1
|
Wang C, Zha YL, Wang H, Sun B, Qiang WG, Yuan Y, Shi HB, Hu WW. Carfilzomib promotes Iodine-125 seed radiation-induced apoptosis, paraptosis, and ferroptosis in esophageal squamous cell carcinoma by aggravating endoplasmic reticulum stress. Transl Oncol 2025; 57:102393. [PMID: 40315760 PMCID: PMC12098164 DOI: 10.1016/j.tranon.2025.102393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 02/09/2025] [Accepted: 04/07/2025] [Indexed: 05/04/2025] Open
Abstract
Iodine-125 (125I) seed brachytherapy has been applied to treat various malignant tumors such as esophageal cancer, however, radioresistance can reduce its efficacy. Endoplasmic reticulum stress (ERS) and subsequent unfolded protein response (UPR) is one of the core mechanisms of 125I seed radiation-induced cell death, thus aggravating ERS has been considered a promising sensitization strategy. Herein, we show that combination therapy of an irreversible proteasome inhibitor carfilzomib (CFZ) and 125I seed radiation displayed strong anti-tumor effect on esophageal squamous cell carcinoma (ESCC). Mechanistically, ERS and UPR regulated multiple cell death modalities induced by the combination therapy, including apoptosis, paraptosis, and ferroptosis. 125I seed radiation induced reactive oxygen species (ROS) production, DNA damage, p53 activation, and apoptosis. CFZ promoted ROS production, and augmented 125I seed radiation-induced apoptosis via the mitochondrial pathway, which was mediated by the UPR-C/EBP homologous protein (CHOP) pathway and was independent of the p53 pathway. CFZ enhanced 125I seed radiation-induced intracellular Ca2+ overload, protein ubiquitination, ERS, and UPR, consequently promoting paraptosis. 125I seed radiation induced accumulation of intracellular Fe2+ and lipid peroxides but upregulated the expression of ferroptosis inhibitors, SLC7A11 and glutathione peroxidase 4 (GPX4). The combination therapy promoted ferroptosis by enhancing the accumulation of intracellular Fe2+ and downregulating GPX4 expression. The mouse experiment demonstrated that CFZ can promote the efficacy of 125I seed radiation with good tolerance. Our findings suggest that combination therapy of 125I seed radiation and CFZ is associated with multiple cell death modalities and may serve as a promising therapeutic strategy for ESCC.
Collapse
Affiliation(s)
- Chao Wang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Yin-Lin Zha
- Department of Radiation Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Hao Wang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Bai Sun
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Wei-Guang Qiang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Ye Yuan
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Hong-Bing Shi
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China.
| | - Wen-Wei Hu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China.
| |
Collapse
|
2
|
Xing HJ, Hu MY, Jiang YQ, Li XH, Zhu B, Wang ZQ. Remission of type 2 diabetes one year after esophagectomy with gastric conduit reconstruction: A prospective cohort study. World J Gastrointest Surg 2025; 17:105514. [DOI: 10.4240/wjgs.v17.i6.105514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/22/2025] [Accepted: 04/29/2025] [Indexed: 05/30/2025] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) remission has been widely reported after bariatric surgery, but rarely reported after esophagectomy.
AIM To explore the incidence and predictors of T2D remission 1 year after esophagectomy with gastric conduit reconstruction.
METHODS In this prospective study, consecutive patients from 2 tertiary hospitals who had esophageal cancer and T2D and underwent esophagectomy with gastric conduit reconstruction were studied preoperatively and at 3 months, 6 months, and 12 months postoperatively. Remission of T2D is defined as glycated hemoglobin (HbA1c) values below 6.5% without glucose-lowering medications. Related clinical information were recorded and analyzed.
RESULTS A total of 187 patients were included. Of these patients, 24 (12.8%) discontinued antidiabetic drugs and maintained HbA1c values below 6.5% 1 year after surgery. At baseline, patients with T2D remission were younger (63.0 ± 5.2 years vs 67.0 ± 6.1 years, P = 0.002), had higher body mass index values (body weight 68.6 ± 11.1 kg vs 61.2 ± 9.3 kg, P = 0.001; body mass index 25.5 ± 2.4 kg/m2vs 23.8 ± 3 kg/m2, P = 0.011), shorter duration of T2D (4.9 ± 3.9 years vs 7.1 ± 3.7 years, P = 0.008) and higher preoperative HbA1c (8.5% ± 1.7% vs 7.7% ± 1.3%, P = 0.042). Multivariate logistic regression analysis showed that younger age and greater body weight were independent predictors of T2D remission after surgery.
CONCLUSION This study reveals a significant incidence of T2D remission after esophagectomy with gastric conduit reconstruction, and remission is more frequent in patients with younger age and greater body weight.
Collapse
Affiliation(s)
- Hua-Jie Xing
- Department of Thoracic Surgery, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Meng-Yu Hu
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yue-Quan Jiang
- Department of Thoracic Surgery, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Xin-Hua Li
- Department of Thoracic Surgery, People’s Hospital of Dali District, Dali 671000, Yunnan Province, China
| | - Bin Zhu
- Department of Thoracic Surgery, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Zhi-Qiang Wang
- Department of Thoracic Surgery, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
3
|
Feng X, Xu D, Xing Z, Zhang Q. Apatinib Mesylate Inhibits Cell Proliferation and the Metastasis of Esophageal Squamous Cell Carcinoma Through ERK/ELK-1/Snail Pathway. Cell Biochem Biophys 2025; 83:2201-2211. [PMID: 39709316 DOI: 10.1007/s12013-024-01631-z] [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: 09/15/2024] [Accepted: 11/19/2024] [Indexed: 12/23/2024]
Abstract
This study aimed to evaluate the impact of apatinib (APT) mesylate on the growth, migration ability, and underlying mechanisms in esophageal squamous cell carcinoma (ESCC) cell lines Kyse30 and Kyse150. Additionally, the anti-metastatic effects of APT mesylate were further validated in a nude mouse xenograft metastasis model. In vitro, APT mesylate treatment significantly reduced cell viability and migration ability in both cell lines in a dose- and time-dependent manner. Western blot analysis showed that APT mesylate inhibited the expression of proteins involved in the ERK/ELK-1/Snail signaling pathway, including ERK1/2, Snail, N-cadherin, and Vimentin, while upregulating E-cadherin expression. In vivo, APT mesylate administration notably decreased the number of pulmonary metastatic nodules in nude mice, with higher doses showing more pronounced effects. The 200 mg/kg high-dose group exhibited a significantly lower number of metastatic nodules compared to the cisplatin (CIS) group. The results suggest that APT mesylate inhibits ESCC cell proliferation and migration primarily by suppressing the ERK/ELK-1/Snail signaling pathway, which mediates epithelial-mesenchymal transition (EMT) and reduces metastasis and invasiveness. This study provides experimental evidence for the potential clinical application of APT mesylate in targeted therapy for ESCC, indicating its promising clinical value.
Collapse
Affiliation(s)
- Xiang Feng
- Department of Pharmacy, Dangtu People's Hospital, Ma'anshan, Anhui Province, China.
| | - Di Xu
- Department of Medical Equipment, Dangtu People's Hospital, Ma'anshan, Anhui Province, China
| | - Zhuqin Xing
- Department of Oncology, Dangtu People's Hospital, Ma'anshan, Anhui Province, China
| | - Qian Zhang
- Department of Pharmacy, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui Province, China.
| |
Collapse
|
4
|
Zhang X, Kang H, Li B, Xiong Y, Zheng S, Zhang D, Liu Y, Li S, Liu Y, Liu H, Gao Y, Ma L. Structural Optimization of 1,3-Diaryl-1,2,4-triazole-Capped Histone Deacetylase 6 Inhibitors to Obtain Novel Antiesophageal Cancer Candidates. J Med Chem 2025. [PMID: 40382720 DOI: 10.1021/acs.jmedchem.4c03231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Esophageal cancer, a leading global cancer, lacks effective therapies. Inhibition of histone deacetylase 6 (HDAC6) is a promising antitumor strategy, yet its role in esophageal cancer remains underexplored. Through structural optimization of our previously developed 1,3-diaryl-1,2,4-triazole-capped HDAC6 inhibitors, we identified compound 38k, exhibiting remarkably enhanced HDAC6 inhibition (IC50 = 3.12 nM) and 352-fold selectivity over HDAC1. Molecular docking analysis, CETSA, and BLI confirmed its strong HDAC6 binding. Moreover, 38k displayed robust in vitro and in vivo antiesophageal cancer efficacy, along with an advantageous pharmacokinetic and safety profile. Notably, combining 38k with a PI3K inhibitor synergistically enhanced the efficacy (75.02% tumor growth inhibition vs 50.94% monotherapy), likely by counteracting HDAC6 inhibition-induced PI3K/AKT activation. These findings validate HDAC6 as a therapeutic target and highlight 38k as a promising candidate for esophageal cancer treatment, particularly in combination regimens.
Collapse
Affiliation(s)
- Xinhui Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
- Newland Pharmaceutical Co., Ltd., Xuchang, Henan 461500, China
| | - Huiqin Kang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Bingqian Li
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yuhan Xiong
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Shuxian Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Di Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yuanfan Liu
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
| | - Shiyu Li
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
| | - Ying Liu
- The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou, Henan 450001, China
| | - Hongmin Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ya Gao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Liying Ma
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
- Key Laboratory of Cardio-cerebrovascular Drug, China Meheco Topfond Pharmaceutical Co., Zhumadian 463000, China
| |
Collapse
|
5
|
Wang M, Wang X, Liu Z, Shen H, Qi B, Fang M, Wang J, Ji Y, Zeng J. Recurrence patterns following neoadjuvant chemotherapy combined with immunotherapy for esophageal squamous cell carcinoma. Dis Esophagus 2025; 38:doaf034. [PMID: 40386863 DOI: 10.1093/dote/doaf034] [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: 03/03/2025] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/20/2025]
Abstract
This study aims to examine the recurrence patterns in patients with locally advanced esophageal squamous cell carcinoma who underwent surgery following neoadjuvant chemotherapy combined with immunotherapy. Retrospective analysis of patients with esophageal squamous cell carcinoma who received neoadjuvant Chemo-IO before surgery at Zhejiang Cancer Hospital between 2019 and 2023. The clinicopathological features, recurrence patterns, overall survival (OS), and disease-free survival (DFS) were analyzed, and the impact of postoperative adjuvant radiotherapy on prognosis was evaluated. In total, 422 patients were included in the study. After a median follow-up of 22 months, disease recurrence was observed in 89 (21.1%) patients, comprising 37 cases (8.8%) of locoregional recurrence, 30 cases (7.1%) of distant metastasis, and 22 cases (5.2%) of combined recurrence. Patients who achieved a pathologic complete response demonstrated significantly higher 3-year OS rates (90.0% vs. 72.5%; P = 0.01) and DFS rates (73.2% vs. 61.8%; P = 0.046). Univariable and multivariable analyses identified pathological lymph node staging (ypN0 vs. ypN+; HR: 1.73; 95% CI: 1.01-2.99; P = 0.047) as an independent prognostic factor for locoregional recurrence. Kaplan-Meier curves for OS and DFS demonstrated that postoperative radiotherapy (PORT) significantly improved OS and DFS in ypN+ patients after propensity score matching. Additionally, PORT significantly enhanced locoregional recurrence-free survival and distant recurrence-free survival in ypN+ patients. In patients receiving neoadjuvant Chemo-IO, locoregional recurrence is the predominant recurrence pattern. For ypN+ patients, PORT significantly improved survival outcomes. However, long-term outcomes require further investigation through randomized controlled trials.
Collapse
Affiliation(s)
- Miao Wang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xinyi Wang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhourong Liu
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Haixia Shen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Baojia Qi
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Min Fang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jin Wang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yongling Ji
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jian Zeng
- Department of Thoracic Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
6
|
Wang C, Ju C, Du D, Zhu P, Yin J, Jia J, Wang X, Xu X, Zhao L, Wan J, Sun T, Yang L, Li H, He F, Zhou M, He J. CircNF1 modulates the progression and immune evasion of esophageal squamous cell carcinoma through dual regulation of PD-L1. Cell Mol Biol Lett 2025; 30:37. [PMID: 40158127 PMCID: PMC11955112 DOI: 10.1186/s11658-025-00712-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 03/03/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Tumor immune escape is a pivotal gateway for esophageal squamous cell carcinoma (ESCC) development. Immune checkpoint-blocking therapies, represented by programmed cell death receptor-1/ligand 1 (PD-1/PD-L1) inhibitors, have achieved remarkable breakthroughs in ESCC treatment. However, not all patients with ESCC receive satisfactory clinical benefit. Therefore, identifying novel biomarkers for predicting the efficacy of immunotherapy in ESCC is of great importance. METHODS CircNF1 was screened from the circRNAs microarray, and its expression was measured by droplet digital polymerase chain reaction (ddPCR) and quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) assays in ESCC tissues and serum. Functional experiments were conducted to demonstrate the role of circNF1 in ESCC proliferation, metastasis, and tumor evasion. High-throughput RNA sequencing, chromatin immunoprecipitation (ChIP), co-immunoprecipitation (co-IP), and chromatin isolation by RNA purification-mass spectrometry (ChIRP-MS) were performed to clarify the underlying mechanisms of circNF1-mediated tumor progression. RESULTS The upregulation of circNF1 was closely associated with the response of anti-PD-L1 immunotherapy. Functionally, circNF1 promoted ESCC cell malignant phenotypes and regulated CD8+ T-cell-mediated antitumor immunity. Mechanistically, circNF1 drove the IL-6-induced oncogenic activation of the JAK-STAT3 pathway, which stimulated p-STAT3 binding of the promoter regions of PD-L1. Furthermore, circNF1 physically interacted with annexin A1 (ANXA1), blocking the ANXA1 deubiquitination induced by ubiquitin-specific protease 7 (USP7), resulting in increased interaction between USP7 and PD-L1 and augmented PD-L1 stability. CONCLUSIONS Our findings provide novel insights into the specific regulatory mechanism of PD-L1 in ESCC cells, which offer a new strategy for synergizing with anti-PD-L1 therapy.
Collapse
Affiliation(s)
- Chang Wang
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Chenxi Ju
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Dan Du
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Peiyu Zhu
- Key Laboratory of Carcinogenesis and Translational Research, Center of Gastrointestinal Cancer, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Jie Yin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jinlin Jia
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xue Wang
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xinyu Xu
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Li Zhao
- Department of Research and Development, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Junhu Wan
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Ting Sun
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Lijun Yang
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Hongle Li
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Fucheng He
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Mingxia Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Jing He
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
7
|
Bonfill Cosp X, Savall-Esteve O, Bracchiglione J, Requeijo C, Santero M. Mismatch between evidence and related clinical recommendations about the treatment of advanced esophageal cancer patients with anticancer drugs: A critical historical review. J Cancer Policy 2025; 44:100580. [PMID: 40147630 DOI: 10.1016/j.jcpo.2025.100580] [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: 07/04/2024] [Revised: 02/19/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
PURPOSE to analyze the most robust research and recommendations that have informed the potential superiority of treatments with anticancer drugs over any type of supportive care for advanced esophageal cancer (EC). METHODS We conducted a critical historical review. First, we identified randomized clinical trials (RCTs) from a previous scoping review conducted by our research group, ASTAC, updating the search strategy. Second, we searched for the most important and recognized international clinical practice guidelines (CPGs) in advanced EC. Finally, we performed a systematic document analysis to compare whether the recommendations proposed in the CPGs were supported by the previously identified relevant evidence. RESULTS We identified and assessed 15 RCTs and 11 CPGs from ESMO (eight), ASCO (two), and NICE (one) published over the last 40 years. There is a clear mismatch between these guidelines' recommendations and the available RCTs regarding the efficacy of anticancer drugs compared to best supportive care (BSC). CONCLUSION There is a lack of consistent evidence to support the treatment of advanced EC patients with anticancer drugs, and a notable mismatch exists between the available evidence and the recommendations made by relevant CPGs. As a result, these guidelines may be biased in favoring the use of anticancer drugs over supportive care and in consequence it is advisable to be very prudent when proposing systemic treatments to patients with advanced EC. Further rigorous and independent research is needed to better evaluate the true benefits of anticancer treatments in advanced EC and to update the CPGs accordingly.
Collapse
Affiliation(s)
- Xavier Bonfill Cosp
- Iberoamerican Cochrane Centre, Barcelona, Spain; Universitat Autònoma Barcelona (UAB), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain; Institut de Recerca Sant Pau (IR Sant Pau), Barcelona, Spain
| | - Olga Savall-Esteve
- Iberoamerican Cochrane Centre, Barcelona, Spain; Institut de Recerca Sant Pau (IR Sant Pau), Barcelona, Spain
| | - Javier Bracchiglione
- Iberoamerican Cochrane Centre, Barcelona, Spain; Universitat Autònoma Barcelona (UAB), Barcelona, Spain; Interdisciplinary Centre for Health Studies (CIESAL), Universidad de Valparaíso, Viña del Mar, Chile; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain; Institut de Recerca Sant Pau (IR Sant Pau), Barcelona, Spain
| | - Carolina Requeijo
- Iberoamerican Cochrane Centre, Barcelona, Spain; Institut de Recerca Sant Pau (IR Sant Pau), Barcelona, Spain
| | - Marilina Santero
- Iberoamerican Cochrane Centre, Barcelona, Spain; Universitat Autònoma Barcelona (UAB), Barcelona, Spain; Institut de Recerca Sant Pau (IR Sant Pau), Barcelona, Spain.
| |
Collapse
|
8
|
Aftab M, Mehmood F, Sahibzada KI, Zhang C, Jiang Y, Liu K. Attention-Enhanced Multi-Task Deep Learning Model for Classification and Segmentation of Esophageal Lesions. ACS OMEGA 2025; 10:10468-10479. [PMID: 40124037 PMCID: PMC11923690 DOI: 10.1021/acsomega.4c10763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/25/2025]
Abstract
Accurate detection and segmentation of esophageal lesions are crucial for diagnosing and treating gastrointestinal diseases. However, early detection of esophageal cancer remains challenging, contributing to a reduced five-year survival rate among patients. This paper introduces a novel multitask deep learning model for automatic diagnosis that integrates classification and segmentation tasks to assist endoscopists effectively. Our approach leverages the MobileNetV2 deep learning architecture enhanced with a mutual attention module, significantly improving the model's performance in determining the locations of esophageal lesions. Unlike traditional models, the proposed model is designed not to replace endoscopists but to empower them to correct false predictions when provided with additional Supporting Information. We evaluated the proposed model on three well-known data sets: Early Esophageal Cancer (EEC), CVC-ClinicDB, and KVASIR. The experimental results demonstrate promising performance, achieving high classification accuracies of 98.72% (F1-score: 98.08%) on CVC-ClinicDB, 98.95% (F1-score: 98.32%) on KVASIR, and 99.12% (F1-score: 99.00%) on our generated EEC data set. Compared to state-of-the-art models, our classification results show significant improvement. For the segmentation task, the model attained a Dice coefficient of 92.73% and an Intersection over Union (IoU) of 91.54%. These findings suggest that the proposed multitask deep learning model can effectively assist endoscopists in evaluating esophageal lesions, thereby alleviating their workload and enhancing diagnostic precision.
Collapse
Affiliation(s)
- Muhammad Aftab
- Pathophysiology
Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Tianjian
Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
- China-US
(Henan) Hormel Cancer Institute, Zhengzhou, Henan 450000, China
| | - Faisal Mehmood
- Department
of Civil and Transportation Engineering, Shenzhen University, Shenzhen 518060, China
| | - Kashif Iqbal Sahibzada
- College
of Bioengineering, Henan University of Technology, Zhengzhou 450001, China
- Department
of Health Professional Technologies, The
University of Lahore, Lahore 54000, Pakistan
| | - Chengjuan Zhang
- Center
of Bio-Repository, The Affiliated Cancer
Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Yanan Jiang
- Pathophysiology
Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Tianjian
Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
- China-US
(Henan) Hormel Cancer Institute, Zhengzhou, Henan 450000, China
- State
Key Laboratory of Metabolic Dysregulation & the Prevention and
Treatment of Esophageal Cancer, Zhengzhou, Henan 450000, China
- The
Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan 450000, China
| | - Kangdong Liu
- Pathophysiology
Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Tianjian
Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
- China-US
(Henan) Hormel Cancer Institute, Zhengzhou, Henan 450000, China
- State
Key Laboratory of Metabolic Dysregulation & the Prevention and
Treatment of Esophageal Cancer, Zhengzhou, Henan 450000, China
- The
Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan 450000, China
| |
Collapse
|
9
|
Luo L, Jiang C, Xie S. The mechanism of high mobility group box-1 in the proliferation and macrophage polarization in esophageal squamous cell carcinoma cells. Eur J Med Res 2025; 30:144. [PMID: 40022250 PMCID: PMC11869724 DOI: 10.1186/s40001-025-02390-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/17/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Previous studies showed that high mobility group box-1 (HMGB1) facilitates the initiation and progression of esophageal squamous cell carcinoma (ESCC), and the current research investigated the detailed mechanisms implicated. METHODS The impact of HMGB1 and IGFBP3 levels on the survival of ESCC was examined by plotting Kaplan-Meier (KM) curves based on the data collected from The Cancer Genome Atlas (TCGA). Quantitative real-time PCR (qRT-PCR) was performed to detect the expressions of HMGB1 in both human esophageal epithelial cells (HEEC) and ESCC cells. After cell transfection, the proliferation of ESCC cells was measured, and the cell metastasis was determined based on the levels of cadherins (CDHs) and Vimentin (VIM). Macrophage polarization was determined by calculating the mean fluorescence intensity (MFI) of CD206 and CD86. In addition, co-immunoprecipitation and immunoblotting were applied to evaluate the interaction between insulin-like growth factor binding protein 3 (IGFBP3)/DNA-dependent protein kinase catalytic subunit (DNA-PKcs) and HMGB1. RESULTS A high level of HMGB1 was predictive of an unfavorable prognosis of ESCC (p < 0.05). HMGB1 showed a higher expression in ESCC cells (p < 0.05), while knockdown of HMGB1 inhibited ESCC cell proliferation, downregulated the levels of CDH2 and VIM and upregulated the level of CDH1 (p < 0.05). In contrast, overexpressed HMGB1 showed the opposite effects (p < 0.05), suggesting the role of HMGB1 in the epithelial-mesenchymal transition (EMT) of ESCC. After the knockout of HMGB1, the MFI of CD86 was increased but that of CD206 was reduced, indicating the polarization towards M1 macrophages (p < 0.05). However, the results were reversed when HMGB1 was overexpressed (p < 0.05). Meanwhile, HMGB1 could interact with the IGFBP3/DNA-PKcs complex (p < 0.05). Low-expressed IGFBP3 was predictive of an unfavorable prognosis of ESCC, and IGFBP3 silencing promoted the proliferation of ESCC cells (p < 0.05). Besides, HMGB1 and IGFBP3 could act antagonistically in influencing the proliferation of ESCC cells and macrophage polarization. CONCLUSIONS Through in vitro experiments, this study found that HMGB1 was linked to the proliferation and polarization of macrophages in ESCC, providing novel evidence for the role of HMGB1 in ESCC development.
Collapse
Affiliation(s)
- Liling Luo
- Department of Radiation Oncology, Guangdong Provincial People'S Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510000, China
| | - Chao Jiang
- Department of Radiation Oncology, The People's Hospital of Shenzhen Baoan District, the Second Affiliated Hospital of Shenzhen University, Shenzhen, 518100, China
| | - Songxi Xie
- Department of Radiation Oncology, Guangdong Provincial People'S Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510000, China.
| |
Collapse
|
10
|
Tian W, Liu W, Wang C, Liu L, Zhang L, Chen L. Developing a health education program for home enteral nutrition after esophageal cancer surgery based on the Delphi method. Medicine (Baltimore) 2025; 104:e41586. [PMID: 39993103 PMCID: PMC11856940 DOI: 10.1097/md.0000000000041586] [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: 09/22/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/26/2025] Open
Abstract
To develop a health education program for home enteral nutrition (HEN) after Esophageal cancer (EC) surgery based on Delphi method, providing reference opinions for clinical nursing education and patient home care. The health education program for HEN after EC surgery was constructed through literature research, theoretical analysis and group discussion. From February to June 2024, experts in clinical nursing, clinical medicine, and nutrition of EC were invited to conduct 2 rounds of Delphi expert consultation to initially determine the items of the program. The weight and assignment of each items were determined through analytic hierarchy process (AHP), and then the final health education program for HEN after EC surgery was established. The authoritative coefficients of the 2 rounds consulting experts were 0.918 and 0.929 and the positive coefficients were 100% and 95%, respectively; The mean range of importance assignment for the second round of consultation indicators was 4.26 to 5.00 points, with a full score range of 40.90% to 95.45%. The mean harmony coefficients for expert opinions from 2 rounds were 0.206 and 0.218 (P < .01), respectively. The final health education program for postoperative HEN in EC includes 6 primary items, 27 secondary items, and 18 tertiary items. The results of the AHP showed that the consistency coefficients (CR values) of each matrix for the tertiary items were all < 0.1, meeting the requirements of consistency testing. The constructed health education program for postoperative HEN patients with EC in this study has high scientific and practical value, and can provide reference for the health education of postoperative HEN patients with EC.
Collapse
Affiliation(s)
- Weiran Tian
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wei Liu
- Department of Breast Surgery, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Cuirong Wang
- Department of Otolaryngology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lei Liu
- Department of Breast Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Liming Zhang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lu Chen
- Department of Nursing, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
11
|
Zhang M, Su Y, Wen P, Shao X, Yang P, An P, Jing W, Liu L, Yang Z, Yang M. Subtype cluster analysis unveiled the correlation between m6A- and cuproptosis-related lncRNAs and the prognosis, immune microenvironment, and treatment sensitivity of esophageal cancer. Front Immunol 2025; 16:1539630. [PMID: 40034693 PMCID: PMC11872909 DOI: 10.3389/fimmu.2025.1539630] [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: 12/04/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Objective Esophageal cancer (EC) is characterized by a high degree of malignancy and poor prognosis. N6-methyladenosine (m6A), a prominent post-transcriptional modification of mRNA in mammalian cells, plays a pivotal role in regulating various cellular and biological processes. Similarly, cuproptosis has garnered attention for its potential implications in cancer biology. This study seeks to elucidate the impact of m6A- and cuproptosis-related long non-coding RNAs (m6aCRLncs) on the prognosis of patients with EC. Methods The EC transcriptional data and corresponding clinical information were retrieved from The Cancer Genome Atlas (TCGA) database, comprising 11 normal samples and 159 EC samples. Data on 23 m6A regulators and 25 cuproptosis-related genes were sourced from the latest literature. The m6aCRLncs linked to EC were identified through co-expression analysis. Differentially expressed m6aCRLncs associated with EC prognosis were screened using the limma package in R and univariate Cox regression analysis. Subtype clustering was performed to classify EC patients, enabling the investigation of differences in clinical outcomes and immune microenvironment across patient clusters. A risk prognostic model was constructed using least absolute shrinkage and selection operator (LASSO) regression. Its robustness was evaluated through survival analysis, risk stratification curves, and receiver operating characteristic (ROC) curves. Additionally, the model's applicability across various clinical features and molecular subtypes of EC patients was assessed. To further explore the model's utility in predicting the immune microenvironment, single-sample gene set enrichment analysis (ssGSEA), immune cell infiltration analysis, and immune checkpoint differential expression analysis were conducted. Drug sensitivity analysis was performed to identify potential therapeutic agents for EC. Finally, the mRNA expression levels of m6aCRLncs in EC cell lines were validated using reverse transcription quantitative polymerase chain reaction (RT-qPCR). Results We developed a prognostic risk model based on five m6aCRLncs, namely ELF3-AS1, HNF1A-AS1, LINC00942, LINC01389, and MIR181A2HG, to predict survival outcomes and characterize the immune microenvironment in EC patients. Analysis of molecular subtypes and clinical features revealed significant differences in cluster distribution, disease stage, and N stage between high- and low-risk groups. Immune profiling further identified distinct immune cell populations and functional pathways associated with risk scores, including positive correlations with naive B cells, resting CD4+ T cells, and plasma cells, and negative correlations with macrophages M0 and M1. Additionally, we identified key immune checkpoint-related genes with significant differential expression between risk groups, including TNFRSF14, TNFSF15, TNFRSF18, LGALS9, CD44, HHLA2, and CD40. Furthermore, nine candidate drugs with potential therapeutic efficacy in EC were identified: Bleomycin, Cisplatin, Cyclopamine, PLX4720, Erlotinib, Gefitinib, RO.3306, XMD8.85, and WH.4.023. Finally, RT-qPCR validation of the mRNA expression levels of m6aCRLncs in EC cell lines demonstrated that ELF3-AS1 expression was significantly upregulated in the EC cell lines KYSE-30 and KYSE-180 compared to normal esophageal epithelial cells. Conclusion This study elucidates the role of m6aCRLncs in shaping the prognostic outcomes and immune microenvironment of EC. Furthermore, it identifies potential therapeutic agents with efficacy against EC. These findings hold significant promise for enhancing the survival of EC patients and provide valuable insights to inform clinical decision-making in the management of this disease.
Collapse
Affiliation(s)
- Ming Zhang
- Department of General Practice, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yani Su
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Pengfei Wen
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Xiaolong Shao
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Peng Yang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Peng An
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Wensen Jing
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Lin Liu
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Zhi Yang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Mingyi Yang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
12
|
Safari MH, Rahimzadeh P, Alaei E, Alimohammadi M, Esfandiari N, Daneshi S, Malgard N, Farahani N, Taheriazam A, Hashemi M. Targeting ferroptosis in gastrointestinal tumors: Interplay of iron-dependent cell death and autophagy. Mol Cell Probes 2025; 79:102013. [PMID: 39837469 DOI: 10.1016/j.mcp.2025.102013] [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: 11/22/2024] [Revised: 01/06/2025] [Accepted: 01/18/2025] [Indexed: 01/23/2025]
Abstract
Ferroptosis is a regulated cell death mechanism distinct from apoptosis, autophagy, and necroptosis, marked by iron accumulation and lipid peroxidation. Since its identification in 2012, it has developed into a potential therapeutic target, especially concerning GI disorders like PC, HCC, GC, and CRC. This interest arises from the distinctive role of ferroptosis in the progression of diseases, presenting a new avenue for treatment where existing therapies fall short. Recent studies emphasize the promise of focusing on ferroptosis to fight GI cancers, showcasing its unique pathophysiological mechanisms compared to other types of cell death. By comprehending how ferroptosis aids in the onset and advancement of GI diseases, scientists aim to discover novel drug targets and treatment approaches. Investigating ferroptosis in gastrointestinal disorders reveals exciting possibilities for novel therapies, potentially revolutionizing cancer treatment and providing renewed hope for individuals affected by these tumors.
Collapse
Affiliation(s)
- Mohamad Hosein Safari
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elmira Alaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Negin Esfandiari
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Neda Malgard
- Department of Internal Medicine, Firoozgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
13
|
Wang Z, Xiao Z, Zhang T, Lu M, Li H, Cao J, Zheng J, Zhou Y, Dai J, Wang C, Chen L, Xu J. Development and validation of a novel artificial intelligence algorithm for precise prediction the postoperative prognosis of esophageal squamous cell carcinoma. BMC Cancer 2025; 25:134. [PMID: 39849452 PMCID: PMC11756118 DOI: 10.1186/s12885-025-13520-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 01/14/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a highly aggressive malignancy, and current postoperative prognostic assessment methods remain unsatisfactory, underlining the urgent to develop a reliable approach for precision medicine. Given the similarities with gametogenesis, cancer/testis genes (CTGs) are acknowledged for regulation unrestrained multiplication and immune microenvironment during oncogenic processes. These processes are associated with advanced disease and poorer prognosis, indicating that CTGs could serve as ideal prognostic biomarkers in ESCC. The purpose of this study is to develop a novel clinically prognostic prediction system to facilitate the individualized postoperative care. METHODS We conducted LASSO regression analysis of protein-coding CTGs and clinical characteristics from 119 pathologically confirmed ESCC patients to recognize powerful predictive variables. We employed nine supervised machine learning classifiers and integrated best predictive machine learning classifiers by weighted voting method to construct an ensemble model called PPMESCC. Additionally, functional assay was conducted to examine the potential effect of top-ranking CTG HENMT1 in ESCC. RESULTS LASSO regression identified five CTGs and TNM stage as optimized prognostic features. Six machine learning classifiers were integrated to construct an ensemble model, PPMESCC, which exhibited outstanding performance in ESCC prediction. The AUC for PPMESCC was 0.9828 (95% confidence interval: 0.9608 to 0.9926), with an accuracy of 98.32% (95% CI: 96.64-99.16%) in the discovery cohort and 0.9057 (95% CI: 0.8897 to 0.9583) of AUC with an accuracy of 90% (95% CI: 89.08-93.28%) in validation cohort. In addition, the top-ranking CTG HENMT1 encodes 2'-O-methyltransferase of piRNAs that was confirmed positively correlated with the proliferation capacity of ESCC cells. Then we systematically screen piRNAs associated with esophageal carcinoma based on GWAS, eQTL-piRNA, and i2OM databases, and successfully discovered 8 piRNAs potentially regulated by HENMT1. CONCLUSION The study highlights the clinical utility of PPMESCC algorithm in prognostic prediction that may facilitate to establish the personalized screening and management strategies for postoperative ESCC patients.
Collapse
Affiliation(s)
- Zichen Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Zhihan Xiao
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Tongyu Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Meiyou Lu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Hai Li
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Jing Cao
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Jianan Zheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Yichan Zhou
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Juncheng Dai
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, No. 101, Longmian Avenue, Nanjing, 211166, Jiangsu, China
| | - Cheng Wang
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, No. 101, Longmian Avenue, Nanjing, 211166, Jiangsu, China
| | - Liang Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Jing Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
14
|
Wang Y, Sun H, Li G, Xu J, Wang S, Zhang S, Zhou T, Han T, Sun C, Ma J, Wang X, Yin H. Impact of maternal and offspring smoking and breastfeeding on oesophageal cancer in adult offspring. Nat Commun 2025; 16:938. [PMID: 39843420 PMCID: PMC11754477 DOI: 10.1038/s41467-025-56252-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025] Open
Abstract
Numerous risk factors for oesophageal cancer are linked to lifestyle habits, but the role of early-life factors in its incidence and mortality is unclear. Using UK Biobank data, we explore the association among breastfeeding, maternal smoking, smoking in offspring, and oesophageal cancer risk in adult offspring via multivariable Cox regression. Here, we show that being breastfed, compared with not being breastfed, is associated with a lower risk of oesophageal cancer incidence (HR: 0.83, 95% CI: 0.70-0.98) and mortality (HR: 0.74, 95% CI: 0.61-0.89) in adult offspring. Additionally, it is associated with a reduced impact of smoking in offspring on oesophageal cancer incidence (HR: 0.79, 95% CI: 0.64-0.96) and mortality (HR: 0.73, 95% CI: 0.59-0.91). We subsequently construct a polygenic risk score for oesophageal cancer to explore the influence of genetic factors. Our findings emphasize the importance of breastfeeding, and smoking cessation to prevent oesophageal cancer.
Collapse
Affiliation(s)
- Yixue Wang
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Hongru Sun
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Gen Li
- Department of Radiation Therapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jingxue Xu
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Siyu Wang
- Department of Radiation Therapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Shijie Zhang
- Department of Radiation Therapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Tianle Zhou
- Department of Radiation Therapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Tianshu Han
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China
| | - Changhao Sun
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jianqun Ma
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| | - Xiaoyuan Wang
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| | - Hang Yin
- Department of Radiation Therapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| |
Collapse
|
15
|
Cao W, Jin D, Min W, Li H, Wang R, Zhang J, Gou Y. Prognostic values of intracellular cell-related genes in esophageal cancer and their regulatory mechanisms. BMC Cancer 2025; 25:105. [PMID: 39833728 PMCID: PMC11744837 DOI: 10.1186/s12885-025-13483-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Esophageal cancer is a grave malignant condition. While radiotherapy, often in conjunction with chemotherapy, serves as a cornerstone in the management of locally advanced or metastatic cases, patient tolerance and treatment resistance frequently hinder its efficacy. Cell-in-cell structures, prevalent in various tumors, have been linked to prognosis. Hence, investigating the prognostic significance and regulatory mechanisms of genes related to these intracellular structures in esophageal cancer is imperative. The Cancer Genome Atlas (TCGA) Esophageal Cancer (ESCA) dataset served as the training set for the analysis. Differentially expressed genes (DEGs) in ESCA samples were identified, with those related to intercellular structures designated cell-in-cell-related differential expression genes (CIC-related DEGs). Cox regression analysis was employed to identify prognostic genes, categorizing samples into high- and low-risk groups based on median risk scores. Validation was conducted using the GSE53624 risk model. Established methodologies included morphological mapping, enrichment analysis, immune infiltration analysis, prognostic gene expression validation, molecular docking, and Reverse Transcription Polymerase Chain Reaction (RT-PCR) validation. Thirty-eight intersecting genes were identified between the disease and normal groups in ESCA samples. Stepwise multivariate Cox analysis pinpointed three prognostic genes: androgen receptor (AR), C-X-C motif chemokine ligand 8 (CXCL8), and epidermal growth factor receptor (EGFR). The risk model's applicability was confirmed in the GSE53624 dataset, revealing eight significantly different immune-related gene sets. Prognostic gene expression validation demonstrated significant differences between the disease and normal groups in both datasets. The proteins corresponding to the three prognostic genes interacted with gefitinib and osimertinib. RT-PCR results corroborated the differential expression of prognostic genes in esophageal cancer tissues. This study identified AR, CXCL8, and EGFR as prognostic genes and demonstrated their molecular interactions with gefitinib and osimertinib, providing a foundation for ESCA diagnosis and treatment.
Collapse
Affiliation(s)
- Wei Cao
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Dacheng Jin
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Weirun Min
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Haochi Li
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Rong Wang
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
| | - Jinlong Zhang
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Yunjiu Gou
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China.
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China.
| |
Collapse
|
16
|
Yu SS, Zheng X, Li XS, Xu QJ, Zhang W, Liao ZL, Lei HK. Development of a nomogram for overall survival in patients with esophageal carcinoma: A prospective cohort study in China. World J Gastrointest Oncol 2025; 17:96686. [PMID: 39817137 PMCID: PMC11664612 DOI: 10.4251/wjgo.v17.i1.96686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Esophageal carcinoma (EC) presents a significant public health issue in China, with its prognosis impacted by myriad factors. The creation of a reliable prognostic model for the overall survival (OS) of EC patients promises to greatly advance the customization of treatment approaches. AIM To create a more systematic and practical model that incorporates clinically significant indicators to support decision-making in clinical settings. METHODS This study utilized data from a prospective longitudinal cohort of 3127 EC patients treated at Chongqing University Cancer Hospital between January 1, 2018, and December 12, 2020. Utilizing the least absolute shrinkage and selection operator regression alongside multivariate Cox regression analyses helped pinpoint pertinent variables for constructing the model. Its efficacy was assessed by concordance index (C-index), area under the receiver operating characteristic curve (AUC), calibration curves, and decision curve analysis (DCA). RESULTS Nine variables were determined to be significant predictors of OS in EC patients: Body mass index (BMI), Karnofsky performance status, TNM stage, surgery, radiotherapy, chemotherapy, immunotherapy, platelet-to-lymphocyte ratio, and albumin-to-globulin ratio (ALB/GLB). The model demonstrated a C-index of 0.715 (95%CI: 0.701-0.729) in the training cohort and 0.711 (95%CI: 0.689-0.732) in the validation cohort. In the training cohort, AUCs for 1-year, 3-year, and 5-year OS predictions were 0.773, 0.787, and 0.750, respectively; in the validation cohort, they were 0.772, 0.768, and 0.723, respectively, illustrating the model's precision. Calibration curves and DCA verified the model's predictive accuracy and net benefit. CONCLUSION A novel prognostic model for determining the OS of EC patients was successfully developed and validated to help clinicians in devising individualized treatment schemes for EC patients.
Collapse
Affiliation(s)
- Shi-Shi Yu
- Department of Gastroenterology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Xi Zheng
- Department of Gastroenterology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Xiao-Sheng Li
- Chongqing Cancer Multi-omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Qian-Jie Xu
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Wei Zhang
- Chongqing Cancer Multi-omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Zhong-Li Liao
- Department of Gastroenterology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Hai-Ke Lei
- Chongqing Cancer Multi-omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
17
|
Leonhardi J, Schnarkowski B, Mehdorn M, Höhn AK, Niebisch S, Plum P, Seehofer D, Tiepolt S, Denecke T, Meyer HJ. Diagnostic Accuracy and Reliability of CT-based Node-RADS for Esophageal Cancer. In Vivo 2025; 39:353-359. [PMID: 39740864 PMCID: PMC11705101 DOI: 10.21873/invivo.13835] [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: 10/06/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND/AIM The recently published Node-Reporting and Data System (Node-RADS) can aid the characterization of lymph nodes in cross-sectional imaging. This study investigated the Node-RADS system in computed tomography (CT) to characterize lymph nodes in esophageal cancer. PATIENTS AND METHODS Overall, 126 patients (15 female, 11.9%) with a mean age of 62.1±10.4 years comprised the patient sample. All patients underwent resection with curative intent and the lymph nodes were histopathologically analyzed during clinical routine. For every patient, the locoregional lymph nodes were scored in accordance with the Node-RADS classification. For statistical analysis, receiver-operating characteristics (ROC) with area under the curve (AUC) were used to test for diagnostic accuracy; inter-reader variability was assessed with Cohen's kappa. RESULTS Overall, 54 patients were nodal positive (42.9%), 72 patients were nodal negative (57.1%). Inter-reader agreement was substantial for the overall Node-RADS scoring ([Formula: see text]=0.65, p<0.001). ROC curve analysis for lymph node discrimination (N0 versus N1-3) showed an AUC of 0.69 (95% confidence interval=0.59-0.79). A threshold score of more than 2 resulted in a sensitivity of 0.77 and a specificity of 0.55 for correctly predicting nodal positivity. Node-RADS 1 category had a malignancy rate of 30%, Node-RADS 2 of 14%, Node-RADS 3 of 81%, Node-RADS 4 of 90.1% and Node-RADS 5 of 86.5%. CONCLUSION The Node-RADS score on staging CT is associated with the malignancy rate of lymph nodes in patients with EC with only moderate diagnostic accuracy. The inter-reader variability is moderate, which could pose difficulties for translation into clinical routine.
Collapse
Affiliation(s)
- Jakob Leonhardi
- Department of Diagnostic and Interventional Radiology, University of Leipzig, Leipzig, Germany
| | - Benedikt Schnarkowski
- Department of Diagnostic and Interventional Radiology, University of Leipzig, Leipzig, Germany
| | - Matthias Mehdorn
- Department of Visceral and Transplantation Surgery, University Hospital Leipzig, University of Leipzig, Leipzig, Germany
| | - Anne-Kathrin Höhn
- Department of Pathology, University Hospital Leipzig, University of Leipzig, Leipzig, Germany
| | - Stefan Niebisch
- Department of Visceral and Transplantation Surgery, University Hospital Leipzig, University of Leipzig, Leipzig, Germany
| | - Patrick Plum
- Department of Visceral and Transplantation Surgery, University Hospital Leipzig, University of Leipzig, Leipzig, Germany
| | - Daniel Seehofer
- Department of Visceral and Transplantation Surgery, University Hospital Leipzig, University of Leipzig, Leipzig, Germany
| | - Solveig Tiepolt
- Department of Nuclear Medicine, University Hospital Leipzig, University of Leipzig, Leipzig, Germany
| | - Timm Denecke
- Department of Diagnostic and Interventional Radiology, University of Leipzig, Leipzig, Germany
| | - Hans-Jonas Meyer
- Department of Diagnostic and Interventional Radiology, University of Leipzig, Leipzig, Germany;
| |
Collapse
|
18
|
Huang T, Wei L, Zhou H, Liu J. Macrophage Infiltration and ITGB2 Expression in ESCC: A Novel Correlation. Cancer Med 2025; 14:e70604. [PMID: 39825491 PMCID: PMC11742006 DOI: 10.1002/cam4.70604] [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: 05/28/2024] [Revised: 09/22/2024] [Accepted: 01/03/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the most prevalent and lethal malignancies worldwide. Despite progress in immunotherapy for cancer treatment, its application and efficacy in ESCC remain limited. Therefore, there is an ongoing need to explore potential molecules and therapeutic strategies related to tumor immunity in ESCC. METHODS In this study, we integrated high-throughput sequencing data, gene chip data, single-cell sequencing data, and various bioinformatics analysis methods along with experimental approaches to identify key genes involved in immune infiltration in ESCC and investigate their relationship with immune cell development, as well as the potential of these key genes in immunotherapy. RESULTS We discovered and validated a positive correlation between macrophage infiltration and ITGB2 expression in ESCC. ITGB2 is overexpressed in ESCC and has potential as a prognostic biomarker for the disease. We present for the first time the finding that the expression of ITGB2 in infiltrating macrophages increases as these macrophages polarize toward a tumor-promoting phenotype in ESCC. Moreover, during the progression of ESCC, ITGB2 expression in infiltrating macrophages is upregulated. The higher the expression of ITGB2, the more feasible it is to target macrophages. Additionally, we found that evaluating immune therapy responses in ESCC patients through ITGB2 expression is a viable approach. Furthermore, we identified three miRNAs associated with abnormal ITGB2 expression, providing insights into the upstream molecular interactions of ITGB2. CONCLUSIONS Macrophage infiltration in ESCC is closely associated with ITGB2, which holds significant potential for immunotherapy applications in ESCC. Based on our findings and prior studies, we propose a novel hypothesis: inducing M1 macrophages in vitro, knocking out ITGB2, and then reinfusing these ITGB2-knockout M1 macrophages into ESCC patients may represent a promising new immunotherapy strategy, providing a new avenue for ESCC immunotherapy.
Collapse
Affiliation(s)
- Tao Huang
- Department of Cardiothoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningPeople's Republic of China
| | - Longqian Wei
- Department of Cardiothoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningPeople's Republic of China
| | - Huafu Zhou
- Department of Cardiothoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningPeople's Republic of China
| | - Jun Liu
- Department of Cardiothoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningPeople's Republic of China
| |
Collapse
|
19
|
Wu Y, Xian D, Liu Y, Huang D, Liu Q, Yang S. USP8-Dependent Family Tyrosine Kinase Promotes the Malignant Progression of Esophageal Squamous Cell Carcinoma by Upregulating Protein Tyrosine Kinase 2 Expression. Thorac Cancer 2025; 16:e15489. [PMID: 39702934 PMCID: PMC11739124 DOI: 10.1111/1759-7714.15489] [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: 07/24/2024] [Revised: 09/03/2024] [Accepted: 11/03/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a lethal malignancy, and the molecular underpinnings of its aggressive behavior are not fully understood. FYN proto-oncogene, Src family tyrosine kinase (FYN) has been linked to cancer progression, yet its role in ESCC remains elusive. This study investigated the influence of FYN on ESCC malignancy. METHODS Quantitative real-time polymerase chain reaction was used to assess the mRNA expression of FYN, while western blotting and immunohistochemistry (IHC) assays were performed to detect the protein expression of FYN, ubiquitin specific peptidase 8 (USP8) and protein tyrosine kinase 2 (PTK2). Cell viability was measured with a cell counting kit-8 assay, and cell apoptosis was evaluated using flow cytometry. RESULTS FYN expression was increased in ESCC tissues and cells when compared with normal esophageal tissues and normal esophageal epithelial cells. Knockdown of FYN inhibited cell invasion, migration, stem-like traits, and glycolysis, while promoting apoptosis. USP8 was shown to stabilize FYN protein expression through its deubiquitinating activity in ESCC cells. Overexpression of FYN reversed the effects of USP8 silencing on the malignant phenotypes of ESCC cells in vitro and in vivo. FYN upregulated PTK2 expression in both TE1 and KYSE150 cell lines. Furthermore, PTK2 overexpression reversed the effects of FYN silencing on the malignant phenotypes of ESCC cells. Further, USP8 silencing-induced inhibitory effect on PTK2 protein expression was counteracted after FYN overexpression. CONCLUSION USP8-dependent FYN contributed to the malignant progression of ESCC by interacting with PTK2. Targeting this pathway may offer a novel therapeutic strategy for ESCC treatment.
Collapse
Affiliation(s)
- Yuechang Wu
- Department of Cardiothoracic SurgeryThe First Affiliated Hospital of Hainan Medical CollegeHaikouChina
| | - Dubiao Xian
- Department of Cardiothoracic SurgeryThe First Affiliated Hospital of Hainan Medical CollegeHaikouChina
| | - Yunzhong Liu
- Department of Cardiothoracic SurgeryThe First Affiliated Hospital of Hainan Medical CollegeHaikouChina
| | - Ding Huang
- Department of Cardiothoracic SurgeryThe First Affiliated Hospital of Hainan Medical CollegeHaikouChina
| | - Qingfeng Liu
- Department of Cardiothoracic SurgeryThe First Affiliated Hospital of Hainan Medical CollegeHaikouChina
| | - Shubo Yang
- Department of Cardiothoracic SurgeryThe First Affiliated Hospital of Hainan Medical CollegeHaikouChina
| |
Collapse
|
20
|
Shen S, Liu B, Guan W, Liu Z, Han Y, Hu Y, Chen Y, Liu S, He J, Li Z, Tang W, Zhang P, Ren W, Qiu Y, Zheng H, Li J. Advancing precision medicine in esophageal squamous cell carcinoma using patient-derived organoids. J Transl Med 2024; 22:1168. [PMID: 39741269 PMCID: PMC11686971 DOI: 10.1186/s12967-024-05967-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Patient-derived organoids (PDOs) represent a promising approach for replicating the characteristics of original tumors and facilitating drug testing for personalized treatments across diverse cancer types. However, clinical evidence regarding their application to esophageal cancer remains limited. This study aims to evaluate the efficacy of implementing PDOs in clinical practice to benefit patients with esophageal squamous cell carcinoma (ESCC). METHODS Fresh surgical biopsies were obtained from patients with esophageal cancer for the establishment of PDOs. These PDOs were subsequently characterized through histological analysis. A customized drug panel, based on standard-of-care chemotherapy regimens, was applied to the PDOs. The resulting drug sensitivity profiles were then correlated with the clinical responses observed in individual patients undergoing actual treatment. RESULTS A total of 34 PDOs were successfully established with a 61.8% success rate. The classification method based on chemotherapy sensitivity closely corresponded to clinical responses. The paclitaxel plus cisplatin (TP)-sensitive group demonstrated significantly longer progression-free survival (PFS) compared to the resistant groups, Hazard ratio (HR), 5.12; 95% confidence intervals (CI 0.58-44.71; p < 0.05), thus illustrating the potential of this approach for guiding personalized treatment strategies. CONCLUSION Organoid biobanks were established across multiple institutes to facilitate PDOs-based functional precision medicine. The findings demonstrate that this framework offers robust predictive value in clinical settings, enhances precision therapeutics, and advances drug discovery for esophageal cancer.
Collapse
Affiliation(s)
- Suya Shen
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Bing Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Wenyan Guan
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Ziyao Liu
- Department of Precision Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Yuqing Han
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yingzhe Hu
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Yiqiang Chen
- Department of Precision Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Siyuan Liu
- Department of Precision Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Zhiwen Li
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Weifeng Tang
- Department of Esophageal Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, NanjingJiangsu, 210008, China
| | - Pengju Zhang
- Zhejiang Honray Medical Technology Co., LTD, Taizhou, 318001, Zhejiang, China
| | - Wei Ren
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, Jiangsu, China.
| | - Yudong Qiu
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Hongping Zheng
- Zhejiang Honray Medical Technology Co., LTD, Taizhou, 318001, Zhejiang, China.
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, 200032, China.
| | - Jingjing Li
- Department of Precision Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
21
|
Yu Y, Jin B, Jia R, Shi L, Chen Y, Ge J, Xu C. Exosomes loaded with the anti-cancer molecule mir-1-3p inhibit intrapulmonary colonization and growth of human esophageal squamous carcinoma cells. J Transl Med 2024; 22:1166. [PMID: 39741298 DOI: 10.1186/s12967-024-05997-9] [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: 07/07/2024] [Accepted: 12/15/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND The overall prognosis of patients with esophageal cancer (EC) is extremely poor. There is an urgent need to develop innovative therapeutic strategies. This study will investigate the anti-cancer effects of exosomes loaded with specific anti-cancer microRNAs in vivo and in vitro. METHODS Specific miRNAs that were significantly down-regulated in EC tissues were screened using the miRNAs profiling data of human EC tissue samples in TCGA, and the role of their exogenous expression in the proliferation and migration of human EC cell lines, KYSE150 and Eca109, were detected using CCK-8 and Transwell assays. Exosomes were loaded with miRNAs using electroporation. RESULTS The expression of miR-1-3p was significantly down-regulated in human EC tissues with potential anti-cancer effects. Exosomes loaded with miR-1-3p significantly inhibited the proliferation, migration and invasion of KYSE150 and Eca109 cells in vitro, as well as the intrapulmonary colonization and growth of KYSE150 cells in vivo. In addition, miR-1-3p could directly bind to the 3'UTR of the transcription factor E2F5 mRNA, down-regulate the protein expression of E2F5, and inhibit the activation of the MAPK/ERK signaling pathway. CONCLUSION Exosomes loaded with miR-1-3p may be applicable to the treatment of EC.
Collapse
Affiliation(s)
- Yanmei Yu
- Ultrasonography Department, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Bingjie Jin
- Gastroenterology Department, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324 JingwuWeiqi Road, Jinan, Shandong, 250021, China
| | - Ruzhen Jia
- Gastroenterology Department, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324 JingwuWeiqi Road, Jinan, Shandong, 250021, China
| | - Lei Shi
- Gastroenterology Department, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324 JingwuWeiqi Road, Jinan, Shandong, 250021, China
| | - Yong Chen
- Gastroenterology Department, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324 JingwuWeiqi Road, Jinan, Shandong, 250021, China
| | - Jian Ge
- Gastroenterology Department, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324 JingwuWeiqi Road, Jinan, Shandong, 250021, China
| | - Changqin Xu
- Gastroenterology Department, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324 JingwuWeiqi Road, Jinan, Shandong, 250021, China.
| |
Collapse
|
22
|
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
|
23
|
Ye C, Xu C, Tang Y, Qi Y, Peng X, Wei G, Jiang L. A novel disulfidptosis-related LncRNA prognostic risk model: predicts the prognosis, tumor microenvironment and drug sensitivity in esophageal squamous cell carcinoma. BMC Gastroenterol 2024; 24:437. [PMID: 39604874 PMCID: PMC11603746 DOI: 10.1186/s12876-024-03530-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Disulfidptosis is a newly discovered type of cell death that differs from apoptosis, necrosis, ferroptosis and other death modes and is closely related to the occurrence and progression of tumors. However, the predictive potential and biological characteristics of disulfidptosis-related lncRNAs (DRGs-lncRNAs) in esophageal squamous cell carcinoma (ESCC) are unclear. METHODS RNA transcriptome data, clinical information and mutation data for ESCC patients were obtained from The Cancer Genome Atlas (TCGA) database. Pearson correlation and Cox regression analyses were used to identify the DRGs-lncRNAs associated with overall survival (OS). LASSO regression analysis was used to construct the prognostic model. A nomogram was created to predict the prognosis of patients with ESCC. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA) were used to identify the signaling pathways associated with the model. TIMER, CIBERSORT, ESTIMATE and other methods were used to analyze immune infiltration, immune function, immune checkpoints and drug sensitivity. The tumor mutation burden (TMB) were assessed between different risk groups. Real-time polymerase chain reaction (RT‒PCR) was used to detect the expression of DRGs-lncRNAs in ESCC cell lines. RESULTS A total of 155 lncRNAs significantly associated with disulfidptosis were identified. Through univariate Cox regression analysis, LASSO regression analysis and multivariate Cox regression analysis, 9 lncRNAs with independent prognostic significance were selected, and a prognosis model was established. Survival analysis with the prognostic model revealed that there were obvious differences in survival between the high- and low-risk groups. Further analysis revealed that the immune microenvironment, immune infiltration, immune function, immune checkpoints, and drug sensitivity significantly differed between the high-risk and low-risk groups. Patients who exhibited both high risk and high tumor mutation burden (TMB) survived shorter, while those who fell into the low risk and low TMB categories survived longer. In addition, RT‒PCR analysis revealed differential expression of DRG lncRNAs between ESCC cell lines and esophageal epithelial cell lines. CONCLUSIONS We established a DRG-lncRNA prognostic model that can be used to predict the prognosis, tumor mutation burden, immune cell infiltration, and drug sensitivity of ECSS patients. The results of this study provide valuable insights into the understanding of ESCC and provide valuable assistance for the individualized treatment of ESCC patients.
Collapse
Affiliation(s)
- Chunlin Ye
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China
| | - Chuan Xu
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China
| | - Yongchao Tang
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China
| | - Yingcheng Qi
- Department of Gastroenterological Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Xiaoyue Peng
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China
| | - Guangxia Wei
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China.
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China.
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China.
| | - Lei Jiang
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China.
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China.
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China.
| |
Collapse
|
24
|
Zhang Y, Zhu Z, Ren N, Wang D, Zeng X, Zhang N. Implication of CCNG1 in radiosensitivity via the Wnt/β-catenin pathway in esophageal squamous cells. Sci Rep 2024; 14:27037. [PMID: 39511268 PMCID: PMC11544242 DOI: 10.1038/s41598-024-77811-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: 08/24/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024] Open
Abstract
Esophageal cancer (EC) poses a substantial threat to human health. The development of radioresistance in esophageal cancer cells is a critical factor contributing to local treatment failure and an unfavorable prognosis in affected patients. A comprehensive analysis was performed using bulk RNA sequencing (RNA-seq) and single-cell RNA sequencing (scRNA-seq) data from esophageal squamous cell carcinoma (ESCC) samples. Radioresistant ESCC cell lines were generated to explore the functional role of CCNG1. Various techniques, including gene knockdown, flow cytometry, and apoptosis assays, were utilized to evaluate alterations in radiosensitivity, cell cycle progression, and cell survival in response to CCNG1 modulation. Elevated CCNG1 expression was associated with poor clinical outcomes in ESCC patients and contributed to various malignant phenotypes in ESCC cells. In radioresistant ESCC cell lines, CCNG1 knockdown markedly increased radiosensitivity, as demonstrated by enhanced G2/M phase arrest and apoptosis following radiation exposure. CellChat analysis indicated a correlation between CCNG1 and the Wnt/β-catenin signaling pathway, while western blot (WB) analysis confirmed that CCNG1 functions as a downstream effector of Wnt/β-catenin. Our study has identified CCNG1 as a key regulator of radiosensitivity in ESCC, mediated through its interaction with the Wnt/β-catenin signaling pathway. Targeting the Wnt/β-catenin/CCNG1 axis presents a promising therapeutic strategy to enhance the efficacy of radiotherapy in ESCC, potentially overcoming radioresistance and improving patient outcomes.
Collapse
Affiliation(s)
- Yuxiang Zhang
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China
- Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhiman Zhu
- Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Naihan Ren
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China
| | - Dongni Wang
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China
| | - Xue Zeng
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China
| | - Na Zhang
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China.
| |
Collapse
|
25
|
Guo D, Jin J, Li D, He Y, Lin Y. Analysis of the incidence and mortality trends of esophageal cancer in cancer registry areas of China and Japan. Int J Cancer 2024; 155:1376-1386. [PMID: 38771567 DOI: 10.1002/ijc.35003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/22/2024]
Abstract
This study aims to analyze the prevalence trend of esophageal cancer in Japan and China to provide suggestions for the prevention and treatment of esophageal cancer. The results showed that the incidence rate for the years 2010-2018 significantly decreased with an APC of 5.66%, and the mortality rate from 2010 to 2015 had an APC of -5.87% in China. However, the incidence rate of Japanese women showed an upward trend, with an APC of 4.09% from 2010 to 2019. The mortality rate of esophageal cancer in Japan showed a downward trend, with an APC of -2.96% from 2010 to 2021. From 2010 to 2018, Chinese esophageal squamous cell carcinoma accounted for the highest proportion, accounting for 85.96%, with the largest distribution in the middle, accounting for 47.25%. Patients are mostly diagnosed at stage III, and the relative survival rate from 2012 to 2015 was 30.3%. Japan also has the highest proportion of squamous cell carcinoma, and the lesions are also mostly located in the middle segment. While Japanese esophageal cancer patients are mostly diagnosed at stage I, and the relative survival rate was 41.5% in Japan from 2009 to 2011. The results of this article indicate that the current prevalence of esophageal cancer in China and Japan is generally declining, and the quality of life of patients is gradually improving, but effective screening and prevention strategies are still needed to reduce the burden of this disease.
Collapse
Affiliation(s)
- Dongli Guo
- Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing Jin
- Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Daojuan Li
- Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yutong He
- Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yingsong Lin
- Department of Public Health, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| |
Collapse
|
26
|
Zhou Y, Meyle J, Groeger S. Periodontal pathogens and cancer development. Periodontol 2000 2024; 96:112-149. [PMID: 38965193 PMCID: PMC11579836 DOI: 10.1111/prd.12590] [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: 01/23/2024] [Revised: 05/03/2024] [Accepted: 06/11/2024] [Indexed: 07/06/2024]
Abstract
Increasing evidence suggests a significant association between periodontal disease and the occurrence of various cancers. The carcinogenic potential of several periodontal pathogens has been substantiated in vitro and in vivo. This review provides a comprehensive overview of the diverse mechanisms employed by different periodontal pathogens in the development of cancer. These mechanisms induce chronic inflammation, inhibit the host's immune system, activate cell invasion and proliferation, possess anti-apoptotic activity, and produce carcinogenic substances. Elucidating these mechanisms might provide new insights for developing novel approaches for tumor prevention, therapeutic purposes, and survival improvement.
Collapse
Affiliation(s)
- Yuxi Zhou
- Department of PeriodontologyJustus‐Liebig‐University of GiessenGiessenGermany
| | - Joerg Meyle
- Department of PeriodontologyJustus‐Liebig‐University of GiessenGiessenGermany
| | - Sabine Groeger
- Department of PeriodontologyJustus‐Liebig‐University of GiessenGiessenGermany
- Department of OrthodonticsJustus‐Liebig‐University of GiessenGiessenGermany
| |
Collapse
|
27
|
Ding X, Shao L, Wang J, Jin Y, Chen H, Li B. HADHA promotes esophageal cancer progression by activating mTOR signaling and the SP1/MDM2 axis. Acta Biochim Biophys Sin (Shanghai) 2024; 57:378-388. [PMID: 39327932 PMCID: PMC11986453 DOI: 10.3724/abbs.2024139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/30/2024] [Indexed: 09/28/2024] Open
Abstract
Esophageal cancer (EC) is one of the most recalcitrant cancers, with a 5-year survival rate of < 30%. The hydroxyacyl-CoA dehydrogenase alpha subunit (HADHA) plays an essential role in long-chain fatty acid metabolism, and dysregulation of HADHA has been demonstrated to be involved in a series of metabolic diseases and cancers. However, its role in cancers remains controversial. HADHA has seldom been investigated in EC, and little is known about how HADHA regulates the malignant progression of EC. In this study, we find that HADHA is significantly upregulated in EC tissues and is correlated with poor survival. HADHA knockdown markedly inhibits EC cell proliferation both in vitro and in vivo. The loss of HADHA also induces EC cell apoptosis, causes cell cycle arrest and inhibits cell migration. Additionally, RNA profiling reveals that mTOR signaling is significantly suppressed after HADHA knockdown. Mechanistically, HADHA interacts with SP1 and induces MDM2 expression. In conclusion, both mTOR signaling and the SP1-MDM2 axis participate in the HADHA-induced malignant behavior of EC cells.
Collapse
Affiliation(s)
- Xusheng Ding
- Departments of Thoracic Surgery and State Key Laboratory of Genetic EngineeringFudan University Shanghai Cancer CenterShanghai200032China
- Institute of Thoracic OncologyFudan UniversityShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Longlong Shao
- Departments of Thoracic Surgery and State Key Laboratory of Genetic EngineeringFudan University Shanghai Cancer CenterShanghai200032China
- Institute of Thoracic OncologyFudan UniversityShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Jie Wang
- Departments of Thoracic Surgery and State Key Laboratory of Genetic EngineeringFudan University Shanghai Cancer CenterShanghai200032China
- Institute of Thoracic OncologyFudan UniversityShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Yongwei Jin
- Department of Thoracic SurgeryFudan University Shanghai Cancer Center Xiamen HospitalXiamen361026China
| | - Haiqing Chen
- Departments of Thoracic Surgery and State Key Laboratory of Genetic EngineeringFudan University Shanghai Cancer CenterShanghai200032China
- Institute of Thoracic OncologyFudan UniversityShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Bin Li
- Departments of Thoracic Surgery and State Key Laboratory of Genetic EngineeringFudan University Shanghai Cancer CenterShanghai200032China
- Institute of Thoracic OncologyFudan UniversityShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| |
Collapse
|
28
|
Nemati S, Islami F, Kamangar F, Poustchi H, Roshandel G, Shakeri R, Domingues A, Khoshnia M, Gharavi A, Brennan P, Abnet CC, Dawsey SM, Boffetta P, Malekzadeh R, Sheikh M. Improvement of esophageal cancer survival in Northeast Iran: A two-decade journey in a high-risk, low- resource region. PLoS One 2024; 19:e0310842. [PMID: 39321196 PMCID: PMC11423987 DOI: 10.1371/journal.pone.0310842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 09/07/2024] [Indexed: 09/27/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Two decades ago, an international initiative (GEMINI) was launched in a high-risk, low-resource region in Northeast Iran, aiming to investigate incidence, etiology, early detection, and treatment of esophageal squamous cell carcinoma (ESCC). An earlier report from this area, highlighted poor ESCC survival rates, with a 5-year survival probability of 3.3% and the median survival time of 7 months. Our study assesses whether ESCC survival has improved since the implementation of the GEMINI initiative in this region. MATERIAL AND METHODS 490 adult patients with histologically-confirmed ESCC were recruited from the Atrak clinic, Golestan, Iran, between 2007 and 2018. At recruitment, information on demographics and various exposures were collected. Active (telephone surveys) and passive (linkage to Golestan population-based cancer and death registries) follow-up methods were used to determine patients' vital status though March 2019. Survival estimates were obtained by Kaplan-Meier method and Cox proportional hazards regression models. RESULTS Over the study period 340 deaths were recorded. Five-year ESCC survival probability was 23% (95% Confidence Interval: 19% to 28%), and the median survival time was 19 months. Five-year survival probability was higher among individuals who were younger (35% in <60-year-olds vs. 12% for >70-year-olds, p<0.001), educated (34% vs. 21% for no formal education, p = 0.027), never used opium (28% vs. 15%, p = 0.0016), and received cancer treatment (37% vs. 4%, p<0.001). In the adjusted models, a higher hazard of death was associated with older age [HR for each 10-year increase = 1.36 (95% CI = 1.22 to 1.51)], Turkman ethnicity [HR = 1.35 (95%CI: 1.07 to 1.70)], opium use [HR = 1.53 (95%CI: 1.20 to 1.94)],and receiving no cancer treatment [HR = 5.81 (95%CI: 3.97 to 8.52)]. CONCLUSION Over the last two decades, ESCC survival in this population has significantly improved, highlighting the potential of enhancing healthcare infrastructure and ensuring access to affordable medical care in resource-limited, high-risk regions. Older age at diagnosis, Turkman ethnicity, opium use, and untreated cases (indicative of advanced disease at diagnosis) were identified as the main ESCC prognostic factors in this population.
Collapse
Affiliation(s)
- Saeed Nemati
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC—WHO), Lyon, France
| | - Farhad Islami
- Surveillance and Health Services Equity Research, American Cancer Society, Atlanta, GA, United States of America
| | - Farin Kamangar
- Department of Biology, School of Computer, Mathematical, and Natural Sciences, Morgan State University, Baltimore, MD, United States of America
| | - Hossein Poustchi
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Roshandel
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ramin Shakeri
- Digestive Oncology Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Allison Domingues
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC—WHO), Lyon, France
| | - Masoud Khoshnia
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Abdolsamad Gharavi
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Paul Brennan
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC—WHO), Lyon, France
| | - Christian C. Abnet
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, United States of America
| | - Sanford M. Dawsey
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, United States of America
| | - Paolo Boffetta
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, United States of America
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Reza Malekzadeh
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
- Digestive Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Sheikh
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC—WHO), Lyon, France
| |
Collapse
|
29
|
Hu K, Huang T, Zhang Y, Ye Z, Guo J, Zhou H. A causal association between esophageal cancer and the oral microbiome: a Mendelian randomization study based on an Asian population. Front Cell Infect Microbiol 2024; 14:1420625. [PMID: 39346897 PMCID: PMC11427439 DOI: 10.3389/fcimb.2024.1420625] [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: 04/20/2024] [Accepted: 08/21/2024] [Indexed: 10/01/2024] Open
Abstract
Background Previous studies have suggested a crosstalk between the oral microbiome and esophageal cancer (EC), but the exact relationship is unclear. This study aimed to investigate the causal relationship between changes in the oral microbiome and EC by Mendelian randomization (MR). Materials and methods In the study, bidirectional MR analyses were conducted using genome-wide association study data from the oral microbiomes from the 4D-SZ cohort and EC data from the BioBank Japan cohort. Multiple sensitivity tests, including Cochrane's Q statistic, MR-Egger intercept, and MR-PRESSO, were used to assess and validate the relative stability of the resulting data at various levels. Results Among the 3,117 samples studied, 73 oral microbiomes were found to be statistically causally associated with EC, 38 of which were considered protective factors. According to species analyses, positive results were concentrated in three phyla: Firmicutes (29 species), Patescibacteria (18 species), and Actinobacteria (9 species). It was also determined that Parvimonas micra, Aggregatibacter, and Clostridia had a negative causal relationship, implying that EC caused a decrease in the counts. Following p-value correction, periodonticum_C, unclassified_mgs_3234, and unclassified_mgs_45 were identified as having a strong evidence-grade causal relationship with EC. There was no strong evidence in the results of the inverse MR analyses of EC to the oral microbiome. The sensitivity analysis confirmed the robustness of the findings. Conclusion This study discovered a bidirectional causal relationship between the oral microbiome and EC, which may provide new insights into the future use of the microbiome for early screening and probiotic therapy.
Collapse
Affiliation(s)
- Keke Hu
- Department of Oncology, Hangzhou Traditional Chinese Medicine (TCM) Hospital Affiliated
to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ting Huang
- Department of Oncology, Hangzhou Traditional Chinese Medicine (TCM) Hospital Affiliated
to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yiming Zhang
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Shandong
First Medical University, Jining, Shandong, China
| | - Zhifeng Ye
- Department of Oncology, Hangzhou Traditional Chinese Medicine (TCM) Hospital Affiliated
to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Junhua Guo
- Department of Oncology, Hangzhou Traditional Chinese Medicine (TCM) Hospital Affiliated
to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Heran Zhou
- Department of Oncology, Hangzhou Traditional Chinese Medicine (TCM) Hospital Affiliated
to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
30
|
Sha Y, Reyimu A, Liu W, He C, Kaisaier A, Paerhati P, Li L, Zou X, Xu A, Cheng X, Abuduaini M. Construction and validation of a prognostic model for esophageal cancer based on prognostic-related RNA-binding protein. Medicine (Baltimore) 2024; 103:e39639. [PMID: 39287291 PMCID: PMC11404941 DOI: 10.1097/md.0000000000039639] [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: 12/29/2023] [Revised: 04/12/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Construction of a prognostic model for esophageal cancer (ESCA) based on prognostic RNA-binding proteins (RBPs) and preliminary evaluation of RBP function. METHODS RNA-seq data of ESCA was downloaded from The Cancer Genome Atlas database and mRNA was extracted to screen differentially expressed genes using R. After screening RBPs in differentially expressed genes, R packages clusterProfiler and pathview were used to analyze the RBPs for Gene Ontology enrichment and Kyoto Encyclopedia of Genes and Genomes pathway. Based on the prognosis-related RBPs, COX regression was used to establish the prognostic risk model of ESCA. Risk model predictive ability was assessed using calibration analysis, receiver operating characteristic curves, Kaplan-Meier curves, decision curve analysis, and Harrell consistency index (C-index). A nomogram was established by combining the risk model with clinicopathological features. RESULTS A total of 105 RBPs were screened from ESCA. A prognostic risk model consisting of 6 prognostic RBPs (ARHGEF28, BOLL, CIRBP, DKC1, SNRPB, and TRIT1) was constructed by COX regression analysis. The prognosis was worse in the high-risk group, and the receiver operating characteristic curve showed (area under the curve = 0.90) that the model better predicted patients' 5-year survival. In addition, 6 prognostic RBPs had good diagnostic power for ESCA. In addition, a total of 39 mRNAs were identified as predicted target molecules for DKC1. CONCLUSION ARHGEF28, BOLL, CIRBP, DKC1, SNRPB, and TRIT1, as RBPs, are associated with the prognosis of ESCA, which may provide new ideas for targeted therapy of ESCA.
Collapse
Affiliation(s)
- Yinzhong Sha
- Department of Laboratory Medicine, The First People’s Hospital of Kashi, Kashi City, China
| | - Abdusemer Reyimu
- Department of Laboratory Medicine, The First People’s Hospital of Kashi, Kashi City, China
| | - Wen Liu
- Department of Laboratory Medicine, The First People’s Hospital of Kashi, Kashi City, China
| | - Chuanjiang He
- Department of Laboratory Medicine, The First People’s Hospital of Kashi, Kashi City, China
| | | | - Pawuziye Paerhati
- Department of Laboratory Medicine, The First People’s Hospital of Kashi, Kashi City, China
| | - Li Li
- The First People’s Hospital of Kashi, Kashi City, China
| | - Xiaoguang Zou
- The First People’s Hospital of Kashi, Kashi City, China
| | - Aimin Xu
- Department of Laboratory Medicine, The First People’s Hospital of Kashi, Kashi City, China
| | - Xiang Cheng
- Department of Laboratory Medicine, The First People’s Hospital of Kashi, Kashi City, China
| | | |
Collapse
|
31
|
Bao H, Bao H, Lin L, Wang Y, Zhang L, Zhang L, Zhang H, Liu L, Cao X. Radical chemoradiotherapy for superficial esophageal cancer complicated with liver cirrhosis. PeerJ 2024; 12:e18065. [PMID: 39282115 PMCID: PMC11401512 DOI: 10.7717/peerj.18065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024] Open
Abstract
Background Although chemoradiotherapy is an effective treatment for esophageal cancer, its feasibility in esophageal cancer with cirrhosis remains largely unclear. Methods We retrospectively studied 11 patients with superficial esophageal cancer with liver cirrhosis (Child-Pugh score ≤8) who underwent radical chemoradiotherapy from four centers, and the overall survival rate, local control rate and adverse events at 1 and 3 years were explored. Results The median age of the included patients was 67 years (Inter-Quartile Range 60-75 years). Complete response was observed in most patients (n = 10, 90.9%), and the remaining patient was unevaluable. The 1- and 3-year overall survival and local control rates were 90.9% and 90.9%, and 72.7% and 63.6%, respectively. Hematotoxicity was a common adverse reaction, and seven patients developed radiation esophagitis, with grade 3-4 observed in two cases. All cases of radiation dermatitis (n = 4) and radiation pneumonia (n = 2) were grade 1-2. Gastrointestinal bleeding occurred in two patients, including one with grade 1-2 bleeding, and one died. Conclusion Radical chemoradiotherapy is a potential treatment option for patients with superficial esophageal cancer complicated with cirrhosis. However, it can increase the risk of bleeding, which warrants prompt recognition and intervention.
Collapse
Affiliation(s)
- Hejing Bao
- Department of Oncology, The Affiliated Panyu Center Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Cancer Institute of Panyu, Guangzhou, China
| | - Hehong Bao
- Department of Psychosomatic Medicine, Chongqing University Three Gorges Hospital, Wanzhou, Chongqing, China
| | - Liping Lin
- Department of Oncology, The Affiliated Panyu Center Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Cancer Institute of Panyu, Guangzhou, China
| | - Yuhuan Wang
- Department of Oncology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Longbin Zhang
- Department of Oncology, Chongqing University Three Gorges Hospital, Wanzhou, Chongqing, China
| | - Li Zhang
- Department of Oncology, Chongqing University Three Gorges Hospital, Wanzhou, Chongqing, China
| | - Han Zhang
- Department of Oncology, Chongqing University Three Gorges Hospital, Wanzhou, Chongqing, China
| | - Lingxiang Liu
- Department of Oncology, The Affiliated Panyu Center Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Cancer Institute of Panyu, Guangzhou, China
| | - Xiaolong Cao
- Department of Oncology, The Affiliated Panyu Center Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Cancer Institute of Panyu, Guangzhou, China
| |
Collapse
|
32
|
Zhang X, An Y, Mai D, Huang W, Zeng W. Modulation of esophageal squamous cell carcinoma progression: the impact of CCR7 on JAK2/STAT3 signaling pathway. Discov Oncol 2024; 15:421. [PMID: 39254762 PMCID: PMC11387284 DOI: 10.1007/s12672-024-01289-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/29/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Existing studies have already revealed the involvement of C-C chemokine receptor type 7 (CCR7) in diverse human cancers, including esophageal cell squamous carcinoma (ESCA). Our current study, aims to explore the relevant mechanisms implicated. METHODS ESCA cell lines were collected for CCR7 expression quantification using western blot. Following the transfection, the viability, migration and invasion of ESCA cells were evaluated via cell counting kit-8 and Transwell assays. The specific molecular mechanisms underlying the effects of CCR7 in ESCA cells were explored via calculating the expressions of proteins related to metastasis and Janus kinase 2/signal transduction and transcription activation 3 (JAK2/STAT3) signaling pathway via western blot. The correlation between CCR7 and metastasis-related proteins was explored via Pearson's correlation test. RESULTS CCR7 was high-expressed in ESCA cells and CCR7 knockdown repressed the viability, migration and invasion of ESCA cells, concurrent with the increased expression of E-cadherin (E-cad, which was also known as CDH1 and lowly expressed in ESCA cells) and the decreased expressions of vimentin (Vim, which was highly expressed in ESCA cells) and matrix metalloproteinase-9 (MMP-9, which was also highly expressed in ESCA cells). Meanwhile, CCR7 was positively correlated with Vim and MMP-9 yet negatively correlated with E-cad in ESCA cells, which indicated that CCR7 has a role in promoting tumor progression in ESCA cells. Besides, the phosphorylation of STAT3 and JAK2 in ESCA cells was elevated, which was diminished following CCR7 knockdown. CONCLUSION This study proves the modulation of CCR7 on ESCA in vitro, which was achieved via JAK2/STAT3 signaling pathway. Our discovery will provide new therapeutic basis and insights for ESCA.
Collapse
Affiliation(s)
- Xuewen Zhang
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Yuji An
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Five Wards of Oncology Department, The Third Affiliated Hospital of Shandong First Medical University, Jinan, 250031, China
| | - Dongmei Mai
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Wan Huang
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Weian Zeng
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
33
|
Ke J, Xie Y, Huang S, Wang W, Zhao Z, Lin W. Comparison of esophageal cancer survival after neoadjuvant chemoradiotherapy plus surgery versus definitive chemoradiotherapy: A systematic review and meta-analysis. Asian J Surg 2024; 47:3827-3840. [PMID: 38448293 DOI: 10.1016/j.asjsur.2024.02.099] [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: 08/10/2023] [Revised: 12/31/2023] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Abstract
Surgery after neoadjuvant chemoradiotherapy remains the gold standard for the treatment of resectable esophageal cancer (EC); however, chemoradiotherapy without surgery has been recommended in specific cases. The aim of this meta-analysis is to analyse the survival between surgeries after neoadjuvant chemoradiotherapy compared with definitive chemoradiotherapy in order to provide a theoretical basis for clinically individualised differential treatment. We conducted an initial search of MEDLINE (PubMed), the Cochrane Library, and Embase for English-only articles that compared treatment regimens and provided survival data. According to the final I2 value of the two survival indicators, the random effect model or fixed effect model was used to calculate the overall hazard ratio (HR) and 95% confidence intervals (CI). Cochrane's Q test was used to judge the heterogeneity of the studies, and a funnel plot was used to evaluate for publication bias. A sensitivity analysis was performed to verify the stability of the included studies. A total of 38 studies involving 29161 patients (neoadjuvant therapy: 15401, definitive chemoradiotherapy: 13760) were included in the analysis. The final pooled results (HR = 0.74, 95% CI: 0.67-0.82) showed a statistically significant increase in overall survival with neoadjuvant chemoradiotherapy plus surgery compared with definitive chemoradiotherapy. Subgroup analyses were performed to determine the effects of heterogeneity, additional treatment regimens, study types, and geographic regions, as well as histologic differences, complications, and recurrence, on the overall results. For people with esophageal cancer that can be removed, neoadjuvant chemoradiotherapy combined with surgery improves survival compared to definitive chemoradiotherapy. However, more research is needed to confirm these results and help doctors make decisions about treatment.
Collapse
Affiliation(s)
- Junli Ke
- Department of Thoracic Surgery, Gaozhou People's Hospital Affiliated to Guangdong Medical University, Maoming, China
| | - Yujie Xie
- Department of Thoracic Surgery, Gaozhou People's Hospital Affiliated to Guangdong Medical University, Maoming, China
| | - Shenyang Huang
- Department of Cardiothoracic Surgery, Guangdong Medical University, Zhanjiang, China
| | - Wei Wang
- Graduate School of Guangdong Medical University, Zhanjiang, China
| | - Zhengang Zhao
- Department of Cardiothoracic Surgery, Guangdong Medical University, Zhanjiang, China
| | - Wanli Lin
- Department of Thoracic Surgery, Gaozhou People's Hospital Affiliated to Guangdong Medical University, Maoming, China.
| |
Collapse
|
34
|
Chen M, Qi Y, Zhang S, Du Y, Cheng H, Gao S. Screening of genes related to programmed cell death in esophageal squamous cell carcinoma and construction of prognostic model based on transcriptome analysis. Expert Rev Anticancer Ther 2024; 24:905-915. [PMID: 38975629 DOI: 10.1080/14737140.2024.2377184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/19/2024] [Indexed: 07/09/2024]
Abstract
OBJECTIVES To screen programmed cell death (PCD)-related genes in esophageal squamous cell carcinoma (ESCC) based on transcriptomic data and to explore its clinical value. METHODS Differentially expressed PCD genes (DEPCDGs) were screened from ESCC transcriptome and clinical data in TCGA database. Univariate COX and LASSO COX were performed on prognostically DEPCDGs in ESCC to develop prognostic model. Differences in immune cell infiltration in different RiskScore groups were determined by ssGSEA and CIBERSORT. The role of RiskScore in immunotherapy response was explored using Tumor Immune Dysfunction and Exclusion (TIDE) and IMvigor210 cohorts. RESULTS Fourteen DEPCDGs associated with prognosis were tapped in ESCC. These DEPCDGs form a RiskScore with good predictive performance for prognosis. RiskScore demonstrated excellent prediction accuracy in three data sets. The abundance of M2 macrophages and Tregs was higher in the high RiskScore group, and the abundance of M1 macrophages was higher in the low RiskScore group. The RiskScore also showed good immunotherapy sensitivity. RT-qPCR analysis showed that AUP1, BCAP31, DYRK2, TAF9 and UBQLN2 were higher expression in KYSE-150 cells. Knockdown BCAP31 inhibited migration and invasion. CONCLUSION A prognostic risk model can predict prognosis of ESCC and may be a useful biomarker for risk stratification and immunotherapy assessment.
Collapse
Affiliation(s)
- Min Chen
- School of Information Engineering, Henan University of Science and Technology, Luoyang, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Cancer Hospital, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
- Medical College, Henan University of Science and Technology, Luoyang, China
| | - Yijun Qi
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Cancer Hospital, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
- Medical College, Henan University of Science and Technology, Luoyang, China
| | - Shenghua Zhang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Cancer Hospital, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
- Medical College, Henan University of Science and Technology, Luoyang, China
| | - Yubo Du
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Cancer Hospital, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
- Medical College, Henan University of Science and Technology, Luoyang, China
| | - Haodong Cheng
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Cancer Hospital, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
- Medical College, Henan University of Science and Technology, Luoyang, China
| | - Shegan Gao
- School of Information Engineering, Henan University of Science and Technology, Luoyang, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- Cancer Hospital, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
- College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
- Medical College, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
35
|
Shao L, Li B. Synaptotagmin 13 Could Drive the Progression of Esophageal Squamous Cell Carcinoma Through Upregulating ACRV1. DNA Cell Biol 2024; 43:452-462. [PMID: 39046915 DOI: 10.1089/dna.2024.0106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024] Open
Abstract
SYT13 is one of the atypical members of the synaptotagmin (SYT) family whose function has attracted considerable attention in recent years. Although SYT13 has been studied in several types of human cancers, such as lung cancer, its role in esophageal squamous cell carcinoma (ESCC) is still unclear. It was demonstrated that SYT13 is significantly upregulated in ESCC tissues compared with normal ones and correlated with higher degree of malignancy. Knockdown of SYT13 could inhibit ESCC cell proliferation and migration, while promoting cell apoptosis. Meanwhile, ESCC cells with relatively lower SYT13 expression grew slower in vivo and finally formed smaller xenografts. Furthermore, acrosomal vesicular protein 1 was identified as a potential downstream target of SYT13, which regulates cell phenotypes of ESCC cells in cooperation with SYT13. All the in vitro and in vivo results in this study identified that SYT13 silencing could be an effective strategy to inhibit the development of ESCC, which could be considered as a promising therapeutic target in the treatment of ESCC.
Collapse
Affiliation(s)
- Longlong Shao
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Bin Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
36
|
Lu Q, Yang Q, Zhao J, Li G, Zhang J, Jia C, Wan Y, Chen Y. The identification of heterogeneous reactive oxygen subtypes in esophageal squamous cell carcinoma to aid patient prognosis and immunotherapy. Heliyon 2024; 10:e35235. [PMID: 39165982 PMCID: PMC11334838 DOI: 10.1016/j.heliyon.2024.e35235] [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: 01/20/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
INTRODUCTION Esophageal cancer is increasingly recognized as a significant global malignancy. The main pathological subtype of this cancer is esophageal squamous cell carcinoma (ESCC), which displays a higher degree of malignancy and a poorer prognosis. Reactive oxygen species (ROS) play a critical role in modulating the immune response to tumors, and understanding the regulation of ROS in ESCC could lead to novel and improved therapeutic strategies for ESCC patients. METHODS A consensus matrix derived from genes involved in the ROS pathway revealed two subtypes of ROS. These subtypes were categorized as ROS-active or ROS-suppressive based on their level of ROS activity. The heterogeneity among the different ROS subtypes was then explored from various perspectives, including gene function, immune response, genomic stability, and immunotherapy. In order to assess the prognosis and the potential benefits of immunotherapy, a ROS activity score (RAS) was developed using the identified ROS subtypes. In vitro experiments were performed to confirm the impact of core RAS genes on the proliferative activity of esophageal cancer cell lines. RESULTS Two distinctive subtypes of ROS were identified. The first subtype, referred to as ROS-active, exhibited elevated ROS activity, enhanced involvement in cancer-associated immune pathways, and increased infiltration of effector immune cells. The second subtype, named ROS-suppressive, demonstrated weaker ROS activity but displayed more pronounced dysregulation in the cell cycle and a denser extracellular matrix, indicating malignant characteristics. Genomic stability, particularly in terms of copy number variation (CNV) events, differed between the two ROS subtypes. By developing a RAS model, reliable risk assessment for overall survival (OS) in patients with ESCC was achieved, and the model demonstrated strong predictive capabilities in real-world immunotherapy cohorts. Moreover, the core gene LDLRAD1 within the RAS model was found to enhance proliferative activity in esophageal cancer cell lines. CONCLUSION Based on the ROS pathway, we successfully identified two distinct subtypes in ESCC: the ROS-active subtype and the ROS-suppressive subtype. These subtypes were utilized to evaluate prognosis and the sensitivity to immunotherapy.
Collapse
Affiliation(s)
- Qiang Lu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, China
| | - Qi Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Jinbo Zhao
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, China
| | - Guizhen Li
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, China
| | - JiPeng Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, China
| | - Chenghui Jia
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, China
| | - Yi Wan
- Department of Health Service, Air Force Medical University, No.169 Changle West Road, Xi'an, 710032, China
| | - Yan Chen
- Department of Oncology, Xijing Hospital, Air Force Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| |
Collapse
|
37
|
Li N, Chen S, Wang X, Zhang B, Zeng B, Sun C, Zheng K, Chen Q, Wang S. Identification of POU4F1 as a novel prognostic biomarker and therapeutic target in esophageal squamous cell carcinoma. Cancer Cell Int 2024; 24:280. [PMID: 39123235 PMCID: PMC11316379 DOI: 10.1186/s12935-024-03471-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Esophageal cancer is a significant global health concern, ranking seventh in incidence and sixth in mortality. It encompasses two pathological types: esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma, with ESCC being more prevalent globally and associated with higher mortality rates. The POU (Pit-Oct-Unc) domain family transcription factors, comprising 15 members, play important roles in embryonic development and organ formation. Aberrant expression of POUs has been observed in several human cancers, influencing cell proliferation, tumor invasion, and drug resistance. However, their specific role in ESCC remains unknown. METHODS We analyzed TCGA and GEO databases to assess POUs expression in ESCC tissues. Kaplan-Meier and ROC analyses were used to evaluate the prognostic value of POUs. Gene Set Enrichment Analysis and Protein-Protein interaction network were used to explore the potential pathway. Functional assays (Cell Counting Kit-8, EdU Staining assay, and cloning formation assay) and mechanism analyses (RNA-seq, flow cytometry, and Western blot) were conducted to determine the effects of POU4F1 knockdown on ESCC cell phenotypes and signaling pathways. RESULTS POU4F1 and POU6F2 were upregulated in various cancer tissues, including ESCC, compared to normal tissues. POU4F1 expression was significantly correlated with patient survival and superior to previous models (AUC = 0.776). Knockdown of POU4F1 inhibited ESCC cell proliferation and affected cell cycle, autophagy, and DNA damage pathways in ESCC cells. CONCLUSION POU4F1 is a novel and promising prognostic and therapeutic target for ESCC patients, providing insights into potential treatment strategies.
Collapse
Affiliation(s)
- Nan Li
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, Guangdong, China
- Department of Pharmacy, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Siying Chen
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiao Wang
- Department of Pharmacy, The Second Clinical Medical College, The First Affiliated Hospital, Shenzhen People's Hospital, Jinan University, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Boqing Zhang
- Department of Pharmacy, The Second Clinical Medical College, The First Affiliated Hospital, Shenzhen People's Hospital, Jinan University, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Boning Zeng
- Department of Pharmacy, The Second Clinical Medical College, The First Affiliated Hospital, Shenzhen People's Hospital, Jinan University, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Chao Sun
- Department of Pharmacy, The Second Clinical Medical College, The First Affiliated Hospital, Shenzhen People's Hospital, Jinan University, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Kai Zheng
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, Guangdong, China
| | - Qiuling Chen
- Department of Pharmacy, The Second Clinical Medical College, The First Affiliated Hospital, Shenzhen People's Hospital, Jinan University, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Shaoxiang Wang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
38
|
Wang M, Miao H. Disease burden and related risk factors of esophageal cancer in China and globally from 1990 to 2021, with forecast to 2035: An analysis and comparison. Tob Induc Dis 2024; 22:TID-22-140. [PMID: 39091891 PMCID: PMC11292605 DOI: 10.18332/tid/191389] [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: 05/16/2024] [Revised: 07/18/2024] [Accepted: 07/21/2024] [Indexed: 08/04/2024] Open
Abstract
INTRODUCTION In this study we estimate the burden of esophageal cancer (EC) in China and globally from 1990 to 2021, with a forecast to 2035, using Global Burden of Disease (GBD) data. We also analyze the related risk factors to investigate burden trends. METHODS Mortality, disability-adjusted life years (DALYs), crude rates, and age-standardized rates of EC were analyzed in China and globally from 1990 to 2035, utilizing GBD open data as a secondary dataset analysis of GBD data. Temporal change trends of EC risk factors were analyzed from 1990 to 2021. Joinpoint regression determined average annual percentage change (AAPC) of age-standardized rates. Descriptive analysis compared mortality and DALYs by age groups. Bayesian age-period-cohort (BAPC) predicted age-standardized mortality and DALYs rates for the next 14 years. RESULTS The ASMR and ASDR fluctuations in EC were significant in China, showing an overall downward trend. Globally, although there was also a downward trend, the fluctuations were relatively mild. The number of deaths and DALYs related to EC in China and globally showed a significant upward trend. Age-specific burden trends in China for EC indicated that the age group with the peak number of EC deaths shifted to the 70-74 years age group in 2021, while DALYs peaked in the 65-69 years age group. The crude mortality rate (CMR) peaked consistently in 1990 and 2021, both within the 90-94 years age range, while the crude DALY rate (CDR) shifted to the 85-89 years age group. Overall, the burden of EC deaths and DALYs in the population aged <40 years was relatively low, increasing rapidly after the age of 40 years, reaching a peak and gradually declining, and reaching a lower level after the age of 85 years. The predictive results of the BAPC model indicated that over the next 14 years, both ASMR and ASDR for EC in China and globally would show a slight overall increase. The GBD 2021 study identified smoking, high alcohol use, chewing tobacco, and diet low in vegetables as the main risk factors affecting EC mortality rate and DALYs. Among these, smoking and alcohol use were the most significant risk factors, with a higher impact on EC in China compared to the global level. From 1990 to 2021, the overall changes in ASMR and ASDR indicate a decreasing trend in the impact of these four risk factors on EC mortality rate and DALYs. CONCLUSIONS The burden of EC is expected to steadily increase in China and globally until 2035, posing a significant challenge. Targeted prevention and control policies, such as calling on people to quit smoking and reduce alcohol use, may help curb this upward trend.
Collapse
Affiliation(s)
- Mimi Wang
- Science and Education Information Section, Hangzhou Center for Disease Control and Prevention, Hangzhou, China
| | - Huiwen Miao
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, China
| |
Collapse
|
39
|
Qi JC, Zhi L, Li H, Huang Y, Ye Y, Li H, Wang T, Lin L, Zhuang Y. Prognostic factors for esophageal respiratory fistula in unresectable esophageal squamous cell carcinoma treated with radiotherapy. Sci Rep 2024; 14:17144. [PMID: 39060397 PMCID: PMC11282182 DOI: 10.1038/s41598-024-67859-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Limited studies have focused on the prognostic factors of esophageal respiratory fistula (ERF) associated with radiotherapy in patients with unresectable esophageal squamous cell carcinoma (ESCC). Between January 1st, 2014 and January 1st, 2021, we included patients who were initially diagnosed with unresectable ESCC and underwent radiotherapy. All patients were followed up for a period of 2 years after completing their radiotherapy treatment. The primary outcomes of the study were defined as death or severe adverse events. The survival curves of ERF were calculated using the Kaplan-Meier method. Cox proportional hazards model was employed to calculated the prognostic factors. A cohort of 232 patients underwent radiotherapy, of whom 32 patients experienced ERF. The median period from initial diagnosis of ESCC to ERF was 5.75 months, and the median period from ERF to the primary outcome was 4.6 weeks. Neck + upper chest location (odds ratio [OR] 3.305), high T stage (OR 1.765), esophageal stenosis (OR 1.073), high neutrophil to lymphocyte ratio (NLR) (OR 1.384) and platelet to lymphocyte ratio (PLR) (OR 1.765) were risk factors for the occurrence of ERF. Cox regression analysis suggested that tumor location (hazards ratio [HR] 3.572, 95% confidence interval [CI] 2.467-5.1), high T stage (HR 4.050, 95% CI 2.812-5.831), esophageal stenosis (HR 2.643, 95% CI 1.753-3.983), high PLR (HR 2.541, 95% CI 1.868-3.177) were independent prognostic factors for poor survival. Esophageal stenosis, neck + upper chest tumor location, high T stage and PLR predicted the prognosis of ERF in ESCC patients undergoing radiotherapy.
Collapse
Affiliation(s)
- Jia-Chao Qi
- Department of Respiratory and Critical Care Medicine, Zhangzhou Affiliated Hospital of Fujian Medical University, No. 59, Shengli Rd, Xiangcheng District, Zhangzhou, 363000, Fujian, People's Republic of China
| | - Lijia Zhi
- Department of Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39, Twelve Bridges Rd, Jinniu District, Chengdu, 610075, Sichuan, People's Republic of China
| | - Huangyu Li
- Department of Respiratory and Critical Care Medicine, Zhangzhou Affiliated Hospital of Fujian Medical University, No. 59, Shengli Rd, Xiangcheng District, Zhangzhou, 363000, Fujian, People's Republic of China
| | - Yanping Huang
- Department of Respiratory and Critical Care Medicine, Zhangzhou Affiliated Hospital of Fujian Medical University, No. 59, Shengli Rd, Xiangcheng District, Zhangzhou, 363000, Fujian, People's Republic of China
| | - Yuming Ye
- Department of Respiratory and Critical Care Medicine, Zhangzhou Affiliated Hospital of Fujian Medical University, No. 59, Shengli Rd, Xiangcheng District, Zhangzhou, 363000, Fujian, People's Republic of China
| | - Hao Li
- Department of Respiratory and Critical Care Medicine, Zhangzhou Affiliated Hospital of Fujian Medical University, No. 59, Shengli Rd, Xiangcheng District, Zhangzhou, 363000, Fujian, People's Republic of China
| | - Tiezhu Wang
- Department of Respiratory and Critical Care Medicine, Zhangzhou Affiliated Hospital of Fujian Medical University, No. 59, