lymphocytes, synergistically enhancing the anti-PD-1 efficacy in the 4T1 tumor-bearing mice. In summary, this prodrug enhanced deep drug penetration and therapeutic sensitivity of anti-PD-1 by regulating CAFs, providing new insights into optimizing immunotherapy in treating fibrotic tumors via nanomedicine.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Zhou
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yiyan Wang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaping Wu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yunkun Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bing Wang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guohao Liu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, NHC Key Laboratory of Transplant Engineering and Immunology, Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China; Xiamen Key Lab of Psychoradiology and Neuromodulation, Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen 361021, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, NHC Key Laboratory of Transplant Engineering and Immunology, Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China.
| | - Jing Jing
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
15
|
Liu Q, Xue J, Ouyang W, Sheng J, He L. Toripalimab-Associated Arthritis and Peripheral Neuropathy. Am J Ther 2025:00045391-990000000-00303. [PMID: 40366363 DOI: 10.1097/mjt.0000000000001846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Affiliation(s)
- Qingru Liu
- Department of Pharmacy, Xiangya Changde Hospital, Changde, Hunan, China
| | - Jiao Xue
- Department of Endocrinology, Xiangya Changde Hospital, Changde, Hunan, China
| | - Wenjuan Ouyang
- Department of Pharmacy, Xiangya Changde Hospital, Changde, Hunan, China
| | - Ju Sheng
- Department of Endocrinology, Xiangya Changde Hospital, Changde, Hunan, China
| | - Leiyan He
- Department of Pharmacy, Xiangya Changde Hospital, Changde, Hunan, China
| |
Collapse
|
16
|
Cai L, Chen H, Wang Y, Zhang J, Song D, Tan Y, Guo Z, Wang X. Platinum(IV) Complexes Trigger Death Receptors and Natural Killer Cells to Suppress Breast Cancer. J Med Chem 2025; 68:9162-9175. [PMID: 39886904 DOI: 10.1021/acs.jmedchem.4c02509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Chemoimmunotherapy is an alternative treatment against cancers. Platinum(IV) complexes FMP and DFMP, coupling formononetin derivative as axial ligand(s), were designed to suppress triple-negative breast cancer (TNBC) by activating death receptors (DRs) and natural killer (NK) cells. These complexes show great potential to overcome the resistance of TNBC to chemotherapy by inducing both intrinsic and extrinsic apoptosis in cancer cells. Particularly, FMP with one axial formononetin derivative not only induced the caspase-3-dependent intrinsic apoptosis but also upregulated the expression of DRs and caspase-8, triggered the extrinsic apoptosis, and enhanced the cytotoxic ability of NK92 cells. Moreover, FMP increased the release of granzyme B, restrained the proliferation and differentiation of myeloid-derived suppressor cells, and the secretion of IL-10, thus inhibiting the TNBC in vitro and in vivo. The results demonstrate that FMP overcomes the chemoresistance and immune escape of TNBC through a new mechanism involving the synergy of chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Linxiang Cai
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Hanhua Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Ying Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Jingwen Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Dongfan Song
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Yehong Tan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
17
|
Jünemann W, Bley I, Rekowski L, Klokow M, Herppich S, Müller I, Cornils K. GD2-CAR NK-92 cell activity against neuroblastoma cells is insusceptible to TIGIT knockout. Cancer Immunol Immunother 2025; 74:191. [PMID: 40317320 PMCID: PMC12049354 DOI: 10.1007/s00262-025-04010-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/03/2025] [Indexed: 05/07/2025]
Abstract
Immunotherapy by inhibition of immune checkpoint (IC) molecules has emerged as an important cancer therapy. Among these lC, the poliovirus receptor/poliovirus receptor-like 2 protein (PVR/PVRL2)-TIGIT axis was discovered as potential target for various cancers. For neuroblastoma (NB), the most common extracranial solid cancer in children, no effective IC therapy has been established yet. To investigate the PVR/PVRL2-TIGIT IC axis as a new target for the treatment of NB, we analysed whether PVR and PVRL2 influence the survival of patients and verified the expression of the receptors on NB cell lines. To disrupt the checkpoint axis, we performed single and double knockouts of these receptors on NB cell lines and subsequently removed TIGIT, an inhibitory receptor on immune effector cells, from NK-92 cells. Finally, we combined checkpoint inhibition with GD2-CAR NK-92 cells and investigated changes in cytotoxicity. Using RNA-Seq data we showed that the expression of PVR and PVRL2 on NB cells correlates to a lower event-free survival of patients. CRISPR/Cas9 knockouts of PVR and PVRL2 showed no improved cytotoxic activity of NK-92 cells. We observed enhanced lysis of NB cells using TIGIT-deficient NK-92 cells. However, the cytotoxicity of GD2-CAR NK-92 was not significantly enhanced. In summary, we have shown that in addition to the interaction of PVR/PVRL2 and TIGIT on engineered immune effector cells against NB, pleiotropic ligands appear to be relevant. Deletion of TIGIT from immune effector cells is a promising approach to protect these cells from tumour-associated inhibitory signals but cannot enhance the effect of GD2-CAR-NK-92 cells.
Collapse
MESH Headings
- Humans
- Neuroblastoma/immunology
- Neuroblastoma/therapy
- Neuroblastoma/pathology
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/immunology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Gangliosides/immunology
- Cell Line, Tumor
- Immunotherapy, Adoptive/methods
- Gene Knockout Techniques
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Virus/genetics
Collapse
Affiliation(s)
- Wiebke Jünemann
- Children's Cancer Centre Research Institute Hamburg, Hamburg, Germany
- Division of Paediatric Stem Cell Transplantation and Immunology, Department of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Isabelle Bley
- Children's Cancer Centre Research Institute Hamburg, Hamburg, Germany
- Division of Paediatric Stem Cell Transplantation and Immunology, Department of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Laura Rekowski
- Children's Cancer Centre Research Institute Hamburg, Hamburg, Germany
- Division of Paediatric Stem Cell Transplantation and Immunology, Department of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Marianne Klokow
- Children's Cancer Centre Research Institute Hamburg, Hamburg, Germany
- Division of Paediatric Stem Cell Transplantation and Immunology, Department of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Susanne Herppich
- Children's Cancer Centre Research Institute Hamburg, Hamburg, Germany
- Division of Paediatric Stem Cell Transplantation and Immunology, Department of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Ingo Müller
- Children's Cancer Centre Research Institute Hamburg, Hamburg, Germany
- Division of Paediatric Stem Cell Transplantation and Immunology, Department of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Kerstin Cornils
- Children's Cancer Centre Research Institute Hamburg, Hamburg, Germany.
- Division of Paediatric Stem Cell Transplantation and Immunology, Department of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
18
|
Tong J, Tan Y, Ouyang W, Chang H. Targeting immune checkpoints in hepatocellular carcinoma therapy: toward combination strategies with curative potential. Exp Hematol Oncol 2025; 14:65. [PMID: 40317077 PMCID: PMC12046748 DOI: 10.1186/s40164-025-00636-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/07/2025] [Indexed: 05/04/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a primary liver cancer characterized by poor immune cell infiltration and a strongly immunosuppressive microenvironment. Traditional treatments have often yielded unsatisfactory outcomes due to the insidious onset of the disease. Encouragingly, the introduction of immune checkpoint inhibitors (ICIs) has significantly transformed the approach to HCC treatment. Moreover, combining ICIs with other therapies or novel materials is considered the most promising opportunity in HCC, with some of these combinations already being evaluated in large-scale clinical trials. Unfortunately, most clinical trials fail to meet their endpoints, and the few successful ones also face challenges. This indicates that the potential of ICIs in HCC treatment remains underutilized, prompting a reevaluation of this promising therapy. Therefore, this article provides a review of the role of immune checkpoints in cancer treatment, the research progress of ICIs and their combination application in the treatment of HCC, aiming to open up avenues for the development of safer and more efficient immune checkpoint-related strategies for HCC treatment.
Collapse
Affiliation(s)
- Jing Tong
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
| | - Yongci Tan
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
| | - Wenwen Ouyang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
| | - Haocai Chang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
19
|
Appleman VA, Matsuda A, Ganno ML, Zhang DM, Rosentrater E, Maldonado Lopez AE, Porciuncula A, Hatten T, Christensen CL, Merrigan SA, Lee HM, Lee MY, Wang CI, Dong L, Huang J, Iartchouk N, Wang J, Xu H, Yoneyama T, Piatkov KI, Haridas S, Harbison CE, Gregory RC, Parent A, Lineberry N, Arendt C, Schalper KA, Abu-Yousif AO. Selective STING Activation in Intratumoral Myeloid Cells via CCR2-Directed Antibody-Drug Conjugate TAK-500. Cancer Immunol Res 2025; 13:661-679. [PMID: 39918395 PMCID: PMC12046323 DOI: 10.1158/2326-6066.cir-24-0103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 08/06/2024] [Accepted: 02/05/2025] [Indexed: 05/03/2025]
Abstract
The tumor microenvironment in solid tumors contains myeloid cells that modulate local immune activity. Stimulator of IFN gene (STING) signaling activation in these myeloid cells enhances local type-I IFN production, inducing an innate immune response that mobilizes adaptive immunity and reprograms immunosuppressive myeloid populations to drive antitumor immunity. In this study, we generated TAK-500, an immune cell-directed antibody-drug conjugate, to deliver a STING agonist to CCR2+ human cells and drive enhanced antitumor activity relative to nontargeted STING agonists. Preclinically, TAK-500 triggered dose-dependent innate immune activation in vitro. In addition, a murine TAK-500 immune cell-directed antibody-drug conjugate surrogate enhanced innate and adaptive immune responses both in in vitro and murine tumor models. Spatially resolved analysis of CCR2 and immune cell markers in the tumor microenvironment of >1,000 primary human tumors showed that the CCR2 protein was predominantly expressed in intratumoral myeloid cells. Collectively, these data highlight the clinical potential of delivering a STING agonist to CCR2+ cells.
