1
|
Xie W, Wei S, Feng C, Fu Y, Zhang Z, Dai S, Zhang C, Zhao L, Shan B. IFI30 Knockdown Inhibits ESCC Progression by Promoting Apoptosis and Senescence via Activation of JNK and P21/P16 Pathways. Thorac Cancer 2025; 16:e70063. [PMID: 40186402 PMCID: PMC11971534 DOI: 10.1111/1759-7714.70063] [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: 11/04/2024] [Revised: 03/11/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a prevalent and deadly cancer, making it essential to understand the molecular mechanisms influencing its development and prognosis. The role of interferon-gamma-inducible protein 30 (IFI30) in antigen processing is well-established, but its impact on the progression of ESCC remains unclear. This study aimed to investigate the biological function and potential mechanisms of IFI30 in ESCC progression. METHODS Public databases, proteomics, and immunohistochemistry (IHC) were employed to analyze IFI30 expression. Cell proliferation, migration, and invasion were evaluated using MTS, colony formation, wound healing, and transwell assays. Nude mouse xenograft models were established to assess the effects of IFI30 knockdown in vivo. Quantitative proteomics was utilized to identify differentially expressed proteins (DEPs) and pathways altered by IFI30 knockdown. Cell apoptosis and senescence were evaluated by flow cytometry, SA-β-gal staining, and reactive oxygen species (ROS) analysis. RESULTS IFI30 was highly expressed in ESCC and was correlated with advanced stage and poor prognosis. IFI30 knockdown inhibited ESCC cell proliferation, migration, and invasion in vitro and suppressed tumor growth in vivo. DEPs were mainly enriched in biological pathways related to apoptosis, mitophagy, cellular senescence, and lysosome. Furthermore, IFI30 knockdown in ESCC cells upregulated HRAS expression, increased ROS production, activated the JNK signaling pathway, and elevated the expression of P16 and P21, thereby promoting apoptosis and senescence. CONCLUSIONS This study suggests that IFI30 may regulate the JNK and P21/P16 pathways, exerting pro-tumorigenic effects in ESCC. IFI30 could serve as a potential novel target for ESCC treatment.
Collapse
Affiliation(s)
- Wenyao Xie
- Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
- Department of OncologyHandan Central HospitalHandanChina
| | - Sisi Wei
- Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
- Key Laboratory of Tumor Prevention and Precision Diagnosis and Treatment of Hebei, Clinical Oncology Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebei ProvinceChina
| | - Caiting Feng
- Department of Thoracic SurgeryHandan First HospitalHandanChina
| | - Yuhui Fu
- Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
- Key Laboratory of Tumor Prevention and Precision Diagnosis and Treatment of Hebei, Clinical Oncology Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebei ProvinceChina
| | - Zhe Zhang
- Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
- Key Laboratory of Tumor Prevention and Precision Diagnosis and Treatment of Hebei, Clinical Oncology Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebei ProvinceChina
| | - Suli Dai
- Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
- Key Laboratory of Tumor Prevention and Precision Diagnosis and Treatment of Hebei, Clinical Oncology Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebei ProvinceChina
| | - Cong Zhang
- Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
- Key Laboratory of Tumor Prevention and Precision Diagnosis and Treatment of Hebei, Clinical Oncology Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebei ProvinceChina
| | - Lianmei Zhao
- Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
- Key Laboratory of Tumor Prevention and Precision Diagnosis and Treatment of Hebei, Clinical Oncology Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebei ProvinceChina
| | - Baoen Shan
- Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
- Key Laboratory of Tumor Prevention and Precision Diagnosis and Treatment of Hebei, Clinical Oncology Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebei ProvinceChina
| |
Collapse
|
2
|
Yi Z, Li X, Li Y, Wang R, Zhang W, Wang H, Ji Y, Zhao J, Song J. Multi-cohort validation based on a novel prognostic signature of anoikis for predicting prognosis and immunotherapy response of esophageal squamous cell carcinoma. Front Oncol 2025; 15:1530035. [PMID: 40165896 PMCID: PMC11955476 DOI: 10.3389/fonc.2025.1530035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Immunotherapy is recognized as an effective and promising treatment modality that offers a new approach to cancer treatment. However, identifying responsive patients remains challenging. Anoikis, a distinct form of programmed cell death, plays a crucial role in cancer progression and metastasis. Thus, we aimed to investigate prognostic biomarkers based on anoikis and their role in guiding immunotherapy decisions for esophageal squamous cell carcinoma (ESCC). By consensus clustering, the GSE53624 cohort of ESCC patients was divided into two subgroups based on prognostic anoikis-related genes (ARGs), with significant differences in survival outcomes between the two subgroups. Subsequently, we constructed an ARGs signature with four genes, and its reliability and accuracy were validated both internally and externally. Additional, different risk groups showed notable variances in terms of immunotherapy response, tumor infiltration, functional enrichment, immune function, and tumor mutation burden. Notably, the effectiveness of the signature in predicting immunotherapy response was confirmed across multiple cohorts, including GSE53624, GSE53625, TCGA-ESCC, and IMvigor210, highlighting its potential utility in predicting immunotherapy response. In conclusion, the ARGs signature has the potential to serve as an innovative and dependable prognostic biomarker for ESCC, facilitating personalized treatment strategies in this field, and may represent a valuable new tool for guiding ESCC immunotherapy decision-making.
