1
|
Lin R, Jiang Z, Lin J, Tong F, Wu Y, Hong J, Qiu S, Wu Q. Nasopharyngeal cancer screening and immunotherapy efficacy evaluation based on plasma separation combined with label-free SERS technology. Anal Chim Acta 2025; 1358:344070. [PMID: 40374239 DOI: 10.1016/j.aca.2025.344070] [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: 12/17/2024] [Revised: 03/17/2025] [Accepted: 04/12/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND In recent years, significant progress has been made in the treatment of nasopharyngeal carcinoma (NPC). The application of immunotherapy, especially the use of Programmed Cell Death Protein 1 inhibitors, has demonstrated excellent therapeutic effects. However, inconsistent immunotherapy outcomes due to individual differences in patients remain a major challenge, making the search for specific biomarkers to screen for the population who may benefit from immunotherapy a priority. Surface-enhanced Raman spectroscopy (SERS) has shown great potential as a highly sensitive and specific optical analytical tool for identifying and monitoring tumor-related markers in NPC. RESULTS In this study, plasma samples from Nasopharyngeal cancer patients before and after immunotherapy, along with those from healthy volunteers, were tested using label-free SERS combined with plasmapheresis. Especially, the components with varying molecular weight sizes were analyzed via the separation process, thereby preventing the potential loss of diagnostic information that could result from competitive adsorption. Subsequently, a robust machine learning algorithm based on principal component analysis and linear discriminant analysis (PCA-LDA) was used to extract features from plasma SERS data and establish an effective discriminant model. The results showed that in the upper plasma layer, the most optimal discrimination was found between normal patients and those post-treatment, with sensitivity and specificity of 88.5 % and 92.3 %, respectively. In the lower plasma layer, the most optimal discrimination was found between the pre-treatment and post-treatment patients, with sensitivity and specificity of 84.6 % and 80.8 %, respectively. SIGNIFICANCE AND NOVELTY Plasmapheresis can reveal latent diagnostic information in the plasma and holds promise for treating NPC, the screening of the population who may benefit from immunotherapy, and the postoperative evaluation of immunotherapy. Further exploration of the feasibility of using SERS to detect tumor markers in the population who may benefit from immunotherapy for NPC will help improve the therapeutic efficacy, optimize clinical practice, and promote the development of individualized treatment strategies.
Collapse
Affiliation(s)
- Ruiying Lin
- Department of Radiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Zhangying Jiang
- Fuzhou Hospital of Traditional Chinese Medicine, Fuzhou, 350001, China
| | - Jinyong Lin
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Feifei Tong
- College of Physics and Electronic Information Engineering, Minjiang University, Fuzhou, 350108, China
| | - Yangmin Wu
- Key Laboratory of OptoElectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou, 350117, China
| | - Jinquan Hong
- College of Physics and Electronic Information Engineering, Minjiang University, Fuzhou, 350108, China
| | - Sufang Qiu
- Department of Radiation Oncology Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China.
| | - Qiong Wu
- College of Physics and Electronic Information Engineering, Minjiang University, Fuzhou, 350108, China.
| |
Collapse
|
2
|
Lin S, Ma Y, Wang Y, Yang Y, Xue J, Huang Y, Zhao Y, Fang W, Hong S, Zhang Y, Liu Q, Liu G, She X, Zha J, Zheng S, Li Y, Luo P, Zhang L, Zhao H. Phase 1b/2 study of ADG106, a 4-1BB/CD137 agonist, in combination with toripalimab in patients with advanced solid tumors. iScience 2025; 28:112497. [PMID: 40421181 PMCID: PMC12104636 DOI: 10.1016/j.isci.2025.112497] [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: 11/18/2024] [Revised: 01/27/2025] [Accepted: 04/17/2025] [Indexed: 05/28/2025] Open
Abstract
This phase 1b/2 clinical trial (NCT04775680) evaluated the safety, efficacy, pharmacokinetics and pharmacodynamics of ADG106, a ligand-blocking agonistic antibody targeting CD137 (4-1BB), combined with toripalimab in patients with advanced malignancies. ADG106 0.75-3 mg/kg plus toripalimab 240 mg were administered every 3 weeks. One dose-limiting toxicity occurred in 1 subject at 1.5 mg/kg and 2 in another subject at 3 mg/kg. Grade ≥ 3 treatment related adverse events occurred in 4/25 patients (16%). The overall disease control rate was 29.2% (7/24), including 1 partial response (PR) patient with a duration of response and a progression-free survival of 17.6 and 24.5 months. Circulating biomarkers suggested increased soluble CD137, CD3-CD16+CD56+ natural killer (NK) cells, interferon γ (IFN-γ), TNFα, and IL-6 after therapy. Elevated baseline memory T cells and PD-L1, activation of immune-related pathways, along with enhanced T cell proliferation and increased IFN-γ following treatment were observed in the PR patient. ADG106 in combination with toripalimab demonstrated a manageable safety profile but no efficacy conclusions could be drawn.
Collapse
Affiliation(s)
- Shaoyan Lin
- Department of Clinical Research, 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, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | - Yuxiang Ma
- Department of Clinical Research, 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, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | - Yongsheng Wang
- Department of Thoracic Medical Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yunpeng Yang
- Department of Medical Oncology, 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, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | - Jinhui Xue
- Department of Clinical Research, 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, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | - Yan Huang
- Department of Medical Oncology, 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, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | - Yuanyuan Zhao
- Department of Medical Oncology, 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, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | - Wenfeng Fang
- Department of Medical Oncology, 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, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | - Shaodong Hong
- Department of Medical Oncology, 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, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | - Yang Zhang
- Department of Clinical Research, 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, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | - Qianwen Liu
- Department of Clinical Research, 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, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | | | | | | | | | - Yan Li
- Adagene, Inc., San Diego, CA, USA
| | | | - Li Zhang
- Department of Medical Oncology, 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, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | - Hongyun Zhao
- Department of Clinical Research, 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, No. 651 Dongfeng East Road, Guangzhou 510060, China
| |
Collapse
|
3
|
Liu Z, Deng X, Wang Z, Guo Y, Hameed MMA, El-Newehy M, Zhang J, Shi X, Shen M. A biomimetic therapeutic nanovaccine based on dendrimer-drug conjugates coated with metal-phenolic networks for combination therapy of nasopharyngeal carcinoma: an in vitro investigation. J Mater Chem B 2025; 13:5440-5452. [PMID: 40241472 DOI: 10.1039/d5tb00226e] [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: 04/18/2025]
Abstract
Developing a minimally invasive and potent therapy for nasopharyngeal carcinoma is still challenging. In this study, we report a photothermal nanovaccine based on phenylboronic acid (PBA)-modified poly(amidoamine) dendrimers of generation 5 (G5) attached with indocyanine green (ICG) as a photothermal agent, toyocamycin (Toy) as an endoplasmic reticulum stress (ERS) drug, and Mn2+-coordinated metal-phenolic networks. The developed nanocomplexes are camouflaged with homologous apoptotic cancer cell membranes, leveraging membrane proteins as an antigenic reservoir and incorporating the immune adjuvant cytosine-guanine (CpG) oligonucleotide to obtain the final nanovaccine formulation. The prepared nanovaccine with a size of 72.4 nm displays satisfactory colloidal stability and photothermal conversion efficiency (36.7%), and is capable of targeting cancer cells and inducing apoptosis under laser irradiation through combined ICG-mediated photothermal therapy, Toy-enabled chemotherapy and Mn2+-mediated chemodynamic therapy. Meanwhile, the combined therapeutic effects can elicit immune responses to mature dendritic cells through the immunogenic cell death of cancer cells and the inserted CpG adjuvant/apoptotic cancer cell membranes, and polarize tumor-associated macrophage cells to the antitumor M1 phenotype. The antitumor efficacy of the nanomedicine platform was proven by the test of the penetration and therapeutic inhibition of 3-dimensional tumor spheroids in vitro. The developed functional nanomedicine integrated with different therapeutic modes may be developed as a biomimetic therapeutic nanovaccine for nasopharyngeal carcinoma treatment.
Collapse
Affiliation(s)
- Zhiyun Liu
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| | - Xiaochun Deng
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| | - Zhiqiang Wang
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| | - Yunqi Guo
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| | - Meera Moydeen Abdul Hameed
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Mohamed El-Newehy
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Jianjun Zhang
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China.
| | - Xiangyang Shi
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| | - Mingwu Shen
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| |
Collapse
|
4
|
Pei Y, Li C, Zhang B, Zheng Q, Chen S, Li J. Single-cell transcriptomics and metabolomics reveal the potential role of ASRGL1 in metabolic reprogramming and invasion of nasopharyngeal carcinoma cells. Int J Biochem Cell Biol 2025; 185:106788. [PMID: 40320045 DOI: 10.1016/j.biocel.2025.106788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Nasopharyngeal carcinoma (NPC) is an aggressive and highly metastatic malignancy, presenting significant challenges for early diagnosis and treatment. Asparaginase-like protein 1 (ASRGL1) is an important enzyme involved in amino acid metabolism, and previous studies have linked it to the progression of various tumors. However, the specific role of ASRGL1 in NPC remains unclear. This study analyzed multiple publicly available datasets related to NPC. We conducted single-cell RNA sequencing (scRNA-seq) analysis on the GSE150430 dataset to identify different cell subpopulations and examine ASRGL1 expression and its functional implications. The expression of ASRGL1 and its correlation with EMT were validated using transcriptomic data. The expression of ASRGL1 in C666-1 cells was interfered with by siRNA, cell proliferation and invasion were detected by CCK8, EdU, plate cloning, Transwell and scratch method, and EMT was evaluated by detecting the expression of E-cadherin and N-cadherin. Amino acid metabolomics and GC-MS headspace metabolomics were used to analyze the effects of ASRGL1 knockdown on the metabolic pattern of NPC cells. This study found that ASRGL1 was mainly expressed in fibroblasts, epithelial cells and myeloid cells in nasopharyngeal carcinoma (NPC). The ASRGL1-cell gene was significantly enriched in the epithelial-mesenchymal transition pathway. Knocking down ASRGL1 can further inhibit the proliferation, invasion and EMT of C666-1 cells. At the same time, the utilization of various amino acids was significantly reduced, and further GC-MS metabolomics analysis showed that the cell metabolism was unbalanced. This study elucidates the expression characteristics and potential functional roles of asparaginase-like protein 1 (ASRGL1) in nasopharyngeal carcinoma (NPC), providing new insights into its potential as a diagnostic marker and therapeutic target.
Collapse
Affiliation(s)
- YingYing Pei
- Department of Otorhinolaryngology, The first affiliated hospital of Ningbo University, Ningbo, Zhejiang, China.
| | - Chunlin Li
- Department of Otorhinolaryngology, The first affiliated hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Bin Zhang
- Department of Otorhinolaryngology, The first affiliated hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Qi Zheng
- Department of Otorhinolaryngology, The first affiliated hospital of Ningbo University, Ningbo, Zhejiang, China
| | | | - Ji Li
- Department of Otorhinolaryngology, The first affiliated hospital of Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|
5
|
Zheng Z, Han J. The Evaluation of the Adjunctive Therapeutic Value of Mindfulness-Based Stress Reduction Therapy for Patients With Nasopharyngeal Carcinoma Induced Moderate Depression. ACTAS ESPANOLAS DE PSIQUIATRIA 2025; 53:516-525. [PMID: 40356009 PMCID: PMC12069915 DOI: 10.62641/aep.v53i3.1749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/07/2024] [Accepted: 08/15/2024] [Indexed: 05/15/2025]
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) presents a substantial challenge for patients, impacting their physical and psychological well-being. Patients may experience moderate depression, anxiety, and reduced quality of life due to the disease and its treatments. Therefore, we retrospectively analyzed the adjunctive therapeutic potential of mindfulness-based stress reduction (MBSR) therapy for NPC patients with moderate depression. METHODS Psychological parameters were assessed using standardized scales, including the Hamilton Depression Scale-17 (HAMD-17), Self-Rating Depression Scale (SDS), Perceived Social Support Scale (PSSS), the Short-From-12 Health Survey (SF-12), and Mindfulness Attention Awareness Scale (MAAS). Statistical analyses were performed to compare the two groups. RESULTS A total of 131 patients including 67 patients with control group and 64 patients with Mindfulness-Based Stress Reduction therapy group were included. After 8 weeks of treatment, the MBSR therapy group showed significant improvements in psychological parameters, including depression, anxiety, perceived stress, quality of life, and mindfulness attention awareness (p < 0.05), compared to the control group. Additionally, the MBSR therapy group reported significantly higher overall satisfaction with treatment, willingness to recommend treatment, and perceived benefit from treatment (p < 0.05). CONCLUSION The study findings support the adjunctive therapeutic value of MBSR therapy in improving psychological outcomes and patient satisfaction among individuals with NPC-induced moderate depression.
Collapse
Affiliation(s)
- Zeyu Zheng
- Department of Otolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Harbin Medical University, 150000 Harbin, Heilongjiang, China
| | - Jing Han
- Department of Otolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Harbin Medical University, 150000 Harbin, Heilongjiang, China
| |
Collapse
|
6
|
An D, Jiang X, Yang Y. Sesamin Exerts Anti-Tumor Activity in Nasopharyngeal Carcinoma Through Inducing Autophagy and Reactive Oxygen Species Production. FRONT BIOSCI-LANDMRK 2025; 30:26038. [PMID: 40302321 DOI: 10.31083/fbl26038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/15/2024] [Accepted: 11/26/2024] [Indexed: 05/02/2025]
Abstract
BACKGROUND Sesamin can suppress many cancers, but its effect on nasopharyngeal carcinoma (NPC) is unclear. Herein, we set out to pinpoint the possible changes in NPC due to Sesamin. METHODS The biological function of NPC cells exposed to Sesamin/N-acetyl-L-cysteine (NAC)/3-Methyladenine (3-MA) was detected, followed by evaluation of reactive oxygen species (ROS) production (dichlorodihydrofluorescein diacetate staining) and mitochondrial membrane potential (MMP) (flow cytometry). Proteins pertinent to apoptosis (cleaved caspase-3, cleaved poly (ADP-ribose) polymerase 1 (PARP1)), cell cycle (Cyclin B1), and autophagy (microtubule-associated protein light chain 3 (LC3)-I, LC3-II, Beclin-1, P62) were quantified by Western blot. After the xenografted tumor model in mice was established, the tumor volume and weight were recorded, and Ki-67 and cleaved caspase-3 levels were determined by immunohistochemical analysis. RESULTS Sesamin inhibited viability, proliferation, cell cycle progression and migration, induced apoptosis, increased ROS production, and decreased MMP in NPC cells. Sesamin elevated cleaved caspase-3/caspase-3, cleaved PARP1/PARP1, and Beclin-1 expressions as well as LC3-II/LC3-I ratio, while diminishing Cyclin B1 and P62 levels. NAC and 3-MA abrogated Sesamin-induced changes as above in NPC cells. Sesamin inhibited the increase of the xenografted tumor volume and weight, down-regulated Ki-67, and up-regulated cleaved caspase-3 in xenografted tumors. CONCLUSION Sesamin exerts anti-tumor activity in NPC, as demonstrated by attenuated tumor proliferation and xenografted tumor volume and weight, as well as induced apoptosis in tumor tissues, consequent upon the promotion of autophagy and reactive oxygen species production.