Collapse
Affiliation(s)
- Vicky A. Appleman
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Atsushi Matsuda
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Michelle L. Ganno
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Dong Mei Zhang
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Emily Rosentrater
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Angel E. Maldonado Lopez
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Angelo Porciuncula
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Tiquella Hatten
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Camilla L. Christensen
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Samantha A. Merrigan
- Translational Sciences, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Hong Myung Lee
- Drug Discovery Science, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Min Young Lee
- Oncology Cell Therapy and Therapeutic Area Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Charlotte I. Wang
- Clinical Science, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Linlin Dong
- Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Jian Huang
- Global Biologics, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Natasha Iartchouk
- OTAU/PTM, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | | | - He Xu
- Drug Discovery Science, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Tomoki Yoneyama
- Quantitative Solutions, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Konstantin I. Piatkov
- Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | | | - Carole E. Harbison
- DSRE, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Richard C. Gregory
- Precision and Translational Medicine, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Alexander Parent
- OTAU, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Neil Lineberry
- Department of Oncology, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Chris Arendt
- Oncology Cell Therapy and Therapeutic Area Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| | - Kurt A. Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Adnan O. Abu-Yousif
- Oncology Drug Discovery Unit, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts
| |
Collapse
|
20
|
Tian C, Ren Z, Chang S, Cui W, Zhao P, Wang Y. Effect of infectious bursal disease virus infection on the expression of immune checkpoint molecules in SPF chickens. Vet Immunol Immunopathol 2025; 283:110933. [PMID: 40203670 DOI: 10.1016/j.vetimm.2025.110933] [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: 01/04/2025] [Revised: 03/23/2025] [Accepted: 04/02/2025] [Indexed: 04/11/2025]
Abstract
Infectious bursal disease (IBD) is an acute, highly contagious, and immunosuppressive avian disease caused by infectious bursal disease virus (IBDV). Immune checkpoint molecules are vital for regulating immune equilibrium, preventing autoimmune responses, and controlling the scale and duration of immune reactions. These molecules are implicated in the immunosuppressive mechanisms initiated by viral infections. Currently, the correlation between IBDV infection and the expression of immune checkpoint molecules is not clearly defined. In this study, White Leghorn SPF chickens were inoculated with very virulent (vvIBDV), attenuated (attIBDV), or novel variant (nvIBDV) IBDV strains. Using real-time PCR, we quantified immune checkpoint genes and cytokines in the bursa of fabricius (BF), thymus, spleen, and peripheral blood mononuclear cells (PBMCs). The findings revealed a significant upregulation of immune checkpoint genes (PD-1, PD-L1, PD-L2, LAG3, CTLA4) in the BF, thymus, spleen, and PBMCs following infection with various virulent strains of IBDV compared to control tissues. Specifically, PD-L1 in BF surged to 7.32-fold (vvIBDV, 3 dpi), 7.44-fold (attIBDV, 3 dpi), and 6.47-fold (nvIBDV, 3 dpi) compared to controls. PD-L2 in spleen surged to 7.66-fold (attIBDV, 21 dpi), and 7.82-fold (nvIBDV, 21 dpi) compared to controls. Moreover, the expression levels of inhibitory and pro-inflammatory cytokines (TGF-β2, IL-10, IL-6, IL-1β, TNF-α) were notably elevated in infected tissues compared to control tissues, with IL-1β upregulated by 7.47-fold in BF at 3 dpi. TGF-β2 in spleen surged to 8.16-fold (vvIBDV, 21 dpi), 6.33-fold (attIBDV, 21 dpi), and 7.61-fold (nvIBDV, 21 dpi). These results suggest that IBDV infection in SPF chickens induces robust upregulation of immune checkpoint molecules (PD-1, PD-L1, PD-L2, LAG3, CTLA4) and cytokines (TGF-β2, IL-10, IL-6, IL-1β, TNF-α), correlating with viral virulence and immunosuppression. Our findings suggest that immune checkpoint dysregulation may contribute to IBDV pathogenesis, particularly in mediating sustained immunosuppression in poultry.
Collapse
Affiliation(s)
- Chundi Tian
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Zoonoses, Tai'an, China
| | - Zhihao Ren
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Zoonoses, Tai'an, China
| | - Shang Chang
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Zoonoses, Tai'an, China
| | - Wenping Cui
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Zoonoses, Tai'an, China
| | - Peng Zhao
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Zoonoses, Tai'an, China.
| | - Yixin Wang
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Zoonoses, Tai'an, China.
| |
Collapse
|
21
|
Pan Y, Dai J, Liu Y, Wang Y, Zhang Q, Lou Y, Qiu Y. NAE1 protein: a prognostic, immunomodulatory, and therapeutic biomarker associated with neddylation in hepatocellular carcinoma. Int J Biol Macromol 2025; 310:143539. [PMID: 40300298 DOI: 10.1016/j.ijbiomac.2025.143539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 05/01/2025]
Abstract
Current predictive biomarkers for clinical outcomes and treatment in hepatocellular carcinoma (HCC) are not reliable enough. Neddylation, a novel post-translational modification, plays a crucial role in the immunomodulation, metabolism, and pathogenesis of HCC. However, whether it can function as a powerful predictive biomarker for HCC remains unknown. In current research, we first identified NAE1 as the most significant neddylation-related gene affecting the prognosis of HCC patients mainly through weighted gene co-expression network (WGCNA) and machine learning. Subsequently, we determined NAE1 expression as an independent risk factor for HCC using univariate and multivariate Cox regression and constructed a nomogram integrating NAE1 expression with clinical characteristics to predict survival probabilities in HCC patients. Bulk and single-cell RNA sequencing analyses revealed that NAE1 expression was primarily positively connected with immune cell infiltration in HCC, as assessed by the six latest immune algorithms. In addition, drug sensitivity and molecular docking collectively revealed the influence of NAE1 expression on the IC50 values of the four agents and the binding interactions between NAE1 protein and these drugs. Furthermore, we found that NAE1 depletion suppressed proliferation, migration, and invasion of HCC cells in vitro experiments. In conclusion, NAE1 protein holds considerable potential as a valuable biomarker for predicting clinical outcomes, immune landscapes, and drug sensitivity in HCC, as well as a promising therapeutic target.
Collapse
Affiliation(s)
- Yong Pan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Clinical Research Center for Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jinyao Dai
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Clinical Research Center for Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yi Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Clinical Research Center for Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yujing Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Clinical Research Center for Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qiudan Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Clinical Research Center for Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yan Lou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Clinical Research Center for Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China..
| | - Yunqing Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Clinical Research Center for Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China..
| |
Collapse
|
22
|
Takahashi H, Matsuyama T, Kawabata-Iwakawa R, Kawamoto T, Chikamatsu K. Comprehensive profiling of the heterogeneity of molecular endotypic traits in chronic rhinosinusitis. Hum Immunol 2025; 86:111267. [PMID: 39986126 DOI: 10.1016/j.humimm.2025.111267] [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: 09/06/2024] [Revised: 01/26/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025]
Abstract
Chronic rhinosinusitis (CRS) is a common clinical disease with molecular endotypes. In the present study, we performed a comprehensive transcriptomic profiling to investigate the heterogeneity of endotypes in CRS with nasal polyps (CRSwNP). The GSE23552 dataset, which includes microarray, was acquired from the Gene Expression Omnibus database. Additionally, surgical specimens were collected at Gunma University Hospital, and reverse transcription-quantitative PCR was performed. We performed gene expression analysis, Gene Set Enrichment Analysis (GSEA), deconvolution analysis, and hierarchical clustering of samples. Gene expression analysis and GSEA revealed that type 1, type 2, and Treg-related responses, were upregulated in nasal polyp tissues when compared with those in controls. Deconvolution analysis indicated the enrichment of type 1-related cells and generation of memory T cells. Furthermore, nasal polyps exhibited higher expression of effector function- and immune checkpoint-related genes than controls. In addition, hierarchical clustering revealed the heterogeneity in patients with type 2-inflamed CRSwNP. Notably, type 1 and type 2 scores correlated with the duration from surgery to biopharmaceuticals initiation. In conclusion, our study demonstrated the heterogeneity of molecular endotypes in CRSwNP. Further characterisation and stratification are required to develop a new endotype-based precision medicine for patients with CRS.
Collapse
Affiliation(s)
- Hideyuki Takahashi
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan.
| | - Toshiyuki Matsuyama
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Reika Kawabata-Iwakawa
- Division of Integrated Oncology Research, Gunma University, Initiative for Advanced Research, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Takayuki Kawamoto
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Kazuaki Chikamatsu
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
23
|
Tian RF, Feng LL, Liang X, Shi Y, Wang H, Fan J, Fan XY, Zhang JJ, Ke Y, Yang T, Huo F, Fu X, Cui HY, Chen ZN, Li L. Carnitine palmitoyltransferase 2 as a novel prognostic biomarker and immunoregulator in colorectal cancer. Int J Biol Macromol 2025; 309:142945. [PMID: 40210071 DOI: 10.1016/j.ijbiomac.2025.142945] [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: 10/31/2024] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Metabolic interventions are critical for enhancing immunotherapy efficacy, but reliable metabolic targets remain absent for colorectal cancer (CRC). This study aims to investigate the interplay between metabolic and immunological factors in CRC, identify metabolic immunoregulatory molecules, and propose targets for prognostic and therapeutic applications. METHODS Immune infiltration and metabolic pathways in CRC were analyzed using CIBERSORT and gene set variation analyses. Cox regression identified survival-related metabolic genes, forming a metabolic-related gene prognostic index (MRGPI), which was validated through survival analysis, timeROC, GSEA, CIBERSORT, and TIDE. Hub genes in the MRGPI were assessed using enrichment and co-expression network analyses. The expression of carnitine palmitoyltransferase 2 (CPT2) was validated through multiplex immunofluorescence of tissue microarrays. While its role was examined by western blot, CCK-8 assay, flow cytometry, qRT-PCR, Elisa, chemotaxis assays, etc. RESULTS: Fatty acid oxidation (FAO) pathways were significantly altered in CRC and correlated with immune cell infiltration and patient survival. The MRGPI, constructed from five survival-related metabolic genes, demonstrated strong prognostic and immunotherapeutic predictive value. Moreover, CPT2, a key hub gene in the MRGPI, whose lower expression in plasma cells predicts unfavorable patients' survival and could be an independent prognostic indicator, while its knockout in tumor cells significantly increases the infiltrating levels of CD8+ T cells via promoting the release of CCL25. CONCLUSION The FAO-dominated MRGPI is a promising biomarker for predicting patient outcomes and immunotherapy response. CPT2 holds potential as a prognostic marker and therapeutic target for CRC metabolic immunotherapy.
