1
|
Huang R, Wu Y, Shen F, Chen S, Yang X, Lin Y, Fang Y, Shen J. Manganese-coordinated nanoparticles loaded with CHK1 inhibitor dually activate cGAS-STING pathway and enhance efficacy of immune checkpoint therapy. Biomaterials 2025; 319:123199. [PMID: 40009899 DOI: 10.1016/j.biomaterials.2025.123199] [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: 11/03/2024] [Revised: 01/26/2025] [Accepted: 02/16/2025] [Indexed: 02/28/2025]
Abstract
Notable advancements have been made in utilizing immune checkpoint blockade (ICB) for the treatment of various cancers. However, the overall response rates and therapeutic effectiveness remain unsatisfactory. One cause is the inadequate immune environment characterized by poor T cell infiltration in tumors. To address these limitations, enhancing immune infiltration is crucial for optimizing the therapeutic efficacy of ICB. Activating the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway is essential for initiating immune response and has become a potential target for developing combination therapies with ICB. In this study, we designed and fabricated manganese-containing nanoparticles loaded with the CHK1 inhibitor PF477736, which were subsequently encapsulated with macrophage membrane (PF/MMSN@MPM). This innovative design achieved excellent tumor targeting and demonstrated potent antitumor effects. The combination therapy dually amplified the cGAS-STING pathway, causing a cascade of enhanced therapeutic effects against tumors. Furthermore, single-cell mass cytometry (CyTOF) analysis revealed that PF/MMSN@MPM enhanced the activation and infiltration of immune cells. Moreover, the combination of PF/MMSN@MPM with anti-PD-1 (αPD-1) exhibited a stronger therapeutic effect compared to αPD-1 alone. PF/MMSN@MPM precisely and synergistically activated the cGAS-STING pathway, significantly improving therapeutic efficacy of ICB, and offering promising potential for tumor therapy.
Collapse
Affiliation(s)
- Rui Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yijia Wu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Feiyang Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shuai Chen
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoyu Yang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yao Lin
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yan Fang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Jianfeng Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, China; Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
2
|
Chua MLK, Zhang X, Wong KCW, Grégoire M, Spreafico A, Ma B. Updates on Treatments and Management of Nasopharyngeal Carcinoma. Am Soc Clin Oncol Educ Book 2025; 45:e472460. [PMID: 40209143 DOI: 10.1200/edbk-25-472460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
Nasopharyngeal carcinoma (NPC) is a unique head and neck cancer, where the endemic subtype is strongly associated with Epstein-Barr virus (EBV) infection, whereas emerging data suggest that a subset of nonendemic NPC may be associated with human papillomavirus (HPV) infection. Nonetheless, treatment advances have been driven by clinical trials conducted in endemic NPC, investigating optimal sequencing of chemotherapy and immune checkpoint inhibitors with radiotherapy for locoregionally advanced disease. The preference for induction chemotherapy (IC) in these patients has also led to evolution in the concept of radiotherapy target delineation. Because of its association with EBV, plasma EBV DNA is an archetypal biomarker for endemic NPC, and it is being explored for precise stratification and treatment individualization in several ongoing trials. In the space of recurrent or metastatic-NPC, with the advent of platinum-doublet chemotherapy and anti-PD-1 antibody as the new standard of care, several trials are investigating new immunotherapeutic combinations, bispecific antibodies, and antibody-drug conjugates that have demonstrated promise in early phase trials. An important advance for NPC in 2025 is the update of the 9th version of the TNM staging system, which has introduced several key changes, including downgrading of the TNM stage groupings for localized disease, and splitting of metastatic NPC into IVA and IVB based on the number of metastatic lesions. These revisions would have implications for the treatment and design of future trials. These advances are also relevant to nonendemic NPC, where evidence is inconclusive whether this disease responds differently to current treatments compared with endemic NPC.
Collapse
Affiliation(s)
- Melvin L K Chua
- Divisions of Radiation Oncology and Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
- Oncology Academic Program, Duke-NUS Medical School, Singapore, Singapore
| | - Xin Zhang
- Radiation Oncology Centre, Chongqing University Cancer Hospital, Chongqing, China
| | - Kenneth C W Wong
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir Y.K. Pao Centre for Cancer, Hong Kong Cancer Institute, The Chinese University of Hong Kong, Hong Kong SAR
| | - Marret Grégoire
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Anna Spreafico
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Brigette Ma
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir Y.K. Pao Centre for Cancer, Hong Kong Cancer Institute, The Chinese University of Hong Kong, Hong Kong SAR
- Charlie Lee Precision Immuno-Oncology Program, The Chinese University of Hong Kong, Hong Kong SAR
| |
Collapse
|
3
|
Wang Q, Yin X, Liu H, Wang Q, Zhang L, Wang Y, Lu H. Mitochondrial function changes in T cell subsets during radiotherapy for patients with nasopharyngeal carcinoma. Oncol Lett 2025; 29:273. [PMID: 40235681 PMCID: PMC11998068 DOI: 10.3892/ol.2025.15019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 03/21/2025] [Indexed: 04/17/2025] Open
Abstract
Mitochondrial dysfunction-mediated T cell exhaustion is associated with the efficacy of tumor therapy; however, the effect of radiotherapy (RT) on the mitochondrial function of peripheral blood immune cells remains still unclear. Therefore, the current study aimed to determine mitochondrial function indicators in immune cells, in particular mitochondrial mass (MM) and mitochondrial membrane potential (MMP), to assess the dynamic changes of immune status in patients with nasopharyngeal carcinoma (NPC) during RT. Peripheral venous blood was collected from patients with locally advanced NPC at day 1 pre-RT, at the 10th fraction of RT and within 2 days after RT. Based on a novel immunofluorescence technique, flow cytometry was used to assess the proportion of lymphocytes and their subsets in peripheral blood and the mitochondrial indexes, MM and low MMP (MMPlow). Univariate and multivariate logistic regression analyses were performed to evaluate the clinical factors associated with the efficacy of RT. A total of 27 patients were enrolled. After RT, lymphocyte count (P<0.05) and the proportion of CD4+ T cells (P<0.05) demonstrated a downward trend. In addition, the proportion of CD4+ memory-effector T (Tem; P<0.05) cells and CD8+ Tem cells (P=0.005) significantly increased during RT. No significant changes were demonstrated for MM in CD4+ effector T (Te) cells, whilst MMPlow was significantly reduced (P=0.047). However, the mitochondrial function of CD8+ T cells did not significantly change. Multivariate logistic regression analysis revealed that lymphocyte count [odds ratio (OR), 47.317; 95% confidence interval (CI), 1.240-1806.065] and MMPlow in CD4+ Te cells (OR, 0.889; 95% CI, 0.792-0.997) were independent factors that could affect clinical efficacy. Receiver operating characteristic curve analysis demonstrated that the area under the curve values for MMPlow in CD4+ T cells, lymphocyte count and their combination were 0.72 (P=0.13), 0.69 (P=0.19) and 0.89 (P=0.0073), respectively. These findings suggest that RT could inhibit immune cells in peripheral blood. However, this treatment approach could activate the memory cell subsets of immune cells and enhance the MMP of effector CD4+ T cells. Therefore, the evaluation of mitochondrial function in lymphocytes could be used as a predictor of RT efficacy in patients with locally advanced NPC.
Collapse
Affiliation(s)
- Quan Wang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266035, P.R. China
| | - Xiangzhi Yin
- Department of Orthopaedics, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266035, P.R. China
| | - Hongbo Liu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266035, P.R. China
| | - Qing Wang
- Department of Clinical Laboratory, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266035, P.R. China
| | - Lu Zhang
- Department of Radiation Oncology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| | - Yifan Wang
- Department of Emergency, Laizhou People's Hospital, Yantai, Shandong 261400, P.R. China
| | - Haijun Lu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266035, P.R. China
| |
Collapse
|
4
|
Gao T, Zhao P, Han S. Integrating bulk RNA-seq and scRNA-seq analyses with machine learning to predict platinum response and prognosis in ovarian cancer. Sci Rep 2025; 15:19123. [PMID: 40450069 DOI: 10.1038/s41598-025-99930-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/23/2025] [Indexed: 06/03/2025] Open
Abstract
Platinum-based therapy is an integral part of the standard treatment for ovarian cancer. However, despite extensive research spanning several decades, the identification of dependable predictive biomarkers for platinum response in clinical practice has proven to be a formidable challenge. Recently, the development of single-cell technology has enabled more precise investigations into the heterogeneity of cancer. In this study, we isolated cancer cells from the single-cell transcriptomic data of platinum-sensitive and platinum-resistant patients with ovarian cancer. Differential gene analysis of platinum-sensitive and platinum-resistant cancer cells revealed that several of the differentially expressed genes had previously been reported in other studies to be associated with platinum resistant. Gene set enrichment analysis revealed the up-regulation of pathways involved in processes such as autophagy, cell cycle regulation, and DNA damage repair, which are known to promote platinum resistance in ovarian cancer. Based on these findings, we hypothesized that these differentially expressed genes could be used to predict the response of ovarian cancer patients to platinum-based chemotherapy. To validate this hypothesis, we explored 7 different machine learning models for predicting platinum chemotherapy response at varying feature gene counts. Ultimately, the random forest model performed the best, with 5 genes (PAX2, TFPI2, APOA1, ADIRF and CRISP3) and achieve an AUC of 0.993 in test cohort and 0.989 in GSE63885 independent validation cohorts. We named this model GPPS (Genes to Predict Platinum response Signature). Furthermore, we discovered that the GPPS model can also predict patient prognosis.
Collapse
Affiliation(s)
- Tingting Gao
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, China
| | - Peng Zhao
- Oncology Department of Xi'an Daxing Hospital, Xi'an, China
| | - Suxia Han
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
5
|
Liu XF, Song B, Sun CB, Zhu Q, Yue JH, Liang YJ, He J, Zeng XL, Qin YC, Chen QY, Mai HQ, Zhang X, Li J. Tumor-infiltrated double-negative regulatory T cells predict outcome of T cell-based immunotherapy in nasopharyngeal carcinoma. Cell Rep Med 2025; 6:102096. [PMID: 40315843 DOI: 10.1016/j.xcrm.2025.102096] [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: 10/13/2024] [Revised: 01/24/2025] [Accepted: 04/04/2025] [Indexed: 05/04/2025]
Abstract
Adoptive cell therapy (ACT) using tumor-infiltrating lymphocytes (TILs) has demonstrated clinical success in solid tumors. We analyze 47 TIL infusion products and 62 pretreatment tumor microenvironments (TMEs) from a randomized phase 2 clinical study of concurrent chemoradiotherapy plus TIL-ACT (NCT02421640). Using single-cell and bulk RNA sequencing along with flow cytometry, we identify 14 CD3+ T cell clusters within 26 TIL infusion products: 11 CD3+CD8+ TILs, 2 CD3+CD4+ TILs, and 1 CD3+CD8-CD4- double-negative (DN) TIL. (DN) TILs, significantly associated with poor TIL-ACT outcomes, exhibit an activated regulatory T cell-like phenotype and include two CD56+ and four CD56- subsets. Among them, CD56-KZF2+ (DN) TILs are predominantly suppressive. (DN) TILs inhibit CD8+ TIL expansion via Fas-FasL, transforming growth factor β (TGF-β), and interleukin (IL)-10 signaling. Distinct CD8+ T subsets differentially impact on TIL-ACT outcomes, while 9 baseline TME gene signatures and 14 intracellular T cell genes hold prognostic value. Our findings identify predictive TIL subsets and biomarkers for TIL-ACT outcomes.
Collapse
Affiliation(s)
- Xiu-Feng Liu
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Bin Song
- BGI, Shenzhen 518083, P.R. China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Chang-Bin Sun
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, P.R. China
| | - Qian Zhu
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | | | - Yu-Jing Liang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Jia He
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Xi-Liang Zeng
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | | | - Qiu-Yan Chen
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| | - Hai-Qiang Mai
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| | - Xi Zhang
- BGI, Shenzhen 518083, P.R. China.
| | - Jiang Li
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| |
Collapse
|
6
|
Li X, Qiu X, Lin C, Liu Y, Wang Y, Tang L, Tong Y, Tang L. Intratumoral CD8+ tumor-infiltrating lymphocytes as prognostic predictors in radio-chemoradiotherapy-treated nasopharyngeal carcinoma. Front Oncol 2025; 15:1551980. [PMID: 40444078 PMCID: PMC12119675 DOI: 10.3389/fonc.2025.1551980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 04/28/2025] [Indexed: 06/02/2025] Open
Abstract
Background The prognostic value of tumor-infiltrating lymphocytes (TILs) in nasopharyngeal carcinoma (NPC) has been established. However, the prognostic significance of CD4+ and CD8+ TIL subtypes in NPC remains unclear. Methods We collected 214 tissue samples diagnosed with NPC for immunohistochemical staining. The density of CD4+ and CD8+ TILs was evaluated in intratumoral (within tumor cell nests) and stromal (the surrounding stroma of tumor cell nests) areas. Correlations between TIL density and progression-free survival (PFS) and overall survival (OS) were analyzed. Results High levels of intratumoral CD8+ TILs were significantly associated with reduced risk of disease progression (HR 0.382; 95% CI, 0.178-0.819, P = 0.013) and death (HR 0.265; 95% CI, 0.104-0.675, P = 0.005). Although high stromal CD8+ TIL levels were linked to higher PFS and OS, these differences did not reach statistical significance (P = 0.114 and P = 0.079, respectively). CD4+ TILs showed no significant correlation with PFS or OS. In multivariate analysis, intratumoral CD8+ TILs remained an independent prognostic factor for PFS and OS. Subgroup analysis revealed that in patients with locally advanced disease, high intratumoral CD8+ TILs were significantly associated with improved PFS (HR 0.329; 95% CI, 0.129-0.843, P = 0.021) and OS (HR 0.209; 95% CI, 0.064-0.681, P = 0.009). Conversely, in early-stage patients, neither CD8+ nor CD4+ TILs were significantly associated with PFS or OS. Conclusion Our findings suggest that intratumoral CD8+ TILs serve as a reliable prognostic biomarker for NPC, with their prognostic value particularly pronounced in patients with locally advanced disease.
Collapse
Affiliation(s)
- Xinjing Li
- Department of Pathology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Xiaoming Qiu
- Department of Pathology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Cuihong Lin
- Department of Pathology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Yuanying Liu
- Department of Pathology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Yongbin Wang
- Department of Radiation Oncology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Langlang Tang
- Department of Radiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Yuanhe Tong
- Department of Radiation Oncology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Linbo Tang
- Department of Radiation Oncology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| |
Collapse
|
7
|
Chung DLS, Leung GHD, Liu S, Lok SWY, Xin Y, Xia Y, Zhavoronkov A, Pun FW, Ng WT, Dai W. AI-Driven Drug Target Screening Platform Identified Oncogene CACNA2D1 Activated by Enhancer Infestation in Epstein-Barr Virus-Associated Nasopharyngeal Carcinoma. Int J Mol Sci 2025; 26:4697. [PMID: 40429844 PMCID: PMC12111453 DOI: 10.3390/ijms26104697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 05/08/2025] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
The management of nasopharyngeal cancer (NPC) is rapidly evolving, with immune checkpoint inhibitors emerging as a prominent treatment approach. However, drug development targeting specific molecular and cellular abnormalities in NPC has slowed. Recent advancements in artificial intelligence (AI) and bioinformatics, particularly those integrating multi-omics data, offer a more effective alternative to traditional in vitro screening methods for identifying clinically actionable targets in NPC. Through a combination of multi-omics analyses and AI-driven screening, we identified CACNA2D1 as a novel cancer-cell-specific therapeutic target in NPC. Our research indicates that exploiting Epstein-Barr virus (EBV) tethering increases H3K27 acetylation near the CACNA2D1 promoter. Analysis of clinical specimens revealed significant upregulation of CACNA2D1 at both the transcriptional and translational levels (p-value < 0.01). Functional studies demonstrated that the mouse tumour size shrank by one-third upon the depletion of CACNA2D1, and there was an 85% reduction in cancer cell growth through the blockage of enhancers, while the presence of CACNA2D1 conferred a survival advantage during NPC tumour development. These findings highlight the potential of CACNA2D1 as a promising target for therapeutic intervention in NPC.
Collapse
Affiliation(s)
- Dittman Lai-Shun Chung
- Department of Clinical Oncology, University of Hong Kong, Hong Kong SAR, China; (D.L.-S.C.); (S.L.); (W.-T.N.)
| | - Geoffrey Ho Duen Leung
- Insilico Medicine Hong Kong Ltd., Unit 310, 3/F, Building 8W, Hong Kong Science and Technology Park, Hong Kong SAR, China; (G.H.D.L.); (S.W.Y.L.); (Y.X.); (A.Z.)
| | - Songran Liu
- Department of Clinical Oncology, University of Hong Kong, Hong Kong SAR, China; (D.L.-S.C.); (S.L.); (W.-T.N.)
| | - Sarah Wing Yan Lok
- Insilico Medicine Hong Kong Ltd., Unit 310, 3/F, Building 8W, Hong Kong Science and Technology Park, Hong Kong SAR, China; (G.H.D.L.); (S.W.Y.L.); (Y.X.); (A.Z.)
| | - Ying Xin
- Insilico Medicine Hong Kong Ltd., Unit 310, 3/F, Building 8W, Hong Kong Science and Technology Park, Hong Kong SAR, China; (G.H.D.L.); (S.W.Y.L.); (Y.X.); (A.Z.)
| | - Yunfei Xia
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Centre, Guangzhou 510060, China;
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Alex Zhavoronkov
- Insilico Medicine Hong Kong Ltd., Unit 310, 3/F, Building 8W, Hong Kong Science and Technology Park, Hong Kong SAR, China; (G.H.D.L.); (S.W.Y.L.); (Y.X.); (A.Z.)