Collapse
Affiliation(s)
- Zhongquan Yi
- Department of Central Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, China
| | - Xia Li
- Department of General Medicine, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, China
| | - Yangyang Li
- Department of Cardiothoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Rui Wang
- Department of Cardiothoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Weisong Zhang
- Department of Cardiothoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Hao Wang
- Department of Cardiothoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Yanan Ji
- Department of Central Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, China
| | - Jing Zhao
- Department of Central Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, China
| | - JianXiang Song
- Department of Cardiothoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| |
Collapse
|
3
|
Zheng J, Huang B, Chen Y, Zeng B, Xiao L, Wu M. Exploratory analyses of the associations between Ki-67 expression, lymph node metastasis, and prognosis in patients with esophageal squamous cell cancer. PeerJ 2025; 13:e19062. [PMID: 40028218 PMCID: PMC11871893 DOI: 10.7717/peerj.19062] [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: 09/06/2024] [Accepted: 02/06/2025] [Indexed: 03/05/2025] Open
Abstract
Background The relationships between Ki-67/MKI67 expression, lymph node metastasis (LNM), vascular invasion (VI), and perineural invasion (PI) in esophageal squamous cell cancer (ESCC) remain unclear. This retrospective cohort study was performed to evaluate the prognostic value of Ki-67 expression and its association with LNM in patients with resected ESCC. Methods The analysis included 168 patients with ESCC with available Ki-67 protein expression data. The patients were divided into Ki-67 high-expression group (Ki-67 High, 93 cases) and Ki-67 low-expression (Ki-67 Low, 75 cases) groups. Associations between Ki-67 expression and ESCC pathological features was assessed using chi-square test. Overall survival (OS) was compared between the two groups using Kaplan-Meier survival analysis and Cox proportional hazards model. Results Median follow-up duration was 33.5 months (range 3.0-60.0 months). High Ki-67 expression was significantly associated with poor OS in patients with ESCC compared to that of the low-expression in both univariate (hazard ratios (HR) = 3.42, 95% CI [2.22-5.27], P < 0.001) and multivariate analyses (HR = 1.98, 95% CI [1.33-2.94], P < 0.001). Furthermore, high Ki-67 expression was significantly associated with an increased risk of LNM (χ 2 = 11.219, P = 0.011), VI (χ 2 = 6.359, P = 0.012), and PI (χ 2 = 8.877, P = 0.003). Conclusions High Ki-67 protein expression is associated with poor prognosis in ESCC. Increased Ki-67 expression significantly increases the risk of LNM, VI, and PI in ESCC, and thus may serve as an indication for adjuvant therapy in ESCC management.
