51
|
Li YH, Huang XJ, Zhao XY. Translational study of the regulatory mechanism by which immune synapses enhance immune cell function. Cancer Lett 2025; 614:217542. [PMID: 39924076 DOI: 10.1016/j.canlet.2025.217542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/29/2025] [Accepted: 02/07/2025] [Indexed: 02/11/2025]
Abstract
Immune synapses, which were initially discovered at the interface between antigen-presenting cells (APCs) and T cells, are special structures formed at the contact site between antigen-presenting cells and immune cells and constitute the structural basis for immune cells to kill tumours and synthesise antibodies. Their structures are very similar to those of neural synapses in the nervous system, and they contain different functional structural regions. With the development of cell visualization research, scientists have increasingly conducted in-depth research on immune synapses. At present, it is known that T cells, B cells, and NK cells can form different immune synapses with target cells. Immune synapses formed by different cell subsets as well as CAR-T cells have their own characteristics, mainly in terms of their structure, formation process and regulatory mechanism. Therefore, how to enhance immune cell killing function by enhancing immune synaptic function has long been a research hotspot. At present, the killing function of immune cells can be enhanced by influencing the signalling molecules of immune synapses and the cell microenvironment and modifying the structure of immune synapses. Through a review of the factors affecting immune synapses, we can better explore the target for enhancing immune system function.
Collapse
Affiliation(s)
- Ya-Hui Li
- Peking University People's Hospital, Peking University Institute of Haematology, National Clinical Research Center for Haematologic Disease, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, Peking University, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Haematology, National Clinical Research Center for Haematologic Disease, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, Peking University, Beijing, China
| | - Xiang-Yu Zhao
- Peking University People's Hospital, Peking University Institute of Haematology, National Clinical Research Center for Haematologic Disease, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, Peking University, Beijing, China.
| |
Collapse
|
52
|
Galluzzi L. T cell exhaustion: early or late in tumour progression? Nat Rev Immunol 2025; 25:227-228. [PMID: 40038448 DOI: 10.1038/s41577-025-01158-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Affiliation(s)
- Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
53
|
Ahvati H, Roudi R, Sobhani N, Safari F. CD47 as a potent target in cancer immunotherapy: A review. Biochim Biophys Acta Rev Cancer 2025; 1880:189294. [PMID: 40057140 DOI: 10.1016/j.bbcan.2025.189294] [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: 09/24/2024] [Revised: 02/22/2025] [Accepted: 03/02/2025] [Indexed: 03/22/2025]
Abstract
Cancer is the second-highest cause of death worldwide. Accordingly, finding new cancer treatments is of great interest to researchers. The current platforms to fight cancer such as chemotherapy, radiotherapy, and surgery are limited in efficacy, especially in the metastatic setting. In this war against cancer, the immune system is a powerful ally, but tumor cells often outsmart it through alternative pathways. Cluster of differentiation 47 (CD47), a protein that normally prevents healthy cells from being attacked by immune cells, is often overexpressed on cancer cells. This makes CD47 a prime target for immunotherapy. Blocking of CD47 has the potential to unleash the immune system's cell populations-such as myeloid cells, macrophages, and T cells-to allow the immune system to discover and destroy cancer cells more successfully. In this review, we aimed to provide the latest information and findings about the roles of CD47 in the regulation of various cellular pathways and, thus, the importance of CD47 as a potential target in cancer therapy.
Collapse
Affiliation(s)
- Hiva Ahvati
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Raheleh Roudi
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA.
| | - Navid Sobhani
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fatemeh Safari
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| |
Collapse
|
54
|
Li Z, Zhao D, Zhu C. Predicting colorectal adenoma recurrence: the role of systemic inflammatory markers and insulin resistance. Scand J Gastroenterol 2025; 60:300-306. [PMID: 40009759 DOI: 10.1080/00365521.2025.2469801] [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: 12/07/2024] [Revised: 02/01/2025] [Accepted: 02/16/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Colorectal adenomas (CRA) exhibit high recurrence rates following endoscopic resection. Insulin resistance (IR) and chronic inflammation, increasingly prevalent due to unhealthy lifestyles, are key factors in CRA development. This study aimed to evaluate the predictive power of combining the inflammation score with the triglyceride-glucose (TyG) index for CRA recurrence. METHODS We conducted a comprehensive analysis of the clinical characteristics of 847 CRA patients who underwent endoscopic resection. Postoperative recurrence of CRA was assessed using logistic regression analyses to determine odds ratios (ORs) and 95% confidence intervals (CIs). The receiver operating characteristic (ROC) curve analysis was utilized to predict the risk of CRA recurrence based on the inflammation score and TyG index. RESULTS Among the 847 CRA included in the study, 126 experienced recurrences. Logistic regression analysis identified NLR (OR 2.641, 95% CI 1.982-3.549), TyG (OR 1.494, 95% CI 1.146-1.956), three or more adenomas (OR 2.182, 95% CI 1.431-3.322) and CRA larger than 10 mm (OR 1.917, 95% CI 1.267-2.921) as independent risk factors for CRA recurrence. ROC curves demonstrated the efficacy of NLR (AUC 0.701, 95% CI 0.652-0.750) and TyG (AUC 0.607, 95% CI 0.553-0.660) in predicting CRA recurrence. The combination of NLR, TyG and adenoma characteristics showed improved performance in predicting CRA recurrence (AUC 0.762, 95% CI 0.718-0.805). CONCLUSIONS Elevated NLR and TyG were associated with an increased risk of CRA recurrence. The integration of NLR and TyG with CRA characteristics significantly enhanced the predictive power for CRA recurrence.
Collapse
Affiliation(s)
- Zhiqiang Li
- Department of Gastroenterology, Beijing Zhongguancun Hospital, Beijing, China
| | - Danyun Zhao
- Department of Gastroenterology, Beijing Zhongguancun Hospital, Beijing, China
| | - Chunyan Zhu
- Department of Gastroenterology, Beijing Zhongguancun Hospital, Beijing, China
| |
Collapse
|
55
|
Yu Z, Cao L, Shen Y, Chen J, Li H, Li C, Yin JY, Li Y, Meng Y, Li X. Inducing Cuproptosis with Copper Ion-Loaded Aloe Emodin Self-Assembled Nanoparticles for Enhanced Tumor Photodynamic Immunotherapy. Adv Healthc Mater 2025; 14:e2404612. [PMID: 39998287 DOI: 10.1002/adhm.202404612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/05/2025] [Indexed: 02/26/2025]
Abstract
Immunotherapy has fundamentally transformed the clinical treatment landscape for non-small cell lung cancer (NSCLC). While its effectiveness is ultimately limited by patient heterogeneity and immunosuppressive tumor microenvironment. Photodynamic therapy (PDT), as an emerging antitumor immunotherapy, has shown its unique therapeutic advantages. However, previous studies often overlooked the potential toxicity of photosensitizers (PS), making the discovery of safe and effective PS a pressing clinical need. In this study, Aloe Emodin (AE), a medicinal plant natural compound, was loaded with copper ions (Cu), and self-assembled into nanoparticles (NPs) under the modification of PEG2k-DSPE-FA. NPs can target, accumulate, and reside within tumor sites, responsively releasing copper ions and AE, thus dual-functioning by inducing tumor cell death via cuproptosis and enhancing PDT effects. The LLC tumor-bearing mouse model demonstrated that NPs induce the maturation of dendritic cells (DCs) in vivo, promote lymphocyte infiltration, transform "cold tumors" into "hot tumors" and significantly enhance the efficacy of immune checkpoint blockade (ICB). This study provides experimental evidence of AE as a clinically promising PDT agent and offers a novel perspective for the synergistic treatment of clinical NSCLC.
Collapse
Affiliation(s)
- Zhen Yu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
| | - Lei Cao
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410008, P. R. China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410008, P. R. China
| | - Yue Shen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
| | - Jieqi Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
| | - Huizhen Li
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
| | - Chengmin Li
- Department of pathology and Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
| | - Ji-Ye Yin
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410008, P. R. China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410008, P. R. China
| | - Yueqin Li
- Institute of Integrative Medicine, Department of Integrated Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, P. R. China
| | - Yingcai Meng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
| | - Xiangping Li
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
| |
Collapse
|
56
|
Manini C, Larrinaga G, Angulo JC, López JI. Hot Spots in Urogenital Basic Cancer Research and Clinics. Cancers (Basel) 2025; 17:1173. [PMID: 40227699 PMCID: PMC11987958 DOI: 10.3390/cancers17071173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/28/2025] [Accepted: 03/29/2025] [Indexed: 04/15/2025] Open
Abstract
Urogenital cancer is very common in the male population of Western countries, a problem of major concern for public health systems, and a frequent test subject for oncological research. In this narrative, we identify the main hot topics for clinics and the basic science of urological cancer in the last few years (from 2021 onwards), considering the information given in the abstracts of almost 300 original articles published in outstanding journals of pathology, urology, and basic science. Once defined, for the top ten list of hot topics (the 2022 WHO update on the classification of urinary and male genital tumors, new entities in kidney cancer, urinary cancer-omics, update on the Gleason grading system, targeted therapies and other novel therapies in renal cancer, news on non-muscle invasive urothelial carcinoma, artificial intelligence in urologic cancer, intratumor heterogeneity influence in therapeutic failures in urologic neoplasms, intratumor microbiome and its influence in urologic tumor aggressiveness, and ecological principles and mathematics applied to urogenital cancer study), each issue is independently reviewed in an attempt to put together the most relevant updates and/or useful features accompanied by selected illustrations. This review article addresses some of the most interesting and current hot spots in urogenital basic cancer research and clinics and is mainly aimed toward clinicians, including pathologists, urologists, and oncologists. Readers are invited to explore each topic for further, more detailed information, in addition to the references provided.
Collapse
Affiliation(s)
- Claudia Manini
- Department of Pathology, San Giovanni Bosco Hospital, ASL Città di Torino, 10154 Turin, Italy;
| | - Gorka Larrinaga
- Department of Nursing, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Biobizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Javier C. Angulo
- Clinical Department, Faculty of Medical Sciences, European University of Madrid, 28905 Getafe, Spain;
| | - José I. López
- Biobizkaia Health Research Institute, 48903 Barakaldo, Spain
| |
Collapse
|
57
|
Zhong X, Zhang Z, Gao R, Ren S, Li S, Zhang M, Fang J, Hou Y. Multi-omics pan-cancer analysis reveals the diagnostic and prognostic value of C8orf76, with experimental validation of its impact on lung adenocarcinoma cell proliferation. Front Genet 2025; 16:1524422. [PMID: 40206508 PMCID: PMC11979282 DOI: 10.3389/fgene.2025.1524422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Background Chromosome 8 open reading frame 76 (C8orf76) is a nuclear protein-encoding gene, has received limited attention in current study. Multi-omics pan-cancer analysis focused on the diagnosis, prognosis, immune cell infiltration, methylation, and anti-cancer drug sensitivity remains an enigma. The effect of C8orf76 on lung adenocarcinoma (LUAD) is unknown. Methods Multi-omics pan-cancer analysis by utilizing datasets including UALCAN, TIMER 2.0, Human Protein Atlas (HPA), The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), cBioPortal, Gene Expression Profiling Interactive Analysis (GEPIA), OncoDB, and MethSurv datasets, were conducted to analyze C8orf76 across 33 cancer types. Furthermore, differential R packages were uesd for an in-depth analysis of C8orf76. The correlation between C8orf76 expression and diagnostic, prognosis, genetic alteration, mRNA modification, DNA methylation, lncRNA-miRNA-C8orf76 regulatory network, immune cell infiltration, and anti-tumor drugs response were explored to evaluate the potential roles of C8orf76. Most importantly, experiments including quantitative polymerase chain reaction (qPCR), RNA interference (RNAi), Western blotting (WB), and Edu staining, were performed for experimental verification. Results It was noted that the C8orf76 expression was markedly elevated across multiple tumor types. Moreover, C8orf76 showed potential as a diagnostic and prognostic biomarker. Besides, it was confirmed that the expression of C8orf76 was related to DNA methylation, mRNA modification, and the infiltration of immune cells. The lncRNA-miRNA-C8orf76 network was established in the study of LUAD. Experimental validation in LUAD A549 cells demonstrated that the knockdown of C8orf76 significantly inhibited cell proliferation in LUAD. Conclusion The present study is the first to report that the multi-omics pan-cancer analysis predicts C8orf76 as a promising target in cancer prognosis, diagnosis, immunology, and chemotherapy, highlighting its influence on cell proliferation in LUAD with experimental validation.
Collapse
Affiliation(s)
- Xiaohong Zhong
- Department of Clinical Laboratory, Qilu Hospital of Shandong University Dezhou Hospital (Dezhou People’s Hospital), Dezhou, Shandong, China
| | - Zhiyong Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University Dezhou Hospital (Dezhou People’s Hospital), Dezhou, Shandong, China
| | - Rongjing Gao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University Dezhou Hospital (Dezhou People’s Hospital), Dezhou, Shandong, China
| | - Shiqi Ren
- Department of Clinical Laboratory, Qilu Hospital of Shandong University Dezhou Hospital (Dezhou People’s Hospital), Dezhou, Shandong, China
| | - Shifang Li
- Department of Clinical Laboratory, Qilu Hospital of Shandong University Dezhou Hospital (Dezhou People’s Hospital), Dezhou, Shandong, China
| | - Miao Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University Dezhou Hospital (Dezhou People’s Hospital), Dezhou, Shandong, China
| | - Jie Fang
- Pneumology Department, Qilu Hospital of Shandong University Dezhou Hospital (Dezhou People’s Hospital), Dezhou, Shandong, China
| | - Yanjiao Hou
- Department of Clinical Laboratory, Qilu Hospital of Shandong University Dezhou Hospital (Dezhou People’s Hospital), Dezhou, Shandong, China
| |
Collapse
|
58
|
Chang J, Lu J, Liu Q, Xiang T, Zhang S, Yi Y, Li D, Liu T, Liu Z, Chen X, Dong Z, Li C, Yi H, Yu S, Huang L, Qu F, Wang M, Wang D, Dong H, Cheng G, Zhu L, Li J, Li C, Wu P, Xie X, Teschendorff AE, Lin D, Wang X, Wu C. Single-cell multi-stage spatial evolutional map of esophageal carcinogenesis. Cancer Cell 2025; 43:380-397.e7. [PMID: 40068596 DOI: 10.1016/j.ccell.2025.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 01/09/2025] [Accepted: 02/10/2025] [Indexed: 05/13/2025]
Abstract
Cancer development involves the co-evolution of cancer cells and their surrounding microenvironment, yet the dynamics of this interaction within the physical architecture remains poorly understood. Here, we present a spatial transcriptomic map at single-cell resolution, encompassing 127 multi-stage fields of view from 43 patients, to chart the evolutionary trajectories of human esophageal squamous cell carcinoma (ESCC). By analyzing 6.4 million cells, we reveal that ESCC progression is driven by a proliferative epithelial cell subpopulation that acquires dedifferentiated and invasive characteristics. At the late precancerous stage, these cells disrupt the epithelial-stromal interface and recruit normal fibroblasts via JAG1-NOTCH1 signaling, transforming them into cancer-associated fibroblasts (CAFs). This interaction leads to the formation of a "CAF-Epi" (CAF and epithelial cell) niche at the tumor edge that shields the tumor from immune surveillance. The CAF-Epi niche formation is a key indicator of progression in ESCC and other squamous cell carcinomas and patient outcomes.
Collapse
Affiliation(s)
- Jiang Chang
- Department of Health Toxicology, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Junting Lu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qingyi Liu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Tao Xiang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shaosen Zhang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yonglin Yi
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Dongxu Li
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Tianyuan Liu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Zeyuan Liu
- Changping Laboratory, Beijing 102206, China
| | - Xinjie Chen
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhenghao Dong
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Cainan Li
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - HanZhang Yi
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Siqi Yu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Luwei Huang
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100875, China
| | - Fangfei Qu
- Changping Laboratory, Beijing 102206, China
| | - Mengdi Wang
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100875, China
| | - Dehe Wang
- Changping Laboratory, Beijing 102206, China
| | - Hao Dong
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100875, China
| | - Guoyu Cheng
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Liang Zhu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jiachen Li
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Chenying Li
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Pujie Wu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiaoting Xie
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Dongxin Lin
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou 510060, China.
| | - Xiaoqun Wang
- Changping Laboratory, Beijing 102206, China; State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100875, China; State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, New Cornerstone Science Laboratory, Beijing Normal University, Beijing 100875, China.
| | - Chen Wu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China; CAMS Oxford Institute, Chinese Academy of Medical Sciences, Beijing 100006, China.
| |
Collapse
|
59
|
Song H, Chen L, Pan X, Shen Y, Ye M, Wang G, Cui C, Zhou Q, Tseng Y, Gong Z, Zhong B, Cui H, Mo S, Zheng J, Jin B, Zheng W, Luo F, Liu J. Targeting tumor monocyte-intrinsic PD-L1 by rewiring STING signaling and enhancing STING agonist therapy. Cancer Cell 2025; 43:503-518.e10. [PMID: 40068600 DOI: 10.1016/j.ccell.2025.02.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/29/2024] [Accepted: 02/11/2025] [Indexed: 05/13/2025]
Abstract
STING is an important DNA sensing machinery in initiating immune response, yet therapies targeting STING have shown poor outcomes in clinical trials. Here, we reveal that STING signaling induces PD-L1hi tumor monocytes (Tu.Mons) that dominate the resistance against STING agonist therapy. Cell-intrinsic PD-L1, induced by the STING-IRF3-IFN-I axis, is identified as the driving factor for protumoral PD-L1hi Tu.Mons. Notably, TLR2-activated Tu.Mons resist STING-induced upregulation of cell-intrinsic PD-L1 and the associated protumoral functions. Mechanistically, TLR2 stimulation remodels STING signaling by facilitating STING and TRAF6 interaction, which suppresses the IRF3-IFN-I response and enhances NF-κB activation. Moreover, we demonstrate that combining STING agonists with TLR2 agonist pretreatment significantly improves antitumor efficacy in murine syngeneic and humanized models. Our findings uncover a protumoral aspect of STING activation mediated by cell-intrinsic PD-L1 and propose a promising strategy to boost antitumor immunity by fine-tuning STING signaling outputs.
Collapse
Affiliation(s)
- Huan Song
- Department of Digestive Diseases, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lin Chen
- Department of Digestive Diseases, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China; State Key Laboratory of Genetic Engineering, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - Xuanxuan Pan
- Department of Digestive Diseases, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yuru Shen
- Department of Digestive Diseases, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Maolin Ye
- Department of Digestive Diseases, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Guohong Wang
- Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Can Cui
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Qi Zhou
- Department of Digestive Diseases, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yujen Tseng
- Department of Digestive Diseases, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zheng Gong
- Department of Digestive Diseases, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Bin Zhong
- Department of Digestive Diseases, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Haoshu Cui
- Department of Digestive Diseases, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shaocong Mo
- Department of Digestive Diseases, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jiayue Zheng
- Department of Digestive Diseases, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Bryan Jin
- Department of Digestive Diseases, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wanwei Zheng
- Department of Digestive Diseases, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Feifei Luo
- Department of Digestive Diseases, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Jie Liu
- Department of Digestive Diseases, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China; State Key Laboratory of Genetic Engineering, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200438, China.
| |
Collapse
|
60
|
Zhang X, Fang H, Wu W, Jiang C, Wang H, Shi Y. LPIN3 promotes colorectal cancer growth by dampening intratumoral CD8 + T cell effector function. Cancer Immunol Immunother 2025; 74:135. [PMID: 40042548 PMCID: PMC11883066 DOI: 10.1007/s00262-025-03989-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025]
Abstract
LPIN3 has emerged as a key factor in a variety of malignancies, although its precise role in colorectal cancer (CRC) remains unclear. By analyzing the data from The Cancer Genome Atlas, we discovered that the expression pattern of LPIN3 and the relevant makeup of the immune microenvironment were immensely diverse among tumors. LPIN3 is abundantly expressed in CRC and may enhance tumor growth by activating the β-catenin signaling pathway. In addition, we discovered that LPIN3 might reduce tumor antigen presentation signals, hence suppressing CD8+ T cell-mediated cytotoxicity. Furthermore, high expression of LPIN3 predicts decreased CD8+ T cell infiltration and effector function via bioinformatics analysis. Indeed, CD8+ T cell-mediated cytotoxicity as well as CD8+ T cell infiltration and activation in vivo were strengthened by LPIN3 knockdown. To sum up, our results highlight the part that LPIN3 plays in driving the progression of CRC by regulating β-catenin signaling and CD8+ T cell activity.
Collapse
Affiliation(s)
- Xiaoming Zhang
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hao Fang
- Department of Gastroenterology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Wenliang Wu
- Division of Gastrointestinal Surgery, Department of Surgery, Wuhan No.1 Hospital, Wuhan, 430022, China
| | - Congqing Jiang
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Haizhou Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Yifei Shi
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
61
|
Zhang L, Qian J, Zhang X, Lv Y, Zhao J, Wang S, Xu H. Dual‐Responsive PPy‐AIPH@LA Nanoplatform for Synergistic Photothermal and Thermodynamic Therapy of Colorectal Cancer. ChemistrySelect 2025; 10. [DOI: 10.1002/slct.202500263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 02/26/2025] [Indexed: 04/02/2025]
Abstract
AbstractIn this study, we introduce a novel nanoplatform, polypyrrole (PPy)‐2,2′‐Azobis[2‐(2‐imidazolin‐2‐yl)propane] dihydrochloride (AIPH)@lauric acid (LA) (PPy‐AIPH@LA) nanoparticles (NPs), designed to overcome these limitations through synergistic photothermal therapy (PTT) and photodynamic therapy (PDT). This dual‐responsive system incorporates PPy for efficient photothermal conversion, AIPH for thermos‐responsive and oxygen‐independent free radical generation, and LA as a thermally responsive encapsulation layer. The LA coating melts upon 808 nm near‐infrared laser irradiation, releasing AIPH and free radicals to enable precise spatiotemporal activation of therapeutic effects. PPy‐AIPH@LA demonstrates exceptional photothermal conversion efficiency (55.74%) and generates sufficient radicals to enhance PDT efficacy, even in hypoxic tumor microenvironments. In vitro studies revealed concentration‐dependent tumor cell ablation and inhibition of migration, while in vivo experiments showed that the combined PTT‐PDT treatment achieved an impressive 90.7% tumor growth inhibition rate in a mouse colon cancer cells CT‐26 murine model, with no significant systemic toxicity. Molecular analyses further revealed modulations in pathways associated with tumor metabolism, apoptosis, and immune escape, highlighting the comprehensive therapeutic potential of this nanoplatform. These findings underscore the potential of PPy‐AIPH@LA as a safe, effective, and minimally invasive nanotherapeutic platform for combating CRC and other solid tumors.
Collapse
Affiliation(s)
- Liang Zhang
- School of Materials and Chemistry University of Shanghai for Science and Technology No. 516 Jungong Road Shanghai 200093 P. R. China
- Department of Gastroenterology Changhai Hospital Naval Medical University No. 168 Changhai Road Shanghai 200433 P. R. China
| | - Jiahao Qian
- Department of Gastroenterology Changhai Hospital Naval Medical University No. 168 Changhai Road Shanghai 200433 P. R. China
| | - Xinyuan Zhang
- Department of Gastroenterology Changhai Hospital Naval Medical University No. 168 Changhai Road Shanghai 200433 P. R. China
| | - Yanwei Lv
- Department of Gastroenterology Changhai Hospital Naval Medical University No. 168 Changhai Road Shanghai 200433 P. R. China
| | - Jiulong Zhao
- Department of Gastroenterology Changhai Hospital Naval Medical University No. 168 Changhai Road Shanghai 200433 P. R. China
| | - Shige Wang
- School of Materials and Chemistry University of Shanghai for Science and Technology No. 516 Jungong Road Shanghai 200093 P. R. China
| | - Hao Xu
- Department of Gastroenterology Changhai Hospital Naval Medical University No. 168 Changhai Road Shanghai 200433 P. R. China
| |
Collapse
|
62
|
Dong S, Li X, Huang Q, Li Y, Li J, Zhu X, Xue C, Chen R, Zeng Y, Wu J, Zhong Y, Hu S. Resistance to immunotherapy in non-small cell lung cancer: Unraveling causes, developing effective strategies, and exploring potential breakthroughs. Drug Resist Updat 2025; 81:101215. [PMID: 40081220 DOI: 10.1016/j.drup.2025.101215] [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: 08/21/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 03/15/2025]
Abstract
Over the last two decades, advancements in deciphering the intricate interactions between oncology and immunity have fueled a meteoric rise in immunotherapy for non-small cell lung cancer, typified by an explosive growth of immune checkpoint inhibitors. However, resistance to immunotherapy remains inevitable. Herein we unravel the labyrinthine mechanisms of resistance to immunotherapy, characterized by their involvement of nearly all types of cells within the body, beyond the extrinsic cancer cells, and importantly, such cells are not only (inhibitory or excitatory, or both) signal recipients but also producers, acting in a context-dependent manner. At the molecular level, these mechanisms underlie genetic and epigenetic aberrations, which are regulated by or regulate various protein kinases, growth factors, and cytokines with inherently dynamic and spatially heterogeneous properties. Additionally, macroscopic factors such as nutrition, comorbidities, and the microbiome within and around organs or tumor cells are involved. Therefore, developing therapeutic strategies combined with distinct action informed by preclinical, clinical, and real-world evidence, such as radiotherapy, chemotherapy, targeted therapy, antibody-drug conjugates, oncolytic viruses, and cell-based therapies, may stand as a judicious reality, although the ideality is to overcome resistance point-by-point through a novel drug. Notably, we highlight a realignment of treatment aims, moving the primary focus from eliminating cancer cells -- such as through chemotherapy and radiotherapy -- to promoting immune modulation and underscore the value of regulating various components within the host macro- or micro-environment, as their effects, even if seemingly minimal, can cumulatively contribute to visible clinical benefit when applied in combination with ICIs. Lastly, this review also emphasizes the current hurdles scattered throughout preclinical and clinical studies, and explores evolving directions in the landscape of immunotherapy for NSCLC.
Collapse
Affiliation(s)
- Shuang Dong
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Xiaoyu Li
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Qing Huang
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yuanxiang Li
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | | | - Xianmin Zhu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Chang Xue
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Runzhi Chen
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yuan Zeng
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Jingyi Wu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Yi Zhong
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China.
| | - Sheng Hu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China.
| |
Collapse
|
63
|
He S, Luo S, Cai B, Chen J, Zhang Y, Zhao F, Liu Q, Liu T, Wang W, Peng T, Lu X, Zheng S. Divergent roles of PKM2 in regulating PD-L1 and PD-L2 expression and their implications in human and mouse cancer models. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 39980348 DOI: 10.3724/abbs.2025019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025] Open
Abstract
Cancer cells evade immune detection through checkpoint molecules like PD-L1 and PD-L2 which suppress T-cell activation. While PD-L1 is well-studied, the role of PD-L2 remains unclear. Pyruvate kinase M2 (PKM2), a metabolic enzyme, influences immune checkpoint regulation, but its role in PD-L1 and PD-L2 modulation is not well defined. Here, we investigate the role of pyruvate kinase M2 (PKM2) in modulating the immune checkpoint molecules PD-L1 and PD-L2 via GATA3 in cancer cells, with insights from both human and mouse models. We find that PKM2 enhances PD-L1 expression while inhibiting PD-L2, a dual regulatory mechanism that facilitates immune evasion. Knockdown and overexpression experiments revealed GATA3 as a key mediator. PKM2 knockout reduced GATA3 level, leading to decreased PD-L1 and increased PD-L2 expression. Chromatin immunoprecipitation (ChIP)-qPCR demonstrates that GATA3 functions as a direct transcription factor capable of binding to the promoters of PD-L1 and PD-L2. In silico analyses of 81 esophageal squamous cell carcinoma (ESCC) cases from the TCGA database demonstrate that PKM2 mRNA is unrelated to PD-L1 and PD-L2 expression but is negatively correlated with CD8 + T-cell infiltration in ESCC. To further validate these findings, we establish a xenograft model using immune-competent C57/BL6N mice, where knockdown of PKM2 results in significant downregulation of both PD-L1 and PD-L2 expression. Collectively, these findings underscore the divergent roles of PKM2 in regulating immune checkpoint expression in human and mouse cancer models and suggest that targeting the PKM2-GATA3 axis could enhance cancer immunotherapy by fine-tuning PD-L1 and PD-L2 levels.
Collapse
Affiliation(s)
- Shuo He
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
- Department of Pathology, Basic Medicine College, Xinjiang Medical University, Urumqi 830017, China
| | - Shujuan Luo
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
- Department of Pathology, Basic Medicine College, Xinjiang Medical University, Urumqi 830017, China
| | - Bangwu Cai
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
- Department of Pathology, Basic Medicine College, Xinjiang Medical University, Urumqi 830017, China
| | - Jiao Chen
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
| | - Yao Zhang
- Beijing Beanstalk International Bilingual School, Beijing, 100016
| | - Feng Zhao
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Disease, Operation Management Department, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011
| | - Qing Liu
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
| | - Tao Liu
- Department of Clinical Laboratory, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
| | - Wei Wang
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
| | - Tianyuan Peng
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
| | - Xiaomei Lu
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
| | - Shutao Zheng
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
- Department of Pathology, Basic Medicine College, Xinjiang Medical University, Urumqi 830017, China
| |
Collapse
|
64
|
Cheng X, Zhao H, Li Z, Yan L, Min Q, Wu Q, Zhan Q. Integrative analysis of T cell-mediated tumor killing-related genes reveals KIF11 as a novel therapeutic target in esophageal squamous cell carcinoma. J Transl Med 2025; 23:197. [PMID: 39966857 PMCID: PMC11834232 DOI: 10.1186/s12967-025-06178-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/25/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are emerging promising agents for the treatment of patients with esophageal squamous cell carcinoma (ESCC), however, there are only a small proportion respond to ICI therapy. Therefore, selecting candidate patients who will benefit the most from these drugs is critical. However, validated biomarkers for predicting immunotherapy response and overall survival are lacking. As the fundamental principle of ICI therapy is T cell-mediated tumor killing (TTK), we aimed to develop a unique TTK-related gene prognostic index (TTKPI) for predicting survival outcomes and responses to immune-based therapy in ESCC patients. METHODS Transcriptomic and clinical information of ESCC patients were from the GSE53625, GSE53624, GSE47404 and TCGA datasets. TTK-related genes were from the TISIDB database. The LASSO Cox regression model was employed to create the TTKPI. The prediction potential of the TTKPI was evaluated using the KM curve and time-dependent ROC curve analysis. Finally, the relationship between TTKPI and immunotherapy efficacy was investigated in clinical trials of ICIs (GSE91061, GSE135222, IMvigor210 cohort). The role of KIF11 in accelerating tumor progression was validated via a variety of functional experiments, including western blot, CCK-8, colony formation, wound healing scratch, and xenograft tumor model. The KIF11 expression was detected by multiplex fluorescent immunohistochemistry on tissue microarray from ESCC patients. RESULTS We constructed the TTKPI based on 8 TTK-related genes. The TTKPI low-risk patients exhibited better overall survival. TTKPI was significantly and positively correlated with the main immune checkpoint molecules levels. Furthermore, the low-risk patients were more prone to reap the benefits of immunotherapy in the cohort undergoing anti-PD-L1 therapy. Moreover, we performed functional experiments on KIF11, which ranked as the most significant prognostic risk gene among the 8 TTK-related genes. Our findings identified that KIF11 knockdown significantly hindered cell proliferation and mobility in ESCC cells. The KIF11 expression was negatively related with CD8+ T cell infiltration in ESCC patient samples. CONCLUSIONS The TTKPI is a promising biomarker for accurately determining survival and predicting the effectiveness of immunotherapy in ESCC patients. This risk indicator can help patients receive timely and precise early intervention, thereby advancing personalized medicine and facilitating precise immuno-oncology research. KIF11 plays a crucial role in driving tumor proliferation and migration and may act as a potential tumor biomarker of ESCC.
Collapse
Affiliation(s)
- Xinxin Cheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Huihui Zhao
- Department of Medical Oncology and Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhangwang Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Liping Yan
- Institute of Cytology and Genetics, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Qingjie Min
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Qingnan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Qimin Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
- Peking University International Cancer Institute, Beijing, 100142, China.
| |
Collapse
|
65
|
Galluzzi L, Guilbaud E, Garg AD. Mitochondrial succinate feeds T cell exhaustion in cancer. Cancer Cell 2025; 43:168-170. [PMID: 39933894 DOI: 10.1016/j.ccell.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/10/2025] [Accepted: 01/10/2025] [Indexed: 02/13/2025]
Abstract
Mitochondrial fitness is critical for effector CD8+ T cell responses against cancer. In this issue of Cancer Cell, Ma et al. delineate a novel mechanism linking defects in mitochondrial metabolism as elicited by prolyl 4-hydroxylase subunit alpha 1 (P4HA1) to T cell exhaustion and reduced tumor sensitivity to immunotherapy.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Emma Guilbaud
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Abhishek D Garg
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium.
| |
Collapse
|
66
|
Chen MH, Jiang J, Chen H, Wu RH, Xie W, Dai SZ, Zheng WP, Tan GH, Huang FY. Reinforcing cancer immunotherapy with engineered porous hollow mycobacterium tuberculosis loaded with tumor neoantigens. J Immunother Cancer 2025; 13:e010150. [PMID: 39915006 PMCID: PMC11804190 DOI: 10.1136/jitc-2024-010150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 01/20/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Enhancing antigen cross-presentation is essential for the development of a tumor neoantigen vaccine. One approach is to stimulate antigen-presenting cells (APCs) to uptake neoantigens. Mycobacterium tuberculosis (MTb) contains pathogen-associated molecular patterns (PAMPs) recognized by APCs and adhesion molecules that facilitate MTb invasion of APCs. Therefore, we suggest using MTb as a carrier to enhance APC phagocytosis of neoantigens, thereby promoting antigen cross-presentation. METHODS The successful preparation of the MTb carrier (phMTb) was confirmed through electron and confocal microscopy. Fluorescence microscopy was used to detect PAMPs and adhesion molecules on phMTb as well as to observe its role in aiding dendritic cells (DCs) in antigen uptake into endosomes or lysosomes. Flow cytometry was used to assess the retention of PAMPs and adhesion molecules on phMTb, investigate antigen uptake by DCs, evaluate their activation and maturation status, examine the presentation of tumor neoantigens, and analyze immune cells in draining lymph nodes and tumor tissues. The efficacy of phMTb vaccine formulations in combination with anti-programmed cell death protein 1 (PD-1) antibody therapy was assessed using the MC38 mouse tumor models. Adverse effects were evaluated through H&E staining of major organs, assessment of reproductive capability and detection of biochemical indices. RESULTS The engineered porous hollow phMTb carrier successfully encapsulated model tumor neoantigens, with or without the adjuvant CpG. The phMTb retained PAMPs and adhesion molecules on its surface, similar to the parental MTb, thereby enhancing DC uptake of phMTb and its formulations containing tumor neoantigens and CpG. Vaccines formulated with phMTb facilitated DC maturation, activation, cross-presentation of tumor neoantigens, and promoted migration of phMTb-laden DCs to lymph nodes, enhancing effector and memory CD8+ T lymphocyte function. In murine tumor models, immunization with phMTb-formulated neoantigen vaccines elicited a robust tumor-specific cytotoxic T lymphocyte immune response with minimal adverse effects. Additionally, vaccination with phMTb-formulated neoantigen vaccines effectively reversed the tumor's immune-suppressive microenvironment. Concurrent administration of the PD-1 antibody with the phMTb-formulated neoantigen vaccine exhibited significant synergistic therapeutic effects. CONCLUSIONS The results of our study highlight the potential clinical translation of personalized tumor neoantigen vaccines using the phMTb carrier.
Collapse
Affiliation(s)
- Ming-Hui Chen
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Jie Jiang
- Public Research Center, Hainan Medical University, Haikou, Hainan, China
| | - Hengyu Chen
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Ri-Hong Wu
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Weijing Xie
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Shu-Zhen Dai
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Wu-Ping Zheng
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Guang-Hong Tan
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Feng-Ying Huang
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
67
|
Lv J, Zhang Y, Wu Q, Jiang P, Lin Y. Inhibition of SIRT4 promotes bladder cancer progression and immune escape via attenuating CD8 + T cells function. Int Immunopharmacol 2025; 147:113906. [PMID: 39756164 DOI: 10.1016/j.intimp.2024.113906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Bladder cancer (BCa) is one of the most common malignancies of the urinary system and is characterized by a high recurrence rate and significant mortality. Sirtuin 4 (SIRT4), a member of the NAD+-dependent deacetylase and ADP-ribosyltransferase family, is involved in regulating cellular metabolism, DNA repair, and longevity, potentially influencing tumor progression and immune escape. This study aimed to elucidate the role of SIRT4 in BCa. METHODS The correlation between the sirtuin family and immunotherapy sensitivity in BCa patients was analyzed via IMvigor210 data. The clinical significance and immunological role of SIRT4 across multiple cancer types were assessed by evaluating its associations with clinicopathologic features, prognosis, tumor mutation burden (TMB), microsatellite instability (MSI), immune cell infiltration, and immune response genes across 33 datasets from The Cancer Genome Atlas (TCGA). SIRT4 expression was confirmed in BCa tissues, and its functions were examined via proliferation and migration assays. CD8+ T cells were isolated from the peripheral blood of healthy individuals and activated with CD3 and CD28 antibodies and recombinant IL2. Coculture assays involving BCa cells and activated CD8+ T cells, alongside ELISA, were conducted to evaluate the immunological function of SIRT4. RESULTS SIRT4 was positively associated with the immunotherapy response of BCa patients on the basis of IMvigor210 data. Its expression was downregulated in 11 tumor types but upregulated in 3. SIRT4 was significantly correlated with tumor stage in 2 tumor types and showed varying associations with overall survival, progression-free survival, and disease-specific survival. Additionally, SIRT4 was correlated with TMB in 10 tumor types and with MSI in 8. GSEA indicated that SIRT4 was negatively associated with the immune response in 9 tumor types, excluding BCa. It was positively correlated with immune cell infiltration in 2 tumor types and negatively correlated in 6. The TCGA data revealed that SIRT4 was positively associated with activated NK cell infiltration but negatively associated with M1 macrophages, neutrophils, resting NK cells, and activated memory CD4 T cells. Enrichment analyses revealed positive correlations with various chemokines, immunoinhibitors, immunostimulators, lymphocytes, MHC molecules, and MHC receptors, suggesting that SIRT4 may enhance the immune response in BCa. Further experiments confirmed that SIRT4 was downregulated in BCa tissues compared with adjacent normal tissues. Inhibition of SIRT4 promoted BCa cell proliferation and migration, whereas knockdown of SIRT4 impaired the chemotaxis and tumor-killing ability of CD8+ T cells in the BCa tumor microenvironment. CONCLUSIONS In summary, SIRT4 inhibits the progression and immune escape of BCa, indicating its potential as a novel biomarker and immune checkpoint for immunotherapy.
Collapse
Affiliation(s)
- Jiancheng Lv
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yu Zhang
- Department of Urology, Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| | - Qikai Wu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Peng Jiang
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Yiwei Lin
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
68
|
Ning B, Chiu DJ, Pfefferkorn RM, Kefella Y, Kane E, Reyes-Ortiz V, Liu G, Zhang S, Liu H, Sultan L, Green E, Constant M, Spira AE, Campbell JD, Reid ME, Varelas X, Burks EJ, Lenburg ME, Mazzilli SA, Beane JE. Epithelial miR-149-5p up-regulation is associated with immune evasion in progressive bronchial premalignant lesions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636307. [PMID: 39975222 PMCID: PMC11838605 DOI: 10.1101/2025.02.03.636307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The molecular drivers bronchial premalignant lesion progression to invasive lung squamous cell carcinoma are not well defined. Prior work profiling longitudinally collected bronchial premalignant lesion biopsies by RNA sequencing defined a proliferative subtype, enriched with bronchial dysplasia. We found that a gene co-expression module associated with interferon gamma signaling and antigen processing/presentation was down-regulated in progressive/persistent versus regressive lesions within the proliferative subtype, suggesting a functional impact of these genes on immune evasion. RNA from these same premalignant lesions was profiled by microRNA (miRNA) sequencing and a miRNA-gene network analysis identified hsa-miR-149-5p as a potential regulator of this antigen presentation gene co-expression module associated with lesion progression. hsa-miR-149-5p was found to be predominantly expressed in the epithelium and up-regulated in progressive/persistent versus regressive proliferative lesions while targets of this miRNA, the transcriptional coactivator of MHC-I gene expression, NLRC5 , and the genes it regulates were down-regulated. MicroRNA in situ hybridization of hsa-miR-149-5p in tissue from adjacent fixed biopsies showed that hsa-miR-149-5p was increased in areas of bronchial dysplasia in progressive/persistent versus regressive lesions. Imaging mass cytometry showed that NLRC5 protein expression was decreased in progressive/persistent versus regressive lesions within areas of hyperplasia, metaplasia, and dysplasia. Additionally, basal cells with high versus low levels of NLRC5 were found to be in close spatial proximity to CD8 T cells, suggesting that these cells exhibit increased functional MHC-I gene expression in lesions with low hsa-miR-149-5p expression. Collectively, our data suggests a functional role for hsa-miR-149-5p in bronchial premalignant lesions and may serve as a therapeutic target for PML immunomodulation. STATEMENT OF SIGNIFICANCE Integrative analysis across bronchial premalignant lesions has identified and localized a potential regulator of immune evasion in progressive/persistent lesions that could be a novel therapeutic target.
Collapse
|
69
|
Baumann AA, Buribayev Z, Wolkenhauer O, Salybekov AA, Wolfien M. Epigenomic Echoes-Decoding Genomic and Epigenetic Instability to Distinguish Lung Cancer Types and Predict Relapse. EPIGENOMES 2025; 9:5. [PMID: 39982247 PMCID: PMC11843950 DOI: 10.3390/epigenomes9010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/17/2025] [Accepted: 02/02/2025] [Indexed: 02/22/2025] Open
Abstract
Genomic and epigenomic instability are defining features of cancer, driving tumor progression, heterogeneity, and therapeutic resistance. Central to this process are epigenetic echoes, persistent and dynamic modifications in DNA methylation, histone modifications, non-coding RNA regulation, and chromatin remodeling that mirror underlying genomic chaos and actively influence cancer cell behavior. This review delves into the complex relationship between genomic instability and these epigenetic echoes, illustrating how they collectively shape the cancer genome, affect DNA repair mechanisms, and contribute to tumor evolution. However, the dynamic, context-dependent nature of epigenetic changes presents scientific and ethical challenges, particularly concerning privacy and clinical applicability. Focusing on lung cancer, we examine how specific epigenetic patterns function as biomarkers for distinguishing cancer subtypes and monitoring disease progression and relapse.
Collapse
Affiliation(s)
- Alexandra A. Baumann
- Department of Systems Biology and Bioinformatics, Institute of Computer Science, University of Rostock, 18051 Rostock, Germany; (A.A.B.)
- Faculty of Medicine Carl Gustav Carus, Institute for Medical Informatics and Biometry, TUD Dresden University of Technology, 01069 Dresden, Germany
| | - Zholdas Buribayev
- Department of Computer Science, Faculty of Information Technologies, Al-Farabi Kazakh National University, 050040 Almaty, Kazakhstan
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, Institute of Computer Science, University of Rostock, 18051 Rostock, Germany; (A.A.B.)
- Leibniz-Institute for Food Systems Biology, Technical University of Munich, 80333 Freising, Germany
- Stellenbosch Institute of Advanced Study, Wallenberg Research Centre, Stellenbosch University, Stellenbosch 7535, South Africa
| | - Amankeldi A. Salybekov
- Regenerative Medicine Division, Cell and Gene Therapy Department, Qazaq Institute of Innovative Medicine, 010000 Astana, Kazakhstan
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan
| | - Markus Wolfien
- Faculty of Medicine Carl Gustav Carus, Institute for Medical Informatics and Biometry, TUD Dresden University of Technology, 01069 Dresden, Germany
- Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI), 01069 Dresden, Germany
| |
Collapse
|
70
|
Kelly JJ, Newkirk SE, Chordia MD, Pires MM. Evaluation and In Situ Library Expansion of Small Molecule MHC-I Inducers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.635109. [PMID: 39975032 PMCID: PMC11838524 DOI: 10.1101/2025.01.31.635109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Immunotherapy has emerged as a powerful strategy for combating cancer by harnessing the patient immune system to recognize and eliminate malignant cells. The major histocompatibility complex class I (MHC-I) has a pivotal role in the recognition step. These surface proteins present cancer-specific neoantigens to CD8+ T cells, which triggers activation and T cell-mediated killing. However, cancer cells can often evade immune detection by downregulating MHC-I surface expression, which renders the immune response less effective. In turn, this resistance mechanism offers an opportunity to bolster MHC-I surface expression via therapeutic interventions. Here, we conducted an initial comprehensive evaluation of previously purported small molecule MHC-I inducers and identified heat shock protein 90 (Hsp90) inhibitors as privileged inducers of MHC-I surface expression. With a core scaffold in hand, we employed an in situ click chemistry-based derivatization strategy to generate 380 novel compounds in the same family. New agents from this library showed high levels of induction, with one of the triazole-based analogs, CliMB-325, also enhancing T cell activation and exhibiting lower toxicity, which could potentiate some immunotherapeutic modalities. Moreover, we demonstrated the potential of a click chemistry-based diversification strategy for the discovery of small molecules to counter immune evasion.
Collapse
Affiliation(s)
- Joey J. Kelly
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States 22904
- Department of Microbiology, Immunology, and Cancer, University of Virginia, Charlottesville, VA, United States 22904
| | - Sarah E. Newkirk
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States 22904
- Department of Microbiology, Immunology, and Cancer, University of Virginia, Charlottesville, VA, United States 22904
| | - Mahendra D. Chordia
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States 22904
- Department of Microbiology, Immunology, and Cancer, University of Virginia, Charlottesville, VA, United States 22904
| | - Marcos M. Pires
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States 22904
- Department of Microbiology, Immunology, and Cancer, University of Virginia, Charlottesville, VA, United States 22904
| |
Collapse
|
71
|
Galluzzi L, Manic G, Vitale I. Effective immunosurveillance of clonally expanded mammary aneuploid cells. Cell Mol Immunol 2025; 22:131-133. [PMID: 39668162 PMCID: PMC11782682 DOI: 10.1038/s41423-024-01250-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024] Open
Affiliation(s)
- Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Gwenola Manic
- Italian Institute for Genomic Medicine, c/o IRCSS Candiolo, Torino, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Ilio Vitale
- Italian Institute for Genomic Medicine, c/o IRCSS Candiolo, Torino, Italy.
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
| |
Collapse
|
72
|
Sirera R, Beltrán-Visiedo M, Galluzzi L. A novel pharmacological entity toward integrated multimodal immunotherapy. Trends Pharmacol Sci 2025; 46:95-97. [PMID: 39721827 DOI: 10.1016/j.tips.2024.12.001] [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: 11/16/2024] [Revised: 12/04/2024] [Accepted: 12/04/2024] [Indexed: 12/28/2024]
Abstract
Most solid tumors are insensitive to single-agent immunotherapy, calling for the development of combinatorial treatment regimens. Recently, Lin and collaborators developed a pharmacological platform enabling the combination of different immunotherapies into a single chemical entity. This approach may effectively circumvent obstacles associated with the simultaneous delivery of multiple immunotherapeutic agents.
Collapse
Affiliation(s)
- Rafael Sirera
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA; Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain; Unidad Mixta TRIAL, Centro Investigación Príncipe Felipe-Fundación Investigación, Hospital General Universitario de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red Cáncer, CIBERONC, Madrid, Spain
| | - Manuel Beltrán-Visiedo
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
73
|
Shi S, Ou X, Liu C, Li R, Zheng Q, Hu L. NF-κB signaling and the tumor microenvironment in osteosarcoma: implications for immune evasion and therapeutic resistance. Front Immunol 2025; 16:1518664. [PMID: 39949765 PMCID: PMC11821961 DOI: 10.3389/fimmu.2025.1518664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/10/2025] [Indexed: 02/16/2025] Open
Abstract
Osteosarcoma, a highly aggressive malignancy with a generally poor prognosis, is characterized by tumor cells' ability to evade immune responses and resist treatment. The nuclear transcription factor NF-κB signaling pathway is crucial in regulating inflammatory and immune reactions. It occupies a central position in the development of the osteosarcoma tumor microenvironment. This research aimed to explore how NF-κB influences the recruitment and polarization of tumor-associated macrophages and myeloid-derived suppressor cells, both of which contribute to immunosuppression. Furthermore, NF-κB facilitates immune surveillance evasion in osteosarcoma cells by altering the expression of immune checkpoint molecules, such as PD-L1. It also enhances tumor cell resistance to chemotherapy and radiotherapy by activating anti-apoptotic signaling pathways and exacerbating treatment-induced inflammation. Potential therapeutic approaches include using NF-κB inhibitors, possibly in combination with immune checkpoint inhibitors, to overcome tumor cell resistance mechanisms and reshape antitumor immune responses. A thorough examination of NF-κB's role in osteosarcoma development is expected to yield novel clinical treatment strategies, and significantly improve patient prognosis by targeting this key signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | - Leiming Hu
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, XI’an, China
| |
Collapse
|
74
|
Chen W, Baker T, Zhang Z, Ogilvie HA, Van Loo P, Gu S(S. Evolutionary trajectories of immune escape across cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.632799. [PMID: 39868264 PMCID: PMC11761017 DOI: 10.1101/2025.01.17.632799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Immune escape is a critical hallmark of cancer progression and underlies resistance to multiple immunotherapies. However, it remains unclear when the genetic events associated with immune escape occur during cancer development. Here, we integrate functional genomics studies of immunomodulatory genes with a tumor evolution reconstruction approach to infer the evolution of immune escape across 38 cancer types from the Pan-Cancer Analysis of Whole Genomes dataset. Different cancers favor mutations in different immunomodulatory pathways. For example, the antigen presentation machinery is highly mutated in colorectal adenocarcinoma, lung squamous cell carcinoma, and chromophobe renal cell carcinoma, and the protein methylation pathway is highly mutated in bladder transitional cell carcinoma and lung adenocarcinoma. We also observe different timing patterns in multiple immunomodulatory pathways. For instance, mutations impacting genes involved in cellular amino acid metabolism were more likely to happen late in pancreatic adenocarcinoma. Mutations in the glucocorticoid receptor regulatory network pathway tended to occur early, while mutations in the TNF pathways were more likely to occur late in B-cell non-Hodgkin lymphoma. Mutations in the NOD1/2 signaling pathway and DNA binding transcription factor activity tended to happen late in breast adenocarcinoma and ovarian adenocarcinoma. Together, these results delineate the evolutionary trajectories of immune escape in different cancer types and highlight opportunities for improved immunotherapy of cancer.
Collapse
Affiliation(s)
- Wenjie Chen
- Department of Hematopoietic Biology & Malignancy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Toby Baker
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The Francis Crick Institute, London, United Kingdom
- Department of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Zhihui Zhang
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Huw A. Ogilvie
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Peter Van Loo
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The Francis Crick Institute, London, United Kingdom
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shengqing (Stan) Gu
- Department of Hematopoietic Biology & Malignancy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
75
|
Błaszczak E, Miziak P, Odrzywolski A, Baran M, Gumbarewicz E, Stepulak A. Triple-Negative Breast Cancer Progression and Drug Resistance in the Context of Epithelial-Mesenchymal Transition. Cancers (Basel) 2025; 17:228. [PMID: 39858010 PMCID: PMC11764116 DOI: 10.3390/cancers17020228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 12/30/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the most difficult subtypes of breast cancer to treat due to its distinct clinical and molecular characteristics. Patients with TNBC face a high recurrence rate, an increased risk of metastasis, and lower overall survival compared to other breast cancer subtypes. Despite advancements in targeted therapies, traditional chemotherapy (primarily using platinum compounds and taxanes) continues to be the standard treatment for TNBC, often with limited long-term efficacy. TNBC tumors are heterogeneous, displaying a diverse mutation profile and considerable chromosomal instability, which complicates therapeutic interventions. The development of chemoresistance in TNBC is frequently associated with the process of epithelial-mesenchymal transition (EMT), during which epithelial tumor cells acquire a mesenchymal-like phenotype. This shift enhances metastatic potential, while simultaneously reducing the effectiveness of standard chemotherapeutics. It has also been suggested that EMT plays a central role in the development of cancer stem cells. Hence, there is growing interest in exploring small-molecule inhibitors that target the EMT process as a future strategy for overcoming resistance and improving outcomes for patients with TNBC. This review focuses on the progression and drug resistance of TNBC with an emphasis on the role of EMT in these processes. We present TNBC-specific and EMT-related molecular features, key EMT protein markers, and various signaling pathways involved. We also discuss other important mechanisms and factors related to chemoresistance in TNBC within the context of EMT, highlighting treatment advancements to improve patients' outcomes.
Collapse
Affiliation(s)
- Ewa Błaszczak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland
| | | | | | | | | | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland
| |
Collapse
|
76
|
Nunzi I, D'Achille G, Dhaouadi N, Marcheggiani F, Licini C, Di Vincenzo M, Orciani M, Morroni G, Marchi S. Monitoring cellular dynamics upon infection using a holotomography-based approach. Methods Cell Biol 2025; 194:109-118. [PMID: 40058955 DOI: 10.1016/bs.mcb.2024.12.003] [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: 05/13/2025]
Abstract
Many intracellular bacteria interfere with mitochondrial dynamics or target other organelles, thereby inducing a specific cellular response that could emerge as a strategy of the pathogen to ensure its survival, or as a form of defense employed by the host cell to restrict dissemination. In this context, the concomitant monitoring of both pathogen migration and (intra)cellular dynamics in live cells emerges as a pivotal aspect for the comprehension of the infection sequence and to visualize the pathogen-mediated remodeling that could occur to the entire cellular system. Holotomographic microscopy can be used to achieve this goal, allowing the simultaneous analysis of both bacterial movement and intracellular alteration for extended periods of time, with high spatial resolution and avoiding side-effects due to phototoxicity. Here we provide a holotomography-based approach to detect Listeria monocytogenes dynamics and its effects on the entire cellular system at morphological level.
Collapse
Affiliation(s)
- Ilaria Nunzi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Gloria D'Achille
- Microbiology Unit, Department of Biomedical Sciences and Public Health, Marche Polytechnic University, Ancona, Italy
| | - Nada Dhaouadi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Fabio Marcheggiani
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Caterina Licini
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Mariangela Di Vincenzo
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Monia Orciani
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Gianluca Morroni
- Microbiology Unit, Department of Biomedical Sciences and Public Health, Marche Polytechnic University, Ancona, Italy.
| | - Saverio Marchi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy; Advanced Technology Center for Aging Research, IRCCS INRCA, Ancona, Italy.
| |
Collapse
|
77
|
Sirera R, Beltrán-Visiedo M, Galluzzi L. Targeting immune evasion in hepatocellular carcinoma-initiating cells. Trends Immunol 2025; 46:4-6. [PMID: 39721855 DOI: 10.1016/j.it.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024]
Abstract
Tumor-initiating cells (TICs) are particularly efficient at evading detection and elimination by the human immune system. Recent data from Yang and collaborators demonstrate that - at least in preclinical hepatocellular carcinoma models - the immunological privilege of CD49f+ TICs can be limited by targeting CD155, resulting in restored sensitivity to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Rafael Sirera
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA; Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain; Unidad Mixta TRIAL, Centro Investigación Príncipe Felipe-Fundación Investigación, Hospital General Universitario de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red Cáncer, CIBERONC, Madrid, Spain
| | - Manuel Beltrán-Visiedo
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
78
|
Galluzzi L, Spada S. Circulating biomarkers for diagnosis, prognosis, and treatment response prediction in cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2025; 391:xiii-xvii. [PMID: 39939080 DOI: 10.1016/s1937-6448(25)00031-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
Affiliation(s)
- Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, United States; Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy.
| | - Sheila Spada
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, United States; Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
79
|
Catanzaro E, Beltrán-Visiedo M, Galluzzi L, Krysko DV. Immunogenicity of cell death and cancer immunotherapy with immune checkpoint inhibitors. Cell Mol Immunol 2025; 22:24-39. [PMID: 39653769 PMCID: PMC11685666 DOI: 10.1038/s41423-024-01245-8] [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: 10/17/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024] Open
Abstract
While immunotherapy with immune checkpoint inhibitors (ICIs) has revolutionized the clinical management of various malignancies, a large fraction of patients are refractory to ICIs employed as standalone therapeutics, necessitating the development of combinatorial treatment strategies. Immunogenic cell death (ICD) inducers have attracted considerable interest as combinatorial partners for ICIs, at least in part owing to their ability to initiate a tumor-targeting adaptive immune response. However, compared with either approach alone, combinatorial regimens involving ICD inducers and ICIs have not always shown superior clinical activity. Here, we discuss accumulating evidence on the therapeutic interactions between ICD inducers and immunotherapy with ICIs in oncological settings, identify key factors that may explain discrepancies between preclinical and clinical findings, and propose strategies that address existing challenges to increase the efficacy of these combinations in patients with cancer.
Collapse
Affiliation(s)
- Elena Catanzaro
- Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Manuel Beltrán-Visiedo
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Dmitri V Krysko
- Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.
| |
Collapse
|
80
|
Bergeron P, Milliat F, Deutsch E, Mondini M. Heterogeneous intratumor irradiation: a new partner for immunotherapy. Oncoimmunology 2024; 13:2434280. [PMID: 39589158 PMCID: PMC11601051 DOI: 10.1080/2162402x.2024.2434280] [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: 11/17/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024] Open
Abstract
We recently demonstrated that a heterogeneous tumor irradiation strategy, combining high-dose and low-dose radiotherapy (RT) within the same tumor volume, can synergize with immunotherapy in mice. Our findings indicate that heterogeneous RT doses may promote the spatial diversification of the antitumor immune response. Spatial fractionation of the RT dose has the potential to enhance the therapeutic index of RT/IO combinations, particularly in scenarios where irradiating the entire tumor volume is unfeasible or excessively harmful to the patient.
Collapse
Affiliation(s)
- Paul Bergeron
- INSERM U1030, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Fabien Milliat
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PSE-SANTE/SERAMED/LRMed, Fontenay-aux-Roses, France
| | - Eric Deutsch
- INSERM U1030, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Michele Mondini
- INSERM U1030, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
81
|
Santoso A, Levink I, Pihlak R, Chau I. The Immune Landscape and Its Potential for Immunotherapy in Advanced Biliary Tract Cancer. Curr Oncol 2024; 32:24. [PMID: 39851940 PMCID: PMC11763487 DOI: 10.3390/curroncol32010024] [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: 12/19/2024] [Accepted: 12/25/2024] [Indexed: 01/26/2025] Open
Abstract
Biliary tract cancers (BTC) are a highly heterogeneous group of cancers at the genomic, epigenetic and molecular levels. The vast majority of patients initially present at an advanced (unresectable) disease stage due to a lack of symptoms and an aggressive tumour biology. Chemotherapy has been the mainstay of treatment in patients with advanced BTC but the survival outcomes and prognosis remain poor. The addition of immune checkpoint inhibitors (ICI) to chemotherapy have shown only a marginal benefit over chemotherapy alone due to the complex tumour immune microenvironment of these cancers. This review appraises our current understanding of the immune landscape of advanced BTC, including emerging transcriptome-based classifications, highlighting the mechanisms of immune evasion and resistance to ICI and their therapeutic implications. It describes the shifting treatment paradigm from traditional chemotherapy to immunotherapy combinations as well as the potential biomarkers for predicting response to ICI.
Collapse
Affiliation(s)
- Andry Santoso
- Gastrointestinal Unit, The Royal Marsden Hospital, London SW3 6JJ, UK; (A.S.); (I.L.)
| | - Iris Levink
- Gastrointestinal Unit, The Royal Marsden Hospital, London SW3 6JJ, UK; (A.S.); (I.L.)
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Centre, 3015 GD Rotterdam, The Netherlands
| | - Rille Pihlak
- University Hospitals Sussex NHS Foundation Trust, Brighton BN1 9RW, UK;
| | - Ian Chau
- Gastrointestinal Unit, The Royal Marsden Hospital, London SW3 6JJ, UK; (A.S.); (I.L.)
| |
Collapse
|
82
|
Liu P, Zhao L, Kepp O, Kroemer G. Cytoplasmic HMGB2 orchestrates CALR translocation in the course of immunogenic cell death. Oncoimmunology 2024; 13:2421028. [PMID: 39585160 PMCID: PMC11520571 DOI: 10.1080/2162402x.2024.2421028] [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: 10/17/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/26/2024] Open
Abstract
A recent in vitro study showed that pharmacological inhibition of the nuclear export receptor XPO1 suppresses oxaliplatin-induced nuclear release of HMGB1 and HMGB2, as well as the translocation of CALR to the plasma membrane. Moreover, cell-targeted-HMGB2 protein potently induced CALR exposure, even in the absence of oxaliplatin.
Collapse
Affiliation(s)
- Peng Liu
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Liwei Zhao
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
- Department of Biology, Institut du Cancer Paris CARPEM, Paris, France
| |
Collapse
|
83
|
Arrizabalaga L, García-Torralba E, Galluzzi L, Buqué A. Targeting CDK2 to circumvent treatment resistance in HR + breast cancer. Trends Mol Med 2024:S1471-4914(24)00342-3. [PMID: 39732571 DOI: 10.1016/j.molmed.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/30/2024]
Abstract
Genetic and epigenetic defects of the p53 system have previously been associated with resistance to CDK4/6 inhibitors in women with HR+ breast cancer. Recent data from Kudo et al. demonstrate that CDK2-targeting agents may offer an effective strategy to circumvent such resistance by enforcing cellular senescence downstream of RBL2 dephosphorylation.
Collapse
Affiliation(s)
- Leire Arrizabalaga
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA; Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Esmeralda García-Torralba
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA; Department of Medical Oncology, Hospital Universitario Morales Meseguer, Murcia, Spain; Department of Medicine, Medical School, University of Murcia, Murcia, Spain; IMIB-Arrixaca, Murcia, Spain
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Aitziber Buqué
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
84
|
Liu H, Liu X, Lu Y. The roles of LncRNA CARMN in cancers: biomarker potential, therapeutic targeting, and immune response. Discov Oncol 2024; 15:776. [PMID: 39692999 DOI: 10.1007/s12672-024-01679-6] [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: 09/22/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024] Open
Abstract
Long non-coding RNAs (LncRNAs) are crucial regulators of gene expression and cellular processes, with significant implications for cancer research. This review focuses on the role of LncRNA CARMN (Cardiac Arrest and Regulated Myocyte Nuclear Protein) in various cancers. CARMN, originally identified for its function in cardiac tissues, has shown dysregulated expression in several tumor types, including cervical, breast, colorectal, and esophageal cancers. Its altered expression often correlates with tumor progression, metastasis, and patient prognosis, suggesting its potential as both a biomarker and therapeutic target. In cervical cancer, CARMN's role as a tumor suppressor is highlighted by its ability to inhibit cell proliferation, migration, and invasion through interaction with the miR-92a-3p/BTG2 axis and modulation of the Wnt/β-catenin signaling pathway. In breast cancer, CARMN acts as an enhancer RNA, affecting epithelial-mesenchymal transition and metastasis by regulating MMP2 via DHX9. The downregulation of CARMN in triple-negative breast cancer is associated with enhanced sensitivity to chemotherapy. In colorectal cancer, CARMN's expression is regulated by m6A methylation and mutant p53, influencing tumor growth through miR-5683 and FGF2. Lastly, in esophageal cancer, genetic variations in CARMN affect cancer susceptibility, with certain SNPs and haplotypes associated with either increased or decreased risk. Additionally, the relationship between CARMN and immune cell dynamics highlights its potential role in cancer immune surveillance and therapy. Finally, we found that CARMN may regulate immune cell exhaustion in the tumor microenvironment by influencing the recruitment and activation of NK cells and T cells, as well as modulating macrophage polarization. This review emphasizes the diverse roles of CARMN across different cancers and its potential as a diagnostic and therapeutic tool. Future research should address the mechanistic details of CARMN's involvement in cancer, validate its clinical utility, and explore its therapeutic potential in combination with existing treatments.
Collapse
Affiliation(s)
- Huafeng Liu
- Department of Oncology, Ganzhou People's Hospital, No.16 Meiguan Avenue, Ganzhou, China.
| | - Xuewen Liu
- Department of Oncology, Ganzhou People's Hospital, No.16 Meiguan Avenue, Ganzhou, China
| | - Yanjun Lu
- Department of Oncology, Ganzhou People's Hospital, No.16 Meiguan Avenue, Ganzhou, China
| |
Collapse
|
85
|
Haus-Cohen M, Reiter Y. Harnessing antibody-mediated recognition of the intracellular proteome with T cell receptor-like specificity. Front Immunol 2024; 15:1486721. [PMID: 39650646 PMCID: PMC11621052 DOI: 10.3389/fimmu.2024.1486721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/05/2024] [Indexed: 12/11/2024] Open
Abstract
The clinical success of cancer immunotherapy has driven ongoing efforts to identify novel targets that can effectively guide potent effector functions to eliminate malignant cells. Traditionally, immunotherapies have focused on surface antigens; however, these represent only a small fraction of the cancer proteome, limiting their therapeutic potential. In contrast, the majority of proteins within the human proteome are intracellular, yet they are represented on the cell surface as short peptides presented by MHC class I molecules. These peptide-MHC complexes offer a vast and largely untapped resource for cancer immunotherapy targets. The intracellular proteome, including neo-antigens, presents an exciting opportunity for the development of novel cell-based and soluble immunotherapies. Targeting these intracellular-derived peptide-MHC molecules on malignant cell surfaces can be achieved using specific T-cell receptors (TCRs) or TCR-mimicking antibodies, known as TCR-like (TCRL) antibodies. Current therapeutic strategies under investigation include adoptive cell transfer of TCR-engineered or TCRL-T cells and CAR-T cells that target peptide-MHC complexes, as well as soluble TCR- and TCRL-based agents like bispecific T cell engagers. Recent clinical developments in targeting the intracellular proteome using TCRL- and TCR-based molecules have shown promising results, with two therapies recently receiving FDA approval for the treatment of unresectable or metastatic uveal melanoma and synovial sarcoma. This review focuses on the processes for selecting and isolating TCR- and TCRL-based targeting moieties, with an emphasis on pre-clinical and clinical studies that explore the potential of peptide-MHC targeting agents in cancer immunotherapy.
Collapse
Affiliation(s)
| | - Yoram Reiter
- Laboratory of Molecular Immunology and Immunotherapy, Faculty of Biology Technion
– Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
86
|
Li S, Li W, Malhi NK, Huang J, Li Q, Zhou Z, Wang R, Peng J, Yin T, Wang H. Cannabigerol (CBG): A Comprehensive Review of Its Molecular Mechanisms and Therapeutic Potential. Molecules 2024; 29:5471. [PMID: 39598860 PMCID: PMC11597810 DOI: 10.3390/molecules29225471] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/10/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Cannabigerol (CBG), a non-psychoactive cannabinoid found in cannabis, has emerged as a promising therapeutic agent with a diverse range of potential applications. Unlike its well-known counterpart tetrahydrocannabinol (THC), CBG does not induce intoxication, making it an attractive option in the clinic. Recent research has shed light on CBG's intriguing molecular mechanisms, highlighting its potential to modulate multiple physiological processes. This review delves into the current understanding of CBG's molecular interactions and explores its therapeutic power to alleviate various conditions, including cancer, metabolic, pain, and inflammatory disorders, amongst others. We discuss how CBG interacts with the endocannabinoid system and other key signaling pathways, such as CB1, CB2, TPR channels, and α2-adrenoceptor, potentially influencing inflammation, pain, neurodegeneration, and other ailments. Additionally, we highlight the ongoing research efforts aimed at elucidating the full spectrum of CBG's therapeutic potential and its safety profile in clinical settings. Through this comprehensive analysis, we aim to provide a deeper understanding of CBG's role in promoting human health and pave the way for future research endeavors.
Collapse
Affiliation(s)
- Shijia Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China; (S.L.); (J.H.); (Q.L.); (Z.Z.)
| | - Weini Li
- Department of Biomedical Science, Cedars-Sinai Medical Center, Cedars-Sinai Cancer Institute, Los Angeles, CA 90067, USA; (W.L.); (R.W.)
| | - Naseeb Kaur Malhi
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA;
| | - Junwei Huang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China; (S.L.); (J.H.); (Q.L.); (Z.Z.)
| | - Quanqi Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China; (S.L.); (J.H.); (Q.L.); (Z.Z.)
| | - Ziwei Zhou
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China; (S.L.); (J.H.); (Q.L.); (Z.Z.)
| | - Ruiheng Wang
- Department of Biomedical Science, Cedars-Sinai Medical Center, Cedars-Sinai Cancer Institute, Los Angeles, CA 90067, USA; (W.L.); (R.W.)
| | - Jiangling Peng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China; (S.L.); (J.H.); (Q.L.); (Z.Z.)
| | - Tong Yin
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Honggen Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China;
| |
Collapse
|
87
|
Berquez M, Li AL, Luy MA, Venida AC, O'Loughlin T, Rademaker G, Barpanda A, Hu J, Yano J, Wiita A, Gilbert LA, Bruno PM, Perera RM. A multi-subunit autophagic capture complex facilitates degradation of ER stalled MHC-I in pancreatic cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.27.620516. [PMID: 39554122 PMCID: PMC11565957 DOI: 10.1101/2024.10.27.620516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDA) evades immune detection partly via autophagic capture and lysosomal degradation of major histocompatibility complex class I (MHC-I). Why MHC-I is susceptible to capture via autophagy remains unclear. By synchronizing exit of proteins from the endoplasmic reticulum (ER), we show that PDAC cells display prolonged retention of MHC-I in the ER and fail to efficiently route it to the plasma membrane. A capture-complex composed of NBR1 and the ER-phagy receptor TEX264 facilitates targeting of MHC-I for autophagic degradation, and suppression of either receptor is sufficient to increase total levels and re-route MHC-I to the plasma membrane. Binding of MHC-I to the capture complex is linked to antigen presentation efficiency, as inhibiting antigen loading via knockdown of TAP1 or beta 2-Microglobulin led to increased binding between MHC-I and the TEX264-NBR1 capture complex. Conversely, expression of ER directed high affinity antigenic peptides led to increased MHC-I at the cell surface and reduced lysosomal degradation. A genome-wide CRISPRi screen identified NFXL1, as an ER-resident E3 ligase that binds to MHC-I and mediates its autophagic capture. High levels of NFXL1 are negatively correlated with MHC-I protein expression and predicts poor patient prognosis. These data highlight an ER resident capture complex tasked with sequestration and degradation of non-conformational MHC-I in PDAC cells, and targeting this complex has the potential to increase PDAC immunogenicity.
Collapse
|