- Insilico Medicine US Inc., 1000 Massachusetts Avenue, Suite 126, Cambridge, MA 02138, USA
| | - Frank W. Pun
- Insilico Medicine Hong Kong Ltd., Unit 310, 3/F, Building 8W, Hong Kong Science and Technology Park, Hong Kong SAR, China; (G.H.D.L.); (S.W.Y.L.); (Y.X.); (A.Z.)
| | - Wai-Tong Ng
- Department of Clinical Oncology, University of Hong Kong, Hong Kong SAR, China; (D.L.-S.C.); (S.L.); (W.-T.N.)
| | - Wei Dai
- Department of Clinical Oncology, University of Hong Kong, Hong Kong SAR, China; (D.L.-S.C.); (S.L.); (W.-T.N.)
| |
Collapse
|
8
|
Zhang S, Zhou Y, Liu Z, Wang Y, Zhou X, Chen H, Zhang X, Chen Y, Feng Q, Ye X, Xie S, Zeng MS, Zhai W, Zeng YX, Cao S, Li G, Xu M. Immunosequencing identifies signatures of T cell responses for early detection of nasopharyngeal carcinoma. Cancer Cell 2025:S1535-6108(25)00168-0. [PMID: 40345188 DOI: 10.1016/j.ccell.2025.04.009] [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: 06/22/2024] [Revised: 03/10/2025] [Accepted: 04/19/2025] [Indexed: 05/11/2025]
Abstract
To identify nasopharyngeal carcinoma (NPC)-relevant T cell receptors (TCRs), we profile the repertoires of peripheral blood TCRβ chains from 228 NPC patients, 241 at-risk controls positive for serum Epstein-Barr virus (EBV) VCA-IgA antibody, and 251 seronegative controls. We develop a TCR-based signature (T-score) based on 208 NPC-enriched CDR3β sequences, which accurately diagnoses NPC in both the original and independent validation cohorts. Notably, a higher T-score, associated with a shorter time interval to NPC diagnosis, effectively identifies early-stage NPC among EBV-seropositive at-risk individuals prior to clinical diagnosis. These NPC-enriched TCRs react against not only EBV-specific antigens but also non-EBV antigens expressed by NPC cells, indicating a broad range of specificities. Moreover, the abundance of NPC-enriched CD8+ T cells in blood correlates with the infiltration of non-exhausted T cell counterparts in tumors and predicts prolonged survival, suggesting that these NPC-enriched T cells have significant potential for disease monitoring and therapeutic applications.
Collapse
Affiliation(s)
- Shanshan Zhang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Zhonghua Liu
- Department of Biostatistics, Columbia University, New York, NY, USA
| | - Yuqian Wang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China
| | - Xiang Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510620, China
| | - Haiwen Chen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, P.R. China
| | - Xinyu Zhang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Yanhong Chen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Qisheng Feng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Xiaoping Ye
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Shanghang Xie
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Weiwei Zhai
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yi-Xin Zeng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| | - Sumei Cao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| | - Guideng Li
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China.
| | - Miao Xu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| |
Collapse
|
9
|
Zhang W, Wang C, Meng Y, He L, Dong M. EBV Vaccines in the Prevention and Treatment of Nasopharyngeal Carcinoma. Vaccines (Basel) 2025; 13:478. [PMID: 40432090 PMCID: PMC12115577 DOI: 10.3390/vaccines13050478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Epstein-Barr virus (EBV), a ubiquitous human herpesvirus, has been robustly linked to the pathogenesis of nasopharyngeal carcinoma (NPC). The mechanism of EBV-induced NPC involves complex interactions between viral proteins and host cell pathways. This review aims to comprehensively outline the mechanism of EBV-induced NPC and the latest advances in targeted EBV vaccines for prophylaxis and treatment. This review explores the intricate molecular mechanisms by which EBV contributes to NPC pathogenesis, highlighting viral latency, genetic and epigenetic alterations, and immune evasion strategies. It emphasizes the pivotal role of key viral proteins, including EBNA1, LMP1, and LMP2A, in carcinogenesis. Subsequently, the discussion shifts towards the development of targeted EBV vaccines, including preventive vaccines aimed at preventing primary EBV infection and therapeutic vaccines aimed at treating diagnosed EBV-related NPC. The review underscores the challenges and future directions in the field, stressing the importance of developing innovative vaccine strategies and combination therapies to improve efficacy. This review synthesizes current insights into the molecular mechanisms of EBV-induced NPC and the development of EBV-targeted vaccines, highlighting the potential use of mRNA vaccines for NPC treatment.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital/The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China; (W.Z.)
| | - Chuang Wang
- Chengdu Yunce Medical Biotechnology Co., Ltd., Chengdu 611135, China;
| | - Yousheng Meng
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital/The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China; (W.Z.)
| | - Lang He
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital/The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China; (W.Z.)
| | - Mingqing Dong
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou 325000, China
| |
Collapse
|
10
|
Lai W, Li Y, Luo OJ. MIST: An interpretable and flexible deep learning framework for single-T cell transcriptome and receptor analysis. SCIENCE ADVANCES 2025; 11:eadr7134. [PMID: 40184452 PMCID: PMC11970455 DOI: 10.1126/sciadv.adr7134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 02/28/2025] [Indexed: 04/06/2025]
Abstract
Joint analysis of transcriptomic and T cell receptor (TCR) features at single-cell resolution provides a powerful approach for in-depth T cell immune function research. Here, we introduce a deep learning framework for single-T cell transcriptome and receptor analysis, MIST (Multi-insight for T cell). MIST features three latent spaces: gene expression, TCR, and a joint latent space. Through analyses of antigen-specific T cells, and T cell datasets related to lung cancer immunotherapy and COVID19, we demonstrate MIST's interpretability and flexibility. MIST easily and accurately resolves cell function and antigen specificity by vectorizing and integrating transcriptome and TCR data of T cells. In addition, using MIST, we identified the heterogeneity of CXCL13+ subsets in lung cancer infiltrating CD8+ T cells and their association with immunotherapy, providing additional insights into the functional transition of CXCL13+ T cells related to anti-PD-1 therapy that were not reported in the original study.
Collapse
Affiliation(s)
- Wenpu Lai
- The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Yangqiu Li
- The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou 510632, China
- Key Laboratory of Viral Pathogenesis and Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai 519070, China
| |
Collapse
|
11
|
Cheng XX, Lin GW, Zhou YQ, Li YQ, He S, Liu Y, Zeng YN, Guo YM, Liu SQ, Peng W, Wei PP, Luo CL, Bei JX. A rare KLHDC4 variant Glu510Lys is associated with genetic susceptibility and promotes tumor metastasis in nasopharyngeal carcinoma. J Genet Genomics 2025; 52:559-569. [PMID: 39706520 DOI: 10.1016/j.jgg.2024.12.008] [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: 09/05/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024]
Abstract
Various genetic association studies have identified numerous single nucleotide polymorphisms (SNPs) associated with nasopharyngeal carcinoma (NPC) risk. However, these studies have predominantly focused on common variants, leaving the contribution of rare variants to the "missing heritability" largely unexplored. Here, we integrate genotyping data from 3925 NPC cases and 15,048 healthy controls to identify a rare SNP, rs141121474, resulting in a Glu510Lys mutation in KLHDC4 gene linked to increased NPC risk. Subsequent analyses reveal that KLHDC4 is highly expressed in NPC and correlates with poorer prognosis. Functional characterizations demonstrate that KLHDC4 acts as an oncogene in NPC cells, enhancing their migratory and metastatic capabilities, with these effects being further augmented by the Glu510Lys mutation. Mechanistically, the Glu510Lys mutant exhibits increased interaction with Vimentin compared to the wild-type KLHDC4 (KLHDC4-WT), leading to elevated Vimentin protein stability and modulation of the epithelial-mesenchymal transition process, thereby promoting tumor metastasis. Moreover, Vimentin knockdown significantly mitigates the oncogenic effects induced by overexpression of both KLHDC4-WT and the Glu510Lys variant. Collectively, our findings highlight the critical role of the rare KLHDC4 variant rs141121474 in NPC progression and propose its potential as a diagnostic and therapeutic target for NPC patients.
Collapse
Affiliation(s)
- Xi-Xi Cheng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Guo-Wang Lin
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Ya-Qing Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Yi-Qi Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Shuai He
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Yang Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Yan-Ni Zeng
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Yun-Miao Guo
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang, Guangdong 524045, China
| | - Shu-Qiang Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Wan Peng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Pan-Pan Wei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Chun-Ling Luo
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China.
| | - Jin-Xin Bei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Sun Yat-sen University Institute of Advanced Studies Hong Kong, Science Park, Hong Kong SAR, China; Department of Medical Oncology, National Cancer Centre Singapore, Singapore.
| |
Collapse
|
12
|
Moon CY, Belabed M, Park MD, Mattiuz R, Puleston D, Merad M. Dendritic cell maturation in cancer. Nat Rev Cancer 2025; 25:225-248. [PMID: 39920276 PMCID: PMC11954679 DOI: 10.1038/s41568-024-00787-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 02/09/2025]
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that are present at low abundance in the circulation and tissues; they serve as crucial immune sentinels by continually sampling their environment, migrating to secondary lymphoid organs and shaping adaptive immune responses through antigen presentation. Owing to their ability to orchestrate tolerogenic or immunogenic responses to a specific antigen, DCs have a pivotal role in antitumour immunity and the response to immune checkpoint blockade and other immunotherapeutic approaches. The multifaceted functions of DCs are acquired through a complex, multistage process called maturation. Although the role of inflammatory triggers in driving DC maturation was established decades ago, less is known about DC maturation in non-inflammatory contexts, such as during homeostasis and in cancer. The advent of single-cell technologies has enabled an unbiased, high-dimensional characterization of various DC states, including mature DCs. This approach has clarified the molecular programmes associated with DC maturation and also revealed how cancers exploit these pathways to subvert immune surveillance. In this Review, we discuss the mechanisms by which cancer disrupts DC maturation and highlight emerging therapeutic opportunities to modulate DC states. These insights could inform the development of DC-centric immunotherapies, expanding the arsenal of strategies to enhance antitumour immunity.
Collapse
Affiliation(s)
- Chang Yoon Moon
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meriem Belabed
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Park
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raphaël Mattiuz
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Puleston
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
13
|
Gao Z, Han R, Chen Q, Guo J, Wang Y, Hong Q, Zhao C, Mu J, Li J. IRF8 and CD2 are potential targets of immunotherapy in non-small cell lung cancer. J Thorac Dis 2025; 17:1169-1184. [PMID: 40223995 PMCID: PMC11986741 DOI: 10.21037/jtd-24-1589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/13/2025] [Indexed: 04/15/2025]
Abstract
Background Immune checkpoint inhibitors (ICIs) are clinically effective in the treatment of non-small cell lung cancer (NSCLC), but the response rate in nonselective NSCLC patients is approximately 20%. It is important to expand the pool of benefits of immunotherapy. However, current solution strategies are limited. Our research is to identify new targets for combined immunotherapy and expand the beneficiary population of immunotherapy. Methods Functional enrichment analysis was performed for differentially expressed genes (DEGs) in 175 NSCLC immunotherapy cohorts and 494 non-immunotherapy cohorts, and single-sample gene set enrichment analysis (ssGSEA) was used to quantify the level of infiltration of different immune cell subpopulations. Weighted correlation network analysis (WGCNA), univariate Cox regressions, least absolute shrinkage and selection operator (LASSO) regressions, and gene correlation analysis were applied to identify immune signature genes associated with immune cell infiltration, and a nomogram was constructed to predict the survival rate. Results The DEGs were not enriched in the classical antitumour immune response and the dendritic cells (DCs) infiltration level in the tumour microenvironment (TME) was at a low level in the immunotherapy cohort. The high expression of IRF8 and CD2 was positively correlated with the level of DCs infiltration, the core of tumour immune response regulation, and can bring better survival prognosis for patients. Besides, targeted activation of IRF8 and CD2 can improve the efficacy of ICIs. Conclusions High expression of IRF8 and CD2 enhances the antitumour immune response, and IRF8 and CD2 may be new prognostic indicators and targets of combined ICIs for lung adenocarcinoma in NSCLC.
Collapse
Affiliation(s)
- Zhen Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Thoracic Surgery, Shulan (Jinan) Hospital, Jinan, China
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan, China
| | - Rui Han
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qinghao Chen
- Department of Thoracic Surgery, Shulan (Jinan) Hospital, Jinan, China
| | - Jichao Guo
- Lanshan District People’s Hospital, Department of Thoracic Surgery, Linyi, China
| | - Yancheng Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan, China
| | - Qian Hong
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chenguang Zhao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Juwei Mu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiagen Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
14
|
Wang X, Yu L, Zhou X, Chung GTY, Liu AMT, Chan YY, Wu M, Chau KY, Lo KW, Wu AR. Characterizing resistant cellular states in nasopharyngeal carcinoma during EBV lytic induction. Oncogene 2025:10.1038/s41388-025-03341-z. [PMID: 40133476 DOI: 10.1038/s41388-025-03341-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 02/07/2025] [Accepted: 03/03/2025] [Indexed: 03/27/2025]
Abstract
The pervasive occurrence of nasopharyngeal carcinoma (NPC) is intricately linked to Epstein-Barr virus (EBV) infection, making EBV and its associated pathways promising therapeutic targets for NPC and other EBV-related cancers. Lytic induction therapy, an emerging virus-targeted therapeutic strategy, capitalizes on the presence of EBV in tumor cells to specifically induce cytotoxicity against EBV-associated malignancies. Despite the expanding repertoire of compounds developed to induce EBV lytic reactivation, achieving universal induction across all infected cells remains elusive. The inherent heterogeneity of tumor cells likely contributes to this variability. In this study, we used the NPC43 cell line, an EBV-positive NPC in vitro model, and single-cell transcriptomics to characterize the diverse cellular responses to EBV lytic induction. Our longitudinal monitoring revealed a distinctive lytic induction non-responsive cellular state characterized by elevated expression of SOX2 and NTRK2. Cells in this state exhibit phenotypic similarities to cancer stem cells (CSCs), and we verified the roles of SOX2 and NTRK2 in manifesting these phenotypes. Our findings reveal a significant challenge for lytic induction therapy, as not all tumor cells are equally susceptible. These insights highlight the importance of combining lytic induction with therapies targeting CSC-like properties to enhance treatment efficacy for NPC and other EBV-associated cancers.
Collapse
Affiliation(s)
- Xinlei Wang
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Lei Yu
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Xuemeng Zhou
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Grace Tin-Yun Chung
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Alyssa Ming-Ting Liu
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuk-Yu Chan
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Man Wu
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kin Yung Chau
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Kwok-Wai Lo
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Angela Ruohao Wu
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
- Center for Aging Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
| |
Collapse
|
15
|
Zhou YQ, Jiang JX, He S, Li YQ, Cheng XX, Liu SQ, Wei PP, Guan XY, Ong CK, Wang VYF, Luo CL, Bei JX. Epstein-Barr virus hijacks histone demethylase machinery to drive epithelial malignancy progression through KDM5B upregulation. Signal Transduct Target Ther 2025; 10:83. [PMID: 40059116 PMCID: PMC11891327 DOI: 10.1038/s41392-025-02163-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 01/19/2025] [Accepted: 02/01/2025] [Indexed: 05/13/2025] Open
Abstract
Epstein-Barr virus (EBV) is a significant epigenetic driver in the development of epithelial-origin nasopharyngeal carcinoma (NPC) and gastric cancer (GC), which together represent 80% of EBV-associated malignancies. Despite its known association, the specific mechanisms, particularly those involving EBV-induced histone modifications, remain poorly understood. Through integrative analyses of single-cell and bulk transcriptome data from epithelial tumor tissues and EBV-infected cells, we identified KDM5B as a critical histone-modifying factor consistently upregulated following EBV infection. We demonstrated that EBV stimulates KDM5B expression via interactions of its latent gene EBNA1 with transcription factor CEBPB and through direct binding of its lytic gene BZLF1 to Zta-response elements on the KDM5B promoter. Functional assays revealed that KDM5B acts as an oncogene, correlating with poor survival outcomes in EBV-associated epithelial cancers. Mechanistically, KDM5B inhibited the tumor suppressor gene PLK2 through histone demethylation, thereby activating the PI3K/AKT/mTOR signaling pathway and promoting malignant progression. Furthermore, treatment with the KDM5B inhibitor AS-8351 markedly attenuated this signaling activity and exhibited strong anti-tumor effect in both in vitro and in vivo patient-derived xenograft models from EBV-associated tumors. Together, these findings provide novel insights into how EBV hijacks KDM5B to mediate histone demethylation of PLK2, facilitating tumor progression through the PI3K/AKT/mTOR pathway in epithelial cancers, highlighting promising therapeutic strategies targeting epigenetic alterations in EBV-associated cancers.
Collapse
Affiliation(s)
- Ya-Qing Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Jia-Xin Jiang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Shuai He
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yi-Qi Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Xi-Xi Cheng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Shu-Qiang Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Pan-Pan Wei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, P.R. China
| | - Choon Kiat Ong
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | - Vivien Ya-Fan Wang
- Faculty of Health, University of Macau, Avenida da Universidade, Taipa, Macau SAR, P.R. China
| | - Chun-Ling Luo
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China.
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China.
| | - Jin-Xin Bei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China.
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China.
- Department of Medical Oncology, National Cancer Centre of Singapore, Singapore, Singapore.
- Sun Yat-sen University Institute of Advanced Studies Hong Kong, Science Park, Hong Kong SAR, P.R. China.
- Department of Clinical Oncology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, P.R. China.
| |
Collapse
|
16
|
Wu Z, Lepcha TT, Zhou D, He Z, Fiches GN, Park Y, He J, Chen J, Shanaka K, Oghumu S, Zhao W, Ma A, Ma Q, Zhu J, Santoso NG. Analysis of Head and Neck Cancer scRNA-seq Data Identified PRDM6 Promotes Tumor Progression by Modulating Immune Gene Expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.04.641548. [PMID: 40093183 PMCID: PMC11908237 DOI: 10.1101/2025.03.04.641548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a biologically aggressive and heterogeneous group of cancers with limited treatment options for patients who do not respond to standard therapies. While HPV-related HNSCCs tend to show better therapeutic outcomes, we still had limited understanding of the immune mechanisms underlying these cancers. Immune-responsive genes (IRGs) have emerged as critical factors in regulating both tumor progression and immune response. Recent advances in single-cell RNA sequencing (scRNA-seq) and the development of cell-type specific regulon inference tools, such as IRIS3, have provided new insights into the tumor immune microenvironment. In this study, we leveraged the IRIS3 platform to analyze scRNA-seq data from HNSCC patient samples, identifying novel transcription factor (TF)-IRG networks that contribute to tumor proliferation and immune escape. Specifically, we identified PRDM6, a histone methyltransferase, possesses the previously unknown role in promoting tumor cell proliferation by inducing IRG expression. We further demonstrated that HPV viral oncoproteins (E6/E7) oncoproteins up-regulate the PRDM6 expression, which associates PRDM6 with HPV-positive HNSCC.
Collapse
Affiliation(s)
- Zhenyu Wu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Thurbu Tshering Lepcha
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Dawei Zhou
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Zhixian He
- Department of Microbiology, College of Arts and Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Guillaume N. Fiches
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Youngmin Park
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jinshan He
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jianwen Chen
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - K.A.S.N Shanaka
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Steve Oghumu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Weiqiang Zhao
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Anjun Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Qin Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jian Zhu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Netty G. Santoso
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
17
|
Chen JW, Shen RN, Zhu JQ, Wang YH, Fu LM, Chen YH, Cao JZ, Wei JH, Luo JH, Li JY, Gui CP. Transcriptomic profiling reveals mechanism, therapeutic potential, and prognostic value of cancer stemness characteristic in nasopharyngeal carcinoma. Funct Integr Genomics 2025; 25:56. [PMID: 40053129 DOI: 10.1007/s10142-025-01561-w] [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: 09/12/2024] [Revised: 02/18/2025] [Accepted: 02/23/2025] [Indexed: 05/13/2025]
Abstract
Nasopharyngeal carcinoma (NPC) recurrence, distant metastasis, and drug resistance remain significant obstacles in clinical prognosis. Cancer stemness is hypothesized to be a key contributor, though direct evidence is sparse. We utilized bioinformatics and machine learning techniques on single-cell RNA-seq and bulk transcriptomic datasets, complemented by basic experiments, to investigate stemness-based characteristics in NPC. Our analysis identified two potential developmental trajectories of nasopharyngeal cancer cells, each exhibiting varying levels of stemness. We subsequently identified and validated a cancer stemness-related signature (STEM-signature). Single-cell profiling revealed enrichment of LAYN + CD8 + , CTLA4 + CD4 + , CXCL13 + CD4 + T cells, tumor-associated macrophages, and CD14 + monocytes in NPC patients with high stemness. NicheNet analysis suggested these immune cells regulate cancer stemness. Bulk transcriptomic analysis corroborated these findings, indicating a poor therapeutic response in high-stemness NPC. We predicted 13 potential drugs and identified 13 stemness-related miRNAs for NPC with high stemness. A Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression model, based on this miRNA signature, predicted overall survival with an AUC of 0.71 and 0.72 in validation and testing sets, respectively. The miRNA-based stemness signature outperformed previous established signatures. Multivariate Cox regression analysis indicated that our prognostic signature could serve as an independent prognostic factor (p < 0.001). Basic experiments showed that miR-300, miR-361-5p, miR-1246, and miR-1290 enhanced the stemness characteristics of NPC cells, promoting proliferation, invasion, and migration. These findings suggest that these four stemness-related miRNAs could serve as therapeutic targets, potentially improving therapeutic responses by targeting stemness-related genes.
Collapse
Affiliation(s)
- Jin-Wei Chen
- Department of Urology, Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Run-Nan Shen
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiang-Quan Zhu
- Department of Urology, Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ying-Hang Wang
- Department of Urology, Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Liang-Min Fu
- Department of Urology, Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu-Hang Chen
- Department of Urology, Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jia-Zheng Cao
- Department of Urology, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Jin-Huan Wei
- Department of Urology, Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jun-Hang Luo
- Department of Urology, Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Jia-Ying Li
- Department of Urology, Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Cheng-Peng Gui
- Department of Urology, Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
18
|
Ma L, Luan Y, Lu L. Analyze the Diversity and Function of Immune Cells in the Tumor Microenvironment From the Perspective of Single-Cell RNA Sequencing. Cancer Med 2025; 14:e70622. [PMID: 40062730 PMCID: PMC11891933 DOI: 10.1002/cam4.70622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/14/2024] [Accepted: 01/09/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Cancer development is closely associated with complex alterations in the tumor microenvironment (TME). Among these, immune cells within the TME play a huge role in personalized tumor diagnosis and treatment. OBJECTIVES This review aims to summarize the diversity of immune cells in the TME, their impact on patient prognosis and treatment response, and the contributions of single-cell RNA sequencing (scRNA-seq) in understanding their functional heterogeneity. METHODS We analyzed recent studies utilizing scRNA-seq to investigate immune cell populations in the TME, focusing on their interactions and regulatory mechanisms. RESULTS ScRNA-seq reveals the functional heterogeneity of immune cells, enhances our understanding of their role in tumor antibody responses, and facilitates the construction of immune cell interaction networks. These insights provide guidance for the development of cancer immunotherapies and personalized treatment approaches. CONCLUSION Applying scRNA-seq to immune cell analysis in the TME offers a novel pathway for personalized cancer treatment. Despite its promise, several challenges remain, highlighting the need for further advancements to fully integrate scRNA-seq into clinical applications.
Collapse
Affiliation(s)
- Lujuan Ma
- Department of Medical Oncology, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouGuangdongChina
| | - Yu Luan
- Department of Medical Oncology, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouGuangdongChina
| | - Lin Lu
- Department of Medical Oncology, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouGuangdongChina
| |
Collapse
|
19
|
Kam NW, Lau CY, Lau JYH, Dai X, Liang Y, Lai SPH, Chung MKY, Yu VZ, Qiu W, Yang M, Smith C, Khanna R, Ng KM, Dai W, Che CM, Lee VHF, Kwong DLW. Cell-associated galectin 9 interacts with cytotoxic T cells confers resistance to tumor killing in nasopharyngeal carcinoma through autophagy activation. Cell Mol Immunol 2025; 22:260-281. [PMID: 39910335 PMCID: PMC11868493 DOI: 10.1038/s41423-024-01253-8] [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: 04/27/2024] [Revised: 11/14/2024] [Accepted: 12/24/2024] [Indexed: 02/07/2025] Open
Abstract
Immune effector cells, including cytotoxic T lymphocytes (CTLs) play essential roles in eliminating cancer cells. However, their functionality is often compromised, even when they infiltrate the tumor microenvironment (TME) or are transferred to cancer patients adoptively. In this study, we focused on galectin 9 (G9), an inhibitory ligand that we observed to be predominately positioned on the plasma membrane and readily interacts with CD8 + CTL in the TME of nasopharyngeal carcinoma (NPC). We discovered that cell-cell contact between activated effector CTLs and target tumor cells (TarTC) with G9 overexpression led to cellular death defects. Despite the formation of CTL-TarTC conjugates, there is no impact on the cell number nor viability of CTL, and the release of cytolytic content and associated activity were not completely abrogated. Instead, this interaction promoted autophagy and restricted necrosis in the TarTC. Furthermore, reducing G9 expression in tumor cells enhanced the suppressive effect on tumor growth upon adoptive transfer of activated effector CTL. Additionally, inhibiting autophagy effectively controlled tumor growth in cases of G9 overexpression. Therefore, we highlight the contribution of G9 in facilitating the resistance of NPC to CTL-mediated killing by inducing a selection-cell death state in tumor cells, characterized by increased autophagy and decreased necrosis.
Collapse
Affiliation(s)
- Ngar-Woon Kam
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Synthetic Chemistry and Chemical Biology Limited, Hong Kong, China
| | - Cho Yiu Lau
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Synthetic Chemistry and Chemical Biology Limited, Hong Kong, China
| | | | - Xin Dai
- Laboratory of Synthetic Chemistry and Chemical Biology Limited, Hong Kong, China
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yusi Liang
- LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Syrus Pak Hei Lai
- LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | | | - Valen Zhuoyou Yu
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wenting Qiu
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China
| | - Mengsu Yang
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China
| | - Corey Smith
- QIMR Centre for Immunotherapy and Vaccine Development and Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Rajiv Khanna
- QIMR Centre for Immunotherapy and Vaccine Development and Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Kwan Ming Ng
- Laboratory of Synthetic Chemistry and Chemical Biology Limited, Hong Kong, China
| | - Wei Dai
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chi Ming Che
- Laboratory of Synthetic Chemistry and Chemical Biology Limited, Hong Kong, China
- Department of Chemistry, Faculty of Science, The University of Hong Kong, Hong Kong, China
| | - Victor Ho-Fun Lee
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
| | - Dora L W Kwong
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
20
|
Ruan L, Wang L. Adoptive cell therapy against tumor immune evasion: mechanisms, innovations, and future directions. Front Oncol 2025; 15:1530541. [PMID: 40094019 PMCID: PMC11906336 DOI: 10.3389/fonc.2025.1530541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025] Open
Abstract
Tumors employ a range of strategies to evade detection and eradication by the host's immune system. These include downregulating antigen expression, altering antigen presentation processes, and inhibiting immune checkpoint pathways. etc. Adoptive Cell Therapy (ACT) represents a strategy that boosts anti-tumor immunity. This is achieved by amplifying or genetically engineering immune cells, which are either sourced from the patient or a donor, in a laboratory setting. Subsequently, these cells are reintroduced into the patient to bolster their immune response against cancer. ACT has successfully restored anti-tumor immune responses by amplifying the activity of T cells from patients or donors. This review focuses on the mechanisms underlying tumor escape, including alterations in tumor cell antigens, the immunosuppressive tumor microenvironment (TME), and modulation of immune checkpoint pathways. It further explores how ACT can avddress these factors to enhance therapeutic efficacy. Additionally, the review discusses the application of gene-editing technologies (such as CRISPR) in ACT, highlighting their potential to strengthen the anti-tumor capabilities of T cells. Looking forward, the personalized design of ACT, combined with immune checkpoint inhibitors and targeted therapies, is expected to significantly improve treatment outcomes, positioning this approach as a key strategy in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Liqin Ruan
- Department of Hepatobiliary Surgery, JiuJiang City Key Laboratory of Cell Therapy, JiuJiang No.1 People's Hospital, Jiujiang, Jiangxi, China
| | - Lu Wang
- Department of Oncology, JiuJiang City Key Laboratory of Cell Therapy, JiuJiang No.1 People's Hospital, Jiujiang, Jiangxi, China
| |
Collapse
|
21
|
Liu J, Zhao T, Sun Z, Wang J, Chai Z, Chen G. Single-cell profiling and clinical characteristics analysis of lung squamous carcinoma. Funct Integr Genomics 2025; 25:45. [PMID: 40014154 DOI: 10.1007/s10142-025-01556-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/28/2025]
Abstract
Lung squamous carcinoma (LUSC) is a highly heterogeneous disease. However, the tumor microenvironment (TME) landscape and clinical characteristics for LUSC have not yet been elucidated. To map the TME and clinical characteristics of LUSC, we performed single-cell RNA sequencing for 504 LUSC samples on basis of TCGA and Gene Expression Omnibus. We introduced the computational algorithms "ESTIMATE" and "CIBERSORT" to analyze immune cell infiltration and immune-checkpoint-related gene signatures in various LUSC clusters. Weighted gene co-expression network analysis was used to explore the connections between molecular characteristics and clinical traits in LUSC. A prognostic model was constructed by performing multivariate COX. Two gene clusters exhibiting disparate immune and clinical characteristics were identified. Our findings indicate that patients in cluster 2, who have a more favorable prognosis, exhibit immune characteristics such as elevated levels of immunosuppression-associated M2 macrophages, resting memory CD4 T cells, resting dendritic cells (DC), and TNFRSF4, alongside reduced infiltration of activated DC and lower expression of TNFRSF18.Whereafter, the Risk Score model was built on basis of 3-DEGs signature consisted of cystatin C (CST3), transglutaminase type 2 (TGM2), JUN, which were proved by q-PCR and immunofluorescence. Besides, high-Risk Score may be responsible for poor prognosis in LUSC patients. Our study identified that tumor-infiltrating immune cell subtypes and the Risk Score model might shed light on the heterogeneity in LUSC patients. The TME, three DEGs and Risk Score can effectively serve as biomarkers to elucidate the immune landscape and predict prognosis in LUSC patients. They may provide insights to the investigations on therapeutic strategies for LUSC.
Collapse
Affiliation(s)
- Jie Liu
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P.R. China.
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P.R. China.
| | - Tian Zhao
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P.R. China
| | - Zhengliang Sun
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P.R. China
| | - Jinyi Wang
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P.R. China
| | - Zhengjun Chai
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P.R. China
| | - Guohan Chen
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P.R. China.
| |
Collapse
|
22
|
Chen G, Sun DC, Ba Y, Zhang YX, Zhou T, Zhao YY, Zhao HY, Fang WF, Huang Y, Wang Z, Deng C, Hu DS, Wang W, Lin JG, Li GL, Luo SX, Fu ZC, Zhu HS, Wang HL, Cai SL, Kang XQ, Zhang L, Yang YP. Anti-LAG-3 antibody LBL-007 plus anti-PD-1 antibody toripalimab in advanced nasopharyngeal carcinoma and other solid tumors: an open-label, multicenter, phase Ib/II trial. J Hematol Oncol 2025; 18:15. [PMID: 39920751 PMCID: PMC11806529 DOI: 10.1186/s13045-025-01666-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/25/2025] [Indexed: 02/09/2025] Open
Abstract
PURPOSE Open-label phase Ib/II study to investigate the safety and efficacy of LBL-007, an anti-LAG-3 antibody, plus toripalimab, an anti-PD-1 antibody, in patients with previously treated advanced nasopharyngeal carcinoma (NPC) and other solid tumors. METHODS Patients with advanced tumors refractory to prior standard therapies were enrolled. In phase Ib, patients received LBL-007 200 mg or 400 mg and toripalimab 240 mg intravenously once every 3 weeks. In phase II, all patients received LBL-007 at the recommended phase II dose (RP2D) and toripalimab 240 mg intravenously once every 3 weeks. The primary end points were safety in phase Ib and objective response rate (ORR) in phase II. The exploratory end point was the predictive capability of LAG-3 and PD-L1 expression for efficacy. RESULTS Between November 30, 2021, and December 1, 2023, 80 patients were enrolled, including 30 (37.5%) with NPC and 50 (62.5%) with other tumors. Median follow-up was 26.0 months. In Phase Ib, LBL-007 was administered at 200 mg to four patients and 400 mg to six patients, with no dose-limiting toxicities observed. Therefore, the 400 mg dose of LBL-007 was established as the RP2D and administered to 70 patients in phase II. Nine (11.3%) of 80 patients had grade 3 or 4 treatment-related adverse events, the most common of which included anemia (2.5%), hyponatremia (2.5%), increased alanine aminotransferase (2.5%), increased aspartate aminotransferase (1.3%), and fatigue (1.3%). Eight patients (10.0%) had treatment-related serious adverse events. No treatment-related deaths were reported. In immunotherapy-naive NPC patients (n = 12), ORR was 33.3%, disease control rate (DCR) was 75%, and median progression-free survival (PFS) was 10.8 months (95% CI, 1.3 to not estimated). In IO-treated NPC patients (n = 17), ORR was 11.8%, DCR was 64.7%, and median PFS was 2.7 months (95% CI, 1.4 to 4.9). For other tumors, ORRs were 15.8% in immunotherapy-naive patients and 3.7% in immunotherapy-treated patients. Patients with ≥ 2 + LAG-3 expression had a higher ORR of 28.0%, compared to 7.7% in those with < 2 + LAG-3 expression. CONCLUSION LBL-007 plus toripalimab exhibited a manageable safety profile in patients with advanced solid tumors and demonstrated promising antitumor activity in NPC, especially in immunotherapy-naive patients. These findings warrant further validation in future studies.
Collapse
Affiliation(s)
- Gang Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Dongfeng East Road 651, Guangzhou, 510060, Guangdong, P.R. China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Dong-Chen Sun
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Dongfeng East Road 651, Guangzhou, 510060, Guangdong, P.R. China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yi Ba
- Department of Gastrointestinal Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, P.R. China
| | - Ya-Xiong Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Dongfeng East Road 651, Guangzhou, 510060, Guangdong, P.R. China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Ting Zhou
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Dongfeng East Road 651, Guangzhou, 510060, Guangdong, P.R. China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yuan-Yuan Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Dongfeng East Road 651, Guangzhou, 510060, Guangdong, P.R. China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Hong-Yun Zhao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Clinical Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Wen-Feng Fang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Dongfeng East Road 651, Guangzhou, 510060, Guangdong, P.R. China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yan Huang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Dongfeng East Road 651, Guangzhou, 510060, Guangdong, P.R. China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Zhen Wang
- Department of Oncology, Linyi Cancer Hospital, Linyi, P.R. China
| | - Chao Deng
- Cancer Center, Chong Qing University Three Gorges Hospital, Chongqing, P.R. China
| | - De-Sheng Hu
- Department of Radiation Oncology, Hubei Cancer Hospital, Wuhan, P.R. China
| | - Wei Wang
- Hunan Cancer Hospital, Changsha, P.R. China
| | | | - Gui-Ling Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Su-Xia Luo
- Department of Oncology, Henan Cancer Hospital, Zhengzhou, P.R. China
| | - Zhi-Chao Fu
- Department of Radiotherapy, No. 900 Hospital of the PLA Joint Logistics Support Force, Fuzhou, P.R. China
| | - Hai-Sheng Zhu
- Department of Oncology, The First People's Hospital of Yulin, Yulin, P.R. China
| | - Hui-Li Wang
- Department of Oncology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Sheng-Li Cai
- Nanjing Leads Biolabs Co., Ltd., Nanjing, P.R. China
| | | | - Li Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Dongfeng East Road 651, Guangzhou, 510060, Guangdong, P.R. China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yun-Peng Yang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Dongfeng East Road 651, Guangzhou, 510060, Guangdong, P.R. China.
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China.
| |
Collapse
|
23
|
Wang Y, Li R, Tong R, Chen T, Sun M, Luo L, Li Z, Chen Y, Zhao Y, Zhang C, Wei L, Lin W, Chen H, Qian K, Chen AF, Liu J, Chen L, Li B, Wang F, Wang L, Su B, Pu J. Integrating single-cell RNA and T cell/B cell receptor sequencing with mass cytometry reveals dynamic trajectories of human peripheral immune cells from birth to old age. Nat Immunol 2025; 26:308-322. [PMID: 39881000 PMCID: PMC11785523 DOI: 10.1038/s41590-024-02059-6] [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: 10/28/2022] [Accepted: 12/09/2024] [Indexed: 01/31/2025]
Abstract
A comprehensive understanding of the evolution of the immune landscape in humans across the entire lifespan at single-cell transcriptional and protein levels, during development, maturation and senescence is currently lacking. We recruited a total of 220 healthy volunteers from the Shanghai Pudong Cohort (NCT05206643), spanning 13 age groups from 0 to over 90 years, and profiled their peripheral immune cells through single-cell RNA-sequencing coupled with single T cell and B cell receptor sequencing, high-throughput mass cytometry, bulk RNA-sequencing and flow cytometry validation experiments. We revealed that T cells were the most strongly affected by age and experienced the most intensive rewiring in cell-cell interactions during specific age. Different T cell subsets displayed different aging patterns in both transcriptomes and immune repertoires; examples included GNLY+CD8+ effector memory T cells, which exhibited the highest clonal expansion among all T cell subsets and displayed distinct functional signatures in children and the elderly; and CD8+ MAIT cells, which reached their peaks of relative abundance, clonal diversity and antibacterial capability in adolescents and then gradually tapered off. Interestingly, we identified and experimentally verified a previously unrecognized 'cytotoxic' B cell subset that was enriched in children. Finally, an immune age prediction model was developed based on lifecycle-wide single-cell data that can evaluate the immune status of healthy individuals and identify those with disturbed immune functions. Our work provides both valuable insights and resources for further understanding the aging of the immune system across the whole human lifespan.
Collapse
MESH Headings
- Humans
- Aged
- Single-Cell Analysis/methods
- Adult
- Aged, 80 and over
- Middle Aged
- Infant
- Child
- Child, Preschool
- Male
- Female
- Adolescent
- Aging/immunology
- Aging/genetics
- Infant, Newborn
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Flow Cytometry
- Young Adult
- Receptors, Antigen, T-Cell/genetics
- Sequence Analysis, RNA
- T-Lymphocyte Subsets/immunology
- B-Lymphocytes/immunology
Collapse
Affiliation(s)
- Yufei Wang
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Ronghong Li
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Renyang Tong
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Taiwei Chen
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Mingze Sun
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Lingjie Luo
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Li
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yifan Chen
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yichao Zhao
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Chensheng Zhang
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Lin Wei
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Lin
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Haoyan Chen
- Department of Gastroenterology and Hepatology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun Qian
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
- School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Chen
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bing Su
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Pu
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China.
- Aging Biomarker Consortium, Beijing, China.
| |
Collapse
|
24
|
Zhang J, Mizuuchi Y, Ohuchida K, Hisano K, Shimada Y, Katayama N, Tsutsumi C, Tan BC, Nagayoshi K, Tamura K, Fujimoto T, Ikenaga N, Nakata K, Oda Y, Nakamura M. Exploring the tumor microenvironment of colorectal cancer patients post renal transplantation by single-cell analysis. Cancer Sci 2025; 116:500-512. [PMID: 39623744 PMCID: PMC11786312 DOI: 10.1111/cas.16409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/26/2024] [Accepted: 11/08/2024] [Indexed: 02/02/2025] Open
Abstract
Patients with colorectal cancer (CRC) following renal transplantation require long-term immunosuppressants to prevent graft rejection. However, the impact of these immunosuppressants on the tumor immune microenvironment and the roles of immune cells within it remain poorly understood. We conducted comprehensive single-cell RNA sequencing on tumor and normal tissues from four CRC patients post renal transplantation and compared these with published data from 23 non-transplant CRC patients. We set four groups for detailed comparative analysis based on the renal transplantation status and tissue origin: non-renal transplantation normal (nRT_Normal), non-renal transplantation tumor (nRT_Tumor), renal transplantation normal (RT_Normal), renal transplantation tumor (RT_Tumor). Our analysis revealed significant tumor immune microenvironment landscape alterations in the transplantation group. CD8+effector T cells of RT_Tumor showed significantly diminished cytotoxicity and tumor neoantigen recognition (p < 0.0001), while CD4+FOXP3 regulatory T cells of RT_Tumor displayed a higher inhibitory score (p < 0.05), indicating preserved immunomodulatory potential compared with non-transplant CRC. Notably, significantly increased CTLA4 expression in T cells of RT_Tumor was found and testified (p < 0.05). Our findings provide novel mechanistic insights for understanding the immune landscape in renal transplant recipients with CRC and pave the way for potential immunotherapeutic strategies that may improve survival and quality of life for this patient population.
Collapse
Affiliation(s)
- Jinghui Zhang
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yusuke Mizuuchi
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Kenoki Ohuchida
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Advanced Medical Initiatives, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Kyoko Hisano
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yuki Shimada
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Anatomical Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Naoki Katayama
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Chikanori Tsutsumi
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Bryan C. Tan
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Kinuko Nagayoshi
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Koji Tamura
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Takaaki Fujimoto
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Naoki Ikenaga
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Kohei Nakata
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yoshinao Oda
- Department of Anatomical Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| |
Collapse
|
25
|
Wu X, Pan B, Chu C, Zhang Y, Ma J, Xing Y, Ma Y, Zhu W, Zhong H, Alimu A, Zhou G, Liu S, Chen W, Li X, Puyi S. CXCL16/CXCR6/TGF-β Feedback Loop Between M-MDSCs and Treg Inhibits Anti-Bacterial Immunity During Biofilm Infection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409537. [PMID: 39716908 PMCID: PMC11831521 DOI: 10.1002/advs.202409537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/29/2024] [Indexed: 12/25/2024]
Abstract
Staphylococcus aureus (S. aureus) is a leading cause of Periprosthetic joint infection (PJI), a severe complication after joint arthroplasty. Immunosuppression is a major factor contributing to the infection chronicity of S. aureus PJI, posing significant treatment challenges. This study investigates the relationship between the immunosuppressive biofilm milieu and S. aureus PJI outcomes in both discovery and validation cohorts. This scRNA-seq analysis of synovium from PJI patients reveals an expansion and heightened activity of monocyte-related myeloid-derived suppressor cells (M-MDSCs) and regulatory T cells (Treg). Importantly, CXCL16 is significantly upregulated in M-MDSCs, with its corresponding CXCR6 receptor also elevated on Treg. M-MDSCs recruit Treg and enhance its activity via CXCL16-CXCR6 interactions, while Treg secretes TGF-β, inducing M-MDSCs proliferation and immunosuppressive activity. Interfering with this cross-talk in vivo using Treg-specific CXCR6 knockout PJI mouse model reduces M-MDSCs/Treg-mediated immunosuppression and alleviates bacterial burden. Immunohistochemistry and recurrence analysis show that PJI patients with CXCR6high synovium have poor prognosis. This findings highlight the critical role of CXCR6 in Treg in orchestrating an immunosuppressive microenvironment and biofilm persistence during PJI, offering potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Xiaoyu Wu
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Baiqi Pan
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Chenghan Chu
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Yangchun Zhang
- Department of OrthopedicsThe People's Hospital of Baoan ShenzhenShenzhenGuangdong518101China
- Department of OrthopedicsThe Second Affiliated Hospital of Shenzhen UniversityShenzhenGuangdong518101China
| | - Jinjin Ma
- Technology School of MedicineSouth China University of TechnologyGuangzhouGuangdong510640China
- Shien‐ming Wu School of Intelligent EngineeringSouth China University of TechnologyGuangzhouGuangdong510640China
| | - Yang Xing
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Yuanchen Ma
- Department of OrthopedicsGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouGuangdong519041China
| | - Wengang Zhu
- Department of Joint OrthopedicsYuebei People's HospitalShaoguanGuangdong512099China
| | - Huan Zhong
- Department of Joint SurgeryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong524002China
| | - Aerman Alimu
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Guanming Zhou
- Department of OrthopedicsFoshan Hospital of Traditional Chinese MedicineGuangzhouGuangdong528051China
| | - Shuying Liu
- Department of Histology and EmbryologyZhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Weishen Chen
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| | - Xiang Li
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Department of Spine SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Sheng Puyi
- Department of Joint SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Clinical Research Center for Orthopedic DiseasesThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Orthopaedics and TraumatologyGuangzhouGuangdong510080China
| |
Collapse
|
26
|
Xue Z, Wu L, Tian R, Gao B, Zhao Y, He B, Sun D, Zhao B, Li Y, Zhu K, Wang L, Yao J, Liu W, Lu L. Integrative mapping of human CD8 + T cells in inflammation and cancer. Nat Methods 2025; 22:435-445. [PMID: 39614111 DOI: 10.1038/s41592-024-02530-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/16/2024] [Indexed: 12/01/2024]
Abstract
CD8+ T cells exhibit remarkable phenotypic diversity in inflammation and cancer. However, a comprehensive understanding of their clonal landscape and dynamics remains elusive. Here we introduce scAtlasVAE, a deep-learning-based model for the integration of large-scale single-cell RNA sequencing data and cross-atlas comparisons. scAtlasVAE has enabled us to construct an extensive human CD8+ T cell atlas, comprising 1,151,678 cells from 961 samples across 68 studies and 42 disease conditions, with paired T cell receptor information. Through incorporating information in T cell receptor clonal expansion and sharing, we have successfully established connections between distinct cell subtypes and shed light on their phenotypic and functional transitions. Notably, our approach characterizes three distinct exhausted T cell subtypes and reveals diverse transcriptome and clonal sharing patterns in autoimmune and immune-related adverse event inflammation. Furthermore, scAtlasVAE facilitates the automatic annotation of CD8+ T cell subtypes in query single-cell RNA sequencing datasets, enabling unbiased and scalable analyses. In conclusion, our work presents a comprehensive single-cell reference and computational framework for CD8+ T cell research.
Collapse
Affiliation(s)
- Ziwei Xue
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China
- Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China
| | - Lize Wu
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China
- Institute of Immunology and Department of Rheumatology at Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruonan Tian
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China
| | - Bing Gao
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Zhao
- AI Lab, Tencent, Shenzhen, China
| | - Bing He
- AI Lab, Tencent, Shenzhen, China
| | - Di Sun
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingkang Zhao
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China
- Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Yicheng Li
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China
- Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Kaixiang Zhu
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lie Wang
- Bone Marrow Transplantation Center and Institute of Immunology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | - Wanlu Liu
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China.
- Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK.
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China.
- Zhejiang Key Laboratory of Medical Imaging Artificial Intelligence, Haining, China.
| | - Linrong Lu
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China.
- Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK.
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China.
- Institute of Immunology and Department of Rheumatology at Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
27
|
Chong WQ, Low JL, Tay JK, Le TBU, Goh GSQ, Sooi K, Teo HL, Cheo SW, Wong RTX, Samol J, Lim MY, Li H, Shirgaonkar N, Chia S, Wang L, Gopinathan A, Eu DKC, Tsang RKY, Loh KS, Toh HC, Syn N, Kong LR, Dasgupta R, Tai BC, Lim YC, Goh BC. Pembrolizumab with or without bevacizumab in platinum-resistant recurrent or metastatic nasopharyngeal carcinoma: a randomised, open-label, phase 2 trial. Lancet Oncol 2025; 26:175-186. [PMID: 39826567 DOI: 10.1016/s1470-2045(24)00677-6] [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: 07/30/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) is overexpressed in nasopharyngeal carcinoma and suppresses the anti-tumour immune response. Previous studies have shown that adding anti-VEGF treatment to PD-1 inhibition treatment strategies improves tumour response. We aimed to compare the efficacy of pembrolizumab, a PD-1 inhibitor, with or without bevacizumab, a VEGF inhibitor, in nasopharyngeal carcinoma. METHODS In this randomised, open-label, phase 2 trial done at two hospitals (National University Cancer Institute and Tan Tock Seng Hospital) in Singapore, patients with platinum-resistant recurrent or metastatic nasophayngeal carcinoma were eligible if they were aged 21 years or older and had an Eastern Cooperative Oncology Group (ECOG) performance status of 0-1. Patients were assigned (1:1; using random permuted blocks with varying sizes of 4 and 6) to receive either intravenous pembrolizumab (200 mg) every 21 days or a combination of pembrolizumab with intravenous bevacizumab (7·5 mg/kg) administered 1 week prior to each dose, until radiographic disease progression, unacceptable toxicity, completion of 32 cycles, or withdrawal of consent. The study was open label, therefore no masking of treatment assignment was implemented. The primary endpoint was objective response rate, assessed using RECIST (version 1.1) by independent radiologists and analysed in the intention-to-treat population (ie, all randomly assigned patients). This trial is registered with ClinicalTrials.gov, NCT03813394, and enrolment has closed. FINDINGS Between May 13, 2019, and Dec 6, 2023, we assessed 60 individuals for eligibility, 12 were excluded, and 48 were randomly allocated to pembrolizumab alone (n=24) or a combination of bevacizumab and pembrolizumab (n=24). The median age was 56 years (IQR 48-65), and 40 (83%) of 48 patients were male and eight (17%) were female. The median follow-up was 28·3 months (IQR 15·1-55·9). The objective response rate was significantly higher in the bevacizumab and pembrolizumab group (58·3% [95% CI 36·6-77·9] than in the pembrolizumab group (12·5% [2·7-32·4]; unadjusted RR 4·67 [95% CI 1·54-14·18]; p=0·0010). Grade 3 treatment-related adverse events occurred in two (8%) of 24 patients in the pembrolizumab group and in seven (29%) of 24 patients in the bevacizumab and pembrolizumab group; the most common severe or grade 3-4 treatment-related adverse events were thrombosis or bleeding (four [17%] of 24 patients in the bevacizumab and pembrolizumab group vs none of 24 patients in the pembrolizumab group), and others were transaminitis (none vs 1 [4%]), colitis (1 [4%] vs none]), cytopenias (none vs 1 [4%]), dermatological toxicities (1 [4%] vs none]), hypertension (1 [4%] vs none]), and proteinuria (1 [4%] vs none]). There were no grade 4 treatment-related adverse events or treatment-related deaths in either group. INTERPRETATION Pembrolizumab in combination with bevacizumab was more efficacious than pembrolizumab monotherapy, with manageable toxicities in platinum-resistant nasopharyngeal carcinoma. If validated in a phase 3 trial, the combination therapy could be a new standard of care in this population of patients. FUNDING National Medical Research Council of Singapore, National Research Foundation Singapore, Singapore Ministry of Education under its Research Centres of Excellence initiatives, and Merck Sharp & Dohme.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Bevacizumab/administration & dosage
- Bevacizumab/adverse effects
- Male
- Female
- Middle Aged
- Nasopharyngeal Carcinoma/drug therapy
- Nasopharyngeal Carcinoma/pathology
- Nasopharyngeal Carcinoma/mortality
- Nasopharyngeal Carcinoma/secondary
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Adult
- Drug Resistance, Neoplasm/drug effects
- Aged
- Nasopharyngeal Neoplasms/drug therapy
- Nasopharyngeal Neoplasms/pathology
- Nasopharyngeal Neoplasms/mortality
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/pathology
- Antineoplastic Agents, Immunological
- Immune Checkpoint Inhibitors/adverse effects
- Immune Checkpoint Inhibitors/administration & dosage
Collapse
Affiliation(s)
- Wan-Qin Chong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Jia-Li Low
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Joshua K Tay
- Department of Otolaryngology, Head and Neck Surgery, National University Hospital, Singapore; Department of Otolaryngology, Head and Neck Surgery, National University of Singapore, Singapore
| | - Thi Bich Uyen Le
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Grace Shi-Qing Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Kenneth Sooi
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Hui-Lin Teo
- Department of Medical Oncology, Tan Tock Seng Hospital, Singapore
| | - Seng-Wee Cheo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Regina Tong-Xin Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Jens Samol
- Department of Medical Oncology, Tan Tock Seng Hospital, Singapore; Lee Kong Chian School of Medicine, Singapore; School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Ming-Yann Lim
- Department of Otorhinolaryngology and Head and Neck Surgery, Tan Tock Seng Hospital, Singapore
| | - Hao Li
- Department of Otorhinolaryngology and Head and Neck Surgery, Tan Tock Seng Hospital, Singapore
| | - Niranjan Shirgaonkar
- Genome Institute of Singapore, Agency of Science, Technology and Research, Singapore
| | - Shumei Chia
- Genome Institute of Singapore, Agency of Science, Technology and Research, Singapore
| | - Lingzhi Wang
- Department of Pharmacology, National University of Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Anil Gopinathan
- Department of Diagnostic Imaging, National University Hospital, Singapore
| | - Donovan Kum-Chuen Eu
- Department of Otolaryngology, Head and Neck Surgery, National University Hospital, Singapore
| | - Raymond King-Yin Tsang
- Department of Otolaryngology, Head and Neck Surgery, National University Hospital, Singapore; Department of Otolaryngology, Head and Neck Surgery, National University of Singapore, Singapore
| | - Kwok-Seng Loh
- Department of Otolaryngology, Head and Neck Surgery, National University Hospital, Singapore; Department of Otolaryngology, Head and Neck Surgery, National University of Singapore, Singapore
| | | | - Nicholas Syn
- Department of Pathology, National University of Singapore, Singapore
| | - Li-Ren Kong
- Department of Pharmacology, National University of Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Ramanuj Dasgupta
- Genome Institute of Singapore, Agency of Science, Technology and Research, Singapore
| | - Bee-Choo Tai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Yaw-Chyn Lim
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Boon-Cher Goh
- Department of Haematology-Oncology, National University Cancer Institute, Singapore; Department of Pharmacology, National University of Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore.
| |
Collapse
|
28
|
Neugebauer E, Walter S, Tan J, Drayman N, Franke V, van Gent M, Pennisi S, Veratti P, Stein KS, Welker I, Tay S, Verjans GMGM, Timmers HTM, Akalin A, Landthaler M, Ensser A, Wyler E, Full F. Herpesviruses mimic zygotic genome activation to promote viral replication. Nat Commun 2025; 16:710. [PMID: 39814710 PMCID: PMC11735616 DOI: 10.1038/s41467-025-55928-5] [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: 10/16/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025] Open
Abstract
Zygotic genome activation (ZGA) is crucial for maternal to zygotic transition at the 2-8-cell stage in order to overcome silencing of genes and enable transcription from the zygotic genome. In humans, ZGA is induced by DUX4, a pioneer factor that drives expression of downstream germline-specific genes and retroelements. Here we show that herpesviruses from all subfamilies, papillomaviruses and Merkel cell polyomavirus actively induce DUX4 expression to promote viral transcription and replication. Analysis of single-cell sequencing data sets from patients shows that viral DUX4 activation is of relevance in vivo. Herpes-simplex virus 1 (HSV-1) immediate early proteins directly induce expression of DUX4 and its target genes, which mimics zygotic genome activation. Upon HSV-1 infection, DUX4 directly binds to the viral genome and promotes viral transcription. DUX4 is functionally required for infection, since genetic depletion by CRISPR/Cas9 as well as degradation of DUX4 by nanobody constructs abrogates HSV-1 replication. Our results show that DNA viruses including herpesviruses mimic an embryonic-like transcriptional program that prevents epigenetic silencing of the viral genome and facilitates herpesviral gene expression.
Collapse
Affiliation(s)
- Eva Neugebauer
- Institute of Virology, University Medical Center, and Faculty of Medicine, Albert-Ludwig-University Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Stephanie Walter
- Institute for Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Jiang Tan
- Institute of Virology, University Medical Center, and Faculty of Medicine, Albert-Ludwig-University Freiburg, Freiburg, Germany
| | - Nir Drayman
- The Department of Molecular Biology and Biochemistry, the Center for Virus Research and the Center for Complex Biological Systems, The University of California, Irvine, Irvine, CA, 92697, USA
| | - Vedran Franke
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Helmholtz Society, Berlin, Germany
| | - Michiel van Gent
- HerpesLabNL, Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sandra Pennisi
- Institute of Virology, University Medical Center, and Faculty of Medicine, Albert-Ludwig-University Freiburg, Freiburg, Germany
| | - Pia Veratti
- Institute of Virology, University Medical Center, and Faculty of Medicine, Albert-Ludwig-University Freiburg, Freiburg, Germany
| | - Karla S Stein
- Institute of Virology, University Medical Center, and Faculty of Medicine, Albert-Ludwig-University Freiburg, Freiburg, Germany
| | - Isabelle Welker
- Institute of Virology, University Medical Center, and Faculty of Medicine, Albert-Ludwig-University Freiburg, Freiburg, Germany
| | - Savaş Tay
- The Pritzker School for Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA
| | - Georges M G M Verjans
- HerpesLabNL, Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - H T Marc Timmers
- German Cancer Consortium (DKTK), partner site Freiburg, a partnership between the DKFZ and Medical Center-University of Freiburg, and Department of Urology, Medical Center-University of Freiburg, Freiburg, Germany
| | - Altuna Akalin
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Helmholtz Society, Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Helmholtz Society, Berlin, Germany
| | - Armin Ensser
- Institute for Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Helmholtz Society, Berlin, Germany
| | - Florian Full
- Institute of Virology, University Medical Center, and Faculty of Medicine, Albert-Ludwig-University Freiburg, Freiburg, Germany.
- German Consulting Laboratory for HSV and VZV, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
29
|
Wang H, Zhan Y, Luo J, Wang W, Fan S. Unveiling immune resistance mechanisms in nasopharyngeal carcinoma and emerging targets for antitumor immune response: tertiary lymphoid structures. J Transl Med 2025; 23:38. [PMID: 39789621 PMCID: PMC11721552 DOI: 10.1186/s12967-024-05880-7] [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: 07/31/2024] [Accepted: 11/13/2024] [Indexed: 01/12/2025] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a prevalent malignancy in China, commonly associated with undifferentiated cell types and Epstein-Barr virus (EBV) infection. The presence of intense lymphocytic infiltration and elevated expression of programmed cell death ligand 1(PD-L1) in NPC highlights its potential for immunotherapy, yet current treatment outcomes remain suboptimal. In this review, we explore the tumor microenvironment of NPC to better understand the mechanisms of resistance to immunotherapy, evaluate current therapeutic strategies, and pinpoint emerging targets, such as tertiary lymphoid structures (TLSs), that could enhance treatment outcomes and prognostic accuracy. TLSs have demonstrated positive prognostic value in NPC, making them a promising target for future therapies. This review summarizes the key characteristics of TLSs and latest research in the context of NPC. We are optimistic that targeting TLSs could improve immunotherapy outcomes for NPC patients, ultimately leading to more effective treatment strategies and better patient survival.
Collapse
Affiliation(s)
- Huilin Wang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, 410011, China
| | - Yuting Zhan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, 410011, China
| | - Jiadi Luo
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, 410011, China
| | - Weiyuan Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, 410011, China.
| |
Collapse
|
30
|
Zhang Y, Han S, Sun Q, Liu T, Wen Z, Yao M, Zhang S, Duan Q, Zhang X, Pang B, Kou Z, Jiang X. Single-cell transcriptome atlas of peripheral immune features to Omicron breakthrough infection under booster vaccination strategies. Front Immunol 2025; 15:1460442. [PMID: 39835127 PMCID: PMC11743671 DOI: 10.3389/fimmu.2024.1460442] [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: 07/06/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction The high percentage of Omicron breakthrough infection in vaccinees is an emerging problem, of which we have a limited understanding of the phenomenon. Methods We performed single-cell transcriptome coupled with T-cell/B-cell receptor (TCR/BCR) sequencing in 15 peripheral blood mononuclear cell (PBMC) samples from Omicron infection and naïve with booster vaccination. Results We found that after breakthrough infection, multiple cell clusters showed activation of the type I IFN pathway and widespread expression of Interferon-stimulated genes (ISGs); T and B lymphocytes exhibited antiviral and proinflammatory-related differentiation features with pseudo-time trajectories; and large TCR clonal expansions were concentrated in effector CD8 T cells, and clonal expansions of BCRs showed a preference for IGHV3. In addition, myeloid cells in the BA.5.2 breakthrough infection with the fourth dose of aerosolized Ad5-nCoV were characterized by enhanced proliferation, chemotactic migration, and antigen presentation. Discussion Collectively, our study informs the comprehensive understandings of immune characterization for Omicron breakthrough infection, revealing the positive antiviral potential induced by booster doses of vaccine and the possible "trained immunity" phenomenon in the fourth dose of aerosolized Ad5-nCoV, providing a basis for the selection of vaccination strategies.
Collapse
MESH Headings
- Humans
- Immunization, Secondary
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19/genetics
- Single-Cell Analysis
- Transcriptome
- SARS-CoV-2/immunology
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Leukocytes, Mononuclear/immunology
- Vaccination
- B-Lymphocytes/immunology
- Breakthrough Infections
Collapse
Affiliation(s)
- Yuwei Zhang
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Shanshan Han
- School of Public Health and Health Management, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Qingshuai Sun
- School of Public Health and Health Management, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Tao Liu
- Department of Infectious Disease Control, Yantai Center for Disease Control and Prevention, Yantai, Shandong, China
| | - Zixuan Wen
- School of Public Health, Weifang Medical University, Weifang, Shandong, China
| | - Mingxiao Yao
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Shu Zhang
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Qing Duan
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Xiaomei Zhang
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Bo Pang
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Zengqiang Kou
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Xiaolin Jiang
- School of Public Health and Health Management, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- School of Public Health, Weifang Medical University, Weifang, Shandong, China
- Shandong Provincial Key Laboratory of Infectious Disease Control and Prevention, Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
| |
Collapse
|
31
|
Song X, Zhu Y, Geng W, Jiao J, Liu H, Chen R, He Q, Wang L, Sun X, Qin W, Geng J, Chen Z. Spatial and single-cell transcriptomics reveal cellular heterogeneity and a novel cancer-promoting Treg cell subset in human clear-cell renal cell carcinoma. J Immunother Cancer 2025; 13:e010183. [PMID: 39755578 PMCID: PMC11748785 DOI: 10.1136/jitc-2024-010183] [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: 07/29/2024] [Accepted: 12/06/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is the most common histologic type of RCC. However, the spatial and functional heterogeneity of immunosuppressive cells and the mechanisms by which their interactions promote immunosuppression in the ccRCC have not been thoroughly investigated. METHODS To further investigate the cellular and regional heterogeneity of ccRCC, we analyzed single-cell and spatial transcriptome RNA sequencing data from four patients, which were obtained from samples from multiple regions, including the tumor core, tumor-normal interface, and distal normal tissue. On the basis, the findings were investigated in vitro using tissue and blood samples from 15 patients with ccRCC and validated in the broader samples on tissue microarrays. RESULTS In this study, we revealed previously unreported subsets of both stromal and immune cells, as well as mapped their spatial location at finer resolution. In addition, we validated the clusters of tumor cells after removing batch effects according to six characterized gene sets, including epithelial-mesenchymal transitionhigh clusters, metastatic clusters and proximal tubulehigh clusters. Importantly, we identified a special regulatory T (Treg) cell subpopulation that has the molecular characteristics of terminal effector Treg cells but expresses multiple cytokines, such as interleukin (IL)-1β and IL-18. This group of Treg cells has stronger immunosuppressive function and was associated with a worse prognosis in ccRCC cohorts. They were colocalized with MRC1 + FOLR2 + tumor-associated macrophages (TAMs) at the tumor-normal interface to form a positive feedback loop, maintaining a synergistic procarcinogenic effect. In addition, we traced the origin of IL-1β+ Treg cells and revealed that IL-18 can induce the expression of IL-1β in Treg cells via the ERK/NF-κB pathway. CONCLUSIONS We demonstrated a novel cancer-promoting Treg cell subset and its interactions with MRC1 + FOLR2 +TAMs, which provides new insight into Treg cell heterogeneity and potential therapeutic targets for ccRCC.
Collapse
Affiliation(s)
- Xiyu Song
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
- Xijing Innovation Research Institute, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yumeng Zhu
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wenwen Geng
- Department of Breast Surgery, Shandong University, Jinan, Shandong, China
| | - Jianhua Jiao
- Xijing Innovation Research Institute, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Urology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hongjiao Liu
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ruo Chen
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Qian He
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lijuan Wang
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiuxuan Sun
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Weijun Qin
- Department of Urology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jiejie Geng
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
- Xijing Innovation Research Institute, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xian, Shaanxi, China
| | - Zhinan Chen
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xian, Shaanxi, China
| |
Collapse
|
32
|
Guo C, Sun H, Du Y, Dai X, Pang Y, Han Z, Xiong X, Li S, Zhang J, Zheng Q, Gui X. Specifically blocking αvβ8-mediated TGF-β signaling to reverse immunosuppression by modulating macrophage polarization. J Exp Clin Cancer Res 2025; 44:1. [PMID: 39743547 PMCID: PMC11697059 DOI: 10.1186/s13046-024-03250-1] [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: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Targeting the TGF-β pathway in tumor therapy has proven challenging due to the highly context-dependent functions of TGF-β. Integrin αvβ8, a pivotal activator of TGF-β, has been implicated in TGF-β signaling within tumors, as demonstrated by the significant anti-tumor effects of anti-αvβ8 antibodies. Nevertheless, the expression profile of αvβ8 remains a subject of debate, and the precise mechanisms underlying the anti-tumor effects of anti-αvβ8 antibodies are not yet fully elucidated. METHODS We utilized single-cell RNA sequencing to assess αvβ8 expression across various human tumors. An anti-αvβ8 antibody was developed and characterized for its binding and blocking properties in vitro. Cryo-EM single-particle analysis was employed to study the detailed interaction between αvβ8 and the antibody Fab fragment. The anti-tumor efficacy of the antibody was evaluated in syngeneic mouse models with varying levels of αvβ8 expression, both as a monotherapy and in combination with PD-1 antibodies. Human PBMCs were isolated to investigate αvβ8 expression in myeloid cells, and macrophages were exposed to the antibody to study its impact on macrophage polarization. Pharmacokinetic studies of the αvβ8 antibody were conducted in cynomolgus monkeys. RESULTS Integrin αvβ8 is notably expressed in certain tumor types and tumor-infiltrating macrophages. The specific αvβ8 antibody 130H2 demonstrated high affinity, specificity, and blocking potency in vitro. Cryo-EM analysis further revealed that 130H2 interacts exclusively with the β8 subunit, without binding to the αv subunit. In vivo studies showed that this antibody significantly inhibited tumor growth and alleviated immunosuppression by promoting immune cell infiltration. Furthermore, combining the antibody with PD-1 inhibition produced a synergistic anti-tumor effect. In human PBMCs, monocytes exhibited high αvβ8 expression, and the antibody directly modulated macrophage polarization. Tumors with elevated αvβ8 expression were particularly responsive to 130H2 treatment. Additionally, favorable pharmacokinetic properties were observed in cynomolgus monkeys. CONCLUSIONS In summary, integrin αvβ8 is highly expressed in certain tumors and tumor-infiltrating macrophages. Targeting αvβ8 with a blocking antibody significantly inhibits tumor growth by modulating macrophage polarization and enhancing immune cell infiltration. Combining αvβ8 targeting with PD-1 treatment markedly increases the sensitivity of immune-excluded tumors. These results support further clinical evaluation of αvβ8 antibodies.
Collapse
Affiliation(s)
- Cuicui Guo
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China
| | - Hui Sun
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, China
| | - Yulei Du
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China
| | - Xiaodong Dai
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China
| | - Yu Pang
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China
| | - Zhen Han
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China
| | - Xinhui Xiong
- Nanjing Novoacine Biotechnology Co., Ltd, Nanjing, 210032, China
| | - Shaowei Li
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, China
| | - Junhua Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Qingbing Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China.
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, China.
| | - Xun Gui
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China.
| |
Collapse
|
33
|
Zeng Y, Luo CL, Lin GW, Li F, Bai X, Ko JMY, Xiong L, Liu Y, He S, Jiang JX, Yan WX, Ong EHW, Li Z, Zhou YQ, Zhou YH, Xu AY, Liu SQ, Guo YM, Chen JR, Cheng XX, Cao YL, Yu X, Wu B, Wei PP, Ruan ZH, Chen QY, Tang LQ, McKay JD, Jia WH, Mai HQ, Lim ST, Liu JJ, Lin DX, Khor CC, Chua MLK, Ji M, Lung ML, Zeng YX, Bei JX. Whole-exome sequencing association study reveals genetic effects on tumor microenvironment components in nasopharyngeal carcinoma. J Clin Invest 2025; 135:e182768. [PMID: 39744943 PMCID: PMC11684818 DOI: 10.1172/jci182768] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/24/2024] [Indexed: 02/11/2025] Open
Abstract
Nasopharyngeal carcinoma (NPC) presents a substantial clinical challenge due to the limited understanding of its genetic underpinnings. Here we conduct the largest scale whole-exome sequencing association study of NPC to date, encompassing 6,969 NPC cases and 7,100 controls. We unveil 3 germline genetic variants linked to NPC susceptibility: a common rs2276868 in RPL14, a rare rs5361 in SELE, and a common rs1050462 in HLA-B. We also underscore the critical impact of rare genetic variants on NPC heritability and introduce a refined composite polygenic risk score (rcPRS), which outperforms existing models in predicting NPC risk. Importantly, we reveal that the polygenic risk for NPC is mediated by EBV infection status. Utilizing a comprehensive multiomics approach that integrates both bulk-transcriptomic (n = 356) and single-cell RNA sequencing (n = 56) data with experimental validations, we demonstrate that the RPL14 variant modulates the EBV life cycle and NPC pathogenesis. Furthermore, our data indicate that the SELE variant contributes to modifying endothelial cell function, thereby facilitating NPC progression. Collectively, our study provides crucial insights into the intricate genetic architecture of NPC, spotlighting the vital interplay between genetic variations and tumor microenvironment components, including EBV and endothelial cells, in predisposing to NPC. This study opens new avenues for advancements in personalized risk assessments, early diagnosis, and targeted therapies for NPC.
Collapse
Affiliation(s)
- Yanni Zeng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, and
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chun-Ling Luo
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guo-Wang Lin
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Fugui Li
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, China
| | - Xiaomeng Bai
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, and
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Josephine Mun-Yee Ko
- Department of Clinical Oncology, School of Clinical Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Lei Xiong
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, China
| | - Yang Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shuai He
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jia-Xin Jiang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wen-Xin Yan
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Enya Hui Wen Ong
- Precision Radiotherapeutics Oncology Programme, Division of Medical Sciences, National Cancer Centre Singapore, Singapore
| | - Zheng Li
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Ya-Qing Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yun-He Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - An-Yi Xu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shu-Qiang Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yun-Miao Guo
- Zhanjiang Institute of Clinical Medicine, Central People’s Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang, China
| | - Jie-Rong Chen
- Department of Laboratory Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xi-Xi Cheng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yu-Lu Cao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
| | - Xia Yu
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, China
| | - Biaohua Wu
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, China
| | - Pan-Pan Wei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhao-Hui Ruan
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qiu-Yan Chen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
| | - Lin-Quan Tang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
| | - James D. McKay
- Genomic Epidemiology Branch, International Agency for Research on Cancer/World Health Organization (IARC/WHO), Lyon, France
| | - Wei-Hua Jia
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
| | - Hai-Qiang Mai
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
| | - Soon Thye Lim
- Department of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Jian-Jun Liu
- Laboratory of Human Genomics, Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Dong-Xin Lin
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
| | - Chiea Chuen Khor
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore
- Ophthalmology & Visual Sciences Academic Clinical Programme, Duke-National University of Singapore Medical School, Singapore
- Singapore Eye Research Institute, Discovery Tower, Level 6, The Academia, Singapore
| | - Melvin Lee Kiang Chua
- Precision Radiotherapeutics Oncology Programme, Division of Medical Sciences, National Cancer Centre Singapore, Singapore
- Department of Head and Neck and Thoracic Radiation Oncology, National Cancer Centre Singapore, Singapore
- Oncology Academic Clinical Programme, Duke-NUS Medical School, Singapore
| | - Mingfang Ji
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, China
| | - Maria Li Lung
- Department of Clinical Oncology, School of Clinical Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Yi-Xin Zeng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
| | - Jin-Xin Bei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center - Zhongshan School of Medicine
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, National Cancer Centre Singapore, Singapore
- Sun Yat-sen University Institute of Advanced Studies Hong Kong, Science Park, Hong Kong SAR, China
| |
Collapse
|
34
|
Zheng BW, Guo W. Multi-omics analysis unveils the role of inflammatory cancer-associated fibroblasts in chordoma progression. J Pathol 2025; 265:69-83. [PMID: 39611243 DOI: 10.1002/path.6369] [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: 07/17/2024] [Revised: 09/25/2024] [Accepted: 10/13/2024] [Indexed: 11/30/2024]
Abstract
Cancer-associated fibroblasts (CAFs) constitute the primary cellular component of the stroma in chordomas, characterized by an abundance of mucinous stromal elements, potentially facilitating their initiation and progression; however, this inference has yet to be fully confirmed. In this study, single-cell RNA sequencing (scRNA-seq), spatial transcriptomics (ST), bulk RNA-seq, multiplexed quantitative immunofluorescence (QIF), and in vivo and in vitro experiments were performed to determine the heterogeneity, spatial distribution, and clinical significance of CAFs in chordoma. ScRNA-seq was performed on 87,693 single cells derived from seven tumor samples and four control nucleus pulposus samples. A distinct CAF cluster distinguished by the upregulated expression of inflammatory genes and enriched functionality in activating inflammation-associated cells was identified. Pseudotime trajectory and cell communication analyses suggested that this inflammatory CAF (iCAF) subset originated from normal fibroblasts and interacted extensively with tumors and various other cell types. By integrating the scRNA-seq results with ST, the presence of iCAF in chordoma tissue was further confirmed, indicating their positioning at a distance from the tumor cells. Bulk RNA-seq data analysis from 126 patients revealed a correlation between iCAF signature scores, chordoma invasiveness, and poor prognosis. QIF validation involving an additional 116 patients found that although iCAFs were not in close proximity to tumor cells compared with other CAF subsets, their density correlated with malignant tumor phenotypes and adverse outcomes. In vivo and in vitro experiments further confirmed that iCAFs accelerate the malignant progression of chordomas. These findings could provide insights into the development of novel therapeutic strategies. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Bo-Wen Zheng
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Peking University, Beijing, PR China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, PR China
| | - Wei Guo
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Peking University, Beijing, PR China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, PR China
| |
Collapse
|
35
|
Li J, Ping P, Li Y, Xu X. Fatty acid metabolism: A new target for nasopharyngeal carcinoma therapy. Chin J Cancer Res 2024; 36:652-668. [PMID: 39802901 PMCID: PMC11724175 DOI: 10.21147/j.issn.1000-9604.2024.06.05] [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: 08/12/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
Lipid metabolic reprogramming is considered one of the most prominent metabolic abnormalities in cancer, and fatty acid metabolism is a key aspect of lipid metabolism. Recent studies have shown that fatty acid metabolism and its related lipid metabolic pathways play important roles in the malignant progression of nasopharyngeal carcinoma (NPC). NPC cells adapt to harsh environments by enhancing biological processes such as fatty acid metabolism, uptake, production, and oxidation, thereby accelerating their growth. In addition, the reprogramming of fatty acid metabolism plays a central role in the tumor microenvironment (TME) of NPC, and the phenotypic transformation of immune cells is closely related to fatty acid metabolism. Moreover, the reprogramming of fatty acid metabolism in NPC contributes to immune escape, which significantly affects disease treatment, progression, recurrence, and metastasis. This review explores recent advances in fatty acid metabolism in NPC and focuses on the interconnections among metabolic reprogramming, tumor immunity, and corresponding therapies. In conclusion, fatty acid metabolism represents a potential target for NPC treatment, and further exploration is needed to develop strategies that target the interaction between fatty acid metabolic reprogramming and immunotherapy.
Collapse
Affiliation(s)
- Juan Li
- Department of Radiotherapy Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Pengbin Ping
- Department of Radiotherapy Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Yanhua Li
- Department of International Medical, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Xiaoying Xu
- Department of Radiotherapy Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| |
Collapse
|
36
|
Liu Y, Lui KS, Ye Z, Chen L, Cheung AKL. Epstein-Barr Virus BRRF1 Induces Butyrophilin 2A1 in Nasopharyngeal Carcinoma NPC43 Cells via the IL-22/JAK3-STAT3 Pathway. Int J Mol Sci 2024; 25:13452. [PMID: 39769218 PMCID: PMC11677325 DOI: 10.3390/ijms252413452] [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: 11/20/2024] [Revised: 12/08/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Epstein-Barr virus is highly associated with nasopharyngeal carcinoma (NPC) with genes expressed for tumor transformation or maintenance of viral latency, but there are certain genes that can modulate immune molecules. Butyrophilin 2A1 (BTN2A1) is an important activating protein for presenting phosphoantigens for recognition by Vγ9Vδ2 T cells to achieve antitumor activities. We have previously shown that Vγ9Vδ2 T cells achieve efficacy against NPC when BTN2A1 and BTN3A1 are upregulated by stimulating EBV gene expression, particularly LMP1. While BTN3A1 can be induced by the LMP1-mediated IFN-γ/JNK/NLRC5 pathway, the viral gene that can regulate BTN2A1 remains elusive. We showed that BTN2A1 expression is directly mediated by EBV BRRF1, which can trigger the BTN2A1 promoter and downstream JAK3-STAT3 pathway in NPC43 cells, as shown by RNA-seq data and verified via inhibitor experiments. Furthermore, BRRF1 downregulated IL-22 binding protein (IL-22RA2) to complement the EBNA1-targeting probe (P4)-induced IL-22 expression. Therefore, this study elucidated a new mechanism of stimulating BTN2A1 expression in NPC cells via the EBV gene BRRF1. The JAK3-STAT3 pathway could act in concordance with IL-22 to enhance the expression of BTN2A1, which likely leads to increased tumor cell killing by Vγ9Vδ2 T cells for enhanced potential as immunotherapy against the cancer.
Collapse
Affiliation(s)
- Yue Liu
- Medical School, Fuyang Normal University, Fuyang 236000, China;
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China; (K.S.L.); (Z.Y.)
| | - Ka Sin Lui
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China; (K.S.L.); (Z.Y.)
| | - Zuodong Ye
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China; (K.S.L.); (Z.Y.)
| | - Luo Chen
- Department of Chemistry, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China;
| | - Allen Ka Loon Cheung
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China; (K.S.L.); (Z.Y.)
| |
Collapse
|
37
|
Xu L, Li B, Liu Y, Hu Z, Dan Q, Xu B, Xiang H, Chen Y, Zheng T, Sun D, Liu L. Unveiling KLHL23 as a key immune regulator in hepatocellular carcinoma through integrated analysis. Aging (Albany NY) 2024; 16:13608-13626. [PMID: 39636292 PMCID: PMC11723656 DOI: 10.18632/aging.206167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 10/22/2024] [Indexed: 12/07/2024]
Abstract
Age-related cancers are characterized by impaired protein homeostasis, where Kelch protein superfamily members have showed accumulating clues as critical regulators. In this paper, the cancerous role of Kelch-like family member 23 (KLHL23) was comprehensively analyzed with TCGA and single cell GEO database across overall 33 cancer types. By multi-omics analysis upon the transcriptomic, genomic, and methylation data, the current study explored the association of KLHL23 with patient survival, gene ontology, tumor-infiltrating lymphocytes, and drug responses. The correlation of copy number variations and methylation with dysregulated expression of KLHL23 were also addressed. Notably, KLHL23 levels correlated with survival in cancers such as hepatocellular carcinoma and low-grade glioma. The study also highlighted how reduced KLHL23 expression is linked to increased immune activity and sensitivity to chemotherapy, suggesting its potential as a biomarker for cancer prognosis and treatment responsiveness.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Liver Neoplasms/immunology
- DNA Copy Number Variations
- Gene Expression Regulation, Neoplastic
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Prognosis
- DNA Methylation
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Databases, Genetic
Collapse
Affiliation(s)
- Liangliang Xu
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China China
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Bo Li
- Guangdong-Hong Kong-Macau University Joint Laboratory of Digestive Cancer Research, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Yuchen Liu
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China
| | - Zhengming Hu
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China China
| | - Qing Dan
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China China
| | - Bingxuan Xu
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China China
| | - Hongjin Xiang
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China China
| | - Yun Chen
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China China
| | - Tingting Zheng
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China China
| | - Desheng Sun
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China China
| | - Li Liu
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China China
| |
Collapse
|
38
|
Hu X, Huang X, Yin T, Chen J, Zhao W, Yu M, Liu L, Du M. CX3CL1 (Fractalkine): An important cytokine in physiological and pathological pregnancies. J Reprod Immunol 2024; 166:104392. [PMID: 39577056 DOI: 10.1016/j.jri.2024.104392] [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: 09/09/2024] [Revised: 10/25/2024] [Accepted: 11/10/2024] [Indexed: 11/24/2024]
Abstract
C-X3-C motif chemokine ligand 1 (CX3CL1), commonly known as Fractalkine, is an important chemokine with dual functions of chemotaxis and adhesion. It plays a pivotal role in a variety of physiological processes and pathological conditions, particularly in conjunction with its receptor, C-X3-C motif chemokine receptor 1 (CX3CR1). This review focuses on the expression and intricate regulatory mechanisms of CX3CL1 at the maternal-fetal interface, emphasizing its multifaceted role during pregnancy. CX3CL1 was detected in the trophoblast and decidua tissues, playing a crucial role in recruitment of immune cells, enhancing endometrial receptivity, and modulating trophoblast cell activities. Abnormal expression of CX3CL1 has been correlated with adverse pregnancy outcomes such as spontaneous abortion, gestational diabetes, preeclampsia, and preterm births. By elucidating the complex interplay of CX3CL1 at the maternal-fetal interface, this review aims to shed light on its potential roles in pregnancy-related complications.
Collapse
Affiliation(s)
- Xianyang Hu
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China
| | - Xixi Huang
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China
| | - Tingxuan Yin
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China
| | - Jiajia Chen
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China
| | - Weijie Zhao
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China
| | - Min Yu
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China.
| | - Lu Liu
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China.
| | - Meirong Du
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China; Department of Obstetrics and Gynecology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
39
|
Yang J, Song X, Zhang H, Liu Q, Wei R, Guo L, Yuan C, Chen F, Xue K, Lai Y, Wang L, Shi J, Zhou C, Wang J, Yu Y, Mei Q, Hu L, Wang H, Zhang C, Zhang Q, Li H, Gu Y, Zhao W, Yu H, Wang J, Liu Z, Li H, Zheng S, Liu J, Yang L, Li W, Xu R, Chen J, Zhou Y, Cheng X, Yu Y, Wang D, Sun X, Yu H. Single-cell transcriptomic landscape deciphers olfactory neuroblastoma subtypes and intra-tumoral heterogeneity. NATURE CANCER 2024; 5:1919-1939. [PMID: 39543363 DOI: 10.1038/s43018-024-00855-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 10/07/2024] [Indexed: 11/17/2024]
Abstract
Olfactory neuroblastoma (ONB) is a rare malignancy known to originate from the olfactory epithelium. The complex tumor ecosystem of this pathology remains unclear. Here, we explored the cellular components within ten ONB tumors and one olfactory mucosa sample based on single-cell RNA profiles. We showed the intra-tumoral heterogeneity by identifying five unique expression programs among malignant epithelial cells. A distinct three-classification system (neural, basal, mesenchymal) for ONB was established according to the distinguished gene expression patterns. Biomarkers for categorizing bulk tumors into uncharacterized subtypes were elucidated. Different responses towards certain chemotherapy regimens could be cautiously inferred according to the molecular features representing the three tumor types, thus helping with precision chemotherapy. We also analyzed subclusters of the tumor microenvironment (TME) and the interactions among different cell types within the TME. The relative abundance of immunosuppressive tumor-associated macrophages suggests potential benefits of immunotherapies targeting macrophages.
Collapse
Affiliation(s)
- Jingyi Yang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
- Olfactory Neuroblastoma Diagnosis and Treatment Center, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Xiaole Song
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
- Mucosal Melanoma Diagnosis and Treatment Center, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Huankang Zhang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Quan Liu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Ruoyan Wei
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Luo Guo
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, PR China
| | - Cuncun Yuan
- Department of Pathology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Fu Chen
- Department of Radiation Oncology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Kai Xue
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Yuting Lai
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Li Wang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Junfeng Shi
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Chengle Zhou
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Juan Wang
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Pharmaceutical Industry Research Institute, Shanghai, PR China
| | - Yingxuan Yu
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Pharmaceutical Industry Research Institute, Shanghai, PR China
| | - Qibing Mei
- State Key Laboratory of New Drug and Pharmaceutical Process, Shanghai Pharmaceutical Industry Research Institute, Shanghai, PR China
| | - Li Hu
- Department of Experimental Center, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Huan Wang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Chen Zhang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Qianqian Zhang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Houyong Li
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Ye Gu
- Department of Neurosurgery and Otolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Weidong Zhao
- Department of Neurosurgery and Otolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Huapeng Yu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Jingjing Wang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Zhuofu Liu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Han Li
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Shixing Zheng
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Juan Liu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Lu Yang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Wanpeng Li
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Rui Xu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Jiani Chen
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Yumin Zhou
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Xiankui Cheng
- Department of Pathology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, PR China
| | - Yiqun Yu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
- Olfactory Disorder Diagnosis and Treatment Center, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Dehui Wang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Xicai Sun
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China
- Olfactory Neuroblastoma Diagnosis and Treatment Center, Eye and ENT Hospital, Fudan University, Shanghai, PR China
| | - Hongmeng Yu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, PR China.
- Research Unit of New Technologies of Endoscopic Surgery in Skull Base Tumor (2018RU003), Chinese Academy of Medical Sciences, Shanghai, PR China.
| |
Collapse
|
40
|
Chen HX, Ma YZ, Xie PP, Huang JY, Li LQ, Zhang W, Zhu Y, Zhuang SM, Lin YF. Micropeptide MPM regulates cardiomyocyte proliferation and heart growth via the AKT pathway. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119820. [PMID: 39163918 DOI: 10.1016/j.bbamcr.2024.119820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/26/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024]
Abstract
The role of micropeptide in cardiomyocyte proliferation remains unknown. We found that MPM (micropeptide in mitochondria) was highly expressed in cardiomyocytes. Compared to MPM+/+ mice, MPM knockout (MPM-/-) mice exhibited reduction in left ventricular (LV) mass, myocardial thickness and LV fractional shortening. RNA-sequencing analysis in H9c2, a rat cardiomyocyte cell line, identified downregulation of cell cycle-promoting genes as the most significant alteration in MPM-silencing cells. Consistently, gain- and loss-of-function analyses in H9c2 cells revealed that cardiomyocyte proliferation was repressed by silencing MPM but was promoted by overexpressing MPM. Moreover, the cardiomyocytes in the hearts of MPM-/- mice displayed reduced proliferation rates. Mechanism investigations disclosed that MPM is crucial for AKT activation in cardiomyocytes. We also identified an interaction between MPM and PTPMT1, and found that silencing PTPMT1 attenuated the effect of MPM in activating the AKT pathway, whereas inhibition of the AKT pathway abrogated the role of MPM in promoting cardiomyocyte proliferation. Collectively, these results indicate that MPM may promote cardiomyocyte proliferation and thus heart growth by interacting with PTPMT1 to activate the AKT pathway. Our findings identify the novel function and regulatory network of MPM and highlight the importance of micropeptides in cardiomyocyte proliferation and heart growth.
Collapse
Affiliation(s)
- Hua-Xing Chen
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Yan-Zhen Ma
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Peng-Peng Xie
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Jie-Yi Huang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Lan-Qi Li
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Wei Zhang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Ying Zhu
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Shi-Mei Zhuang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China; Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, PR China.
| | - Yi-Fang Lin
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, State Key Laboratory of Oncology in Southern China, Sun Yat-sen University, Guangzhou 510275, PR China; Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, PR China.
| |
Collapse
|
41
|
Ma J, Wu Y, Wu S, Fang Z, Chen L, Jiang J, Zheng X. CX3CR1 +CD8 + T cells: Key players in antitumor immunity. Cancer Sci 2024; 115:3838-3845. [PMID: 39377122 PMCID: PMC11611776 DOI: 10.1111/cas.16359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/18/2024] [Accepted: 09/17/2024] [Indexed: 10/09/2024] Open
Abstract
CX3CR1 functions as the specific receptor for the chemokine CX3CL1, demonstrating expression across a broad spectrum of immune cells. This underscores its pivotal role in communication and response mechanisms within the immune system. Upon engagement with CX3CL1, CX3CR1 initiates a cascade of downstream signaling pathways that regulate various biological functions. In the context of tumor progression, the intricate and inhibitory nature of the tumor microenvironment presents a significant challenge to current clinical treatment techniques. This review aims to comprehensively explore the tumor-destructive potential shown by CX3CR1+CD8+ T cells. Simultaneously, it investigates the promising prospects of utilizing CX3CR1 in future tumor immunotherapies.
Collapse
Affiliation(s)
- Jiajin Ma
- Department of Tumor Biological TreatmentThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
- Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouChina
- Institute for Cell Therapy of Soochow UniversityChangzhouChina
| | - Yue Wu
- Department of Tumor Biological TreatmentThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
- Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouChina
- Institute for Cell Therapy of Soochow UniversityChangzhouChina
| | - Shaoxian Wu
- Department of Tumor Biological TreatmentThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
- Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouChina
- Institute for Cell Therapy of Soochow UniversityChangzhouChina
| | - Zhang Fang
- Department of Tumor Biological TreatmentThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
- Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouChina
- Institute for Cell Therapy of Soochow UniversityChangzhouChina
| | - Lujun Chen
- Department of Tumor Biological TreatmentThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
- Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouChina
- Institute for Cell Therapy of Soochow UniversityChangzhouChina
| | - Jingting Jiang
- Department of Tumor Biological TreatmentThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
- Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouChina
- Institute for Cell Therapy of Soochow UniversityChangzhouChina
| | - Xiao Zheng
- Department of Tumor Biological TreatmentThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
- Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouChina
- Institute for Cell Therapy of Soochow UniversityChangzhouChina
| |
Collapse
|
42
|
Liang Z, Luo C, Li S, Zhu Y, Huang W, Cao D, Liu Y, Ruan G, Liang S, Chen X, Kou KI, Zhang G, Liu L, Li H. Guiding induction chemotherapy of locoregionally advanced nasopharyngeal carcinoma with ternary classification of predicted individual treatment effect. Radiother Oncol 2024; 201:110571. [PMID: 39393470 DOI: 10.1016/j.radonc.2024.110571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 09/26/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND AND PURPOSE Induction chemotherapy (IC) before concurrent chemoradiotherapy does not universally improve long-term overall survival (OS) in locoregionally advanced nasopharyngeal carcinoma (LANPC). Conventional risk stratification often yields suboptimal IC decisions. Our study introduces a ternary classification of predicted individual treatment effect (PITE) to guide personalized IC decisions. MATERIALS AND METHODS A two-center retrospective analysis of 1,213 patients with LANPC was conducted to develop and validate prognostic models integrating magnetic resonance imaging and clinical data to estimate individual 5-year OS probabilities for IC and non-IC treatments. Differences in these probabilities defined PITE, facilitating patient stratification into three IC recommendation categories. Model effectiveness was validated using Kaplan-Meier estimators, decision curve-like analysis, and evaluations of variable importance and distribution. RESULTS The models exhibited strong predictive performance in both treatments across training and cross-validation sets, enabling accurate PITE calculations and patient classification. Compared with non-IC treatment, IC markedly improved OS in the IC-preferred group (HR = 0.62, p = 0.02), had no effect in the IC-neutral group (HR = 1.00, p = 0.70), and worsened OS in the IC-opposed group (HR = 2.00, p = 0.03). The ternary PITE classification effectively identified 41.7 % of high-risk patients not benefiting from IC, and yielded a 2.68 % higher mean 5-year OS probability over risk-based decisions. Significantly increasing distributions of key prognostic indicators, such as metastatic lymph node number and plasma Epstein-Barr virus DNA level from IC-opposed to IC-preferred groups, further validated the clinical relevance of PITE classification. CONCLUSION The ternary PITE classification offers an accurate and clinically advantageous approach to guide personalized IC decision-making in patients with LANPC.
Collapse
Affiliation(s)
- Zhiying Liang
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chao Luo
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shuqi Li
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yuliang Zhu
- Nasopharyngeal Head and Neck Tumor Radiotherapy Department, Zhongshan City People's Hospital, Zhongshan 528400, China
| | - Wenjie Huang
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Di Cao
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yifei Liu
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Guangying Ruan
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shaobo Liang
- Department of Radiation Oncology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xi Chen
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou 510120, China
| | - Kit-Ian Kou
- Department of Mathematics, Faculty of Science and Technology, University of Macau, Macao Special Administrative Region of China
| | - Guoyi Zhang
- Department of Radiation Oncology, First People's Hospital of Foshan, Foshan 528000, China
| | - Lizhi Liu
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Haojiang Li
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
43
|
Ge J, Liu Y, Chen P, Zeng Z, Li G, Xiong W, Yi M, Xiang B. FOXA1 enhances antitumor immunity via repressing interferon-induced PD-L1 expression in nasopharyngeal carcinoma. J Immunother Cancer 2024; 12:e010091. [PMID: 39542656 PMCID: PMC11575282 DOI: 10.1136/jitc-2024-010091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a distinct subtype of head and neck cancer which is prevalent in south of China and southeastern of Asia. Consistent activation of interferon (IFN) signaling, and impairment of T cell mediated antitumor immunity is frequent in NPC. Forkhead box A1 (FOXA1) is one of the earliest discovered pioneer factors, which can open up compact chromatin structures to facilitate the binding of other proteins to chromatin. METHODS By using RNA sequencing, it was discovered that FOXA1 suppresses the activation of the interferon signaling pathway and the expression of the related interferon-responsive genes in NPC cells. The effect of FOXA1 on programmed death-ligand 1 (PD-L1) expression in C666-1 and HK1 cells under conditions with or without IFN-γ was detected through quantitative PCR (qPCR), western blot, and flow cytometry. After co-culturing T cells with IFN-γ-treated NPC cells in vitro, apoptosis of CD8+ T cells and the expression of cytotoxic cytokines were assessed by flow cytometry. The cytotoxic effects of T cells on tumor cells in nude mice were measured by tumorigenesis in nude mice and adoptive T cell therapy. The effects of IFN-γ on the expression and nuclear localization of STAT1, as well as the colocalization of FOXA1 with STAT1 were detected by immunofluorescence, qPCR, western blot, and co-immunoprecipitation experiments. RESULTS In this study, we reported that loss of FOXA1, a pioneer factor downregulated in NPC, results in activation of IFN signaling in NPC cells. Repression of FOXA1 facilitates IFN-γ induced PD-L1 expression, whereas overexpression of FOXA1 exerts the opposite effect. Mechanistically, FOXA1 interacts with STAT1 and inhibits IRF1 expression and binding to PD-L1 promoter on IFN-γ treatment. Co-culture with FOXA1-silenced NPC cells promotes apoptosis of in vitro activated tumor-specific CD8+T cells and reduces the expression of cytotoxic effector molecules. Furthermore, overexpression of FOXA1 increases the therapeutic efficacy of PD-L1 antibody (atezolizumab) against NPC in nude mice receiving adoptive T-cell therapy. CONCLUSIONS We demonstrated that FOXA1 prevents tumor immune evasion by inhibiting IFN-γ induced PD-L1 expression in NPC cells. Our research findings provide new insights into the immunotherapeutic biomarkers and targets for NPC, which is important for the clinical application of programmed cell death protein-1/PD-L1 antibodies in NPC.
Collapse
Affiliation(s)
- Junshang Ge
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Changsha, Hunan, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, Hunan, China
| | - Ying Liu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Changsha, Hunan, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, Hunan, China
| | - Pan Chen
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Changsha, Hunan, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Changsha, Hunan, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Changsha, Hunan, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, Hunan, China
| | - Mei Yi
- Department of Dermotology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Bo Xiang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Changsha, Hunan, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, Hunan, China
| |
Collapse
|
44
|
Pan Y, Fei L, Wang S, Chen H, Jiang C, Li H, Wang C, Yang Y, Zhang Q, Chen Y. Integrated analysis of single-cell, spatial and bulk RNA-sequencing identifies a cell-death signature for predicting the outcomes of head and neck cancer. Front Immunol 2024; 15:1487966. [PMID: 39575251 PMCID: PMC11578999 DOI: 10.3389/fimmu.2024.1487966] [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: 08/29/2024] [Accepted: 10/16/2024] [Indexed: 11/24/2024] Open
Abstract
Background Cell death plays an essential role in carcinogenesis, but its function in the recurrence and postoperative prognosis of head and neck cancer (HNC), which ranks as the 7th most common malignancy globally, remains unclear. Methods Data from five main subtypes of HNC related single-cell RNA sequencing (scRNA-seq) were recruited to establish a single-cell atlas, and the distribution of cell death models (CDMs) across different tissues as well as cell subtypes were analyzed. Bulk RNA-seq from the Cancer Genome Atlas Program (TCGA) dataset was subjected to a machine learning-based integrative procedure for constructing a consensus cell death-related signature risk score (CDRscore) model and validated by external data. The biofunctions including different expression analysis, immune cell infiltration, genomic mutations, enrichment analysis as well as cellchat analysis were compared between the high- and low- risk score groups categorized by this CDRscore model. Finally, samples from laryngeal squamous cell cancer (LSCC) were conducted by spatial transcriptomics (ST) to further validate the results of CDRscore model. Results T cells from HNC patients manifested the highest levels of cell death while HPV infection attenuates malignant cell death based on single-cell atlas. CDMs are positively correlated with the tumor-cell stemness, immune-related score and T cells are infiltrated. A CDRscore model was established based on the transcription of ten cell death prognostic genes (MRPL10, DDX19A, NDFIP1, PCMT1, HPRT1, SLC2A3, EFNB2, HK1, BTG3 and MAP2K7). It functions as an independent prognostic factor for overall survival in HNC and displays stable and powerful performance validated by GSE41613 and GSE65858 datasets. Patients in high CDRscore manifested worse overall survival, more active of epithelial mesenchymal transition, TGF-β-related pathways and hypoxia, higher transcription of T cell exhausted markers, and stronger TP53 mutation. ST from LSCC showed that spots with high-risk scores were colocalized with TGF-β and the proliferating malignant cells, additionally, the risk scores have a negative correlation with TCR signaling but positive association with LAG3 transcription. Conclusion The CDRscore model could be utilized as a powerful prognostic indicator for HNC.
Collapse
Affiliation(s)
- Yue Pan
- Institute of Immunology, People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
| | - Lei Fei
- Institute of Immunology, People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
| | - Shihua Wang
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Hua Chen
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Changqing Jiang
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Hong Li
- Chongqing Renpin Otolaryngology Head and Neck Surgery Hospital, Chongqing, China
| | - Changsong Wang
- Department of Pathology, People’s Liberation Army Joint Logistic Support Force 989 Hospital, Luoyang, Henan, China
| | - Yao Yang
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Qinggao Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China
| | - Yongwen Chen
- Institute of Immunology, People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
| |
Collapse
|
45
|
Yu Y, Li Y, Zhou L, Cheng X, Gong Z. Hepatic stellate cells promote hepatocellular carcinoma development by regulating histone lactylation: Novel insights from single-cell RNA sequencing and spatial transcriptomics analyses. Cancer Lett 2024; 604:217243. [PMID: 39260669 DOI: 10.1016/j.canlet.2024.217243] [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: 04/25/2024] [Revised: 08/23/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024]
Abstract
This study evaluated the cellular heterogeneity and molecular mechanisms of hepatocellular carcinoma (HCC). Single cell RNA sequencing (scRNA-seq), transcriptomic data, histone lactylation-related genes were collected from public databases. Cell-cell interaction, trajectory, pathway, and spatial transcriptome analyses were executed. Differential expression and survival analyses were conducted. Western blot, Real-time reverse transcription PCR (qRT-PCR), and Cell Counting Kit 8 (CCK8) assay were used to detect the expression of αSMA, AKR1B10 and its target genes, and verify the roles of AKR1B10 in HCC cells. Hepatic stellate cell (HSC) subgroups strongly interacted with tumor cell subgroups, and their spatial distribution was heterogeneous. Two candidate prognostic genes (AKR1B10 and RMRP) were obtained. LONP1, NPIPB3, and ZSWIM6 were determined as AKR1B10 targets. Besides, the expression levels of AKR1B10 and αSMA were significantly increased in LX-2 + HepG2 and LX-2 + HuH7 groups compared to those in LX-2 group, respectively. sh-AKR1B10 significantly inhibited the HCC cell proliferation and change the expression of AKR1B10 target genes, Bcl-2, Bax, Pan Kla, and H3K18la at protein levels. Our findings unveil the pivotal role of HSCs in HCC pathogenesis through regulating histone lactylation.
Collapse
Affiliation(s)
- Yifan Yu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China.
| | - Yongnan Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China.
| | - Long Zhou
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China.
| | - Xiaoli Cheng
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China.
| | - Zheng Gong
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China.
| |
Collapse
|
46
|
Dong L, Hu S, Li X, Pei S, Jin L, Zhang L, Chen X, Min A, Yin M. SPP1 + TAM Regulates the Metastatic Colonization of CXCR4 + Metastasis-Associated Tumor Cells by Remodeling the Lymph Node Microenvironment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400524. [PMID: 39236316 PMCID: PMC11600252 DOI: 10.1002/advs.202400524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 05/06/2024] [Indexed: 09/07/2024]
Abstract
Lymph node metastasis, the initial step in distant metastasis, represents a primary contributor to mortality in patients diagnosed with oral squamous cell carcinoma (OSCC). However, the underlying mechanisms of lymph node metastasis in OSCC remain incompletely understood. Here, the transcriptomes of 56 383 single cells derived from paired tissues of six OSCC patients are analyzed. This study founds that CXCR4+ epithelial cells, identified as highly malignant disseminated tumor cells (DTCs), exhibited a propensity for lymph node metastasis. Importantly, a distinct subset of tumor-associated macrophages (TAMs) characterized by exclusive expression of phosphoprotein 1 (SPP1) is discovered. These TAMs may remodel the metastatic lymph node microenvironment by potentially activating fibroblasts and promoting T cell exhaustion through SPP1-CD44 and CD155-CD226 ligand-receptor interactions, thereby facilitating colonization and proliferation of disseminated tumor cells. The research advanced the mechanistic understanding of metastatic tumor microenvironment (TME) and provided a foundation for the development of personalized treatments for OSCC patients with metastasis.
Collapse
Affiliation(s)
- Liang Dong
- Department of DermatologyHunan Engineering Research Center of Skin Health and DiseaseHunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalCentral South UniversityChangshaHunan410008China
- Clinical Research Center (CRC)Medical Pathology Center (MPC)Cancer Early Detection and Treatment Center (CEDTC)Chongqing University Three Gorges HospitalChongqing UniversityChongqing404100China
- Translational Medicine Research Center (TMRC)School of Medicine Chongqing UniversityChongqing404100China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Shujun Hu
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Department of Oral and Maxillofacial SurgeryCenter of StomatologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- Research Center of Oral and Maxillofacail TumorXiangya HospitalCentral South UniversityChangshaHunan410008China
- Insititute of Oral Cancer and Precancerous LesionsCentral South UniversityChangshaHunan410008China
| | - Xin Li
- Clinical Research Center (CRC)Medical Pathology Center (MPC)Cancer Early Detection and Treatment Center (CEDTC)Chongqing University Three Gorges HospitalChongqing UniversityChongqing404100China
- Translational Medicine Research Center (TMRC)School of Medicine Chongqing UniversityChongqing404100China
| | - Shiyao Pei
- Department of DermatologyHunan Engineering Research Center of Skin Health and DiseaseHunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Department of DermatologyThird Xiangya HospitalCentral South UniversityChangsha410008China
| | - Liping Jin
- Department of DermatologyHunan Engineering Research Center of Skin Health and DiseaseHunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Lining Zhang
- Clinical Research Center (CRC)Medical Pathology Center (MPC)Cancer Early Detection and Treatment Center (CEDTC)Chongqing University Three Gorges HospitalChongqing UniversityChongqing404100China
- Translational Medicine Research Center (TMRC)School of Medicine Chongqing UniversityChongqing404100China
| | - Xiang Chen
- Department of DermatologyHunan Engineering Research Center of Skin Health and DiseaseHunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Anjie Min
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Department of Oral and Maxillofacial SurgeryCenter of StomatologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- Research Center of Oral and Maxillofacail TumorXiangya HospitalCentral South UniversityChangshaHunan410008China
- Insititute of Oral Cancer and Precancerous LesionsCentral South UniversityChangshaHunan410008China
| | - Mingzhu Yin
- Department of DermatologyHunan Engineering Research Center of Skin Health and DiseaseHunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalCentral South UniversityChangshaHunan410008China
- Clinical Research Center (CRC)Medical Pathology Center (MPC)Cancer Early Detection and Treatment Center (CEDTC)Chongqing University Three Gorges HospitalChongqing UniversityChongqing404100China
- Translational Medicine Research Center (TMRC)School of Medicine Chongqing UniversityChongqing404100China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| |
Collapse
|
47
|
Huang SW, Jiang W, Xu S, Zhang Y, Du J, Wang YQ, Yang KY, Zhang N, Liu F, Zou GR, Jin F, Wu HJ, Zhou YY, Zhu XD, Chen NY, Xu C, Qiao H, Liu N, Sun Y, Ma J, Liang YL, Liu X. Systemic longitudinal immune profiling identifies proliferating Treg cells as predictors of immunotherapy benefit: biomarker analysis from the phase 3 CONTINUUM and DIPPER trials. Signal Transduct Target Ther 2024; 9:285. [PMID: 39438442 PMCID: PMC11496634 DOI: 10.1038/s41392-024-01988-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 09/09/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024] Open
Abstract
The identification of predictors for immunotherapy is often hampered by the absence of control groups in many studies, making it difficult to distinguish between prognostic and predictive biomarkers. This study presents biomarker analyses from the phase 3 CONTINUUM trial (NCT03700476), the first to show that adding anti-PD-1 (aPD1) to chemoradiotherapy (CRT) improves event-free survival (EFS) in patients with locoregionally advanced nasopharyngeal carcinoma. A dynamic single-cell atlas was profiled using mass cytometry on peripheral blood mononuclear cell samples from 12 pairs of matched relapsing and non-relapsing patients in the aPD1-CRT arm. Using a supervised representation learning algorithm, we identified a Ki67+ proliferating regulatory T cells (Tregs) population expressing high levels of activated and immunosuppressive molecules including FOXP3, CD38, HLA-DR, CD39, and PD-1, whose abundance correlated with treatment outcome. The frequency of these Ki67+ Tregs was significantly higher at baseline and increased during treatment in patients who relapsed compared to non-relapsers. Further validation through flow cytometry (n = 120) confirmed the predictive value of this Treg subset. Multiplex immunohistochemistry (n = 249) demonstrated that Ki67+ Tregs in tumors could predict immunotherapy benefit, with aPD1 improving EFS only in patients with low baseline levels of Ki67+ Tregs. These findings were further validated in the multicenter phase 3 DIPPER trial (n = 262, NCT03427827) and the phase 3 OAK trial of anti-PD-L1 immunotherapy in NSCLC, underscoring the predictive value of Ki67+ Treg frequency in identifying the beneficiaries of immunotherapy and potentially guiding personalized treatment strategies.
Collapse
Affiliation(s)
- Sai-Wei Huang
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Wei Jiang
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Sha Xu
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Yuan Zhang
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Juan Du
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Ya-Qin Wang
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Kun-Yu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Ning Zhang
- Department of Radiation Oncology, The First People's Hospital of Foshan, Foshan, PR China
| | - Fang Liu
- Department of Pathology, The First People's Hospital of Foshan, Foshan, PR China
| | - Guo-Rong Zou
- Department of Oncology, Panyu Central Hospital, Guangzhou, PR China
| | - Feng Jin
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, PR China
| | - Hai-Jun Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, PR China
| | - Yang-Ying Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, PR China
| | - Xiao-Dong Zhu
- Department of Radiation Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, PR China
| | - Nian-Yong Chen
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Cheng Xu
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Han Qiao
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Na Liu
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Ying Sun
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Jun Ma
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
| | - Ye-Lin Liang
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
| | - Xu Liu
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
| |
Collapse
|
48
|
Li B, Zhang L, Yin Y, Chen A, Seo BR, Lou J, Mooney DJ, Weitz DA. Stiff Hydrogel Encapsulation Retains Mesenchymal Stem Cell Stemness for Regenerative Medicine. MATTER 2024; 7:3447-3468. [PMID: 39553898 PMCID: PMC11567665 DOI: 10.1016/j.matt.2024.05.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Mesenchymal stem cell (MSC) stands as a prominent choice in regenerative medicine, yet their therapeutic potential remains controversial due to challenges in maintaining lineage and viability. As directly injected MSCs are quickly cleared by the host immune system, entrapping viable cells in a 3D semi-permeable hydrogel matrix extends cell retention, showing great promise in enhancing therapeutic effect. However, the effects of hydrogel encapsulation on MSC subpopulations are not fully understood. Here, we fabricate thin-shell alginate hydrogel microcapsules using droplet microfluidics, controlling the shell mechanical properties by adjusting alginate molecular weight. We find that a stiffer shell increases the proliferation and supports the residence of MSCs in vivo than a softer shell. The stiff 3D hydrogel also promotes the maintenance of stemness, as confirmed by single-cell RNA sequencing. Our work demonstrates the potential of hydrogel-encapsulated stem cells for long-term therapeutic applications, offering insight into modulating MSC subpopulations for specific function.
Collapse
Affiliation(s)
- Bo Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, CN, 310003
| | - Liyuan Zhang
- School of Petroleum Engineering, China University of Petroleum (East China), Qingdao, Shandong, CN, 266580
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138
| | - Yuan Yin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, CN, 310003
| | - Anqi Chen
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138
| | - Bo Ri Seo
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138
- Takeda Pharmaceutical Company Limited, Los Angeles, CA, 90039
| | - Junzhe Lou
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02138
| | - David J. Mooney
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02138
| | - David A. Weitz
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138
- Department of Physics, Harvard University, Cambridge, MA, 02138
| |
Collapse
|
49
|
Xu D, Zhang N, Shen Y, Zheng D, Xu Z, Li P, Cai J, Tian G, Wei Q, Wang H, Jiang H, Cao M, Wang B, Li K. Single-cell sequencing analysis reveals the dynamic tumour ecosystems of primary and metastatic lymph nodes in nasopharyngeal carcinoma. J Cell Mol Med 2024; 28:e70137. [PMID: 39392128 PMCID: PMC11467730 DOI: 10.1111/jcmm.70137] [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: 07/04/2024] [Revised: 09/19/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024] Open
Abstract
Lymph node metastasis contributed to the leading cause and treatment failure in nasopharyngeal carcinoma (NPC). The microenvironment and the cellular communications of lymph node metastasized tumours determine the tumour progression and therapeutic effect, but the ecosystems about the lymph node metastasis (LNM) for NPC patients remain poorly characterized. Here, we integrated the transcriptomes of 47,618 single cells from eight samples related to NPC LNM. The dynamic immune ecosystems and immunosuppressive microenvironment including T cells, myeloid cells and B cells were observed in the lymph node metastatic samples compared with primary tumours. Additionally, the heterogeneity of epithelial cells was also revealed, and several clusters with expression programs that were associated with the progression-free survival of NPC patients were identified. Additionally, our data revealed the complex intercellular communications from primary to lymph node metastasis. The rewiring of CCL signalling which plays an important role in tumour metastasis was further identified. Altogether, we systematically characterized the ecosystem of NPC primary and lymph node metastasized tumours, which may shed light on the development of a therapeutic strategy to improve clinical outcomes of NPC patients with lymph node metastasis.
Collapse
Affiliation(s)
- Dahua Xu
- College of Biomedical Information and EngineeringHainan General Hospital and Hainan Affiliated Hospital, Hainan Medical UniversityHaikouChina
- Hainan Engineering Research Center for Health Big DataHainan Medical UniversityHaikouChina
| | - Nihui Zhang
- College of Biomedical Information and EngineeringHainan General Hospital and Hainan Affiliated Hospital, Hainan Medical UniversityHaikouChina
- Hainan Engineering Research Center for Health Big DataHainan Medical UniversityHaikouChina
| | - Yutong Shen
- College of Biomedical Information and EngineeringHainan General Hospital and Hainan Affiliated Hospital, Hainan Medical UniversityHaikouChina
- Hainan Engineering Research Center for Health Big DataHainan Medical UniversityHaikouChina
| | - Dehua Zheng
- College of Biomedical Information and EngineeringHainan General Hospital and Hainan Affiliated Hospital, Hainan Medical UniversityHaikouChina
- Hainan Engineering Research Center for Health Big DataHainan Medical UniversityHaikouChina
| | - Zhizhou Xu
- College of Biomedical Information and EngineeringHainan General Hospital and Hainan Affiliated Hospital, Hainan Medical UniversityHaikouChina
- Hainan Engineering Research Center for Health Big DataHainan Medical UniversityHaikouChina
| | - Peihu Li
- College of Biomedical Information and EngineeringHainan General Hospital and Hainan Affiliated Hospital, Hainan Medical UniversityHaikouChina
- Hainan Engineering Research Center for Health Big DataHainan Medical UniversityHaikouChina
| | - Jiale Cai
- College of Biomedical Information and EngineeringHainan General Hospital and Hainan Affiliated Hospital, Hainan Medical UniversityHaikouChina
- Hainan Engineering Research Center for Health Big DataHainan Medical UniversityHaikouChina
| | - Guanghui Tian
- College of Biomedical Information and EngineeringHainan General Hospital and Hainan Affiliated Hospital, Hainan Medical UniversityHaikouChina
- Hainan Engineering Research Center for Health Big DataHainan Medical UniversityHaikouChina
| | - Qingchen Wei
- College of Biomedical Information and EngineeringHainan General Hospital and Hainan Affiliated Hospital, Hainan Medical UniversityHaikouChina
| | - Hong Wang
- College of Biomedical Information and EngineeringHainan General Hospital and Hainan Affiliated Hospital, Hainan Medical UniversityHaikouChina
- Hainan Engineering Research Center for Health Big DataHainan Medical UniversityHaikouChina
| | - Hongyan Jiang
- College of Biomedical Information and EngineeringHainan General Hospital and Hainan Affiliated Hospital, Hainan Medical UniversityHaikouChina
| | - Meng Cao
- College of Biomedical Information and EngineeringHainan General Hospital and Hainan Affiliated Hospital, Hainan Medical UniversityHaikouChina
- Hainan Engineering Research Center for Health Big DataHainan Medical UniversityHaikouChina
| | - Bo Wang
- College of Biomedical Information and EngineeringHainan General Hospital and Hainan Affiliated Hospital, Hainan Medical UniversityHaikouChina
| | - Kongning Li
- College of Biomedical Information and EngineeringHainan General Hospital and Hainan Affiliated Hospital, Hainan Medical UniversityHaikouChina
- Hainan Engineering Research Center for Health Big DataHainan Medical UniversityHaikouChina
| |
Collapse
|
50
|
Conde-Lopez C, Marripati D, Elkabets M, Hess J, Kurth I. Unravelling the Complexity of HNSCC Using Single-Cell Transcriptomics. Cancers (Basel) 2024; 16:3265. [PMID: 39409886 PMCID: PMC11475296 DOI: 10.3390/cancers16193265] [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: 07/17/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND/OBJECTIVES Head and neck squamous cell carcinoma (HNSCC) is a highly heterogeneous and the most common form of head and neck cancer, posing significant challenges for disease management. The objective of this review is to assess the utility of single-cell RNA sequencing (scRNAseq) in addressing these challenges by enabling a detailed characterization of the tumor microenvironment (TME) at the cellular level. METHODS This review compiles and analyzes current strategies that utilize scRNAseq and other single-cell technologies in HNSCC research. RESULTS For HNSCC etiology, scRNAseq allows for the construction of cellular atlases, characterization of different cell types, and investigation of genes and processes involved in cancer initiation, development, and progression within the TME. In terms of HNSCC diagnosis and prognosis, the resolution offered by scRNAseq enables the identification of cell type-specific signatures, enhancing prognostic models and disease stratifiers for patient outcome assessments. Regarding HNSCC treatment, scRNAseq provides insights into cellular responses to various treatments, including radiotherapy, chemotherapy, and immunotherapy, contributing to a better understanding of treatment efficacy and patient outcomes. CONCLUSIONS This review highlights the contributions of scRNAseq to HNSCC research, addressing its cellular and biological complexity, and emphasizes its potential for advancing research and clinical practice in other cancer types.
Collapse
Affiliation(s)
- Cristina Conde-Lopez
- Division Radiooncology/Radiobiology, German Cancer Research Center (DKFZ) Heidelberg, 69120 Heidelberg, Germany; (J.H.); (I.K.)
| | - Divyasree Marripati
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (D.M.); (M.E.)
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (D.M.); (M.E.)
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Jochen Hess
- Division Radiooncology/Radiobiology, German Cancer Research Center (DKFZ) Heidelberg, 69120 Heidelberg, Germany; (J.H.); (I.K.)
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Ina Kurth
- Division Radiooncology/Radiobiology, German Cancer Research Center (DKFZ) Heidelberg, 69120 Heidelberg, Germany; (J.H.); (I.K.)
| |
Collapse
|