Collapse
Affiliation(s)
- Jianqing Zheng
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Bifen Huang
- Department of Obstetrics and Gynecology, Quanzhou Medical College People’s Hospital Affiliated, Quanzhou, Fujian, China
| | - Ying Chen
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Bingwei Zeng
- Department of Pathology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Lihua Xiao
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Min Wu
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
4
|
Christodoulidis G, Agko SE, Koumarelas KE, Kouliou MN, Zacharoulis D. Advancements and challenges in the treatment of esophageal cancer: A comprehensive review. World J Clin Oncol 2024; 15:1463-1467. [PMID: 39720647 PMCID: PMC11514372 DOI: 10.5306/wjco.v15.i12.1463] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/23/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024] Open
Abstract
Esophageal cancer (EC) is an aggressive malignancy with a poor prognosis, ranking seventh in incidence and sixth cancer-related deaths globally. EC is classified in two main types, the esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC), with ESCC being more common in Eastern Europe, South Asia, and Africa, while EAC is prevalent in Western Europe and North America. Molecular analysis identifies three subgroups of ESCC, each with distinct genetic mutations and treatment responses. Early-stage EC is often difficult to detect, leading to late-stage diagnoses that necessitate systemic drug therapies, including molecular-targeted therapies and immunotherapies. Immunotherapy, particularly immune checkpoint inhibitor, has shown promising results in improving survival rates for metastatic or persistent EC. It is particularly important to target to multidisciplinary combination therapies, integrating surgery, chemoradiotherapy, targeted therapy and immunotherapy. Additionally, radioimmunotherapy is being explored for its potential to enhance treatment efficacy, especially in advanced and metastatic tumors. However, the pathological complete response rate to neoadjuvant chemoradiotherapy remains suboptimal, highlighting the need for novel treatment strategies. Future research should focus on optimizing treatment combinations and identifying predictive biomarkers to improve clinical outcomes for EC patients.
Collapse
Affiliation(s)
| | - Sara Eirini Agko
- Intensive Care Unit, Asklepios Paulinen Clinic Wiesbaden, Wiesbaden 65197, Hesse, Germany
| | | | - Marina Nektaria Kouliou
- Department of General Surgery, University Hospital of Larissa, Larisa 41110, Thessalía, Greece
| | - Dimitris Zacharoulis
- Department of Surgery, University Hospital of Larissa, Larisa GR41334, Thessalía, Greece
| |
Collapse
|
5
|
Yang Y, Li S, Shi W, Jin G, Guo D, Li A, Wang B, Lu B, Feng S. Pterostilbene suppresses the growth of esophageal squamous cell carcinoma by inhibiting glycolysis and PKM2/STAT3/c-MYC signaling pathway. Int Immunopharmacol 2024; 142:113247. [PMID: 39321706 DOI: 10.1016/j.intimp.2024.113247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
Pterostilbene (PTS) is a dietary phytochemical that has shown antitumor activity in many types of cancer, but the molecular mechanism remains unclear. It has also not been adequately studied on PTS against esophageal squamous cell carcinoma (ESCC). Thus, this study investigated the effect of PTS on ESCC in vitro and in vivo and explored the underlying molecular mechanism. We found that PTS can inhibit the proliferation, colony formation, and migration of ESCC cells. According to the bioinformatics analysis of proteomics, PTS had a great influence on the metabolic process of ESCC cells. KEGG analysis showed that PTS down-regulated the pyruvate metabolism pathway. Moreover, PTS can inhibit the PK activity, glucose consumption, and lactate production in ESCC cells. By administration of PTS into xenograft mice, experiment results demonstrated that PTS can suppress tumor progress and the PKM2/STAT3/c-MYC signaling pathway. We found that PTS inhibited the PKM2/STAT3/c-MYC signaling pathway by targeting PKM2 in ESCC cells. Collectively, this study revealed that PTS inhibited ESCC growth by suppressing PKM2 mediated aerobic glycolysis and PKM2/STAT3/c-MYC signaling pathway, which enriching the anti-tumor molecular mechanism of PTS and providing a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Yi Yang
- Medical College, Henan University of Chinese Medicine, Zhengzhou 450046 China; Henan Engineering Research Center for Chinese Medicine Foods for Special Medical Purpose, Zhengzhou 450046 China
| | - Shan Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou 450046 China; Henan Engineering Research Center for Chinese Medicine Foods for Special Medical Purpose, Zhengzhou 450046 China
| | - Wenjie Shi
- The Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou 450046 China
| | - Guoguo Jin
- Henan Key Laboratory of Chronic Disease Management, Fuwai Central China Cardiovascular Hospital, Zhengzhou 450018 China
| | - Dandan Guo
- Medical College, Henan University of Chinese Medicine, Zhengzhou 450046 China; Henan Engineering Research Center for Chinese Medicine Foods for Special Medical Purpose, Zhengzhou 450046 China
| | - Aifang Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou 450046 China; Henan Engineering Research Center for Chinese Medicine Foods for Special Medical Purpose, Zhengzhou 450046 China
| | - Baiyan Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou 450046 China; Henan Engineering Research Center for Chinese Medicine Foods for Special Medical Purpose, Zhengzhou 450046 China
| | - Baoping Lu
- Henan Engineering Research Center for Chinese Medicine Foods for Special Medical Purpose, Zhengzhou 450046 China; The Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou 450046 China.
| | - Shuying Feng
- Medical College, Henan University of Chinese Medicine, Zhengzhou 450046 China; Henan Engineering Research Center for Chinese Medicine Foods for Special Medical Purpose, Zhengzhou 450046 China.
| |
Collapse
|
6
|
Wang Y, Xue L. Decoding the role of FOXP3 in esophageal cancer: Underlying mechanisms and therapeutic implications. Biochim Biophys Acta Rev Cancer 2024; 1879:189211. [PMID: 39532205 DOI: 10.1016/j.bbcan.2024.189211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/05/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Esophageal cancer is a significant contributor to cancer-related mortality, and its poor prognosis is primarily attributed to the aggressive nature of the tumor and challenges in early detection. Currently, there are no ideal drugs developed for treatment, making it crucial to explore potential biomarkers and molecular targets for esophageal cancer. FOXP3, as a transcription factor and major regulator of regulatory T cells, not only plays a role in promoting or inhibiting tumor development in various types of cancer cells including esophageal cancer cells but also influences the function of Treg cells by regulating the expression of multiple genes. This paper provides an in-depth discussion on the functional properties, regulatory mechanisms, key signaling pathways, as well as the role and potential application of FOXP3 in treating esophageal cancer. Furthermore, it comprehensively analyzes the complex role of this transcription factor within the tumor immune microenvironment with an aim to aid in developing new potential targets for esophageal cancer treatment.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Thoracic Surgery, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Military Medical University, 200003 Shanghai, China.
| | - Lei Xue
- Department of Thoracic Surgery, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Military Medical University, 200003 Shanghai, China.
| |
Collapse
|
7
|
Zhang WY, Chang YJ, Shi RH. Artificial intelligence enhances the management of esophageal squamous cell carcinoma in the precision oncology era. World J Gastroenterol 2024; 30:4267-4280. [PMID: 39492825 PMCID: PMC11525855 DOI: 10.3748/wjg.v30.i39.4267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/31/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most common histological type of esophageal cancer with a poor prognosis. Early diagnosis and prognosis assessment are crucial for improving the survival rate of ESCC patients. With the advancement of artificial intelligence (AI) technology and the proliferation of medical digital information, AI has demonstrated promising sensitivity and accuracy in assisting precise detection, treatment decision-making, and prognosis assessment of ESCC. It has become a unique opportunity to enhance comprehensive clinical management of ESCC in the era of precision oncology. This review examines how AI is applied to the diagnosis, treatment, and prognosis assessment of ESCC in the era of precision oncology, and analyzes the challenges and potential opportunities that AI faces in clinical translation. Through insights into future prospects, it is hoped that this review will contribute to the real-world application of AI in future clinical settings, ultimately alleviating the disease burden caused by ESCC.