Collapse
Affiliation(s)
- Ruo-Fei Tian
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Le-Le Feng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Xue Liang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Ying Shi
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Hao Wang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Jing Fan
- Air Force Hospital of the Northern Theater Command of the People's Liberation Army of China, Shenyang 110000, China
| | - Xin-Yu Fan
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Jia-Jia Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Yuan Ke
- Department of Radiation and Medical Oncology, Hubei Province Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ting Yang
- Bayi Orthopedic Hospital, Chengdu 610031, China
| | - Fei Huo
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Xin Fu
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Hong-Yong Cui
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China.
| | - Zhi-Nan Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China.
| | - Ling Li
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
24
|
Wang X, Wang L, Liu Y. Current Status of Immune Checkpoint Inhibitors and Treatment Responsive Biomarkers for Triple-Negative Breast Cancer. Thorac Cancer 2025; 16:e70072. [PMID: 40324951 PMCID: PMC12052518 DOI: 10.1111/1759-7714.70072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/24/2025] [Accepted: 04/15/2025] [Indexed: 05/07/2025] Open
Abstract
Triple-negative breast cancer (TNBC), accounting for about 10%-20% of all breast cancer cases, is characterized by its aggressive nature, high recurrence rates, and poor prognosis. Unlike other breast cancer subtypes, TNBC lacks hormone receptors and specific molecular targets, limiting therapeutic options. In recent years, immune checkpoint inhibitors (ICIs) have shown promise in treating TNBC by targeting immune evasion mechanisms. Despite these advancements, several issues remain unresolved, including low response rates in programmed cell death ligand 1 (PD-L1) negative TNBC subtypes and the challenge of predicting which patients will benefit from ICIs. Consequently, there is growing interest in identifying reliable biomarkers beyond PD-L1 expression. This review synthesizes recent studies to provide a comprehensive perspective on ICI therapy in TNBC, clarifying the status of single-agent ICI therapies and combination strategies, emphasizing the need for further research into biomarkers. These insights provide clues for more personalized and effective treatment approaches, ultimately aiming to improve clinical outcomes for patients with TNBC.
Collapse
Affiliation(s)
- Xinran Wang
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Lingxia Wang
- Value & Implementation, Global Medical & Scientific AffairsMSD ChinaShanghaiChina
| | - Yueping Liu
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| |
Collapse
|
25
|
Hao B, Lin S, Liu H, Xu J, Chen L, Zheng T, Zhang W, Dang Y, Reiter RJ, Li C, Zhai H, Xia Q, Fan L. Baicalein tethers CD274/PD-L1 for autophagic degradation to boost antitumor immunity. Autophagy 2025; 21:917-933. [PMID: 39710370 PMCID: PMC12013432 DOI: 10.1080/15548627.2024.2439657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024] Open
Abstract
Immune checkpoint inhibitors, especially those targeting CD274/PD-L1yield powerful clinical therapeutic efficacy. Thoughmuch progress has been made in the development of antibody-basedCD274 drugs, chemical compounds applied for CD274degradation remain largely unavailable. Herein,baicalein, a monomer of traditional Chinese medicine, isscreened and validated to target CD274 and induces itsmacroautophagic/autophagic degradation. Moreover, we demonstrate thatCD274 directly interacts with MAP1LC3B (microtubule associatedprotein 1 light chain 3 beta). Intriguingly, baicalein potentiatesCD274-LC3 interaction to facilitate autophagic-lysosomal degradationof CD274. Importantly, targeted CD274. degradation via baicaleininhibits tumor development by boosting T-cell-mediated antitumorimmunity. Thus, we elucidate a critical role of autophagy-lysosomalpathway in mediating CD274 degradation, and conceptually demonstratethat the design of a molecular "glue" that tethers the CD274-LC3interaction is an appealing strategy to develop CD274 inhibitors incancer therapy.Abbreviations: ATTECs: autophagy-tethering compounds; AUTACs: AUtophagy-TArgeting Chimeras; AUTOTACs: AUTOphagy-TArgeting Chimeras; AMPK: adenosine 5'-monophosphate (AMP)-activated protein kinase; BiFC: bimolecular fluorescence complementation; BafA1: bafilomycin A1; CD274/PD-L1/B7-H1: CD274 molecule; CQ: chloroquine; CGAS: cyclic GMP-AMP synthase; DAPI: 4'6-diamino-2-phenylindole; FITC: fluorescein isothiocyanate isomer; GFP: green fluorescent protein; GZMB: granzyme B; IHC: immunohistochemistry; ICB: immune checkpoint blockade; KO: knockout; KD: equilibrium dissociation constant; LYTAC: LYsosome-TArgeting Chimera; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MST: microscale thermophoresis; NFAT: nuclear factor of activated T cells; NFKB/NF-kB: nuclear factor kappa B; NSCLC: non-small-cell lung cancer; PDCD1: programmed cell death 1; PROTACs: PROteolysis TArgeting Chimeras; PRF1: perforin 1; PE: phosphatidylethanolamine; PHA: phytohemagglutinin; PMA: phorbol 12-myristate 13-acetate; STAT: signal transducer and activator of transcription; SPR: surface plasmon resonance; TILs: tumor-infiltrating lymphocyte; TME: tumor microenvironment.
Collapse
Affiliation(s)
- Bingjie Hao
- Institute of Energy Metabolism and Health, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shumeng Lin
- Institute of Energy Metabolism and Health, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haipeng Liu
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Junfang Xu
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Li Chen
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tiansheng Zheng
- Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen Zhang
- Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yifang Dang
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Chaoqun Li
- Institute of Energy Metabolism and Health, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hong Zhai
- Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qing Xia
- Institute of Energy Metabolism and Health, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lihong Fan
- Institute of Energy Metabolism and Health, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Respiratory Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| |
Collapse
|
26
|
Hysa E, Camellino D, Dejaco C, Bauckneht M, Pesce G, Morbelli S, Bagnasco M, Cutolo M, Matteson EL, Cimmino MA, Saverino D. Soluble Co-Inhibitory Immune Checkpoint Molecules Are Increased in Patients With Polymyalgia Rheumatica Without Significant Correlations With Clinical Status: A Case-Control Study. ACR Open Rheumatol 2025; 7:e70045. [PMID: 40344345 PMCID: PMC12063068 DOI: 10.1002/acr2.70045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/09/2025] [Accepted: 03/20/2025] [Indexed: 05/11/2025] Open
Abstract
OBJECTIVE A dysregulated immune response is involved in the pathogenesis of polymyalgia rheumatica (PMR) and giant cell arteritis (GCA). These diseases have been reported as immune-related adverse events in patients with cancer treated with immune checkpoints inhibitors. In this cross-sectional study, the relationship between soluble immune checkpoint molecules (sICMs) and clinical/imaging features of PMR and GCA was investigated. METHODS Consecutive patients with PMR diagnosed according to the criteria by Bird et al were compared with age- and sex-matched healthy controls. Patients with PMR and overlapping GCA had to also satisfy the 1990 ACR classification criteria for GCA. All patients underwent standardized clinical, laboratory examination, and 18F-fluorodeoxyglucose positron emission tomography/computed tomography scans. The sICM anticytotoxic T Ly-4, the programmed cell death protein 1 (PD-1), and PD-1 ligands PD-L1 and PD-L2 were measured by enzyme-linked immunosorbent assay. RESULTS Forty patients (80% women, mean age 76 years, and mean disease duration 88 days) were assessed. Of these, 30 had isolated PMR and 10 had PMR with GCA. Patients showed significantly higher concentrations of all sICMs compared with controls (P < 0.001). Conditional logistic regression revealed the strong discriminative capacity of these molecules between patients and healthy controls, with PD-1 showing complete separation among groups (effect size = 0.78) and PD-L1 (odds ratio [OR] 134.33, P < 0.001) and PD-L2 (OR 63.00, P < 0.001) demonstrating the strongest ability to distinguish patients from controls. Correlations between sICM levels and clinical features were generally weak or absent, with no significant differences based on disease phenotype or glucocorticoid exposure. Results were similar in glucocorticoid-naive patients. CONCLUSION sICMs are significantly elevated in PMR and GCA and strongly differentiate patients from healthy controls. Although they do not correlate with clinical or imaging features, their consistent elevation in active disease might suggest a complex interplay between innate and adaptive immunity.