Collapse
Affiliation(s)
- Deqiang An
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Xianyao Jiang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Yucheng Yang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| |
Collapse
|
7
|
Feng Z, Yang Y, Luo W, Li J, Xie Z, Zuo L, Duan M, Zuo D, Mo R, Tang X, Yi S, He X, Liu F, Ma N, He F. Integrative analysis of taurine metabolism-related genes prognostic signature with immunotherapy and identification of ABCB1 and GORASP1 as key genes in nasopharyngeal carcinoma. Amino Acids 2025; 57:21. [PMID: 40272558 PMCID: PMC12021963 DOI: 10.1007/s00726-025-03452-7] [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: 12/18/2024] [Accepted: 03/26/2025] [Indexed: 04/25/2025]
Abstract
Taurine is an amino acid with several physiological functions and has been shown to be involved in the anti-tumor of human nasopharyngeal carcinoma (NPC) cells. However, the role of taurine metabolism-related genes (TMRGs) in NPC has not been reported. We integrated data from the Genecards, Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Expression Omnibus(GEO) databases to identify differentially expressed genes associated with taurine metabolism in NPC patients. Gene Ontology (GO) and KEGG analyses were conducted to investigate the underlying mechanisms. Subsequently, Cox regression and Least Absolute Shrinkage and Selection Operator (LASSO) regression analyses were performed to construct a taurine metabolism-related prognostic signature. Survival, medication sensitivity, and immunological microenvironment evaluations were performed to assess the prognostic utility of the model. Finally, immunohistochemistry (IHC) experiments were performed to validate the model's prognostic reliability. In addition, we further verified the reliability of our research results through molecular docking and single-cell sequencing. Our prognostic model was based on three pivotal TMRGs (ABCB1, GORASP1, and EZH2). Functional analysis revealed a strong association between TMRGs and miRNAs in cancer. Notably, increased risk scores correlated with worsening tumor malignancy and prognosis. Significant disparities in immune microenvironment, immune checkpoints, and drug sensitivity were observed between the high- and low-risk groups. The protein expression patterns of the selected genes in clinical NPC samples were validated using immunohistochemistry. Molecular docking verified the interaction between these three core genes and taurine, which was further supported by single-cell sequencing showing significant expression variation among different cell clusters in NPC. We had elucidated the functions, therapeutic potential, and prognostic significance of three key genes related to taurine metabolism in NPC through multidimensional research and experimental validation. This research provided valuable insights and potential avenues for improved NPC management.
Collapse
Affiliation(s)
- Zhang Feng
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
- Department of Otolaryngology Head and Neck Surgery, Pingnan County People's Hospital, Guigang, 537300, China
| | - Yuhang Yang
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Wenqi Luo
- Department of Pathology, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Jinqing Li
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Zhenlian Xie
- Department of Oncology, Pingnan County People's Hospital, Guigang, 537300, China
| | - Long Zuo
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Meijiao Duan
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Dongzhi Zuo
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Ruwei Mo
- Department of Pathology, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Xuejing Tang
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Shijiang Yi
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Xiaosong He
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Fangxian Liu
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Ning Ma
- Institute of Oriental Medicine, Suzuka University of Medical Science, Suzuka , Mie, 510-0293, Japan.
| | - Feng He
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
| |
Collapse
|
8
|
Ye Y, Wang P, Wu D, Tang F, Shen N, Hou G. Deubiquitinating enzyme UCHL1 stabilizes CAV1 to inhibit ferroptosis and enhance docetaxel resistance in nasopharyngeal carcinoma. Anticancer Drugs 2025:00001813-990000000-00387. [PMID: 40279201 DOI: 10.1097/cad.0000000000001721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
The overexpression of CAV1 in many cancers is linked to chemotherapy resistance, but the exact mechanisms by which CAV1 contributes to resistance in nasopharyngeal carcinoma (NPC) are not fully known. Our research aims to elucidate the potential pathways by which CAV1 contributes to chemotherapy resistance in NPC, providing a basis for developing strategies to overcome resistance. A docetaxel-resistant NPC cell line was established, and CAV1 expression was analyzed in the cell line and the resistant variant using western blot. The sensitivity of the resistant cell line to docetaxel was assessed via cell counting kit-8, colony formation assays, and flow cytometry. Flow cytometry was used to measure lipid reactive oxygen species levels, while kits were employed to determine Fe2+ and malondialdehyde concentrations. The Ubibrowser database helped identify ubiquitination enzymes that interact with CAV1. The binding relationship between UCHL1 and CAV1 was studied using co-immunoprecipitation and immunofluorescence, which also evaluated the deubiquitination activity of UCHL1 on CAV1. CAV1 is overexpressed in NPC tissues and cells, correlating with adverse patient prognoses. In docetaxel-resistant cells, CAV1 expression is elevated compared to standard NPC cells. Silencing CAV1 increased the sensitivity of these resistant cells to docetaxel. Additionally, treatment with the ferroptosis inducer erastin could counteract the effects of CAV1 overexpression on drug resistance. UCHL1 interacted with CAV1 and inhibited its ubiquitin-mediated degradation pathway. By deubiquitinating CAV1, UCHL1 stabilizes and increases its expression, which inhibits ferroptosis and enhances the resistance of NPC cells to docetaxel.
Collapse
Affiliation(s)
- Yixian Ye
- Department of Otorhinolaryngology, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen City, Fujian Province
| | - Peng Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongshan Hospital, Fudan University, Shanghai City
| | - Daquan Wu
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongshan Hospital, Fudan University, Shanghai City
| | - Fengrong Tang
- Department of Nursing, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen City, Fujian Province, China
| | - Na Shen
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongshan Hospital, Fudan University, Shanghai City
| | - Guanghui Hou
- Department of Otorhinolaryngology, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen City, Fujian Province
| |
Collapse
|
9
|
Guo J, Zhang M, Li X, Wang J. PTEN as a prognostic factor for radiotherapy plus immunotherapy response in nasopharyngeal carcinoma. J Nanobiotechnology 2025; 23:303. [PMID: 40259377 PMCID: PMC12010603 DOI: 10.1186/s12951-025-03315-z] [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: 09/27/2024] [Accepted: 03/11/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND In the context of nasopharyngeal carcinoma (NPC) treatment, radiotherapy combined with immunotherapy (IR + RT) is gaining traction. This study focuses on analyzing exosomal proteins, particularly Phosphatase and Tensin Homolog (PTEN), for predicting the efficacy of NPC treatments. Serum samples from NPC patients and IR + RT recipients were utilized for exosome (Exo) extraction and subsequent transcriptomic and proteomic analyses to identify treatment-related proteins. Flow cytometry of cells and exosomal analysis were performed to examine these proteins. In vitro experiments using C666-1 cells and their Exos explored various cellular responses, while a murine subcutaneous NPC model investigated the impact of PTEN modulation on tumor growth and the immune microenvironment. RESULTS The study demonstrated that PTEN serves as a crucial predictive biomarker, with its expression changes correlated with M2 macrophage polarization and CD8+ T cell activity. This highlights the potential significance of PTEN in predicting treatment outcomes and influencing the immune response in NPC. CONCLUSION The findings suggest that PTEN could play a key role in enhancing the efficacy of NPC radiotherapy and immunotherapy. By shedding light on PTEN's impact on tumor growth and the immune microenvironment, this study lays the groundwork for future personalized therapeutic strategies in NPC treatment.
Collapse
Affiliation(s)
- Jiaxing Guo
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Ming Zhang
- Department of Otorhinolaryngology, The 4th Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Xiaoli Li
- Department of Otorhinolaryngology, The 4th Affiliated Hospital of China Medical University, Shenyang, 110032, China.
| | - Jiashuo Wang
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
10
|
Tang X, Ma C, Ren Y, Lv Y, He Y, Han L, Wu J. Revealing the Potential of Solamargine for Anti Metastasis and Angiogenesis Inhibition in Nasopharyngeal Carcinoma. J Inflamm Res 2025; 18:4879-4898. [PMID: 40224395 PMCID: PMC11992990 DOI: 10.2147/jir.s485244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/06/2025] [Indexed: 04/15/2025] Open
Abstract
Background Nasopharyngeal carcinoma (NPC) is a major global health issue, especially in Southeast Asia. Solamargine (SM), an alkaloid from natural plants, inhibits various cancer cells. This study evaluates SM's effects on invasion, migration, EMT markers, angiogenesis, and related pathways in the NPC-specific C666-1 cell line. Methods In vitro assays, including wound healing, Transwell invasion, Western blot, and tube formation, were used to assess SM's impact on C666-1 NPC and HUVEC cells. SM concentrations were 2 µM and 5 µM, with axitinib (4 µM) as the control. Network pharmacology and GO-KEGG enrichment analyses were conducted to explore SM's targets and mechanisms in NPC. Results SM significantly inhibited C666-1 NPC cell invasion and migration by reducing EMT markers Vimentin and Snail. In HUVEC cells, SM decreased viability, invasion, migration, and tube formation, likely through VEGF signaling inactivation, EZH2 inhibition, and miR-203a-3p upregulation. Network pharmacology and GO-KEGG analyses identified key targets and pathways, suggesting SM's anti-NPC effects through multiple mechanisms. Discussion SM inhibits NPC cell invasion and migration by regulating EMT, suppressing angiogenesis, and modulating key pathways. These findings highlight SM's potential as an anti-cancer agent for NPC and provide new insights into its mechanisms. Network pharmacology and GO-KEGG analysis further identify its therapeutic targets, offering valuable directions for future drug development.
Collapse
Affiliation(s)
- Xiaojuan Tang
- Central Laboratory, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, 410006, People’s Republic of China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People’s Republic of China
| | - Changju Ma
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People’s Republic of China
| | - Yuan Ren
- Central Laboratory, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, 410006, People’s Republic of China
| | - Yuan Lv
- Central Laboratory, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, 410006, People’s Republic of China
| | - Yongheng He
- Department of Anorectal Surgery, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, 410006, People’s Republic of China
| | - Ling Han
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People’s Republic of China
| | - Jingjing Wu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People’s Republic of China
| |
Collapse
|
11
|
Huang Z, Li Y, Wu W, Wu L, Chen Z, Xu S, Li Y, Lai J, Qiu S, Lu J. Plasma Epstein-Barr Virus DNA Temporal Clearance Pattern During Induction-Concurrent (Chemo)Radiation Therapy for Risk Stratification in Nasopharyngeal Carcinoma. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00349-9. [PMID: 40204246 DOI: 10.1016/j.ijrobp.2025.03.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 03/17/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025]
Abstract
PURPOSE Plasma Epstein-Barr virus (EBV) DNA is a widely used biomarker for nasopharyngeal carcinoma (NPC). Prior investigations predominantly assessed EBV DNA at a single time point, thus neglecting the differential prognostic implications of the temporal clearance pattern of EBV DNA during induction-concurrent (chemo)radiation therapy (RT). METHODS AND MATERIALS We retrospectively reviewed EBV DNA clearance patterns during induction-concurrent chemoRT in newly diagnosed patients with nonmetastatic NPC. EBV DNA was tested at 3 time points (baseline [T0], end of induction chemotherapy [T1], and end of RT [T2]) and recorded as detectable (D) and undetectable (U). The association between EBV DNA pattern and progression-free survival was analyzed. RESULTS A total of 2203 NPCs were included. Five distinct EBV DNA trajectory patterns were identified: type Ⅰ (negative-stable, U-U-U, 7.3%), type Ⅱ (induction chemotherapy-elimination, D-U-U, 42.8%), type Ⅲ (RT-elimination, D-D-U, 35.0%), type Ⅳ (persistent-positive, D-D-D, 11.7%), and type Ⅴ (resurgence, D-U-D [1.5%], U-D-U [1.2%], U-D-D [0.4%], or U-U-D [0.2%]). The median follow-up was 53.5 months (IQR, 43.1-66.9). Type Ⅱ patients displayed superior 5-year progression-free survival (82.9% [95% CI, 80.4%-85.5%]) versus type Ⅲ (75.9% [72.8%-79.1%], P < .001), type Ⅳ (52.5% [46.4%-59.5%], P < .001), and type Ⅴ (72.5% [62.2%-84.6%], P = .028). The 5-year progression-free survival for type V patients with "D-U-D," "U-D-U," "U-D-D," and "U-U-D" patterns was 62.4% (46.2%-84.3%), 78.6% (63.1%-97.8%), 85.7% (63.3%-100.0%), and 75.0% (42.6%-100.0%), respectively. All 33 patients with the "D-U-D" pattern had stage Ⅲ-Ⅳ disease at diagnosis. CONCLUSIONS Temporal EBV DNA clearance patterns during induction-concurrent chemoRT provide valuable prognostic insights, enabling the identification of patients with high-risk NPC and informing personalized treatment strategies. Resurgence of EBV DNA may occur occasionally (3.3%). Caution is required when considering reduced-intensity therapy in patients with locoeregionally advanced disease when EBV DNA becomes U after induction chemotherapy.
Collapse
Affiliation(s)
- Zongwei Huang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Ying Li
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Wenxi Wu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Lishui Wu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Zihan Chen
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Siqi Xu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Yi Li
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Jinghua Lai
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Sufang Qiu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China; Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, Fujian, China.
| | - Jun Lu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China.
| |
Collapse
|
12
|
Zeng Y, Duan T, Huang J, Wang X. Astragaloside IV inhibits nasopharyngeal carcinoma progression by suppressing the SATB2/Wnt signaling axis. Toxicol Res (Camb) 2025; 14:tfaf047. [PMID: 40177383 PMCID: PMC11964083 DOI: 10.1093/toxres/tfaf047] [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: 10/11/2024] [Revised: 02/10/2025] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
Astragaloside IV (AS-IV), a major bioactive component of Astragalus membranaceus, exhibits anti-cancer and anti-inflammatory properties. However, its precise role in nasopharyngeal carcinoma (NPC) remains unclear. This study investigated the effects of AS-IV on NPC progression and its relationship with Special AT-rich binding protein-2 (SATB2), a diagnostic marker for NPC. AS-IV treatment reduced NPC cell viability in a dose-dependent manner, as assessed by CCK-8 assays. Functional experiments, including transwell, immunofluorescence, and flow cytometry assays, demonstrated that AS-IV inhibited cell migration, invasion, and autophagy while promoting apoptosis. Western blot analysis showed that SATB2 expression was significantly elevated in NPC cells, particularly in C666-1 and HK-1 cells. Overexpression of SATB2 partially reversed AS-IV's inhibitory effects on NPC progression. Further analysis revealed that AS-IV suppressed the Wnt signaling pathway by downregulating SATB2 expression, while SATB2 overexpression restored Wnt pathway activation. This effect was reversed upon treatment with the Wnt pathway inhibitor DKK-1. In vivo, AS-IV administration inhibited tumor growth in a nude mouse subcutaneous xenograft model, reduced Ki-67 positivity, and lowered LC3B expression, indicating decreased proliferation and autophagy. However, these effects were diminished upon SATB2 overexpression. These findings suggest that AS-IV exerts anti-tumor effects in NPC by downregulating SATB2 and suppressing Wnt pathway activation, highlighting its potential as a therapeutic agent for NPC. Highlights Astragaloside IV (AS-IV) reduces nasopharyngeal carcinoma (NPC) cell vitality, suppresses cell migration, invasion and autophagy, and fosters apoptosis.SATB2 exhibits notably high levels in NPC cells.Overexpression of SATB2 counteracts the inhibition of NPC malignant progression by AS-IV.AS-IV impedes NPC progression by decreasing SATB2 and thereby hindering the Wnt pathway.AS-IV deters NPC tumor growth in nude mice.
Collapse
Affiliation(s)
- Yinping Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, 31 Longhua Road, Longhua District, Haikou 570102, Hainan Province, China
| | - Tingting Duan
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, 31 Longhua Road, Longhua District, Haikou 570102, Hainan Province, China
| | - Jiajun Huang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, 31 Longhua Road, Longhua District, Haikou 570102, Hainan Province, China
| | - Xiaofeng Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, 31 Longhua Road, Longhua District, Haikou 570102, Hainan Province, China
| |
Collapse
|
13
|
Zhang X, Yan J, Lei Q, Neo J, Tan SH, Shu X, Huang L, Long B, Xie Y, Wang F, Wang Y, Tu H, Wang C, Zhang L, Yang J, Zhang J, Liu H, Lim DWT, Chua MLK, Sui JD, Wang Y. A Randomized, Multicenter, Phase 2 Trial of Camrelizumab With or Without Metastasis-directed Stereotactic Body Radiation Therapy in Recurrent or Metastatic Nasopharyngeal Carcinoma. Int J Radiat Oncol Biol Phys 2025; 121:980-990. [PMID: 39454735 DOI: 10.1016/j.ijrobp.2024.10.019] [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: 06/04/2024] [Revised: 09/22/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024]
Abstract
PURPOSE To investigate the efficacy of metastasis-directed therapy (MDT) when added to camrelizumab (Cam) in patients with recurrent or metastatic nasopharyngeal carcinoma (R/M-NPC). METHODS AND MATERIALS We conducted a randomized, controlled, multicenter, phase 2 trial in 3 centers from China (NCT04830267). Patients with R/M-NPC, without prior exposure to immunotherapy, who presented with ≥2 lesions, and at least 1 measurable lesion were randomized 1:1 to either Cam alone or Cam plus MDT (Cam+MDT). Patients randomized to the MDT group must have 1 lesion that is amendable to stereotactic body radiation therapy (SBRT) prescribed to 27 Gy in 3 fractions every other day. The primary endpoint was objective response rate (ORR) of unirradiated lesions using Response Evaluation Criteria in Solid Tumors v1.1. RESULTS Between April 2021 and August 2023, 39 patients were randomly assigned to receive either Cam (n = 20) or Cam+MDT (n = 19). In total, 17 out of 39 (43.6%) patients had oligometastatic disease (≤3 lesions), 18 out of 39 (46.2%) had liver involvement, and 3 out of 39 (7.7%) had locoregional recurrent disease. ORR of unirradiated lesions did not differ between the treatment groups (26.3% [Cam+MDT] vs 30.0% [Cam], P = 1.0). The disease control rate of unirradiated lesions was 73.7% in the Cam+MDT group compared with 60.0% in the Cam group (P = .571). After a median follow-up of 25.8 months, median progression-free survival was 9.3 (95% CI, 6.2-not reached [NR]) months in the Cam+MDT group and 8.8 (95% CI, 3.3-NR) months in the Cam group (P = .750). Exploratory analyses suggested a longer overall survival (OS) with Cam+MDT for patients with >3 lesions (HR, 0.23; 95% CI, 0.07-0.77; P = .009). G3 and above adverse events were comparable between the treatment groups (15.8% [Cam+MDT] vs 20.0% [Cam]). The overall rate of capillary proliferation was 17.9% (7/39) for the trial. CONCLUSIONS Our study did not meet its primary endpoint of superior ORR of unirradiated lesions with the addition of MDT to Cam in patients with R/M-NPC.
Collapse
Affiliation(s)
- Xin Zhang
- Radiation Oncology Centre, Chongqing University Cancer Hospital, Chongqing, China
| | - Jin Yan
- Radiation Oncology Centre, Chongqing University Cancer Hospital, Chongqing, China
| | - Qianqian Lei
- Radiation Oncology Centre, Chongqing University Cancer Hospital, Chongqing, China
| | - Jialing Neo
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore
| | - Sze Huey Tan
- Oncology Academic Programme, Duke-NUS Medical School, Singapore; Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore
| | - Xiaolei Shu
- Radiation Oncology Centre, Chongqing University Cancer Hospital, Chongqing, China
| | - Luo Huang
- Radiation Oncology Centre, Chongqing University Cancer Hospital, Chongqing, China
| | - Bin Long
- Radiation Oncology Centre, Chongqing University Cancer Hospital, Chongqing, China
| | - Yue Xie
- Radiation Oncology Centre, Chongqing University Cancer Hospital, Chongqing, China
| | - Feng Wang
- Radiation Oncology Centre, Chongqing University Cancer Hospital, Chongqing, China
| | - Yuwei Wang
- Radiation Oncology Centre, Chongqing University Cancer Hospital, Chongqing, China
| | - Honglei Tu
- Radiation Oncology Centre, Chongqing University Cancer Hospital, Chongqing, China
| | - Chengchen Wang
- Radiation Oncology Centre, Chongqing University Cancer Hospital, Chongqing, China
| | - Lu Zhang
- College of Medicine, Chongqing University, Chongqing, China
| | - Jieying Yang
- College of Medicine, Chongqing University, Chongqing, China
| | - Jianwen Zhang
- The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Huawen Liu
- Chongqing University Three Gorges Hospital, Wanzhou, Chongqing, China
| | - Darren W T Lim
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore; Oncology Academic Programme, Duke-NUS Medical School, Singapore; Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Melvin L K Chua
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore; Oncology Academic Programme, Duke-NUS Medical School, Singapore; Division of Radiation Oncology, National Cancer Centre Singapore, Singapore.
| | - Jiang Dong Sui
- Radiation Oncology Centre, Chongqing University Cancer Hospital, Chongqing, China.
| | - Ying Wang
- Radiation Oncology Centre, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
14
|
Xie W, Yin Q, Fang H, Yang KY, Xue HB, Zheng H, Li P. Diagnosis of nasopharyngeal disease during upper GI endoscopy. Gut 2025:gutjnl-2024-334528. [PMID: 40015950 DOI: 10.1136/gutjnl-2024-334528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/09/2025] [Indexed: 03/01/2025]
Affiliation(s)
- Wei Xie
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qian Yin
- Department of Otorhinolaryngology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hong Fang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kai-Yan Yang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hai-Bo Xue
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huimin Zheng
- Central Laboratory of the Medical Research Center, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Pan Li
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, Sydney, New South Wales, Australia
| |
Collapse
|
15
|
Zhang W, Zhu Z, Liu Y. The impact of the ATP-binding cassette (ABC) transporter family on multidrug resistance in head and neck tumors. Mol Biol Rep 2025; 52:256. [PMID: 39982595 DOI: 10.1007/s11033-025-10321-9] [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: 11/14/2024] [Accepted: 01/30/2025] [Indexed: 02/22/2025]
Abstract
The ATP-binding cassette (ABC) transporter family is among the largest protein superfamilies, consisting of seven subfamilies, and plays an important role in various physiological processes and in the clinical manifestations of many diseases. The early clinical signs of head and neck cancer (HNC) are often subtle, resulting in most patients being diagnosed at more advanced stages. This late diagnosis adversely affects tumor treatment, and the resistance of certain tumors to chemotherapy further poses significant challenges for clinical management. Several previous studies have indicated a correlation between the ABC protein family and multidrug resistance (MDR) in tumors. This article offers a thorough review of the subfamilies, structures, functions, and roles of ABC transporters in MDR related to head and neck tumors, with the aim of providing insights and recommendations for overcoming MDR in this context.
Collapse
Affiliation(s)
- Wanqing Zhang
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhengxin Zhu
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuehui Liu
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
16
|
Zeng X, Jin X, Leng J, Zhang S, Wang Y, Chen J, Zhang S, Teng L, Hu Z, Zhou S, Zeng Z, Long J. High-dose radiation induces dendritic cells maturation by promoting immunogenic cell death in nasopharyngeal carcinoma. Front Immunol 2025; 16:1554018. [PMID: 40040692 PMCID: PMC11876370 DOI: 10.3389/fimmu.2025.1554018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 01/31/2025] [Indexed: 03/06/2025] Open
Abstract
Aim and background Due to the radiosensitivity and deep anatomical location of nasopharyngeal carcinoma (NPC), radiotherapy serves as the cornerstone of standardized treatment for this malignancy. Beyond its cytotoxic effects, radiotherapy can serve as an immunological adjuvant by inducing immunogenic cell death (ICD). Dendritic cells (DCs), as potent antigen-presenting cells, play a critical role in tumor immunotherapy, but their exact role in the ICD process of NPC remains unclear. The effects of high-dose radiation (≥2 Gy) on DCs and the type of immune response it elicits in NPC have not been fully elucidated. Methods An in vitro study was conducted to assess whether ICD of NPC 5-8F cells induced by high-dose radiation could regulate the immune response of DCs. Specifically, the maturation and antigen-presenting capacity of DCs were evaluated following co-culture with NPC cells exposed to high-dose radiation. Results High-dose radiation was found to induce ICD in NPC 5-8F cells, as evidenced by increased pro-inflammatory factor levels and reduced anti-inflammatory factor levels in the cell culture supernatant. Co-culture with NPC cells exposed to high-dose radiation for 15 minutes significantly enhanced the expression of surface molecules on DCs, promoting their immune sensitization. Conclusion High-dose radiation-induced apoptosis of NPC 5-8F cells is a form of ICD, which plays an important role in regulating DC immune function. These findings provide insight into the immunomodulatory effects of radiotherapy in NPC and its potential to enhance tumor immunotherapy through DC activation.
Collapse
Affiliation(s)
- Xianlin Zeng
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Xianhuai Jin
- Department of Oncology, Guiyang Public Health Clinical Center, Guiyang, Guizhou, China
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Ji Leng
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Shuai Zhang
- Department of Interventional Radiology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yun Wang
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Jin Chen
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Shichao Zhang
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Lijing Teng
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Zuquan Hu
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Shi Zhou
- Department of Interventional Radiology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhu Zeng
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| | - Jinhua Long
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Department of Oncology, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
17
|
Ni G, Wang W, Dang Y, Cheng C, Wang Q. The role of SOX18 in nasopharyngeal carcinoma: implications for prognosis and therapy. Am J Transl Res 2025; 17:913-926. [PMID: 40092108 PMCID: PMC11909540 DOI: 10.62347/ytrv6870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/26/2024] [Indexed: 03/19/2025]
Abstract
OBJECTIVE To investigate the cellular function of SOX18 in nasopharyngeal carcinoma (NPC) by analyzing its effects on tumor cell proliferation, apoptosis, migration and invasion, and to verify its expression and prognostic significance by clinical samples, thereby providing a basis for precise diagnosis and treatment. METHODS SOX18 expression was analyzed in NPC cell lines and clinical samples. Gene silencing techniques were utilized to reduce SOX18 expression in NPC cells, followed by assays to evaluate cell proliferation, apoptosis, migration, and invasion. Additionally, changes in the Wnt/β-catenin signaling pathway were examined. RESULTS High SOX18 expression was correlated with poor survival in NPC patients. Silencing SOX18 significantly inhibited cell proliferation, increased apoptosis, and suppressed migration and invasion capabilities. Furthermore, SOX18 silencing downregulated key genes and proteins associated with the Wnt/β-catenin signaling pathway. CONCLUSION SOX18 plays a critical role in NPC progression by affecting key cellular behaviors. Targeting SOX18 may offer new therapeutic strategies and improve prognostic assessments for NPC patients, highlighting its potential as a valuable molecular marker for cancer treatment.
Collapse
Affiliation(s)
- Guoqian Ni
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fujian Cancer HospitalFuzhou 350200, Fujian, China
- College of Biological Science and Engineering, Fuzhou UniversityFuzhou 350108, Fujian, China
| | - Wenbin Wang
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fujian Cancer HospitalFuzhou 350200, Fujian, China
- College of Biological Science and Engineering, Fuzhou UniversityFuzhou 350108, Fujian, China
| | - Yuan Dang
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fujian Cancer HospitalFuzhou 350200, Fujian, China
| | - Cui Cheng
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fujian Cancer HospitalFuzhou 350200, Fujian, China
- College of Biological Science and Engineering, Fuzhou UniversityFuzhou 350108, Fujian, China
| | - Qiaowen Wang
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fujian Cancer HospitalFuzhou 350200, Fujian, China
| |
Collapse
|
18
|
Guo Y, Lu J, Li X, Yan S, Zhou J, Tian Z, Liu Y, Li N, Zhou Q, Li X, Shi L, Jiang S, Li M, Zhou X, Huang D, Zeng Z, Fan S, Xiong W, Zhou M, Li G, Zhang W. BRD7 Inhibited Immune Escape in Nasopharyngeal Carcinoma via Inhibiting PD-L1 Expression. Int J Biol Sci 2025; 21:1914-1931. [PMID: 40083706 PMCID: PMC11900823 DOI: 10.7150/ijbs.103703] [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: 09/14/2024] [Accepted: 01/20/2025] [Indexed: 03/16/2025] Open
Abstract
Therapeutic strategies aimed at harnessing anti-tumor immunity are being intensively investigated as they show promising results in cancer treatment. The PD-1/ PD-L1 pathway is an essential target for restoring functional anti-tumor immune response. BRD7 is a candidate tumor suppressor gene and nuclear transcription factor of nasopharyngeal carcinoma (NPC) which was cloned in our laboratory. In this paper, we reported that the candidate tumor suppressor gene BRD7 was strongly associated with good prognosis of NPC patients and negatively regulated PD-L1 expression. In addition, we found that BRD7 down-regulated PD-L1 expression and enhanced the killing function of CD8+ T lymphocytes in NPC cells through binding to p85α via the 485-651 domain and inhibiting the activity of PI3K, thereby inhibiting the activity of PI3K/AKT/mTOR/STAT3 pathway. In vivo experiments, the results showed that BRD7 could not only inhibit the growth of tumors, but also play a better anti-tumor effect when combined with PD-L1 antibody. These results provided further evidence that BRD7 inhibited immune escape of NPC through down-regulating PD-L1 expression.
Collapse
Affiliation(s)
- Yilin Guo
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Blood Transfusion, Children's Hospital Affiliated to Zhengzhou University,Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, Henan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jiaxue Lu
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiaoxu Li
- Department of Clinical Laboratory, WuHu Hospital, East China Normal University (The Second People's Hospital, WuHu), Wuhu, Anhui, China
| | - Shiqi Yan
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jieyu Zhou
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ziying Tian
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ying Liu
- Department of Clinical Laboratory, Zhengzhou Orthopaedics Hospital, Zhengzhou, China
| | - Nan Li
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qing Zhou
- Department of Clinical Laboratory, First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Shi
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Su Jiang
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Mengna Li
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital/ the Affiliated Cancer Hospital of Xiangya School of Medicine, 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
- Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Xiao Zhou
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Donghai Huang
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital/ the Affiliated Cancer Hospital of Xiangya School of Medicine, 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
- Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Songqing Fan
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital/ the Affiliated Cancer Hospital of Xiangya School of Medicine, 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
- Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Ming Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital/ the Affiliated Cancer Hospital of Xiangya School of Medicine, 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
- Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital/ the Affiliated Cancer Hospital of Xiangya School of Medicine, 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
- Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Wenling Zhang
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
19
|
Xu S, He J, Liu Z, Pei Y, Ge J, Qing Y, Wei Y, Chen Y, Peng X. Immune-related adverse events as prognostic biomarkers in recurrent or metastatic nasopharyngeal carcinoma receiving PD-L1 inhibitor: A post-hoc analysis of the multicenter, single-arm, phase 2 study. Oral Oncol 2025; 161:107161. [PMID: 39746267 DOI: 10.1016/j.oraloncology.2024.107161] [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: 12/15/2024] [Accepted: 12/22/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Immune-related adverse events (irAEs) have been associated with better therapeutic outcomes in patients receiving immune checkpoint inhibitors (ICIs) across various cancers. This study investigates the association between irAEs and ICI outcomes in patients with recurrent or metastatic nasopharyngeal carcinoma (R/M NPC). METHODS A post hoc analysis was performed on 153 patients with R/M NPC who received anti-PD-L1 inhibitors between February 26, 2019, and January 13, 2021. These patients were enrolled in a multicenter, single-arm, phase 2 clinical trial, which represents the largest study to date evaluating anti-PD-L1 therapy in previously treated R/M NPC. RESULTS Patients who experienced irAEs had significantly higher overall response rates (ORR) and diseade control rates (DCR) compared to those without irAEs (31.2 % vs. 17.1 %, p = 0.039; 66.7 % vs. 44.8 %, p = 0.017). Additionally, the median progression-free survival (PFS) was longer in patients with irAEs (129 days vs. 56 days, p = 0.007). Patients with endocrine-related irAEs exhibited higher DCR (71.8 % vs. 46.2 %, p = 0.012), and longer PFS (144 days vs. 69 days, p = 0.02) and overall survival (OS: 746 days vs. 438 days, p = 0.02). In contrast, patients with grade ≥ 3 irAEs or thoserequiring systemic steroid therapy had shorter median OS (179 days vs. 466 days, p = 0.03; 166 days vs. 462 days, p = 0.02). CONCLUSIONS The occurrence of irAEs, particularly those involving the endocrine system, is associated with enhanced efficacy of anti-PD-L1 therapy and extended survival in patients with R/M NPC, highlighting their potential as prognostic biomarkers.
Collapse
Affiliation(s)
- Shihong Xu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Jiagang He
- Department of Medical Education, Kweichow Moutai Hospital, Zunyi, Guizhou, China.
| | - Zheran Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Yiyan Pei
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Junyou Ge
- Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd., Chengdu, China.
| | - Yan Qing
- Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd., Chengdu, China.
| | - Youneng Wei
- Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd., Chengdu, China.
| | - Ye Chen
- Division of Abdominal Tumor Multimodality Treatment, Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
20
|
Yang J, Tan F, Chen Y, Li X, Yuan C. The emerging role of long non-coding RNA SOX2-OT in cancers and non-malignant diseases. J Physiol Biochem 2025; 81:57-83. [PMID: 39702742 DOI: 10.1007/s13105-024-01059-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 11/02/2024] [Indexed: 12/21/2024]
Abstract
SOX2 overlapping transcript (SOX2-OT) is a long non-coding RNA located at chromosome 3q26.33 in humans. Convincing data confirm that SOX2-OT is evolutionarily conserved and plays a significant role in various malignant and non-malignant diseases. In most cancers, the upregulation of SOX2-OT acts as an oncogenic factor, strongly correlating with tumor risk, adverse clinicopathological features, and poor prognosis. Mechanistically, SOX2-OT is regulated by seven transcription factors and influences cellular behavior by modulating SOX2 expression, competitively binding 20 types of miRNAs, stabilizing protein expression, or promoting protein ubiquitination. It also participates in epigenetic modifications and activates multiple signaling pathways to regulate cancer cell proliferation, apoptosis, migration, invasion, autophagy, immune evasion, and resistance to chemotherapy/targeted therapies. Additionally, SOX2-OT triggers apoptosis, oxidative stress, and inflammatory responses, contributing to neurodevelopmental disorders, cardiovascular diseases, and diabetes-related conditions. Genetic polymorphisms of SOX2-OT have also been linked to breast cancer, gastric cancer, recurrent miscarriage, sepsis, and eating disorders in patients with bipolar disorder. This review provides an overview of recent research progress on SOX2-OT in human diseases, highlights its substantial potential as a prognostic and diagnostic biomarker, and explores its future clinical applications.
Collapse
Affiliation(s)
- Jingjie Yang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Fangshun Tan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Yaohui Chen
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Xiaolan Li
- The Second People's Hospital of China Three Gorges University, Yichang, 443002, China.
- The Second People's Hospital of Yichang, Hubei, China.
| | - Chengfu Yuan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002, China.
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
| |
Collapse
|
21
|
Liu Q, Wang H, Chen Z, Xiong J, Huang Y, Zhang S, Zhang Q. Global, regional, and national epidemiology of nasopharyngeal carcinoma in middle-aged and elderly patients from 1990 to 2021. Ageing Res Rev 2025; 104:102613. [PMID: 39626854 DOI: 10.1016/j.arr.2024.102613] [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: 10/10/2024] [Revised: 11/24/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND In recent years, changes in the incidence and mortality rates of nasopharyngeal carcinoma have occurred globally, across various regions, and among different countries. As a high incidence group, it is necessary to study the prevalence trend of middle-aged and elderly people. METHODS Detailed information on NPC in middle-aged and elderly patients from 1990 to 2021 was collected from the Global Burden of Disease Database 2021 (GBD2021). Adopted incidence, mortality, disability-adjusted life-years (DALYs), sociodemographic index (SDI) and corresponding Estimated Annual Percentage Changes (EAPCs) to assess the burden of NPC in middle-aged and elderly patients. Additionally, a global risk attribution analysis was conducted, and a Bayesian age-period-cohort (BAPC) model was applied to project the global burden of NPC in middle-aged and elderly patients from 2021 to 2035. FINDINGS Globally, the incidence cases of NPC in middle-aged and elderly people increased by 58.2 %, the numbers of death increased by 33.8 %, and the DALY increased by 42.1 %. However, the EAPCs values and upper limits in incidence, mortality and DALY rates were all less than 0, indicating a decreasing trend of incidence, mortality and disease burden. Both incidence and mortality rates were decreasing in high-incidence territories. Most regions were negatively correlated with the sociodemographic index. Males had obviously higher incidence and mortality of NPC in middle-aged and elderly patients than females. The highest incidences of nasopharyngeal carcinoma in middle-aged and elderly males were in the 65-69 age group, and the incidences in females did not change much among different age groups. We found that Alcohol use, Occupational risk and Tobacco were the major risk factors for NPC-related mortality in middle-aged and elderly patients. CONCLUSION Controllable etiology should be effectively controlled in the future. DATA AVAILABILITY The data sets generated and/or analyzed during the current study are available in the GBD repository (https://vizhub.healthdata.org/gbd-results/). Data will be made available on request.
Collapse
Affiliation(s)
- Qiqi Liu
- Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu postal code: 610072, PR China
| | - Hanyu Wang
- Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu postal code: 610072, PR China
| | - Ze Chen
- Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu postal code: 610072, PR China
| | - Jiahui Xiong
- Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu postal code: 610072, PR China
| | - Yong Huang
- Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu postal code: 610072, PR China
| | - Shipeng Zhang
- Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu postal code: 610072, PR China.
| | - Qinxiu Zhang
- Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu postal code: 610072, PR China; Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu postal code: 610072, PR China; School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu Postal code: 611137, PR China; World Health Organization Collaborating Centre (WHOCC), CHN-56, Chengdu Postal code:610041, PR China.
| |
Collapse
|
22
|
Mark JKK, Teh AH, Yap BK. Epstein-Barr virus-infected nasopharyngeal carcinoma therapeutics: oncoprotein targets and clinical implications. Med Oncol 2025; 42:59. [PMID: 39888474 DOI: 10.1007/s12032-025-02610-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/13/2025] [Indexed: 02/01/2025]
Abstract
Nasopharyngeal carcinoma (NPC) is a distinctive epithelial cancer closely associated with Epstein-Barr Virus (EBV) infection, posing significant challenges in diagnosis and treatment due to its resistance to conventional therapies and high recurrence rates. Current therapies, including radiotherapy and chemotherapy, exhibit limited efficacy, particularly in recurrent or metastatic cases, highlighting the urgent need for novel therapeutic strategies. Targeting EBV oncoproteins, such as Epstein-Barr Virus encoded Nuclear Antigen 1 (EBNA1), Latent Membrane Protein 1 (LMP1), and Latent Membrane Protein 2 (LMP2), presents a promising therapeutic avenue in NPC treatment. This review discusses the latest advancements in drug discovery targeting EBV oncoproteins, emphasizing the identification of inhibitors for specific functional regions of oncoproteins EBNA1, LMP1, and LMP2. Particular attention is given to the molecular mechanisms of these inhibitors and their preclinical or clinical potential in treating EBV-positive NPC. These developments highlight a promising future for targeted therapies in improving outcomes for NPC patients.
Collapse
Affiliation(s)
- Jacqueline Kar Kei Mark
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia
| | - Aik-Hong Teh
- Centre for Chemical Biology, Universiti Sains Malaysia, 11900, Bayan Lepas, Penang, Malaysia
| | - Beow Keat Yap
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia.
| |
Collapse
|
23
|
Hua X, Wang MD, Ni WQ, Long ZQ, Wang SF, Duan FF, Zhang C, Huang X, Xu F, Xia W, Chen JY, Gao YS. Development and validation of a prognostic nomogram incorporating neutrophil-to-albumin ratio for predicting overall survival in patients with nasopharyngeal carcinoma undergoing concurrent chemoradiotherapy. Heliyon 2025; 11:e40881. [PMID: 39801974 PMCID: PMC11719357 DOI: 10.1016/j.heliyon.2024.e40881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Recent research suggests that the emerging neutrophil-albumin ratio (NAR) has a significant correlation with the survival outcomes across a range of tumors, yet its predictive significance for nasopharyngeal carcinoma (NPC) remains insufficiently investigated. This study aimed to evaluate the relationship between the neutrophil-to-albumin ratio (NAR) and overall survival (OS) in patients with NPC, as well as to develop a corresponding prognostic model. METHODS This retrospective analysis included 861 NPC patients treated with concurrent chemoradiotherapy (CCRT), who were randomly divided into a training group (n = 605) and a validation group (n = 256). To identify factors associated with OS and construct a prognostic nomogram, both univariate and multivariate Cox regression analyses were performed. The nomogram's prognostic accuracy was evaluated and independently validated. RESULTS The NAR score successfully segregated NPC patients into two categories with significantly different OS (HR = 0.536; 95 % CI: 0.296-0.972, P = 0.040). Through multivariate analysis, factors such as age, T stage, N stage, and NAR score were identified as independent predictors of OS, leading to the creation of a prognostic nomogram. This nomogram demonstrated superior predictive capability for OS [C-index = 0.702 (95 % CI: 0.636-0.768)], surpassing that of the conventional staging system [C-index = 0.651 (95 % CI: 0.549-0.752)]. The findings underwent internal validation within an independent cohort. CONCLUSIONS The NAR, an emergent biomarker combining nutritional and inflammatory status, offers a practical, low-cost, and non-invasive prognostic measure for NPC patients treated with CCRT. Additionally, the prognostic nomogram derived from NAR surpasses traditional staging systems in predictive accuracy.
Collapse
Affiliation(s)
- Xin Hua
- Department of Radiation Oncology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Meng-Di Wang
- Department of Radiation Oncology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Wei-Qiong Ni
- Department of Radiation Oncology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Zhi-Qing Long
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Si-Fen Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Fang-Fang Duan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Chao Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Xin Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Fei Xu
- Department of Radiation Oncology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Wen Xia
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Jia-Yi Chen
- Department of Radiation Oncology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Yun-Sheng Gao
- Department of Radiation Oncology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| |
Collapse
|
24
|
Chen Q, Hao Q, Yang Y, Li L, Li D, Zhao R, Wei W, Deng L, Su J, Liang Z, Tang S, Lu Y, Liang Y, Zhang Z, Zhou X, Xiao X, Li P, Huang Y, Zhao W. Carboxylesterase 4A Inhibits the Malignant Biological Behavior of Nasopharyngeal Carcinoma via the PI3K/AKT Pathway. Technol Cancer Res Treat 2025; 24:15330338251319144. [PMID: 39912257 PMCID: PMC11800256 DOI: 10.1177/15330338251319144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/10/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Carboxylesterase 4A (CES4A) belongs to the member of the carboxylesterase family, yet there has been limited research into its malignant biological behavior in malignant tumors. Here, we aim to investigate the expression, cellular biological functions, and the potential underlying mechanism of CES4A in nasopharyngeal carcinoma (NPC). METHOD A standardized mean difference (SMD) analysis was used to analyze the dysregulation of CES4A based on the gene expression omnibus (GEO) database. qRT-PCR and immunohistochemical staining (IHC) were used to identify the mRNA and protein levels of CES4A in NPC cell lines and tissues, respectively. CCK-8, colony formation, wound healing and transwell assays were utilized to estimate cellular growth and metastasis, respectively. Western blot was conducted to evaluate the activity of PI3K/AKT signaling pathway. RESULT Both mRNA and protein expression of CES4A was significantly diminished both in NPC cell lines and primary tumor tissues. Ectopic expression of CES4A restrains the proliferation, colony formation, migration and invasion of NPC. Additionally, KEGG analysis based on GEO data and high-throughput transcriptome sequencing of cell lines all strongly suggested that CES4A was involved in regulating phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway. It was observed that AKT and phosphorylated AKT were remarkably reduced in CES4A overexpressing NPC cells, indicating that PI3K/AKT signaling pathway is hindered by CES4A. CONCLUSION CES4A expression is silenced in NPC, functioning as a tumor suppressor by negatively modulating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Qiaoli Chen
- Department of Pathology, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China
- Key Laboratory of Oral Infectious Disease Prevention and Control, Health Commission of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Regional Key Laboratory of Early Prevention and Treatment of High Incidence Tumors, Nanning, China
| | - Quanxiang Hao
- Department of Pathology, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China
- Key Laboratory of Oral Infectious Disease Prevention and Control, Health Commission of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Regional Key Laboratory of Early Prevention and Treatment of High Incidence Tumors, Nanning, China
| | - Yanping Yang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Limei Li
- Department of Pathology, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China
- Key Laboratory of Oral Infectious Disease Prevention and Control, Health Commission of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Danping Li
- Department of Pathology, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China
- Key Laboratory of Oral Infectious Disease Prevention and Control, Health Commission of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ran Zhao
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Wanqi Wei
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lixian Deng
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Jiaming Su
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ziyuan Liang
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Shiyue Tang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yaomin Lu
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Yushan Liang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhe Zhang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaoying Zhou
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Xue Xiao
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ping Li
- Department of Pathology, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China
- Key Laboratory of Oral Infectious Disease Prevention and Control, Health Commission of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yi Huang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Weilin Zhao
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
25
|
Vickram S, Infant SS, Manikandan S, Jenila Rani D, Mathan Muthu CM, Chopra H. Immune biomarkers and predictive signatures in gastric cancer: Optimizing immunotherapy responses. Pathol Res Pract 2025; 265:155743. [PMID: 39616978 DOI: 10.1016/j.prp.2024.155743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/07/2024] [Accepted: 11/25/2024] [Indexed: 12/11/2024]
Abstract
Gastric cancer is a malignant disease with a poor prognosis and few therapeutic options once it has advanced. Immunotherapy using ICIs has emerged as a viable therapeutic method; nevertheless, reliable immunological biomarkers are required to identify who may benefit from these therapies. It focuses on key immune biomarkers and predictive signatures in gastric cancer, such as PD-L1 expression, microsatellite instability (MSI), tumor mutational burden (TMB), and Epstein-Barr virus (EBV) status, to optimize gastric cancer patients' immunotherapy responses. PD-L1 expression is a popular biomarker for ICI effectiveness. Tumors with high MSI-H and TMB are the most susceptible to ICIs because they are highly immunogenic. EBV-positive stomach tumors are highly immunogenic, and immunotherapy has a high response rate. Combining composite biomarker panels with multi-omics-based techniques improved patient selection accuracy. In recent years, machine learning models have been integrated into next-generation sequencing. Dynamic, real-time-monitorable biomarkers for real-time immune response monitoring are also being considered. Thus, enhancing biomarker-driven immunotherapy is critical for improving clinical outcomes with gastric cancer. There is still more work to be done in this field, and verifying developing biomarkers will be an important component in the future of customized cancer therapy.
Collapse
Affiliation(s)
- Sundaram Vickram
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India.
| | - Shofia Saghya Infant
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - S Manikandan
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - D Jenila Rani
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - C M Mathan Muthu
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Hitesh Chopra
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| |
Collapse
|
26
|
Feng R, Guo Y, Chen M, Tian Z, Liu Y, Jiang S, Zhou J, Liu Q, Li X, Xiong W, Shi L, Fan S, Li G, Zhang W. PLUNC downregulates the expression of PD-L1 by inhibiting the interaction of DDX17/β-catenin in nasopharyngeal carcinoma. J Pathol Transl Med 2025; 59:68-83. [PMID: 39815745 PMCID: PMC11736280 DOI: 10.4132/jptm.2024.11.27] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/31/2024] [Accepted: 11/27/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is characterized by high programmed death-ligand 1 (PD-L1) expression and abundant infiltration of non-malignant lymphocytes, which renders patients potentially suitable candidates for immune checkpoint blockade therapies. Palate, lung, and nasal epithelium clone (PLUNC) inhibit the growth of NPC cells and enhance cellular apoptosis and differentiation. Currently, the relationship between PLUNC (as a tumor-suppressor) and PD-L1 in NPC is unclear. METHODS We collected clinical samples of NPC to verify the relationship between PLUNC and PD-L1. PLUNC plasmid was transfected into NPC cells, and the variation of PD-L1 was verified by western blot and immunofluorescence. In NPC cells, we verified the relationship of PD-L1, activating transcription factor 3 (ATF3), and β-catenin by western blot and immunofluorescence. Later, we further verified that PLUNC regulates PD-L1 through β-catenin. Finally, the effect of PLUNC on β-catenin was verified by co-immunoprecipitation (Co-IP). RESULTS We found that PLUNC expression was lower in NPC tissues than in paracancer tissues. PD-L1 expression was opposite to that of PLUNC. Western blot and immunofluorescence showed that β-catenin could upregulate ATF3 and PD-L1, while PLUNC could downregulate ATF3/PD-L1 by inhibiting the expression of β-catenin. PLUNC inhibits the entry of β-catenin into the nucleus. Co-IP experiments demonstrated that PLUNC inhibited the interaction of DEAD-box helicase 17 (DDX17) and β-catenin. CONCLUSIONS PLUNC downregulates the expression of PD-L1 by inhibiting the interaction of DDX17/β-catenin in NPC.
Collapse
Affiliation(s)
- Ranran Feng
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Laboratory Medicine, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Yilin Guo
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Blood Transfusion, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Meilin Chen
- The First Affiliated Hospital of Sun Yatsen University, Guangzhou, China
| | - Ziying Tian
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Medical Laboratory Science , Xiangya School of Medicine, Central South University, Changsha, China
| | - Yijun Liu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Medical Laboratory Science , Xiangya School of Medicine, Central South University, Changsha, China
| | - Su Jiang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Medical Laboratory Science , Xiangya School of Medicine, Central South University, Changsha, China
| | - Jieyu Zhou
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Medical Laboratory Science , Xiangya School of Medicine, Central South University, Changsha, China
| | - Qingluan Liu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Medical Laboratory Science , Xiangya School of Medicine, Central South University, Changsha, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Lei Shi
- Department of Pathology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Songqing Fan
- Department of Pathology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Wenling Zhang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Medical Laboratory Science , Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
27
|
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
|
28
|
Zhang Y, Gu S, Deng H, Shen Z. Global epidemiological profile in nasopharyngeal carcinoma: a prediction study. BMJ Open 2024; 14:e091087. [PMID: 39658299 PMCID: PMC11647315 DOI: 10.1136/bmjopen-2024-091087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
OBJECTIVES This study delineates the global nasopharyngeal carcinoma's (NPC) incidence and mortality across 185 countries in 2020 and projects the disease's burden by 2040. DESIGN A prediction study. SETTING Countries within the 20 world regions. PARTICIPANTS Global NPC population. PRIMARY AND SECONDARY OUTCOME MEASURES The estimated counts of NPC cases and deaths were retrieved from the GLOBOCAN 2020 database. Age-standardised incidence rates (ASIR) and age-standardised death rates (ASDR) were computed. Projections for NPC by 2040 were derived from global population forecasts. RESULTS In the year 2020, East Asia emerged as the epicentre of both NPC incidences and mortalities, encompassing 49.39% (65 866 of the total 133 354 cases) and 45.56% (36 453 of the total 80 008 deaths), respectively, with China's contribution being the most substantial (46.82% of cases and 43.50% of deaths). The disparity between genders was notable, as the ASIR and ASDR for males were approximately triple those observed in females. The incidence exhibited regional diversity, with South-Eastern Asia and East Asia recording the highest ASIR for males and females (7.7 and 2.5, and 3.9 and 1.5 per 100 000 person-years, respectively). Similarly, South-Eastern Asia also reported the highest ASDR for both genders (5.4 and 1.5 per 100 000 person-years, respectively). Projections for 2040 anticipate a rise in annual cases and deaths to 179 476 (indicating a 34.58% increase from 2020) and 113 851 (reflecting a 42.29% increase), respectively. Further analysis revealed a correlation between the Human Development Index and disease burden. CONCLUSIONS NPC, primarily impacting East Asia and predominantly affecting men, is poised for a significant increase in incidence and mortality by 2040, especially in Asia.
Collapse
Affiliation(s)
- Yuna Zhang
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China
- Department of Operating Room, Ningbo University, Ningbo, China
| | - Shanshan Gu
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China
| | - Hongxia Deng
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China
| | - Zhisen Shen
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China
| |
Collapse
|
29
|
Bicci E, Di Finizio A, Calamandrei L, Treballi F, Mungai F, Tamburrini S, Sica G, Nardi C, Bonasera L, Miele V. Head and Neck Squamous Cell Carcinoma: Insights from Dual-Energy Computed Tomography (DECT). Tomography 2024; 10:1780-1797. [PMID: 39590940 PMCID: PMC11598236 DOI: 10.3390/tomography10110131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Head and neck cancer represents the seventh most common neoplasm worldwide, with squamous cell carcinoma being the most represented histologic variant. The rising incidence of the neoplastic pathology of this district, coupled with the drastic changes in its epidemiology over the past decades, have posed significant challenges to physicians worldwide in terms of diagnosis, prognosis, and treatment. In order to meet these challenges, a considerable amount of effort has been spent by the authors of the recent literature to explore new technologies and their possible employment for the better diagnostic and prognostic definition of head and neck squamous cell carcinoma (HNSCC). Among these technologies, a growing interest has been gathering around the possible applications of dual-energy computed tomography (DECT) in head and neck pathology. Dual-energy computed tomography (DECT) utilizes two distinct X-ray energy spectra to obtain two datasets in a single scan, allowing for material differentiation based on unique attenuation profiles. DECT offers key benefits such as enhanced contrast resolution, reduced beam-hardening artifacts, and precise iodine quantification through monochromatic reconstructions. It also creates material decomposition images, like iodine maps, aiding in tumor characterization and therapy assessment. This paper aims to summarize recent findings on the use of DECT in HNSCC, providing a comprehensive overview to aid further research and exploration in the field.
Collapse
Affiliation(s)
- Eleonora Bicci
- Department of Radiology, Careggi University Hospital, 50134 Florence, Italy; (F.M.); (C.N.); (L.B.); (V.M.)
| | - Antonio Di Finizio
- Department of Radiology, Careggi University Hospital, University of Florence, 50134 Florence, Italy; (A.D.F.); (L.C.); (F.T.)
| | - Leonardo Calamandrei
- Department of Radiology, Careggi University Hospital, University of Florence, 50134 Florence, Italy; (A.D.F.); (L.C.); (F.T.)
| | - Francesca Treballi
- Department of Radiology, Careggi University Hospital, University of Florence, 50134 Florence, Italy; (A.D.F.); (L.C.); (F.T.)
| | - Francesco Mungai
- Department of Radiology, Careggi University Hospital, 50134 Florence, Italy; (F.M.); (C.N.); (L.B.); (V.M.)
| | - Stefania Tamburrini
- Department of Radiology, Ospedale del Mare, ASL NA1 Centro, 80147 Naples, Italy;
| | - Giacomo Sica
- Department of Radiology, Monaldi Hospital, 80131 Naples, Italy;
| | - Cosimo Nardi
- Department of Radiology, Careggi University Hospital, 50134 Florence, Italy; (F.M.); (C.N.); (L.B.); (V.M.)
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Luigi Bonasera
- Department of Radiology, Careggi University Hospital, 50134 Florence, Italy; (F.M.); (C.N.); (L.B.); (V.M.)
| | - Vittorio Miele
- Department of Radiology, Careggi University Hospital, 50134 Florence, Italy; (F.M.); (C.N.); (L.B.); (V.M.)
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| |
Collapse
|
30
|
Weng Y, Cai S, Li C, Xu Y, Pan Y, Huang Z, Li Y, Wu Z, Chen Y, Qiu S. Selection of induction chemotherapy cycles for stage N3 nasopharyngeal carcinoma based on pre-treatment plasma EBV DNA. Sci Rep 2024; 14:24484. [PMID: 39424840 PMCID: PMC11489564 DOI: 10.1038/s41598-024-75396-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024] Open
Abstract
This study aimed to explore the selection of induction chemotherapy (IC) cycles for stage N3 nasopharyngeal carcinoma (NPC). We employed propensity score matching (PSM) to categorize patients into 3-cycle and 4-cycle IC groups (IC = 3 and IC = 4). The log-rank and chi-squared tests were used respectively to evaluate the differences in survival and acute toxicities. Survival outcomes including overall survival (OS), progression-free survival (PFS), distant metastasis-free survival (DMFS), and locoregional relapse-free survival (LRRFS) were evaluated among the two groups. After PSM, each group comprised 99 patients. The IC = 4 group exhibited markedly improved survival outcomes compared with the IC = 3 group. Multivariate analysis revealed that pre-EBV DNA was an independent risk factor affecting PFS and DMFS. For high-risk patients with pre-EBV DNA ≥ 7800 copies/ml, the IC = 4 group demonstrated greater survival compared to the IC = 3 group. Among low-risk patients with pre-EBV DNA < 7800 copies/ml, both groups showed comparable survival outcomes. In terms of acute adverse reactions, the IC = 4 group experienced higher incidences, particularly with grade 2-4 alanine transaminase elevation and thrombocytopenia. For stage N3 NPC, pre-EBV DNA could be a powerful predictor for guiding the selection of IC cycles. The IC = 4 regimen is probably more beneficial to high-risk patients due to superior survival, while for low-risk patients, the IC = 3 regimen may be sufficient.
Collapse
Affiliation(s)
- Youliang Weng
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Sunqin Cai
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Chao Li
- Department of Oncology, Second Hospital of Sanming City, Sanming, China
| | - Yun Xu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yuhui Pan
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Zongwei Huang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Ying Li
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Zijie Wu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yu Chen
- The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China.
| | - Sufang Qiu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
| |
Collapse
|
31
|
Zhou YQ, Cheng XX, He S, Liu SQ, Li YQ, Wei PP, Luo CL, Bei JX. A positive feedback loop between PLD1 and NF-κB signaling promotes tumorigenesis of nasopharyngeal carcinoma. J Genet Genomics 2024; 51:997-1006. [PMID: 38885836 DOI: 10.1016/j.jgg.2024.06.004] [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: 04/17/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Abstract
Phospholipase D (PLD) lipid-signaling enzyme superfamily has been widely implicated in various human malignancies, but its role and underlying mechanism remain unclear in nasopharyngeal carcinoma (NPC). Here, we analyze the expressions of 6 PLD family members between 87 NPC and 10 control samples through transcriptome analysis. Our findings reveal a notable upregulation of PLD1 in both NPC tumors and cell lines, correlating with worse disease-free and overall survival in NPC patients. Functional assays further elucidate the oncogenic role of PLD1, demonstrating its pivotal promotion of critical tumorigenic processes such as cell proliferation and migration in vitro, as well as tumor growth in vivo. Notably, our study uncovers a positive feedback loop between PLD1 and the NF-κB signaling pathway to render NPC progression. Specifically, PLD1 enhances NF-κB activity by facilitating the phosphorylation and nuclear translocation of RELA, which in turn binds to the promoter of PLD1, augmenting its expression. Moreover, RELA overexpression markedly rescues the inhibitory effects in PLD1-depleted NPC cells. Importantly, the application of the PLD1 inhibitor, VU0155069, substantially inhibits NPC tumorigenesis in a patient-derived xenograft model. Together, our findings identify PLD1/NF-κB signaling as a positive feedback loop with promising therapeutic and prognostic potential in NPC.
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
| | - 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
| | - 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
| | - 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
| | - 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
| | - 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
| | - 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.
| | - 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 510060, P. R. China; Department of Medical Oncology, National Cancer Centre of Singapore, Singapore.
| |
Collapse
|
32
|
Shan Y, Fan H, Chai L, Kong X, Xiao H, You M, You Y. Tumor-derived exosomal miR-103a-3p promotes vascular permeability and proliferation by targeting ZO-1 and ACOX-1 in nasopharyngeal carcinoma. Transl Cancer Res 2024; 13:4896-4912. [PMID: 39430846 PMCID: PMC11483361 DOI: 10.21037/tcr-23-2359] [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: 12/24/2023] [Accepted: 07/19/2024] [Indexed: 10/22/2024]
Abstract
Background miR-103a-3p has been reported to be a factor leading to poor prognosis in several human malignancies, including nasopharyngeal carcinoma (NPC). Secreted microRNAs containing exosomes may mediate the communication between cancer and stromal cells. The purpose of the current work was to learn more about miR-103a-3p's function in NPC exosomes. Methods Transmission electron microscopy and NanoSight analysis were used to verify the existence of exosomes. To determine the relationship between exosomal miR-103a-3p and carcinogenesis in NPC, gain- and loss-of-function studies were carried out. Cell Counting Kit-8 (CCK8), 5-ethynyl-2'-deoxyuridine (EdU) cell proliferation assay, colony formation, flow cytometry, trans-endothelial invasion assays, endothelial permeability and cellular immunofluorescence were used to identify roles of exosomal miR-103a-3p in vitro. Zebrafish assay was used to disclose the effect of exosomal miR-103a-3p in vivo. Bioinformatics and dual-luciferase reporter assay were applied to clarify the mechanism of exosomal miR-103a-3p regulating the crosstalk between NPC cells and human umbilical vein endothelial cells (HUVECs). Results In the present study, we first demonstrated that the overexpression of exosomal miR-103a-3p improved NPC cell proliferation, migration, and the epithelial-mesenchymal transition (EMT) progression in vitro. Then, we verified that NPC cell-derived exosomal miR-103a-3p destroyed the integrity of the endothelial monolayer in vitro and in vivo by downregulating zonula occludens 1 (ZO-1) expression. Moreover, we revealed that miR-103a-3p containing exosomes facilitated NPC cell proliferation through lipid droplet accumulation by direct target to metabolic enzyme acyl-CoA oxidase 1 (ACOX-1). Conclusions Our data demonstrate that exosomal miR-103a-3p can facilitate the development of NPC by regulating the crosstalk between NPC cells and HUVECs. Exosomal miR-103a-3p could potentially serve as a therapeutic target for NPC.
Collapse
Affiliation(s)
- Ying Shan
- Suzhou Medical College of Soochow University, Suzhou, China
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Hongmei Fan
- Otolaryngology Head and Neck Surgery, Affiliated Rugao Hospital of Nantong University, Nantong, China
| | - Linlin Chai
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical College of Nantong University, Nantong, China
| | - Xiuzhi Kong
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical College of Nantong University, Nantong, China
| | - Haijuan Xiao
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical College of Nantong University, Nantong, China
| | - Mengdie You
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical College of Nantong University, Nantong, China
| | - Yiwen You
- Suzhou Medical College of Soochow University, Suzhou, China
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
33
|
戴 军, 高 昳, 王 坚, 张 舒, 刘 鹏. [Effects of Ionizing Radiation on Intestinal Bile Acid Metabolism: Mechanism of the Radioprotective Effect of Glycoursodeoxycholic Acid]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:1195-1201. [PMID: 39507959 PMCID: PMC11536250 DOI: 10.12182/20240960403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Indexed: 11/08/2024]
Abstract
Objective Radioactive intestinal injury is a common complication during radiotherapy of tumors. The aim of this study is to observe the effect of ionizing radiation on short-term changes in intestinal bile acids and to investigate the radioprotective effect of bile acids on intestinal cells. Methods A rat model of small intestinal injury was constructed by exposing the abdomen of the rats to daily irradiation at 2 Gy for 4 d in succession. The bile acids were quantified using metabolomics analysis. IEC-6 cells, a small intestinal epithelial cell line, were divided into a dimethyl sulfoxide (DMSO) control group receiving DMSO and 0 Gy irradiation, a glycoursodeoxycholic acid (GUDCA) experimental group receiving GUDCA and 0 Gy irradiation, a DMSO irradiation group receiving DMSO and 10 Gy irradiation, and a GUDCA irradiation group receiving GUDCA and 10 Gy irradiation. Cell viability and cytotoxicity was assessed by CCK-8 assay test. The apoptosis rate of cells was determined by flow cytometry. The colony formation rate and the radiosensitivity of the cells were determined by colony formation assay on solid media. The expression levels of proteins associated with cell death were determined using Western blot. Results After exposure to irradiation, the small intestine tissues of the rats showed typical radioactive intestinal injury. In addition, various bile acids showed fluctuation before and after irradiation. Among the bile acids, GUDCA increased significantly at 3 d after irradiation, but returned to the pre-irradiation level at 7 d after irradiation. Compared with the control group, after GUDCA treatment at 20 μmol/L for 24 h, the cell viability rate after irradiation was significantly higher than that of the DMSO group (P<0.05); the expression levels of the proteins, including PARP, caspase-3, RIP, and GSDMD, were significantly lower than those in the control group (P<0.05). After GUDCA treatment at 20 μmol/L for 24 h and 48 h, the cell apoptosis rate of the cells after irradiation was lower than that of the DMSO group (P<0.05). Compared with the DMSO control group, the colony formation ability of the GUDCA experimental group was stronger than that of the DMSO group after irradiation at 0, 2, 4, and 6 Gy (P<0.05). D0, or the mean lethal dose, of the GUDCA group was 6.374, while that of the DMSO group was 4.572. Compared with the DMSO control group, the D0 value of the GUDCA treatment group increased, and the sensitization enhancement ratio (SER) was 0.717. Conclusion After exposing the abdomen of rats to irradiation, the intestinal bile acid metabolism of the rats will change significantly, and GUDCA can produce radioprotective effects on intestinal cells to a certain extent.
Collapse
Affiliation(s)
- 军 戴
- 徐州医科大学江阴临床学院 (无锡 214400)Jiangyin Clinical College, Xuzhou Medical University, Wuxi 214400, China
| | - 昳 高
- 徐州医科大学江阴临床学院 (无锡 214400)Jiangyin Clinical College, Xuzhou Medical University, Wuxi 214400, China
| | - 坚 王
- 徐州医科大学江阴临床学院 (无锡 214400)Jiangyin Clinical College, Xuzhou Medical University, Wuxi 214400, China
| | - 舒羽 张
- 徐州医科大学江阴临床学院 (无锡 214400)Jiangyin Clinical College, Xuzhou Medical University, Wuxi 214400, China
| | - 鹏飞 刘
- 徐州医科大学江阴临床学院 (无锡 214400)Jiangyin Clinical College, Xuzhou Medical University, Wuxi 214400, China
| |
Collapse
|
34
|
Martins AC, Oshiro MY, Albericio F, de la Torre BG. Food and Drug Administration (FDA) Approvals of Biological Drugs in 2023. Biomedicines 2024; 12:1992. [PMID: 39335511 PMCID: PMC11428688 DOI: 10.3390/biomedicines12091992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/26/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
An increase in total drug (small molecules and biologics) approvals by the Food and Drug Administration (FDA) was seen in 2023 compared with the previous year. Cancer remained the disease most targeted by monoclonal antibodies (mAbs), followed by autoimmune conditions. Our data reveal the prevalence of approvals for biologics even during years when the total number of authorizations was low, such as in 2022. Over half the drugs that received the green light in 2023 benefited from expedited programs, as the incidence of many diseases increased. In addition, over half of the biologics approved received Orphan Drug Designation from the FDA. This narrative review delves into details of the most significant approvals in 2023, including mAbs, enzymes, and proteins, explaining their mechanisms of action, differences from previous drugs, placebo, and standards of care, and outcomes in clinical trials. Given the varying number of drugs authorized annually by the U.S. health authority, this review also examines the limits of external influences over the FDA's decisions and independence regarding drug approvals and withdrawals.
Collapse
Affiliation(s)
- Alexander C Martins
- School of Health Sciences, UAM, Universidade Anhembi-Morumbi, São Paulo 03101-001, Brazil
- Medical Information Department, Thermo Fisher Scientific, São Paulo 4542011, Brazil
| | - Mariana Y Oshiro
- School of Health Sciences, UAM, Universidade Anhembi-Morumbi, São Paulo 03101-001, Brazil
| | - Fernando Albericio
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, Department of Organic Chemistry, University of Barcelona, 08028 Barcelona, Spain
| | - Beatriz G de la Torre
- KRISP, College of Health Sciences, University of KwaZulu-Natal, Durban 4001, South Africa
| |
Collapse
|
35
|
Dharmalingam Jothinathan MK. Letter to the Editor, "Immunochemotherapy alone or immunochemotherapy plus subsequent locoregional radiotherapy in de novo metastatic nasopharyngeal carcinoma". Oral Oncol 2024; 156:106948. [PMID: 39004023 DOI: 10.1016/j.oraloncology.2024.106948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Affiliation(s)
- Mukesh Kumar Dharmalingam Jothinathan
- Centre for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India.
| |
Collapse
|
36
|
Liu Q, Xu J, Dai B, Guo D, Sun C, Du X. Single-cell resolution profiling of the immune microenvironment in primary and metastatic nasopharyngeal carcinoma. J Cancer Res Clin Oncol 2024; 150:391. [PMID: 39158776 PMCID: PMC11333513 DOI: 10.1007/s00432-024-05900-5] [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: 05/22/2024] [Accepted: 07/17/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is an assertive malignancy with partially understood underlying mechanisms, urging further study into its diverse and dynamic tumor microenvironment (TME) to bolster diagnosis, treatment, and prognostic accuracy. AIMS To track the evolutionary route of metastasis, here we perform a yielding scRNA-seq data from 24 primary carcinoma, 7 peripheral blood mononuclear cell (PBMC) nasopharyngeal carcinoma, and 7 metastatic carcinoma patients' sample. MATERIALS AND METHODS Following high quality control and filtration, a total of 292,298 cells from these tumors were classified into 10 clusters: T cells, B cells, Macrophages/Monocytes, Natural Killer (NK) cells, Plasma cells, plasmacytoid Dendritic Cells, Migratory Dendritic Cells, Mast cells, Cancer-Associated Fibroblasts, and Epithelial cells. RESULTS By comparing and analyzing the different functional capacities of cellular entities within primary and metastatic nasopharyngeal carcinoma, coupled with a detailed investigation into the heterogeneity and differential fate trajectories of T cells, B cells, and myeloid cells, as well as assessing the interactions of cell-cell communicative heterogeneity between these carcinogenic states, we established single-cell atlases for primary and metastatic tumors and identified a large number of potential therapeutic targets. CONCLUSION This comprehensive analysis significantly advances our understanding of nasopharyngeal carcinoma (NPC) metastasis by detailing the evolutionary dynamics and the impact of the tumor microenvironment at a single-cell resolution, thereby laying a crucial foundation for future metastatic tumor research and providing new insights into immune heterogeneity, molecular interactions, and potential therapeutic strategies for NPC.
Collapse
Affiliation(s)
- Qiuping Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China
| | - Jingping Xu
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, People's Republic of China
| | - Bingyi Dai
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China
| | - Danni Guo
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China
| | - Changling Sun
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China
| | - Xiaodong Du
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China.
| |
Collapse
|
37
|
Kai J, Huang H, Su J, Chen Q. Identification of shared immune infiltration characteristic molecules in dermatomyositis and nasopharyngeal carcinoma using bioinformatics: Traits in dermatomyositis and nasopharyngeal cancer. Skin Res Technol 2024; 30:e13871. [PMID: 39081134 PMCID: PMC11289422 DOI: 10.1111/srt.13871] [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: 06/02/2024] [Accepted: 07/05/2024] [Indexed: 08/03/2024]
Abstract
BACKGROUND Dermatomyositis (DM) is a kind of dermatologically associated autoimmune disease that is notably associated with an increased risk of concurrent malignancies, although the underlying mechanisms remain to be fully elucidated. The purpose of this investigation was to examine the immunological parallels between DM and nasopharyngeal carcinoma (NPC), with the aim of identifying pivotal biomarkers that could facilitate a deeper understanding and enhance the predictive capabilities of NPC in DM patients. METHOD Data for DM and NPC were sourced from the Gene Expression Omnibus (GEO) database. Immune infiltration was analyzed using the "cibersort" R package, differentially expressed genes (DEGs) were identified with the "limma" package, and functional pathways were investigated through Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses. Characteristic genes were determined by Utilizing Protein-Protein Interaction (PPI) and Least Absolute Shrinkage and Selection Operator (LASSO), and their features were validated using the GSE53819 dataset. RESULTS In comparison to normal samples, significant infiltration of macrophage M1 was observed in both DM and NPC. The analysis revealed 77 DEGs in DM and 1051 DEGs in NPC, with 22 genes found to be co-DEGs. Following PPI and LASSO analysis, six distinctive genes were retained. Notably, CCL8, IFIH1, CXCL10, and CXCL11 exhibited optimal diagnostic efficacy for NPC and displayed significant correlation with macrophage M1 infiltration within the carcinoma. CONCLUSION Four characteristic genes, CCL8, IFIH1, CXCL10, and CXCL11 are risk factors for both DM and NPC. They exhibit a robust correlation with the incidence of NPC and offer a commendable diagnostic efficacy. Furthermore, they may serve as prospective predictive biomarkers for the emergence of NPC in DM.
Collapse
Affiliation(s)
- Jinyan Kai
- Department of Clinical Medical LaboratoryThe Affiliated Second Hospital of Xiamen Medical CollegeXiamenFujianChina
| | - Haitao Huang
- Department of MicrobiologyGuilin Medical UniversityGuilinGuangxiChina
| | - Jiaqi Su
- Department of Clinical Medical LaboratoryThe Affiliated Second Hospital of Xiamen Medical CollegeXiamenFujianChina
| | - Qiong Chen
- Department of Traditional Chinese MedicineShanghai General Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
38
|
Zhao Z, Hu C, Li L, Zhang J, Zhang L. Main chemical constituents and mechanism of anti-tumor action of Solanum nigrum L. Cancer Med 2024; 13:e7314. [PMID: 39155844 PMCID: PMC11331249 DOI: 10.1002/cam4.7314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 08/20/2024] Open
Abstract
OBJECTIVE Solanum nigrum L. (SNL) is a natural drugwith diverse bioactive components and multi-targeted anti-tumor effects, gaining increasing attention in clinical application. METHOD AND RESULTS This paper reviews the studies on SNL by searching academic databases (Google Scholar, PubMed, Science Direct,and Web of Science, among others), analyzing its chemical compositions (alkaloids, saponins, polysaccharides, and polyphenols, among others), andbriefly describes the anti-tumor mechanisms of the main components. DISCUSSION This paper discusses the shortcomings of the current research on SNL and proposes corresponding solutions, providing theoretical support for further research on its biological functions and clinical efficacy.
Collapse
Affiliation(s)
- Zhen‐duo Zhao
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Cheng Hu
- Experiment Center for Science and TechnologyShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Ling Li
- Institute of Vascular Anomalies, Shanghai TCM‐Integrated Hospital, Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Jia‐qi Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Li‐chao Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
39
|
Zeng C, Qiao M, Chen Y, Xie H. EBV-positive glycoproteins associated with nasopharyngeal carcinoma. Pathol Res Pract 2024; 260:155427. [PMID: 38936091 DOI: 10.1016/j.prp.2024.155427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/15/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024]
Abstract
Nasopharyngeal carcinoma (NPC) is closely related to Epstein-Barr virus (EBV) infection, and glycosylation of proteins is associated with precancerous lesions and carcinogenesis of NPC, and viral glycoproteins mediates the fusion of viruses with B cells or epithelial cells in the infection stage, promoting the conversion of normal epithelial cells into cancer cells. In the process of occurrence and development of NPC, various glycoproteins in the body promote or inhibit the proliferation, invasion, metastasis, and drug resistance of tumor cells, such as the tumor inhibitory effect of NGX6 and inhibin B (INHBB); the cancer-promoting effect of tenascin-C (TNC), fibronectin 1 (FN1), insulin-like growth factor binding protein-3 (IGFBP3), serglycin, and its core protein; and some effects of glycosylation of immune proteins on immunotherapy in NPC. This article provides an overview of the research progress on the interaction of glycoproteins associated with EBV infection with the occurrence and development of NPC.
Collapse
Affiliation(s)
- Chenlu Zeng
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, China
| | - Muchuan Qiao
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, China
| | - Yanhua Chen
- Department of Medical Oncology, the Second Affiliated Hospital of the University of South China, Hengyang, Hunan Province, China
| | - Hailong Xie
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, China.
| |
Collapse
|
40
|
Li Z, Lin Y, Qiu T, Liang J, Lan Y, Meng F, Liang C, Zhang Y, Wang Q, Shi D, Zhang C, Shi Y, Liu L, Yang Y, Zhang J. Noninvasive Photothermal Therapy of Nasopharyngeal Cancer Guided by High Efficiency Optical-Absorption Nanomaterial Enhanced by NIR-II Photoacoustic Imaging. Int J Nanomedicine 2024; 19:7817-7830. [PMID: 39099790 PMCID: PMC11298190 DOI: 10.2147/ijn.s457069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 07/25/2024] [Indexed: 08/06/2024] Open
Abstract
Background Photothermal therapy (PTT) guided by photoacoustic imaging (PAI) using nanoplatforms has emerged as a promising strategy for cancer treatment due to its efficiency and accuracy. This study aimed to develop and synthesize novel second near-infrared region (NIR-II) absorption-conjugated polymer acceptor acrylate-substituted thiadiazoloquinoxaline-diketopyrrolopyrrole polymers (PATQ-DPP) designed specifically as photothermal and imaging contrast agents for nasopharyngeal carcinoma (NPC). Methods The PATQ-DPP nanoparticles were synthesized and characterized for their optical properties, including low optical band gaps. Their potential as PTT agents and imaging contrast agents for NPC was evaluated both in vitro and in vivo. The accumulation of nanoparticles at tumor sites was assessed post-injection, and the efficacy of PTT under near-infrared laser irradiation was investigated in a mouse model of NPC. Results Experimental results indicated that the PATQ-DPP nanoparticles exhibited significant photoacoustic contrast enhancement and favorable PTT performance. Safety and non-toxicity evaluations confirmed the biocompatibility of these nanoparticles. In vivo studies showed that PATQ-DPP nanoparticles effectively accumulated at NPC tumor sites and demonstrated excellent tumor growth inhibition upon exposure to near-infrared laser irradiation. Notably, complete elimination of nasopharyngeal tumors was observed within 18 days following PTT. Discussion The findings suggest that PATQ-DPP nanoparticles are a promising theranostic agent for NIR-II PAI and PTT of tumors. This innovative approach utilizing PATQ-DPP nanoparticles provides a powerful tool for the early diagnosis and precise treatment of NPC, offering a new avenue in the management of this challenging malignancy.
Collapse
Affiliation(s)
- Zhaoyong Li
- Department of Radiology, DongGuan Tungwah Hospital, Dongguan Key Laboratory of Radiology and Molecular Imaging, DongGuan, Guangdong, 523000, People’s Republic of China
| | - Yanping Lin
- Department of Radiology, DongGuan Tungwah Hospital, Dongguan Key Laboratory of Radiology and Molecular Imaging, DongGuan, Guangdong, 523000, People’s Republic of China
| | - Ting Qiu
- Department of Radiology, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, People’s Republic of China
| | - Junsheng Liang
- Department of Radiology, DongGuan Tungwah Hospital, Dongguan Key Laboratory of Radiology and Molecular Imaging, DongGuan, Guangdong, 523000, People’s Republic of China
| | - Yintao Lan
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, Guangdong, 510030, People’s Republic of China
| | - Fan Meng
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Chaohao Liang
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Yiqing Zhang
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Qingyun Wang
- Department of Radiology, DongGuan Tungwah Hospital, Dongguan Key Laboratory of Radiology and Molecular Imaging, DongGuan, Guangdong, 523000, People’s Republic of China
| | - Da Shi
- Department of Radiology, DongGuan Tungwah Hospital, Dongguan Key Laboratory of Radiology and Molecular Imaging, DongGuan, Guangdong, 523000, People’s Republic of China
| | - Changli Zhang
- Department of Radiology, DongGuan Tungwah Hospital, Dongguan Key Laboratory of Radiology and Molecular Imaging, DongGuan, Guangdong, 523000, People’s Republic of China
| | - Yanan Shi
- Department of Radiology, DongGuan Tungwah Hospital, Dongguan Key Laboratory of Radiology and Molecular Imaging, DongGuan, Guangdong, 523000, People’s Republic of China
| | - Liujun Liu
- Department of Radiology, DongGuan Tungwah Hospital, Dongguan Key Laboratory of Radiology and Molecular Imaging, DongGuan, Guangdong, 523000, People’s Republic of China
| | - Yanlan Yang
- Department of Radiology, DongGuan Tungwah Hospital, Dongguan Key Laboratory of Radiology and Molecular Imaging, DongGuan, Guangdong, 523000, People’s Republic of China
| | - Jian Zhang
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
- Department of Oncology, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, 511500, People’s Republic of China
| |
Collapse
|
41
|
Peng Z, Hong R, Dunhui Y, Zhen W, Yongjin W, Xianhai Z. Network pharmacology and biological verification of morusin's therapeutic mechanisms in inhibiting nasopharyngeal carcinoma growth. J Cancer 2024; 15:4866-4878. [PMID: 39132159 PMCID: PMC11310868 DOI: 10.7150/jca.97044] [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: 04/07/2024] [Accepted: 06/27/2024] [Indexed: 08/13/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) presents a significant therapeutic challenge due to its aggressive nature and limited treatment options. Although morusin, a compound found in traditional Chinese medicines, exhibits significant tumor-inhibiting properties, its specific effects on NPC proliferation remain unclear. This study aims to elucidate the inhibitory effects of morusin on NPC survival and proliferation while exploring the underlying mechanisms through the utilization of network pharmacology, molecular docking, and experimental validation in vitro and in vivo. Network pharmacology analysis identified 117 potential targets of morusin against NPC, with 8 hub targets including AKT1, BCL2, CASP3, CTNNB1, ESR1, HSP90AA1, MMP9, STAT3, and the IL-17 signaling pathway. Further investigation of public data indicated that the expression levels of BLC2, CASP3, CTNNB1, HSP90AA1, and STAT3 in NPC tissue were significantly elevated compared to normal nasopharyngeal tissue. Docking studies exposed robust binding activity between morusin and key gene molecules. Additionally, biological assays demonstrated that morusin effectively inhibits NPC growth both in vivo and in vitro. Through a comprehensive investigation, this study identified the pharmacological mechanisms essential for morusin-induced inhibition of NPC growth by targeting multiple molecular targets and signaling pathways. These findings show the potential to contribute to the development of novel clinical agents for treating NPC.
Collapse
Affiliation(s)
- Zhang Peng
- ✉ Corresponding authors: Peng Zhang, E-mail: ; Yongjin Wu, E-mail: ; Xianhai Zeng, E-mail:
| | | | | | | | - Wu Yongjin
- Department of Otolaryngology, Longgang Otolaryngology Hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Zeng Xianhai
- Department of Otolaryngology, Longgang Otolaryngology Hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| |
Collapse
|
42
|
Yu YF, Lu GZ, Wang RJ, Song YK, Wu SG. Additional PD-1 inhibitor improves complete response to induction chemotherapy in locally advanced nasopharyngeal carcinoma. Front Immunol 2024; 15:1415246. [PMID: 38911859 PMCID: PMC11191375 DOI: 10.3389/fimmu.2024.1415246] [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: 04/10/2024] [Accepted: 05/24/2024] [Indexed: 06/25/2024] Open
Abstract
Purpose To investigate the treatment response and toxicity of the combination of induction chemotherapy (IC) and PD-1 inhibitor in locally advanced nasopharyngeal carcinoma (LANPC). Methods Patients with stage III-IVA NPC who received IC or IC + PD-1 inhibitor were included. The chi-square test and multivariate logistic regression analysis were used for statistical analysis. Results A total of 225 patients were identified, including 193 (85.8%) and 32 (14.2%) who received IC alone and IC + PD-1 inhibitor, respectively. The addition of PD-1 inhibitor to IC significantly improved the tumor response than those treated with IC alone. The complete response (CR), partial response, stable disease, and progressive disease rates of 4.7% vs. 31.3%, 69.4% vs. 62.5%, 24.9% vs. 6.3%, and 1.0% vs. 0% in patients receiving IC alone and IC + PD-1 inhibitor, respectively (P<0.001). The results of the multivariate logistic regression showed that receiving PD-1 inhibitor was an independent predictor influencing the CR rate of patients (odds ratio 9.814, P<0.001). The most common toxicity by using IC and PD-1 inhibitor was hematological toxicity. In terms of non-hematological toxicity, 7 (21.9%) patients experienced thyroid dysfunction and all of them were hyperthyroidism. No grade 5 toxicities were found. In those who received IC and PD-1 inhibitor, the one-year locoregional recurrence-free survival, distant metastasis-free survival, disease-free survival, and overall survival were 100%, 96.9%, 96.9%, and 100%, respectively. Conclusion The addition of PD-1 inhibitor to IC has promise as an effective treatment approach for LANPC. More studies are expected to provide further insights into the optimal use of this treatment strategy, paving the way for more personalized and effective treatment options for patients with LANPC.
Collapse
Affiliation(s)
- Yi-Feng Yu
- Department of Radiation Oncology, Xiamen Cancer Quality Control Center, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Guan-Zhong Lu
- Department of Radiation Oncology, Xiamen Cancer Quality Control Center, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Run-Jie Wang
- Department of Radiation Oncology, Xiamen Cancer Quality Control Center, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yu-Kun Song
- Department of Radiology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - San-Gang Wu
- Department of Radiation Oncology, Xiamen Cancer Quality Control Center, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
43
|
An Z, He L, Chen T, Liang B, Wu Q. The efficacy and safety of EGFR-TKI in recurrent/metastatic nasopharyngeal carcinoma patients: A systematic review and meta-analysis. Laryngoscope Investig Otolaryngol 2024; 9:e1279. [PMID: 38803463 PMCID: PMC11129551 DOI: 10.1002/lio2.1279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024] Open
Abstract
Objectives EGFR-tyrosine kinase inhibitor (TKI) is used to treat recurrent and metastatic nasopharyngeal carcinoma (rmNPC). This meta-analysis aims to study the efficacy and safety of EGFR-TKI in treating patients with rmNPC. Methods We conducted a systematic search of PubMed, Embase, and Web of Science up to November 2023, and included literature that met the criteria. We extracted objective response rate (ORR), disease control rate (DCR), median progression-free survival (mPFS), median overall survival (mOS), and adverse reaction-related events and performed meta-analysis using Stata 14.0. Results A total of nine articles were included. The summary results showed that the ORR for patients treated with EGFR-TKI for rmNPC was 38% (95% CI = 27%-49%), the DCR was 71% (95% CI = 61%-80%), the mPFS was 6.29 months (95% CI = 5.22-7.35), and the mOS was 15.94 months (95% CI = 14.68-17.20). The most common grade 3-4 adverse reaction events in these patients were mucositis, nasopharyngeal necrosis, and oral ulceration. We found an incidence rate of 49% (95% CI = 38%-61%) for grade 3-4 adverse events (AEs). The anti-PD1 combined with TKI treatment method is more effective than the EGFR-TKI alone for treating rmNPC. Conclusion The study shows that EGFR-TKI has good efficacy in treating rmNPC but does not translate into survival benefits and owns a high incidence of grade 3-4 AEs. More RCT trials are needed in the future to verify the efficacy of anti-PD1 combined with TKI treatment method.
Collapse
Affiliation(s)
- Zeqi An
- Department of Otorhinolaryngology Head and Neck SurgeryShenzhen University General HospitalShenzhenChina
| | - Libin He
- Department of Otorhinolaryngology Head and Neck SurgeryShenzhen University General HospitalShenzhenChina
| | - Tuo Chen
- Department of Otorhinolaryngology Head and Neck SurgeryShenzhen University General HospitalShenzhenChina
| | - Bosen Liang
- Department of Otorhinolaryngology Head and Neck SurgeryShenzhen University General HospitalShenzhenChina
| | - Qiang Wu
- Department of Otorhinolaryngology Head and Neck SurgeryShenzhen University General HospitalShenzhenChina
| |
Collapse
|
44
|
Fang X, Tong W, Wu S, Zhu Z, Zhu J. The role of intratumoral microorganisms in the progression and immunotherapeutic efficacy of head and neck cancer. ONCOLOGIE 2024; 26:349-360. [DOI: 10.1515/oncologie-2023-0511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Abstract
The effectiveness of cancer immunization is largely dependent on the tumor’s microenvironment, especially the tumor immune microenvironment. Emerging studies say microbes exist in tumor cells and immune cells, suggesting that these microbes can affect the state of the immune microenvironment of the tumor. Our comprehensive review navigates the intricate nexus between intratumoral microorganisms and their role in tumor biology and immune modulation. Beginning with an exploration of the historical acknowledgment of microorganisms within tumors, the article underscores the evolution of the tumor microenvironment (TME) and its subsequent implications. Using findings from recent studies, we delve into the unique bacterial compositions across different tumor types and their influence on tumor growth, DNA damage, and immune regulation. Furthermore, we illuminate the potential therapeutic implications of targeting these intratumoral microorganisms, emphasizing their multifaceted roles from drug delivery agents to immunotherapy enhancers. As advancements in next-generation sequencing (NGS) technology redefine our understanding of the tumor microbiome, the article underscores the importance of discerning their precise role in tumor progression and tailoring therapeutic interventions. The review culminates by emphasizing ongoing challenges and the pressing need for further research to harness the potential of intratumoral microorganisms in cancer care.
Collapse
Affiliation(s)
- Xuzhe Fang
- The Fourth School of Clinical Medicine , Zhejiang Chinese Medical University , Hangzhou , China
| | - Weihong Tong
- The Fourth School of Clinical Medicine , Zhejiang Chinese Medical University , Hangzhou , China
| | - Sheng Wu
- The Fourth School of Clinical Medicine , Zhejiang Chinese Medical University , Hangzhou , China
| | - Zhengyong Zhu
- The Fourth School of Clinical Medicine , Zhejiang Chinese Medical University , Hangzhou , China
| | - Jin Zhu
- Department of Otorhinolaryngology and Head Neck Surgery, Affiliated Hangzhou First People’s Hospital , Zhejiang University School of Medicine , Hangzhou , China
| |
Collapse
|
45
|
王 媛, 陈 腾, 从 小, 李 依, 陈 蕊, 张 配, 孙 小, 赵 素. [Pristimerin enhances cisplatin-induced apoptosis in nasopharyngeal carcinoma cells via ROS-mediated deactivation of the PI3K/AKT signaling pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:904-912. [PMID: 38862448 PMCID: PMC11166717 DOI: 10.12122/j.issn.1673-4254.2024.05.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Indexed: 06/13/2024]
Abstract
OBJECTIVE To explore the effect of pristimerin combined with cisplatin on proliferation and apoptosis of nasopharyngeal carcinoma cells. METHODS CCK-8 assay was used to examine the survival rate of HNE-1 and CNE-2Z cells following treatment for 24 h with different concentrations of pristimerin, cisplatin or their combination. The changes in colony formation ability, apoptosis, and intracellular reactive oxygen species (ROS) levels of the treated cells were analyzed using colony formation assay and flow cytometry. Western blotting was performed to detect the changes in protein expressions in the cells. The effects of pre-treatment with NAC on proliferation, apoptosis, and PI3K/AKT signaling pathway were observed in pristimerin- and/or cisplatin-treated cells. RESULTS Both pristimerin and cisplatin significantly lowered the survival rate of HNE-1 and CNE-2Z cells (P < 0.05). Compared with pristimerin or cisplatin alone, their combination more strongly inhibited survival and colony formation ability of the cells, increased cell apoptosis rate and intracellular ROS levels, upregulated the protein expressions of Bax, cleaved caspase-3, and cleaved PARP, and downregulated the protein expressions of Bcl-2, Mcl-1, PARP and p-PI3K and p-AKT (P < 0.05). NAC pretreatment significantly attenuated proliferation inhibition and apoptosis-promoting effects of pristimerin combined with cisplatin, and partially restored the downregulated protein expressions of p-PI3K and p-AKT in HNE-1 and CNE-2Z cells with the combined treatment (P < 0.05). CONCLUSION Pristimerin can enhance cisplatin-induced proliferation inhibition and apoptosis in nasopharyngeal carcinoma cells, the mechanism of which may involve ROS-mediated deactivation of the PI3K/AKT signaling pathway.
Collapse
|
46
|
Li S, Dai W, Kam NW, Zhang J, Lee VHF, Ren X, Kwong DLW. The Role of Natural Killer Cells in the Tumor Immune Microenvironment of EBV-Associated Nasopharyngeal Carcinoma. Cancers (Basel) 2024; 16:1312. [PMID: 38610990 PMCID: PMC11011204 DOI: 10.3390/cancers16071312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/23/2024] [Accepted: 03/23/2024] [Indexed: 04/14/2024] Open
Abstract
Endemic nasopharyngeal carcinoma (NPC) is closely associated with the Epstein-Barr virus (EBV), which contributes to tumor development and influences the tumor immune microenvironment (TIME) in NPC. Natural killer (NK) cells, as part of the innate immune system, play a crucial role in responding to viral infections and malignant cell transformations. Notably, NK cells possess a unique ability to target tumor cells independent of major histocompatibility complex class I (MHC I) expression. This means that MHC I-deficient tumor cells, which can escape from effective T cell attack, are susceptible to NK-cell-mediated killing. The activation of NK cells is determined by the signals generated through inhibitory and activating receptors expressed on their surface. Understanding the role of NK cells in the complex TIME of EBV+ NPC is of utmost importance. In this review, we provide a comprehensive summary of the current understanding of NK cells in NPC, focusing on their subpopulations, interactions, and cytotoxicity within the TIME. Moreover, we discuss the potential translational therapeutic applications of NK cells in NPC. This review aims to enhance our knowledge of the role of NK cells in NPC and provide valuable insights for future investigations.
Collapse
Affiliation(s)
- Shuzhan Li
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; (S.L.); (J.Z.)
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin 300060, 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 999077, China; (W.D.); (N.-W.K.); (V.H.F.L.)
| | - 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 999077, China; (W.D.); (N.-W.K.); (V.H.F.L.)
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, New Territories, Hong Kong 999077, China
| | - Jiali Zhang
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; (S.L.); (J.Z.)
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin 300060, China
| | - Victor H. F. Lee
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China; (W.D.); (N.-W.K.); (V.H.F.L.)
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Xiubao Ren
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; (S.L.); (J.Z.)
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin 300060, China
| | - Dora Lai-Wan Kwong
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China; (W.D.); (N.-W.K.); (V.H.F.L.)
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| |
Collapse
|
47
|
Wu Y, Jia Q, Tang Q, Deng H, He Y, Tang F. Berberine-mediated Ferroptosis through System Xc -/GSH/GPX4 Axis Inhibits Metastasis of Nasopharyngeal Carcinoma. J Cancer 2024; 15:685-698. [PMID: 38213727 PMCID: PMC10777030 DOI: 10.7150/jca.90574] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/23/2023] [Indexed: 01/13/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant tumor that is highly prevalent in Southeast China, and its metastasis remains an unresolved clinical problem. Ferroptosis, a type of nonapoptotic cell death, is a critical pathway in tumor metastasis. Berberine (BBR), a plant alkaloid, has been explored as a potential anti-NPC metastatic agent; however, the underlying mechanisms are unknown. Here, we showed that BBR exerted its anti-metastasis role by inhibiting system Xc-/GSH/GPX4 axis-driven ferroptosis. The present study demonstrated for the first time that BBR induced ferroptosis in NPC cells by increasing reactive oxygen species, lipid peroxidation and cellular Fe2+ and that the ferroptosis inhibitors Ferrostatin-1 and Deferoxamine mesylate rescued BBR-induced NPC cell death. Moreover, the ferroptotic characteristics of BBR-treated NPC cells were observed using transmission electron microscopy. Mechanistically, system Xc- (SLC7A11 and SLC3A2) and GSH levels were found to be suppressed after treatment with BBR. We demonstrated that the system Xc-/GSH/GPX4 axis was a critical mediator of BBR-induced ferroptosis. Furthermore, GPX4, a key inhibitor of lipid peroxidation, was greatly suppressed by BBR at both protein and mRNA levels. Molecular docking results showed a strong interaction between GPX4 and BBR. Notably, GPX4 overexpression reversed the effect of BBR-induced ferroptosis in NPC cells. Finally, BBR-mediated inhibition of NPC metastasis was validated in vivo using a mouse model. Taken together, our data suggest that BBR induced ferroptosis of NPC cells via suppressing the system Xc-/GSH/GPX4 axis, provides new insights into the mechanism of BBR anti-NPC metastasis.
Collapse
Affiliation(s)
- Yao Wu
- The First Clinical College of Traditional Chinese Medicine of Hunan University of Chinese Medicine, and Hunan Cancer Hospital, Changsha, 410007, China
- Hunan Key Laboratory of Oncotarget Gene and Clinical Laboratory, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha 410013, China
| | - Qunying Jia
- Hunan Key Laboratory of Oncotarget Gene and Clinical Laboratory, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha 410013, China
| | - Qi Tang
- The First Clinical College of Traditional Chinese Medicine of Hunan University of Chinese Medicine, and Hunan Cancer Hospital, Changsha, 410007, China
| | - Hongyu Deng
- Hunan Key Laboratory of Oncotarget Gene and Clinical Laboratory, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha 410013, China
| | - Yingchun He
- The First Clinical College of Traditional Chinese Medicine of Hunan University of Chinese Medicine, and Hunan Cancer Hospital, Changsha, 410007, China
| | - Faqing Tang
- The First Clinical College of Traditional Chinese Medicine of Hunan University of Chinese Medicine, and Hunan Cancer Hospital, Changsha, 410007, China
- Hunan Key Laboratory of Oncotarget Gene and Clinical Laboratory, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/ Hunan Cancer Hospital, Changsha 410013, China
| |
Collapse
|
48
|
Zou Z, Li R, Huang X, Chen M, Tan J, Wu M. Identification and validation of immune-related methylated genes as diagnostic and prognostic biomarkers of nasopharyngeal carcinoma. Head Neck 2024; 46:192-211. [PMID: 37929674 DOI: 10.1002/hed.27569] [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: 02/16/2023] [Revised: 09/26/2023] [Accepted: 10/27/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a common malignancy occurring in the head and neck. Identification of immune-related methylated biomarkers might be helpful for NPC detection and prognostic evaluation. METHODS A co-methylation network based on WGCNA was constructed to identify modules associated with NPC and immune cells. In combination with differentially expressed genes (DEGs) and immune-related genes from ImmPort database, the candidate immune-related methylated genes (IRMGs) were obtained. RESULTS Our combined analysis identified 12 IRMGs. Among them, both the methylation and mRNA expression of CCL28, CSK, and PRKCB were correlated with the infiltration of B cells. CD1D, CR2, and GDF10 were favorable markers. Demethylation experiments validated that downregulation of GDF10, PRKCB, SLC40A1, and TGFBR3 in NPC resulted from promoter hypermethylation. Additionally, a diagnostic model was developed and exhibited high discriminative accuracy. CONCLUSIONS These results provided a group of immune-related methylated biomarkers that may help with the diagnosis and prognosis of NPC.
Collapse
Affiliation(s)
- Zhenning Zou
- Department of Pathology, Guangdong Medical University, Zhanjiang, China
| | - Rujia Li
- Department of Pathology, Guangdong Medical University, Zhanjiang, China
| | - Xueshan Huang
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, China
| | - Mei Chen
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, China
| | - Jingyi Tan
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Minhua Wu
- School of Basic Medicine, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
49
|
Lv M, Ding Y, Zhang Y, Liu S. Targeting EBV-encoded products: Implications for drug development in EBV-associated diseases. Rev Med Virol 2024; 34:e2487. [PMID: 37905912 DOI: 10.1002/rmv.2487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/02/2023] [Accepted: 10/22/2023] [Indexed: 11/02/2023]
Abstract
Epstein-Barr virus, a human gamma-herpesvirus, has a close connection to the pathogenesis of cancers and other diseases, which are a burden for public health worldwide. So far, several drugs or biomolecules have been discovered that can target EBV-encoded products for treatment, such as Silvestrol, affinity toxin, roscovitine, H20, H31, curcumin, thymoquinone, and ribosomal protein L22. These drugs activate or inhibit the function of some biomolecules, affecting subsequent signalling pathways by acting on the products of EBV. These drugs usually target LMP1, LMP2; EBNA1, EBNA2, EBNA3; EBER1, EBER2; Bam-HI A rightward transcript and BHRF1. Additionally, some promising findings in the fields of vaccines, immunological, and cellular therapies have been established. In this review, we mainly summarise the function of drugs mentioned above and unique mechanisms, hoping that we can help giving insight to the design of drugs for the treatment of EBV-associated diseases.
Collapse
Affiliation(s)
- Mengwen Lv
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Pathogenic Biology, Qingdao University Medical College, Qingdao, China
| | - Yuan Ding
- Department of Special Examination, Qingdao Women & Children Hospital, Qingdao, China
| | - Yan Zhang
- Department of Pathogenic Biology, Qingdao University Medical College, Qingdao, China
- Department of Clinical Laboratory, Zibo Central Hospital, Zibo, China
| | - Shuzhen Liu
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|