Collapse
Affiliation(s)
- Wan-Yue Zhang
- School of Medicine, Southeast University, Nanjing 221000, Jiangsu Province, China
| | - Yong-Jian Chang
- School of Cyber Science and Engineering, Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Rui-Hua Shi
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing 210009, Jiangsu Province, China
| |
Collapse
|
8
|
Zhong W, Zhang Q. Atractylodin: An Alkyne Compound with Anticancer Potential. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1729-1757. [PMID: 39192675 DOI: 10.1142/s0192415x24500551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Atractylodin is one of the main active ingredients of Atractylodis Rhizoma. It has various pharmacological properties, such as antigastric ulcer, immune regulation, antibacterial, anti-inflammatory, antitumor, anti-oxidant, and neuroprotective properties. In the past few decades, atractylodin has attracted the attention of researchers due to its excellent therapeutic effects. This paper aims to review the pharmacology of atractylodin, focusing mainly on its pharmacological effects in tumor treatment. Atractylodin exerts its antitumor effect by regulating different signaling pathways to induce important biological events such as apoptosis, cell cycle arrest, and autophagy, inhibiting cancer cell invasion and metastasis. In the process of cell apoptosis, atractylodin mainly induces cancer cell apoptosis by downregulating the Notch signaling pathway, affecting multiple upstream and downstream targets. In addition, atractylodin induces autophagy in cancer cells by regulating various signaling pathways such as PI3K/AKT/mTOR, p38MAPK, and hypothalamic Sirt1 and p-AMPK. Atractylodin effectively induces G1/M and G2/M phase arrest under the action of multiple signaling pathways. Among them, the pathways related to G1/M are more widely stagnated. In inhibiting the migration and invasion of cancer cells, atractylodin mainly regulates the Wnt signaling pathway, downregulates the expression of N-cadherin in cancer cells, and then blocks the PI3K/AKT/mTOR signaling pathway, inhibiting the phosphorylation of PI3K, AKT, and mTOR proteins, thereby having a significant impact on the invasion and migration of cancer cells. This paper systematically reviews the research progress on the antitumor effects and mechanisms of atractylodin, hoping to provide a reference and theoretical basis for its clinical application and new drug development.
Collapse
Affiliation(s)
- Wenxia Zhong
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Qi Zhang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| |
Collapse
|
9
|
Ge R, Luan Z, Guo T, Xia S, Ye J, Xu J. The expression and biological role of complement C1s in esophageal squamous cell carcinoma. Open Life Sci 2024; 19:20220915. [PMID: 39071493 PMCID: PMC11282917 DOI: 10.1515/biol-2022-0915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 05/23/2024] [Accepted: 06/06/2024] [Indexed: 07/30/2024] Open
Abstract
The present work focused on investigating the role of the altered expression of complement C1s in proliferation and apoptosis of esophageal squamous cell carcinoma (ESCC) cells and explore its biological functions in ESCC, so as to lay a theoretical foundation and provide certain clinical reference for diagnosing and treating ESCC. Complement C1s expression within ESCC was assessed, and its clinical pathological characteristics in ESCC patients were analyzed. Subsequently, in vitro experiments were performed to further explore the mechanisms by which complement C1s affected ESCC. According to the results, complement C1s expression within ESCC markedly increased relative to adjacent non-cancerous samples. High C1s expression showed positive relation to race, residual lesion, and tumor location of ESCC patients. Complement C1s affected ESCC cell proliferation and apoptosis. Notably, C1s knockdown significantly inhibited ESCC cell proliferation and enhanced their apoptosis. C1s suppressed ESCC cell proliferation via Wnt1/β-catenin pathway and promoted their apoptosis through modulating the expression of Bcl2, Bax, and cleaved-caspase3.
Collapse
Affiliation(s)
- Ruomu Ge
- Central Laboratory, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, 225300, P.R. China
- Anhui Province Key Laboratory of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhengyun Luan
- Department of Clinical Laboratory, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, 225300, P.R. China
| | - Ting Guo
- Central Laboratory, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, 225300, P.R. China
| | - Sheng Xia
- School of Medicine, Jiangsu University School, Zhenjiang, Jiangsu, 212000, P.R. China
| | - Jun Ye
- Central Laboratory, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, 225300, P.R. China
| | - Jie Xu
- Central Laboratory, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, 225300, P.R. China
| |
Collapse
|
10
|
Wang X, Peng W, Zhao Y, Sha J, Li N, Huang S, Wang H. Immune cell related signature predicts prognosis in esophageal squamous cell carcinoma based on single-cell and bulk-RNA sequencing. Front Oncol 2024; 14:1370801. [PMID: 38903709 PMCID: PMC11187079 DOI: 10.3389/fonc.2024.1370801] [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: 01/15/2024] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
Background It has been reported that tumor immune microenvironment performs a vital role in tumor progress. However, acting mechanism of immune cell related genes (IRGs) in esophageal squamous cell carcinoma (ESCC) is uncertain. Methods TCGA-ESCC, GSE23400, GSE26886, GSE75241, and GSE196756 datasets were gained via public databases. First, differentially expressed genes (DEGs) between ESCC and control samples from GSE23400, GSE26886, and GSE75241 were screened out by differential expression analysis, and overlapping DEGs were identified. Single-cell transcriptome data of GSE196756 were applied to explore immune cells that might be involved in regulation of ESCC. Then, weighted gene co-expression network analysis was applied to screen IRGs. Next, differentially expressed IRGs (DE-IRGs) were identified by overlapping IRGs and DEGs, and were incorporated into univariate Cox, least absolute shrinkage and selection operator, and multivariate Cox to acquire prognosis-related genes, and ESCC samples were grouped into high-/low-risk groups on the basis of median risk score. Finally, the role of prognosis model in immunotherapy was analyzed. Results Totally 248 DEGs were yielded by overlapping 3,915 DEGs in GSE26886, 459 DEGs in GSE23400, and 1,641 DEGs in GSE75241. Single-cell analysis found that B cells, dendritic cells, monocytes, neutrophils, natural killer cells, and T cells were involved in ESCC development. Besides, MEred, MEblack, MEpink, MEblue and MEbrown modules were considered as key modules because of their highest correlations with immune cell subtypes. A total of 154 DE-IRGs were yielded by taking intersection of DEGs and genes in key modules. Moreover, CTSC, ALOX12, and RMND5B were identified as prognosis-related genes in ESCC. Obviously, Exclusion and TIDE scores were notably lower in high-risk group than in the other one, indicating that high-risk group was more responsive to immunotherapy. Conclusions Through bioinformatic analysis, we identified a prognosis model consisting of IRGs (CTSC, ALOX12, and RMND5B) in ESCC, providing new ideas for studies related to treatment and prognosis of ESCC.
Collapse
Affiliation(s)
- Xian Wang
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Wei Peng
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yali Zhao
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiming Sha
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Na Li
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shan Huang
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Gastroenterology, The Second People’s Hospital of Hefei, Hefei, China
| |
Collapse
|
11
|
Yan F, Chen L, Ying M, Li J, Fu Q. Efficacy and safety of pembrolizumab combined with albumin-bound paclitaxel and nedaplatin for advanced esophageal squamous cell carcinoma. Immunotherapy 2024; 16:305-317. [PMID: 38197157 DOI: 10.2217/imt-2023-0188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024] Open
Abstract
Objective: This research aimed to assess the efficacy and safety of pembrolizumab (PBL) combined with albumin-bound paclitaxel (ab-Pac) and nedaplatin (NDP) for advanced esophageal squamous cell carcinoma (ESCC). Methods: A total of 47 ESCC patients were administered PBL or NDP on day 1 and ab-Pac on days 1 and 8, every 21 days for one cycle. Tumor and toxicities were evaluated every two cycles and every cycle, respectively. Results: The objective response rate was 68.1% and the disease control rate was 100%. The median follow-up was 16.7 months; median progression-free and overall survival were 12.6 and 19.9 months, respectively. Conclusion: The combination of PBL with ab-Pac and NDP proved to be an effective and safe treatment regimen for advanced ESCC.
Collapse
Affiliation(s)
- Fang Yan
- Department of Oncology, Shanghai Changhai Hospital, No. 168 Changhai Road, Shanghai, 200433, China
| | - Longpei Chen
- Department of Oncology, Shanghai Changhai Hospital, No. 168 Changhai Road, Shanghai, 200433, China
| | - Mingzhen Ying
- Department of Oncology, Shanghai Changhai Hospital, No. 168 Changhai Road, Shanghai, 200433, China
| | - Jie Li
- Department of Oncology, Shanghai Changhai Hospital, No. 168 Changhai Road, Shanghai, 200433, China
| | - Qiang Fu
- Department of Oncology, Shanghai Changhai Hospital, No. 168 Changhai Road, Shanghai, 200433, China
| |
Collapse
|
12
|
Shahid MH, Mithany RH, Aslam S, Daniel N, Gerges F, Gill MU, Wanees A, Abdallah S, Abdelmaseeh M, Hannan A. Journey Through Words: Exploring Esophageal Cancer in Literature. Cureus 2023; 15:e48411. [PMID: 37954625 PMCID: PMC10637758 DOI: 10.7759/cureus.48411] [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] [Accepted: 11/06/2023] [Indexed: 11/14/2023] Open
Abstract
Esophageal cancer is a significant global health challenge, characterized by its aggressive nature and high mortality rates. The disease disproportionately affects males and ranks among the leading causes of cancer-related deaths worldwide. Alarming projections indicate that the prevalence of esophageal cancer is expected to surge by approximately 140% by the year 2025. This trend starkly contrasts with the anticipated decline in incidence observed for many other types of cancers. The cancer manifests primarily in two major subtypes: esophageal squamous cell carcinoma and adenocarcinoma, each with distinct epidemiological and biological characteristics. This review provides an in-depth exploration of the risk factors, anatomy, clinical presentation, diagnosis, staging, prognosis, treatment modalities, recurrence, advancements, and emerging therapies in esophageal cancer. Additionally, preventive and early detection strategies are discussed, focusing on primary, secondary, and tertiary prevention approaches. A comprehensive understanding of esophageal cancer is vital for formulating effective management strategies and improving patient outcomes.