Collapse
Affiliation(s)
- Elvis Hysa
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Department of Experimental Medicine (DIMES), University of GenovaGenovaItaly
| | - Dario Camellino
- DIMES, University of Genova, Division of Rheumatology, “La Colletta” Hospital, Local Health Trust 3GenovaItaly
| | - Christian Dejaco
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria, and Department of Rheumatology, Hospital of Brunico (SABES‐ASDAA), Department of Rheumatology, Teaching Hospital of the Paracelsus Medical UniversityBrunicoItaly
| | - Matteo Bauckneht
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Department of Health Sciences, University of GenovaGenovaItaly
| | - Giampaola Pesce
- Department of Internal Medicine, Immunology Unit, University of Genova, Diagnostic Laboratory of Autoimmunology, IRCSS Ospedale Policlinico San MartinoGenovaItaly
| | - Silvia Morbelli
- Nuclear Medicine Unit, Department of Medical SciencesUniversity of TurinTurinItaly
| | - Marcello Bagnasco
- Department of Internal Medicine and Medical SpecialtiesUniversity of GenovaGenovaItaly
| | - Maurizio Cutolo
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal MedicineUniversity of Genova, IRCSS Ospedale Policlinico San MartinoGenovaItaly
| | | | - Marco A. Cimmino
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal MedicineUniversity of GenovaGenovaItaly
| | - Daniele Saverino
- DIMES, University of Genova, Diagnostic Laboratory of AutoimmunologyIRCSS Ospedale Policlinico San MartinoGenovaItaly
| |
Collapse
|
27
|
Rana M, Liou KC, Thakur A, Nepali K, Liou JP. Advancing glioblastoma therapy: Learning from the past and innovations for the future. Cancer Lett 2025; 617:217601. [PMID: 40037502 DOI: 10.1016/j.canlet.2025.217601] [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: 11/14/2024] [Revised: 02/25/2025] [Accepted: 03/01/2025] [Indexed: 03/06/2025]
Abstract
Marred by a median survival of only around 12-15 months coupled with poor prognosis and effective therapeutic deprived drug armory, treatment/management of glioblastoma has proved to be a daunting task. Surgical resection, flanked by radiotherapy and chemotherapy with temozolomide, stands as the standard of care; however, this trimodal therapy often manifests limited efficacy due to the heterogeneous and highly infiltrative nature of GBM cells. In addition, the existence of the blood-brain barrier, tumor microenvironment, and the immunosuppressive nature of GBM, along with the encountered resistance of GBM cells towards conventional therapy, also hinders the therapeutic applications of chemotherapeutics in GBM. This review presents key insights into the molecular pathology of GBM, including genetic mutations, signaling pathways, and tumor microenvironment characteristics. Recent innovations such as immunotherapy, oncolytic viral therapies, vaccines, nanotechnology, electric field, and cancer neuroscience, as well as their clinical progress, have been covered. In addition, this compilation also encompasses a discussion on the role of personalized medicine in tailoring treatments based on individual tumor profiles, an approach that is gradually shifting the paradigm in GBM management. Endowed with the learnings imbibed from past failures coupled with the zeal to embrace novel/multidisciplinary approaches, researchers appear to be on the right track to pinpoint more effective and durable solutions in the context of GBM treatment.
Collapse
Affiliation(s)
- Mandeep Rana
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Ke-Chi Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan; TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, 110, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan.
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan; TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, 110, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan.
| |
Collapse
|
28
|
Bai S, Wang Y, Zhou Y, Qiao L. Multi-omics pan-cancer analysis of monocyte to macrophage differentiation-associated (MMD) and its significance in hepatocellular carcinoma. Cancer Biomark 2025; 42:18758592251329280. [PMID: 40393675 DOI: 10.1177/18758592251329280] [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: 05/22/2025]
Abstract
BackgroundMalignant tumors are serious diseases that endanger human health. Therefore, it is crucial to identify markers that facilitate tumor diagnosis and prognostic assessment.ObjectiveThis study analyzed the significance of Monocyte to macrophage differentiation-associated (MMD) in various tumors from multiple perspectives, to explore the possibility of using MMD as a novel tumor marker.MethodsUsing the R software, an examination of MMD levels was conducted across diverse human cancers and their influence on cancer outcomes. MMD methylation, mutations, and immune infiltration analyses of various tumors were performed. A Cox regression model was used to predict the survival rates of patients with hepatocellular carcinoma (HCC). Finally, MMD expression and function were validated in Hep-3B cells.ResultsMMD was aberrantly expressed in diverse tumors and can predict patient outcomes. Methylation and functional enrichment studies indicated possible function of MMD in tumor progression, whereas immune infiltration data suggested its involvement in tumor immune evasion. Cox regression analysis revealed that elevated MMD levels were independent predictors of HCC patient outcomes. The quantitative real-time polymerase chain reaction (qPCR) data demonstrated high MMD levels in Hep-3B cells, and its suppression impeded Hep-3B cell growth.ConclusionsMMD was abnormally expressed in various tumors and was closely associated with tumor prognosis. Thus, it had the potential to be used as a novel tumor marker.
Collapse
Affiliation(s)
- Suyang Bai
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yuping Wang
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yongning Zhou
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Liang Qiao
- Storr Liver Centre, the Westmead Institute for Medical Research, The University of Sydney, Westmead, Australia
| |
Collapse
|
29
|
Wong NKY, Kang N, Dourieu C, Wang Y. Exploiting pre-dormancy tumor immune microenvironment induced by androgen deprivation in prostate cancer. Transl Cancer Res 2025; 14:2183-2187. [PMID: 40386281 PMCID: PMC12079259 DOI: 10.21037/tcr-2024-2683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/17/2025] [Indexed: 05/20/2025]
Affiliation(s)
- Nelson K. Y. Wong
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
| | - Ning Kang
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
| | - Claire Dourieu
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver, BC, Canada
| |
Collapse
|
30
|
Zhou JJ, Feng YC, Zhao ML, Guo Q, Zhao XB. Nanotechnology-driven strategies in postoperative cancer treatment: innovations in drug delivery systems. Front Pharmacol 2025; 16:1586948. [PMID: 40371327 PMCID: PMC12075547 DOI: 10.3389/fphar.2025.1586948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 04/23/2025] [Indexed: 05/16/2025] Open
Abstract
Cancer remains a global health challenge, and this challenge comes with a significant burden. Current treatment modalities, such as surgery, chemotherapy, and radiotherapy, have their limitations. The emergence of nanomedicines presents a new frontier in postoperative cancer treatment, offering potential to inhibit tumor recurrence and manage postoperative complications. This review deeply explores the application and potential of nanomedicines in the treatment of cancer after surgery. In particular, it focuses on local drug delivery systems (LDDS), which consist of in situ injection, implantation, and spraying. LDDS can provide targeted drug delivery and controlled release, which enhancing therapeutic efficacy. At the same time, it minimizes damage to healthy tissues and reduces systemic side effects. The nanostructures of these systems are unique. They facilitate the sustained release of drugs, prolong the effects of treatment, and decrease the frequency of dosing. This is especially beneficial in the postoperative period. Despite their potential, nanomedicines have limitations. These include high production costs, concerns regarding long-term toxicity, and complex regulatory approval processes. This paper aims to analyze several aspects. These include the advantages of nanomedicines, their drug delivery systems, how they combine with multiple treatment methods, and the associated challenges. Future research should focus on certain issues. These issues are stability, tumor specificity, and clinical translation. By addressing these, the delivery methods can be optimized and their therapeutic efficacy enhanced. With the advancements in materials science and biomedical engineering, the future design of LDDS is set to become more intelligent and personalized. It will cater to the diverse needs of clinical treatment and offer hope for better outcomes in cancer patients after surgery.
Collapse
Affiliation(s)
- Jun-Jie Zhou
- The Stomatological Hospital, Anyang Sixth People’s Hospital, Anyang, China
| | | | | | | | | |
Collapse
|
31
|
Otaegui J, Sultan D, Heo GS, Liu Y. Positron Emission Tomography Imaging of the Adaptive Immune System in Cardiovascular Diseases. CHEMICAL & BIOMEDICAL IMAGING 2025; 3:209-224. [PMID: 40313531 PMCID: PMC12042138 DOI: 10.1021/cbmi.4c00117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 05/03/2025]
Abstract
Cardiovascular diseases are the leading cause of death around the globe. In recent years, a crucial role of the immune system has been acknowledged in cardiac disease progression, opening the door for immunomodulatory therapies. To this ongoing change of paradigm, positron emission tomography (PET) imaging of the immune system has become a remarkable tool to reveal immune cell trafficking and monitor disease progression and treatment response. Currently, PET imaging of the immune system in cardiovascular disease mainly focuses on the innate immune system such as macrophages, while the immune cells of the adaptive immune system including B and T cells are less studied. This can be ascribed to the lack of radiotracers specifically binding to B and T cell biomarkers compatible with PET imaging within the cardiovascular system. In this review, we summarize current knowledge about the role of the adaptive immune system (e.g., B and T cells) in major cardiovascular diseases and introduce key biomarkers for specific targeting of these immune cells and their subpopulations. Finally, we present available radiotracers for these biomarkers and propose a pathway for developing probes or optimizing those already used in other fields (e.g., oncology) to make them compatible with the cardiovascular system.
Collapse
Affiliation(s)
- Jaume
Ramon Otaegui
- Mallinckrodt Institute of
Radiology, Washington University, St. Louis, Missouri 63110, United States
| | - Deborah Sultan
- Mallinckrodt Institute of
Radiology, Washington University, St. Louis, Missouri 63110, United States
| | - Gyu Seong Heo
- Mallinckrodt Institute of
Radiology, Washington University, St. Louis, Missouri 63110, United States
| | - Yongjian Liu
- Mallinckrodt Institute of
Radiology, Washington University, St. Louis, Missouri 63110, United States
| |
Collapse
|
32
|
Lerch M, Ramanathan S. The pathogenesis of neurological immune-related adverse events following immune checkpoint inhibitor therapy. Semin Immunol 2025; 78:101956. [PMID: 40294474 DOI: 10.1016/j.smim.2025.101956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/30/2025]
Abstract
Cancer is a leading cause of morbidity and mortality worldwide. The development of immune checkpoint inhibitors (ICI) has revolutionised cancer therapy, and patients who were previously incurable can now have excellent responses. These therapies work by blocking inhibitory immune pathways, like cytotoxic T lymphocyte-associated protein 4 (CTLA-4), programmed cell death-1 (PD-1), its ligand PD-L1, and lymphocyte activation gene 3 (LAG-3); which leads to increased anti-tumour immune responses. However, their use can lead to the development of immune-related adverse events (irAEs), which may result in severe disability, interruption of cancer therapy, and even death. Neurological autoimmune sequelae occur in 1-10 % of patients treated with ICIs and can be fatal. They encompass a broad spectrum of diseases, may affect the central and the peripheral nervous system, and include syndromes like encephalitis, cerebellitis, neuropathy, and myositis. In some cases, neurological irAEs can be associated with autoantibodies recognising neuronal or glial targets. In this review, we first describe the key targets in ICI therapy, followed by a formulation of irAEs and their clinical presentations, where we focus on neurological syndromes. We comprehensively formulate the current literature evaluating cell surface and intracellular autoantibodies, cytokines, chemokines, leukocyte patterns, other blood derived biomarkers, and immunogenetic profiles; and highlight their impact on our understanding of the pathogenesis of neurological irAEs. Finally, we describe therapeutic pathways and patient outcomes, and provide an overview on future aspects of ICI cancer therapy.
Collapse
Affiliation(s)
- Magdalena Lerch
- Translational Neuroimmunology Group, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Sudarshini Ramanathan
- Translational Neuroimmunology Group, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia; Department of Neurology and Concord Clinical School, Concord Hospital, Sydney, Australia.
| |
Collapse
|
33
|
Sonar PV, Singh AK, Mandadi S, Sharma NK. Expanding horizons of cancer immunotherapy: hopes and hurdles. Front Oncol 2025; 15:1511560. [PMID: 40352591 PMCID: PMC12061710 DOI: 10.3389/fonc.2025.1511560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/31/2025] [Indexed: 05/14/2025] Open
Abstract
Background Tumor displays various forms of tumor heterogeneity including immune heterogeneity that allow cancer cells to survive during conventional anticancer drug interventions. Thus, there is a strong rationale for overcoming anticancer drug resistance by employing the components of immune cells. Using the immune system to target tumor cells has revolutionized treatment. Recently, significant progress has been achieved at preclinical and clinical levels to benefit cancer patients. Approach A review of literature from the past ten years across PubMed, Scopus, and Web of Science focused on immunotherapy strategies. These include immune checkpoint inhibitors (ICIs), tumor-infiltrating lymphocyte therapy, antibody-drug conjugates (ADCs), cancer vaccines, CAR T-cell therapy, and the role of the gut microbiome. Conclusion While immunotherapy outcomes have improved, particularly for tumor types such as melanoma and non-small cell lung cancer (NSCLC), challenges persist regarding predictive biomarker identification and better management. Ongoing research on modifiers of immune function like gut microbiome-derived metabolites, next-generation ADCs, and new classes of biologics is warranted. Overall, continued investigation toward optimizing synergistic immunotherapeutic combinations through strategic drug delivery systems is imperative for preclinical and clinical success in cancer patients.
Collapse
Affiliation(s)
- Priyanka Vijay Sonar
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Anuj Kumar Singh
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
- Ichnos Glenmark Innovation, Glenmark Pharmaceuticals Limited, Navi Mumbai, Maharashtra, India
| | - Sravan Mandadi
- Ichnos Glenmark Innovation, Glenmark Pharmaceuticals Limited, Navi Mumbai, Maharashtra, India
| | - Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| |
Collapse
|
34
|
Guan J, Gong X, Zeng H, Zhang W, Qin Q, Gou H, Liu X, Song B. Gastrointestinal tumor personalized immunotherapy: an integrated analysis from molecular genetics to imaging biomarkers. Therap Adv Gastroenterol 2025; 18:17562848251333527. [PMID: 40297204 PMCID: PMC12035075 DOI: 10.1177/17562848251333527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
The immunotherapy landscape for gastrointestinal (GI) tumors is rapidly evolving. There is an urgent need for reliable biomarkers capable of predicting treatment outcomes to optimize therapeutic strategies and enhance patient prognosis. This review presents a comprehensive overview of biomarkers associated with the immunotherapy response of GI tumors, covering advances in molecular genetics, histopathological markers, and imaging. Key molecular biomarkers, such as microsatellite instability, tumor mutational burden, and programmed death-ligand 1 expression, remain critical for identifying patients likely to benefit from immune checkpoint inhibitors. The significance of tumor-infiltrating lymphocytes, notably the CD8+ T cell to regulatory T cell ratio, as a predictor of immunotherapy response is explored. In addition, advanced imaging techniques, including computed tomography (CT), magnetic resonance imaging, and positron emission tomography-CT, facilitate the noninvasive evaluation of tumor biology and therapeutic response. By bridging molecular and imaging data, this integrated strategy enhances precision in patient selection, treatment monitoring, and adaptive therapy design. Future studies should aim to validate these biomarkers in larger, multicenter cohorts and focus on clinical translation to advance precision medicine in GI oncology.
Collapse
Affiliation(s)
- Jian Guan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’s Armed Police Forces, Leshan, China
| | - Xiaoling Gong
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hanjiang Zeng
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’s Armed Police Forces, Leshan, China
| | - Qing Qin
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongfeng Gou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xijiao Liu
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya, Hainan, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, China
- Department of Radiology, Sanya People’s Hospital, Sanya, Hainan, China
| |
Collapse
|
35
|
Hsieh HC, Young MJ, Chen KY, Su WC, Lin CC, Yen YT, Hung JJ, Wang YC. Deubiquitinase USP24 activated by IL-6/STAT3 enhances PD-1 protein stability and suppresses T cell antitumor response. SCIENCE ADVANCES 2025; 11:eadt4258. [PMID: 40238877 PMCID: PMC12002121 DOI: 10.1126/sciadv.adt4258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 03/11/2025] [Indexed: 04/18/2025]
Abstract
Persisting programmed cell death-1 (PD-1) signaling impairs T cell effector function, which is highly associated with T cell exhaustion and immunotherapy failure. However, the mechanism responsible for PD-1 deubiquitination and T cell dysfunction remains unclear. Here, we show that ubiquitin-specific peptidase 24 (USP24) promotes PD-1 protein stability by removing K48-linked polyubiquitin. Increased interleukin-6 level transcriptionally activates the USP24 expression, which leads to PD-1 stabilization. Furthermore, USP24 deficiency reduces PD-1 levels in CD8+ T cells and attenuates EgfrL858R-driven lung tumorigenesis in Usp24C1695A catalytic deficient mice. Targeting PD-1 stability with the USP24-specific inhibitor USP24-i-101 boosts cytotoxic T cell activity, restrains lung tumor growth, and achieves superior therapeutic effects when combined with anti-CTLA4 immunotherapy. Clinically, patients with lung cancer exhibiting high USP24 expression in tumor-infiltrating CD8+ T cells display exhausted features and show unfavorable responses to immunotherapy. Our findings dissect the mechanism for regulating enhanced PD-1 stability in tumor-infiltrating CD8+ T cells and reveal USP24 as a potential target of antitumor immunotherapy.
Collapse
Affiliation(s)
- Hung-Chia Hsieh
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Ming-Jer Young
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 70101, Taiwan
| | - Kuan-Yu Chen
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Wu-Chou Su
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Chien-Chung Lin
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yi-Ting Yen
- Department of Surgery, National Cheng Kung University Hospital, College of Medical College, National Cheng Kung University, Tainan 70101, Taiwan
| | - Jan-Jong Hung
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yi-Ching Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| |
Collapse
|
36
|
Ren J, Yan G, Yang L, Kong L, Guan Y, Sun H, Liu C, Liu L, Han Y, Wang X. Cancer chemoprevention: signaling pathways and strategic approaches. Signal Transduct Target Ther 2025; 10:113. [PMID: 40246868 PMCID: PMC12006474 DOI: 10.1038/s41392-025-02167-1] [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: 08/02/2024] [Revised: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 04/19/2025] Open
Abstract
Although cancer chemopreventive agents have been confirmed to effectively protect high-risk populations from cancer invasion or recurrence, only over ten drugs have been approved by the U.S. Food and Drug Administration. Therefore, screening potent cancer chemopreventive agents is crucial to reduce the constantly increasing incidence and mortality rate of cancer. Considering the lengthy prevention process, an ideal chemopreventive agent should be nontoxic, inexpensive, and oral. Natural compounds have become a natural treasure reservoir for cancer chemoprevention because of their superior ease of availability, cost-effectiveness, and safety. The benefits of natural compounds as chemopreventive agents in cancer prevention have been confirmed in various studies. In light of this, the present review is intended to fully delineate the entire scope of cancer chemoprevention, and primarily focuses on various aspects of cancer chemoprevention based on natural compounds, specifically focusing on the mechanism of action of natural compounds in cancer prevention, and discussing in detail how they exert cancer prevention effects by affecting classical signaling pathways, immune checkpoints, and gut microbiome. We also introduce novel cancer chemoprevention strategies and summarize the role of natural compounds in improving chemotherapy regimens. Furthermore, we describe strategies for discovering anticancer compounds with low abundance and high activity, revealing the broad prospects of natural compounds in drug discovery for cancer chemoprevention. Moreover, we associate cancer chemoprevention with precision medicine, and discuss the challenges encountered in cancer chemoprevention. Finally, we emphasize the transformative potential of natural compounds in advancing the field of cancer chemoprevention and their ability to introduce more effective and less toxic preventive options for oncology.
Collapse
Affiliation(s)
- Junling Ren
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Guangli Yan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Le Yang
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China
| | - Ling Kong
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Yu Guan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Hui Sun
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
| | - Chang Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Lei Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Ying Han
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Xijun Wang
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China.
| |
Collapse
|
37
|
Zhang D, Wang W, Tang M, Qu C, Jiang Z, Li X, Luan Y. In Situ Gene Engineering Approach to Overcome Tumor Resistance and Enhance T Cell-Mediated Cancer Immunotherapy. NANO LETTERS 2025; 25:6200-6208. [PMID: 40180619 DOI: 10.1021/acs.nanolett.5c00488] [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: 04/05/2025]
Abstract
T cell-mediated cancer immunotherapy harnesses the power of cytotoxic T lymphocytes (CTLs) to target and eradicate tumor cells. However, tumor cells often evade immune attack through membrane repair mechanisms involving endosomal sorting complexes required for transport (ESCRT) and immune suppression within the tumor microenvironment. Here, we developed a robust TMV@PpCHIL nanomedicine to address these issues by reprogramming tumor cells via in situ gene editing. Using CRISPR/Cas9, we disrupted the Chmp4b gene, a key component of the ESCRT machinery, preventing tumor cells from repairing CTL-induced membrane damage. Simultaneously, we genetically engineered tumor cells to produce interleukin-12 (IL-12), a cytokine that enhances CTL activation. The TMV@PpCHIL nanomedicine, designed by coating tumor membrane vesicles (TMVs) onto polyamidoamine (PAMAM) dendrimer-condensed plasmid complexes, ensured efficient CRISPR/Cas9-based gene editing and sustained IL-12 production. This approach significantly enhanced CTL-mediated tumor cell cytotoxicity, suppressed tumor growth, reduced metastasis, and prolonged survival, providing a promising strategy for durable cancer treatment.
Collapse
Affiliation(s)
- Di Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Wenjuan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Mingtan Tang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Chenghao Qu
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, China
| | - Zeyu Jiang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Xuerui Li
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Yuxia Luan
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| |
Collapse
|
38
|
Hetta HF, Aljohani HM, Sirag N, Elfadil H, Salama A, Al-Twalhy R, Alanazi D, Al-johani MD, Albalawi JH, Al-Otaibi RM, Alsharif RA, Sayad R. Synergizing Success: The Role of Anlotinib Combinations in Advanced Non-Small Cell Lung Cancer Treatment. Pharmaceuticals (Basel) 2025; 18:585. [PMID: 40284020 PMCID: PMC12030542 DOI: 10.3390/ph18040585] [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: 02/24/2025] [Revised: 04/08/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025] Open
Abstract
Anlotinib, a novel receptor tyrosine kinase inhibitor that is taken orally, targets several RTKs and is authorized as a third-line treatment for patients with advanced non-small cell lung cancer (NSCLC). Anlotinib is also used in combination with immunotherapy or chemotherapy for advanced NSCLC. We aimed to explore the efficacy and safety of anlotinib-based regimens in NSCLC treatment, focusing on combination therapies. We also addressed challenges that hinder oncologists from using it, such as toxicity and resistance mechanisms. A systematic approach involves searching the National Institute of Health PubMed, Scopus, MedLine, and Web of Science databases up to April 2024. Relevant studies were identified and analyzed for their methodologies, outcomes, and patient characteristics. Findings revealed that numerous effective combination regimens, such as anlotinib with platinum-based chemotherapy and anlotinib combined with PD-1 blockades, have shown positive results in terms of progression-free survival (PFS), overall survival (OS), and objective response rate (ORR). On the other hand, NSCLC treatment faces hurdles due to drug resistance and its toxicity profile. These challenges underscore the need for continued research and optimization of treatment strategies.
Collapse
Affiliation(s)
- Helal F. Hetta
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Hashim M. Aljohani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Madina 41477, Saudi Arabia;
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Nizar Sirag
- Division of Pharmacognosy, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Hassabelrasoul Elfadil
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Ayman Salama
- Department of Pharmaceutics, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Rand Al-Twalhy
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.-T.); (D.A.); (M.D.A.-j.); (J.H.A.); (R.M.A.-O.); (R.A.A.)
| | - Danah Alanazi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.-T.); (D.A.); (M.D.A.-j.); (J.H.A.); (R.M.A.-O.); (R.A.A.)
| | - Manal D. Al-johani
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.-T.); (D.A.); (M.D.A.-j.); (J.H.A.); (R.M.A.-O.); (R.A.A.)
| | - Jumanah H. Albalawi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.-T.); (D.A.); (M.D.A.-j.); (J.H.A.); (R.M.A.-O.); (R.A.A.)
| | - Rinad M. Al-Otaibi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.-T.); (D.A.); (M.D.A.-j.); (J.H.A.); (R.M.A.-O.); (R.A.A.)
| | - Raghad A. Alsharif
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.-T.); (D.A.); (M.D.A.-j.); (J.H.A.); (R.M.A.-O.); (R.A.A.)
| | - Reem Sayad
- Department of Histology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt;
| |
Collapse
|
39
|
Wang X, Zhang W, Liang K, Wang Y, Zhang J, Wang J, Li A, Yun Y, Liu H, Sun Y. Identification of m6 A-regulated ferroptosis biomarkers for prognosis in laryngeal cancer. BMC Cancer 2025; 25:694. [PMID: 40229748 PMCID: PMC11998228 DOI: 10.1186/s12885-025-14134-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 04/10/2025] [Indexed: 04/16/2025] Open
Abstract
Laryngeal cancer (LC) is a malignant tumor that occurs in the larynx. N6-methyladenosine (m6A) RNA methylation, a pivotal and prevalent epigenetic modification in eukaryotic mRNA, intricately intertwines with ferroptosis, and together, they play a crucial role in the development of LC. Accordingly, further research on related molecular mechanisms and pathology of LC is necessary. Weighted gene co-expression network analysis and correlation analysis were used to identify differentially expressed m6A-related ferroptosis genes in LC. The TCGA-HNSC and GSE65858 datasets were obtained from public databases. The TCGA-HNSC dataset consisted of 110 primary tumor oropharynx samples and 12 control oropharynx samples, while the GSE65858 dataset contained forty-eight primary tumor oropharynx samples. Univariate Cox and least absolute shrinkage and selection operator (LASSO) regression were utilized for feature selection and risk model construction in the TCGA-HNSC dataset. The risk model was validated in the GSE65858 dataset. Then, a nomogram was built based on the independent prognostic factor identified using univariate and multivariate Cox regression in the TCGA-HNSC dataset. Mutation analysis, immune-related analysis, and drug sensitivity prediction were applied to analyze the utility of the risk model in the TCGA-HNSC dataset. Additionally, qRT-PCR and western blot were performed to detect the TFRC, RGS4, and FTH1 expression. Three biomarkers were identified to build a risk model using the univariate Cox and LASSO regression algorithms. Receiver operating characteristic (ROC) analysis verified the accuracy of the risk model. Tumor Immune Dysfunction and Exclusion (TIDE) and Estimation of STromal and Immune cells in MAlignant Tumors using the Expression data (ESTIMATE) algorithm showed a positive relationship between risk score and TIDE or ESTIMATE score. Furthermore, drug sensitivity prediction found that 19 chemotherapy drugs were strongly correlated with a risk score. TFRC, RGS4, and FTH1 exhibited high expression levels in 30 laryngeal carcinoma tissues and cell lines. Notably, TFRC and FTH1 expression levels were significantly associated with patient prognosis. In Conclusion, TFRC, RGS4, and FTH1, were identified as m6A-regulated ferroptosis biomarkers in LC, providing insights into LC treatment and prognosis.
Collapse
Affiliation(s)
- Xin Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, 256 Youyi Road, Xi'an, 710000, China
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710068, China
- Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710068, China
| | - Wen Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, 256 Youyi Road, Xi'an, 710000, China
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710068, China
- Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710068, China
| | - Kun Liang
- Department of Otorhinolaryngology, Head and Neck Surgery, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, 256 Youyi Road, Xi'an, 710000, China
| | - Yujuan Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, 256 Youyi Road, Xi'an, 710000, China
| | - Jin Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, 256 Youyi Road, Xi'an, 710000, China
| | - Jinping Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, 256 Youyi Road, Xi'an, 710000, China
| | - An Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, 256 Youyi Road, Xi'an, 710000, China
| | - Yonggang Yun
- Department of Otorhinolaryngology, Head and Neck Surgery, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, 256 Youyi Road, Xi'an, 710000, China
| | - Hiu Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, 256 Youyi Road, Xi'an, 710000, China.
| | - Yanan Sun
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital, Harbin Medical University, 246 Xuefu Road, Harbin, 150000, China.
| |
Collapse
|
40
|
Hashemi V, Baradaran B, Naseri B, Masoumi J, Baghbani E, Alizadeh N, Haris RS, Hosseini A. The effect of immunomodulatory celecoxsib on the gene expression of inhibitory receptors in dendritic cells generated from monocyte cells. BMC Res Notes 2025; 18:164. [PMID: 40223111 PMCID: PMC11995585 DOI: 10.1186/s13104-025-07226-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: 10/23/2024] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
Autoimmune diseases are characterized by irregular immune responses that disrupt self-tolerance. This research explores the effects of the immunomodulatory drug celecoxib on the expression of immune checkpoint receptors in monocyte-derived dendritic cells (DCs). Key receptors, including CTLA-4, VISTA, BTLA, PDL-1, B7H7, and LAG3, play critical roles in initiating and regulating immune responses and maintaining self-tolerance. Previous studies have highlighted the significance of immune checkpoints in preventing autoimmune conditions, with animal research supporting their effectiveness in immunotherapy. Our findings demonstrate that the upregulation of immune checkpoint receptors can enhance the inhibitory functions of DCs, thereby promoting self-tolerance. As a result, tolerogenic DCs present a promising therapeutic avenue for treating autoimmune diseases. Although these results are promising, further trials are required to validate this approach before it can be applied clinically. This study underscores the potential of targeting immune checkpoint receptors as a therapeutic strategy for autoimmune disorders.
Collapse
Affiliation(s)
- Vida Hashemi
- Department of Laboratory Science, Faculty of Medicine, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahar Naseri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Masoumi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Shiri Haris
- Department of Laboratory Science, Faculty of Medicine, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Arezoo Hosseini
- Department of Immunology and Genetics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
41
|
Lin YZ, Liu CH, Wu WR, Liao TY, Lee CC, Li HW, Chung FC, Shen YC, Zhuo GY, Liu LC, Cheng WC, Wang SC. Memory-promoting function of miR-379-5p attenuates CD8 + T cell exhaustion by targeting immune checkpoints. J Immunother Cancer 2025; 13:e010363. [PMID: 40221151 PMCID: PMC11997822 DOI: 10.1136/jitc-2024-010363] [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: 08/16/2024] [Accepted: 02/16/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are epigenetic regulators of T cell maturation and exhaustion. However, the mechanisms by which miRNAs influence T cell function in tumor environments remain unclear. This study focuses on miR-379-5p, which counteracts T cell exhaustion and enhances antitumor responses. METHODS Native CD8+ T cells were isolated from the blood of healthy donors and subjected to chronic stimulation to induce exhaustion. RNA sequencing and miRNA sequencing were performed to identify differentially expressed miRNAs. These miRNAs underwent bioinformatics analyses, including DESeq enrichment, immune cell infiltration assessment, and patient prognostic outcomes in The Cancer Genome Atlas data sets to assess their potential involvement in T cell exhaustion and antitumor immunity. The biological functions of miRNA on T cell differentiation, cytotoxic killing, and immune checkpoint regulation were investigated using in vitro assays, OT-I B16F10-OVA models, and patient-derived tumor organoids. RESULTS MiR-379-5p is downregulated in exhausted T cells and negatively associated with exhausted tumor-infiltrating lymphocytes in advanced tumors. It correlates positively with better survival outcomes in breast cancer, cervical cancer and melanoma. In CD8+ T cells, miR-379-5p reduces the expression of immune checkpoint proteins T cell immunoglobulin and mucin-domain containing-3 (TIM3) and T cell immunoreceptor with Ig and ITIM domains (TIGIT) by targeting their 3' untranslated region. Overexpression of miR-379-5p in CD8+ T cell promotes differentiation into memory-like T effector cells and enhances cytotoxic killing of cancer cells. The transcription factor nuclear receptor subfamily 4 group A member 1 (NR4A1) with increased expression in exhausted T cells and negatively regulates miR-379, restoring immune checkpoint expression and suppressing cancer-killing ability. In contrast, OT-I T cells expressing ectopic miR-379-5p show increased cytotoxicity against B16F10-OVA tumors in mice. Autologous T cells isolated from patients with breast cancer transduced with miR-379-5p significantly improve killing of tumor organoids derived from the same patients. CONCLUSIONS MiR-379-5p acts as an epigenetic tumor suppressor by enhancing CD8+ T cell effector functions and suppressing T cell exhaustion. MiR-379-5p could represent a novel marker and strategy for cancer immunotherapy, offering promising avenues for enhancing antitumor immune responses.
Collapse
Affiliation(s)
- You-Zhe Lin
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Chia-Hsin Liu
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
| | - Wan-Rong Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Ting-Yi Liao
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chuan-Chun Lee
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Hong-Wei Li
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
| | - Feng-Chi Chung
- Program for Cancer Biology and Drug Discovery, China Medical University, Taichung, Taiwan
| | - Yi-Chun Shen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Guan-Yu Zhuo
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung, Taiwan
| | - Liang-Chih Liu
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Wei-Chung Cheng
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Program for Cancer Biology and Drug Discovery, China Medical University, Taichung, Taiwan
| | - Shao-Chun Wang
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
- Program for Cancer Biology and Drug Discovery, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
42
|
Mou W, Deng Z, Zhu L, Jiang A, Lin A, Xu L, Deng G, Huang H, Guo Z, Zhu B, Wu S, Yang T, Wang L, Liu Z, Wei T, Zhang J, Cheng L, Huang H, Chen R, Shao Y, Cheng Q, Wang L, Yuan S, Luo P. Intratumoral mycobiome heterogeneity influences the tumor microenvironment and immunotherapy outcomes in renal cell carcinoma. SCIENCE ADVANCES 2025; 11:eadu1727. [PMID: 40203108 PMCID: PMC11980860 DOI: 10.1126/sciadv.adu1727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/27/2025] [Indexed: 04/11/2025]
Abstract
The intratumoral mycobiome plays a crucial role in the tumor microenvironment, but its impact on renal cell carcinoma (RCC) remains unclear. We collected and quantitatively profiled the intratumoral mycobiome data from 1044 patients with RCC across four international cohorts, of which 466 patients received immunotherapy. Patients were stratified into mycobiota ecology-depauperate and mycobiota ecology-flourishing (MEF) groups based on fungal abundance. The MEF group had worse prognosis, higher fungal diversity, down-regulated lipid catabolism, and exhausted CD8+ T cells. We developed the intratumoral mycobiota signature and intratumoral mycobiota-related genes expression signature, which robustly predicted prognosis and immunotherapy outcomes in RCC and other cancers. Aspergillus tanneri was identified as a potential key fungal species influencing RCC prognosis. Our findings suggest that the intratumoral mycobiome suppresses lipid catabolism and induces T cell exhaustion in RCC.
Collapse
Affiliation(s)
- Weiming Mou
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
- Donghai County People’s Hospital–Jiangnan University Smart Healthcare Joint Laboratory, Lianyungang 222000, China
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Zhixing Deng
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Lingxuan Zhu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Liling Xu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Gengwen Deng
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Hongsen Huang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zeji Guo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Bang Zhu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Shuqi Wu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Tao Yang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lu Wang
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Ting Wei
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Liang Cheng
- Department of Surgery (Urology), Brown University Warren Alpert Medical School, Providence, RI, USA
- Department of Pathology and Laboratory Medicine, Department of Surgery (Urology), Brown University Warren Alpert Medical School, Lifespan Health, and the Legorreta Cancer Center, Brown University, Providence, RI, USA
| | - Haojie Huang
- Institute of Urologic Science and Technology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311100, China
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311100, China
- Department of Urology, Mayo Comprehensive Cancer Center, Rochester, MN, USA
| | - Rui Chen
- Department of Urology, Shanghai Jiao Tong University School of Medicine Renji Hospital, Shanghai 200127, China
| | - Yi Shao
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Shuofeng Yuan
- Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518009, China
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
- Donghai County People’s Hospital–Jiangnan University Smart Healthcare Joint Laboratory, Lianyungang 222000, China
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
43
|
Xu Y, Yang S, Rao Q, Gao Y, Zhou G, Zhao D, Shi X, Chai Y, Zhao C. A mechanistic quantitative systems pharmacology model platform for translational efficacy evaluation and checkpoint combination design of bispecific immuno-modulatory antibodies. Front Pharmacol 2025; 16:1571844. [PMID: 40276607 PMCID: PMC12018249 DOI: 10.3389/fphar.2025.1571844] [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: 02/06/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
Over the past 2 decades, tumor immunotherapies have witnessed remarkable advancements, especially with the emergence of immune checkpoint-targeting bispecific antibodies. However, a quantitative understanding of the dynamic cross-talking mechanisms underlying different immune checkpoints as well as the optimal dosing and target design of checkpoint-targeting bispecific antibodies still remain challenging to researchers. To address this challenge, we have here developed a multi-scale quantitative systems pharmacology (QSP) model platform that integrates a diverse array of immune checkpoints and their interactive functions. The model has been calibrated and validated against an extensive collection of multiscale experimental datasets covering 20+ different monoclonal and bispecific antibody treatments at over 60 administered dose levels. Based on high-throughput simulations, the QSP model platform comprehensively screened and characterized the potential efficacy of different bispecific antibody target combination designs, and model-based preclinical population-level simulations revealed target-specific dose-response relationships as well as alternative dosing strategies that can maintain anti-tumor treatment efficacy while reducing dosing frequencies. Model simulations also pointed out that combining checkpoint-targeting bispecific antibodies with monoclonal antibodies can lead to significantly enhanced anti-tumor efficacy. Our mechanistic QSP model can serve as an integrated precision medicine simulation platform to guide the translational research and clinical development of checkpoint-targeting immuno-modulatory bispecific antibodies.
Collapse
Affiliation(s)
- Yiyang Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Siyuan Yang
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Qi Rao
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yuan Gao
- QSPMed Technologies, Nanjing, China
| | - Guanyue Zhou
- Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing, China
| | - Dongmei Zhao
- Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing, China
| | - Xinsheng Shi
- Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing, China
| | - Yi Chai
- Phase I Clinical Trial Unit, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chen Zhao
- School of Pharmacy, Nanjing Medical University, Nanjing, China
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
44
|
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
|
45
|
Nemati M, Hsu CY, Nathiya D, Kumar MR, Oghenemaro EF, Kariem M, Kaur P, Bhanot D, Hjazi A, Azam Saedi T. Gemcitabine: immunomodulatory or immunosuppressive role in the tumor microenvironment. Front Immunol 2025; 16:1536428. [PMID: 40270972 PMCID: PMC12014622 DOI: 10.3389/fimmu.2025.1536428] [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: 11/28/2024] [Accepted: 03/21/2025] [Indexed: 04/25/2025] Open
Abstract
Gemcitabine (GEM), a nucleoside analog chemotherapy agent, has been widely used in the treatment of various cancers. In recent years, there has been growing interest in understanding the immunomodulatory or immunosuppressive effects of GEM. The immunomodulatory roles of GEM could influence the anti-tumor immune responses via several mechanisms, such as modulation of antigen presentation, cytokine production, and immune cell population. Furthermore, there is evidence that GEM enhances the therapeutic efficacy of immunotherapies, including oncolytic viruses, immune checkpoint inhibitors, CAR T-cells, and therapeutic vaccines. On the other hand, accumulating evidence also proposed that GEM may act as an immunosuppressive agent within the tumor microenvironment, resulting in immune evasion of tumor cells and tumor growth. These paradoxical roles of GEM in modifying immune responses highlight the complexity of GEM interaction with immune cells and responses within the tumor microenvironment. This review aims to provide an overview of the immunomodulatory and immunosuppressive effects of GEM within the tumor microenvironment and how GEM affects the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Mahnaz Nemati
- Amir Oncology Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University, Phoenix, AZ, United States
| | - Deepak Nathiya
- Department of Pharmacy Practice, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - M. Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | - Deepak Bhanot
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Tayebeh Azam Saedi
- Department of Genetics, Faculty of Science, Islamic Azad University, Tonekabon, Iran
| |
Collapse
|
46
|
Ding Z, Wang L, Sun J, Zheng L, Tang Y, Tang H. Hepatocellular carcinoma: pathogenesis, molecular mechanisms, and treatment advances. Front Oncol 2025; 15:1526206. [PMID: 40265012 PMCID: PMC12011620 DOI: 10.3389/fonc.2025.1526206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/21/2025] [Indexed: 04/24/2025] Open
Abstract
Hepatocellular Carcinoma (HCC), a highly prevalent malignancy, poses a significant global health challenge. Its pathogenesis is intricate and multifactorial, involving a complex interplay of environmental and genetic factors. Viral hepatitis, excessive alcohol consumption, and cirrhosis are known to significantly elevate the risk of developing HCC. The underlying biological processes driving HCC are equally complex, encompassing aberrant activation of molecular signaling pathways, dysregulation of hepatocellular differentiation and angiogenesis, and immune dysfunction. This review delves into the multifaceted nature of HCC, exploring its etiology and the intricate molecular signaling pathways involved in its development. We examine the role of immune dysregulation in HCC progression and discuss the potential of emerging therapeutic strategies, including immune-targeted therapy and tumor-associated macrophage interventions. Additionally, we explore the potential of traditional Chinese medicine (TCM) monomers in inhibiting tumor growth. By elucidating the complex interplay of factors contributing to HCC, this review aims to provide a comprehensive understanding of the disease and highlight promising avenues for future research and therapeutic development.
Collapse
Affiliation(s)
- Zhixian Ding
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Lusheng Wang
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Jiting Sun
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Lijie Zheng
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Yu Tang
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Heng Tang
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| |
Collapse
|
47
|
Gong JR, Lee J, Han Y, Cho KH. DDX54 downregulation enhances anti-PD1 therapy in immune-desert lung tumors with high tumor mutational burden. Proc Natl Acad Sci U S A 2025; 122:e2412310122. [PMID: 40172969 PMCID: PMC12002276 DOI: 10.1073/pnas.2412310122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 02/25/2025] [Indexed: 04/04/2025] Open
Abstract
High tumor mutational burden (TMB-H) is a predictive biomarker for the responsiveness of cancer to immune checkpoint inhibitor (ICI) therapy that indicates whether immune cells can sufficiently recognize cancer cells as nonself. However, about 30% of all cancers from The Cancer Genome Atlas (TCGA) are classified as immune-desert tumors lacking T cell infiltration despite TMB-H. Since the underlying mechanism of these immune-desert tumors has yet to be unraveled, there is a pressing need to transform such immune-desert tumors into immune-inflamed tumors and thereby enhance their responsiveness to anti-PD1 therapy. Here, we present a systems framework for identifying immuno-oncotargets, based on analysis of gene regulatory networks, and validating the effect of these targets in transforming immune-desert into immune-inflamed tumors. In particular, we identify DEAD-box helicases 54 (DDX54) as a master regulator of immune escape in immune-desert lung cancer with TMB-H and show that knockdown of DDX54 can increase immune cell infiltration and lead to improved sensitivity to anti-PD1 therapy.
Collapse
Affiliation(s)
- Jeong-Ryeol Gong
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Republic of Korea
| | - Jungeun Lee
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Republic of Korea
| | - Younghyun Han
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Republic of Korea
| | - Kwang-Hyun Cho
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Republic of Korea
| |
Collapse
|
48
|
Choi AS, Moon TJ, Bhalotia A, Rajan A, Ogunnaike L, Hutchinson DW, Hwang I, Gokhale A, Kim JN, Ma T, Karathanasis E. Lipid Nanoparticles and PEG: Time Frame of Immune Checkpoint Blockade Can Be Controlled by Adjusting the Rate of Cellular Uptake of Nanoparticles. Mol Pharm 2025; 22:1859-1868. [PMID: 40035231 PMCID: PMC11975481 DOI: 10.1021/acs.molpharmaceut.4c01039] [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] [Indexed: 03/05/2025]
Abstract
The engineerability of lipid nanoparticles (LNPs) and their ability to deliver nucleic acids make LNPs attractive tools for cancer immunotherapy. LNP-based gene delivery can be employed for various approaches in cancer immunotherapy, including encoding tumor-associated antigens and silencing of negative immune checkpoint proteins. For example, LNPs carrying small interfering RNAs can offer several advantages, including sustained and durable inhibition of an immune checkpoint protein. Due to their tunable design, modifying the lipid composition of LNPs can regulate the rate of their uptake by immune cells and the rate of gene silencing. Controlling the kinetics of LNP uptake provides additional flexibility and strategies to generate appropriate immunomodulation in the tumor microenvironment. Here, we evaluated the effects of polyethylene glycol (PEG) content ranging from 0.5 to 6 mol % on the cellular uptake of LNPs by immune cells and gene silencing of PD-L1 after intratumoral administration. We evaluated the cellular uptake and PD-L1 blockade in vitro in cell studies and in vivo using the YUMM1.7 melanoma tumor model. Cell studies showed that the rate of cell uptake was inversely correlated to an increasing mol % of PEG in a linear relationship. In the in vivo studies, 0.5% PEG LNP initiated an immediate effect in the tumor with a significant decrease in the PD-L1 expression of immune cells observed within 24 h. In comparison, the gene silencing effect of 6% PEG LNP was delayed, with a significant decrease of PD-L1 expression in immune cell subsets being observed 72 h after administration. Notably, performance of the 6% PEG LNP at 72 h was comparable to that of the 0.5% PEG LNP at 24 h. Overall, this study suggests that PEG modifications and intratumoral administration of LNPs can be a promising strategy for an effective antitumor immune response.
Collapse
Affiliation(s)
- Andrew S Choi
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Taylor J Moon
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Anubhuti Bhalotia
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Aarthi Rajan
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Laolu Ogunnaike
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Diarmuid W Hutchinson
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Inga Hwang
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Aaditya Gokhale
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Justin N Kim
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Timothy Ma
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Efstathios Karathanasis
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
49
|
Novák J, Takács T, Tilajka Á, László L, Oravecz O, Farkas E, Than NG, Buday L, Balogh A, Vas V. The sweet and the bitter sides of galectin-1 in immunity: its role in immune cell functions, apoptosis, and immunotherapies for cancer with a focus on T cells. Semin Immunopathol 2025; 47:24. [PMID: 40178639 PMCID: PMC11968517 DOI: 10.1007/s00281-025-01047-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 02/07/2025] [Indexed: 04/05/2025]
Abstract
Galectin-1 (Gal-1), a member of the β-galactoside-binding soluble lectin family, is a double-edged sword in immunity. On one hand, it plays a crucial role in regulating diverse immune cell functions, including the apoptosis of activated T cells. These processes are key in resolving inflammation and preventing autoimmune diseases. On the other hand, Gal-1 has significant implications in cancer, where tumor cells and the tumor microenvironment (TME) (e.g., tumor-associated fibroblasts, myeloid-derived suppressor cells) secrete Gal-1 to evade immune surveillance and promote cancer cell growth. Within the TME, Gal-1 enhances the differentiation of tolerogenic dendritic cells, induces the apoptosis of effector T cells, and enhances the proliferation of regulatory T cells, collectively facilitating tumor immune escape. Therefore, targeting Gal-1 holds the potential to boost anti-tumor immunity and improve the efficacy of cancer immunotherapy. This review provides insights into the intricate role of Gal-1 in immune cell regulation, with an emphasis on T cells, and elucidates how tumors exploit Gal-1 for immune evasion and growth. Furthermore, we discuss the potential of Gal-1 as a therapeutic target to augment current immunotherapies across various cancer types.
Collapse
Affiliation(s)
- Julianna Novák
- Signal Transduction and Functional Genomics Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
| | - Tamás Takács
- Signal Transduction and Functional Genomics Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, Budapest, 1117, Hungary
| | - Álmos Tilajka
- Signal Transduction and Functional Genomics Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, Budapest, 1117, Hungary
| | - Loretta László
- Signal Transduction and Functional Genomics Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, Budapest, 1117, Hungary
| | - Orsolya Oravecz
- Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, Budapest, 1117, Hungary
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
| | - Emese Farkas
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Károly Rácz Conservative Medicine Division, Doctoral College, Semmelweis University, Budapest, 1091, Hungary
| | - Nándor Gábor Than
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, 1088, Hungary
| | - László Buday
- Signal Transduction and Functional Genomics Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Department of Molecular Biology, Semmelweis University, Budapest, 1094, Hungary
| | - Andrea Balogh
- Systems Biology of Reproduction Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary.
| | - Virág Vas
- Signal Transduction and Functional Genomics Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary.
| |
Collapse
|
50
|
Yan H, Liu Y, Wang M, Shu Z, Fang X, Li Z. Reactive Oxygen Species-Responsive Pyroptosis Nanoinitiators Promote Immune Cell Infiltration and Activate Anti-Tumor Immune Response. Int J Nanomedicine 2025; 20:4069-4084. [PMID: 40191041 PMCID: PMC11972606 DOI: 10.2147/ijn.s503580] [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: 11/18/2024] [Accepted: 03/23/2025] [Indexed: 04/09/2025] Open
Abstract
Background Immunotherapy, particularly immune checkpoint inhibitors, has become the standard treatment strategy for diverse malignant tumors. However, the inadequate infiltration of immune cells in tumors coupled with the immunosuppressive tumor microenvironment severely hinders the efficacy of immunotherapy. Methods A poly(ethylene glycol)-block-poly(lysine) copolymer (mPEG-b-PLL) was prepared through ring-opening polymerization and deprotection, and thioketal (TK) was attached to the amino group of mPEG-b-PLL via the condensation reaction to obtain mPEG-b-PLL-TK. Doxorubicin (DOX) and decitabine (DAC) were encapsulated in mPEG-b-PLL-TK to prepare the pyroptosis nanoinitiator (NP/(DAC+DOX)). The drug release behavior, cellular uptake, pyroptosis-triggering performance, and cytotoxicity of NP/(DAC+DOX) were evaluated in vitro experiments. The in vivo pharmacokinetics and biodistribution of NP/(DAC+DOX) were assessed through fluorescence imaging and high-performance liquid chromatography analysis. CT26 and 4T1 tumor-bearing mouse models were established to evaluate the anti-tumor efficacy, pyroptosis-triggering performance, and immune activation effects of NP/(DAC+DOX). Results NP/(DAC+DOX) exhibited excellent reactive oxygen species (ROS)-responsive drug release behavior and could be effectively taken up by tumor cells. Experiments both in vitro and in vivo demonstrated that NP/(DAC+DOX) effectively triggered pyroptosis in tumor cells, which was attributed to the DOX-induced activation of caspase-3 and the upregulation of GSDME expression caused by DAC. Following intravenous administration, NP/(DAC+DOX) specifically aggregated in tumor tissues. NP/(DAC+DOX) significantly suppressed tumor growth and extended the survival time of tumor-bearing mice. Furthermore, NP/(DAC+DOX) promoted dendritic cell maturation, enhanced the infiltration of cytotoxic T lymphocytes within the tumor, and decreased the proportion of myeloid-derived suppressor cells. Conclusion This study developed a ROS-responsive pyroptosis nanoinitiator to precisely induce the pyroptosis of tumor cells, thereby enhancing intratumoral immune cell infiltration and activating anti-tumor immune responses.
Collapse
Affiliation(s)
- Hongbo Yan
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Yilun Liu
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Maoshan Wang
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Zhenbo Shu
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Xuedong Fang
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Zhongmin Li
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| |
Collapse
|