Collapse
Affiliation(s)
| | - Reda H Mithany
- Laparoscopic Colorectal Surgery, Kingston Hospital National Health Service (NHS) Foundation Trust, Kingston, GBR
| | - Samana Aslam
- General Surgery, Lahore General Hospital, Lahore, PAK
| | - Nesma Daniel
- Medical Laboratory Science, Ain Shams University, Cairo, EGY
| | - Farid Gerges
- General and Emergency Surgery, Kingston Hospital National Health Service (NHS) Foundation Trust, Kingston, GBR
| | - Muhammad Umar Gill
- Accident and Emergency, King's College Hospital National Health Service (NHS) Foundation Trust, London, GBR
| | - Andrew Wanees
- General Surgery, Dar El-Salam General Hospital, Cairo, EGY
| | | | - Mark Abdelmaseeh
- General Surgery, Faculty of Medicine, Assuit University, Assuit, EGY
| | - Abdul Hannan
- Surgery, Glangwili General Hospital, Carmarthen, GBR
| |
Collapse
|
13
|
Zhao X, Zhang Z, Wen C, Huang J, Yang S, Liu J, Geng H, Peng B, Li Z, Zhang Y. The safety and anti-tumor effect of multiple peptides-pulsed dendritic cells combined with induced specific cytotoxic T lymphocytes for patients with solid tumors. Front Immunol 2023; 14:1284334. [PMID: 37942324 PMCID: PMC10628471 DOI: 10.3389/fimmu.2023.1284334] [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: 08/28/2023] [Accepted: 10/10/2023] [Indexed: 11/10/2023] Open
Abstract
Objective The aim of this study was to explore the safety and efficacy of multiple peptide-pulsed autologous dendritic cells (DCs) combined with cytotoxic T lymphocytes (CTLs) in patients with cancer. Methods Five patients diagnosed with cancer between November 2020 and June 2021 were enrolled and received DC-CTLs therapy. Peripheral blood was collected and antigenic peptides were analyzed. The phenotype and function of DC-CTLs and the immune status of patients were detected using flow cytometry or IFN-γ ELISPOT analysis. Results DCs acquired a mature phenotype and expressed high levels of CD80, CD86, CD83, and HLA-DR after co-culture with peptides, and the DC-CTLs also exhibited high levels of IFN-γ. Peripheral blood mononuclear cells from post-treatment patients showed a stronger immune response to peptides than those prior to treatment. Importantly, four of five patients maintained a favorable immune status, of which one patient's disease-free survival lasted up to 28.2 months. No severe treatment-related adverse events were observed. Conclusion Our results show that multiple peptide-pulsed DCs combined with CTLs therapy has manageable safety and promising efficacy for cancer patients, which might provide a precise immunotherapeutic strategy for cancer.
Collapse
Affiliation(s)
- Xuan Zhao
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China
| | - Zhen Zhang
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China
| | - Chunli Wen
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jianmin Huang
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China
| | - Shuangning Yang
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China
| | - Jinyan Liu
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China
| | - Huizhen Geng
- Hebei Bio-High Technology Development Co., LTD. Shijiazhuang, Hebei, China
| | - Bing Peng
- Hebei Bio-High Technology Development Co., LTD. Shijiazhuang, Hebei, China
| | - Zibo Li
- Hebei Bio-High Technology Development Co., LTD. Shijiazhuang, Hebei, China
| | - Yi Zhang
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|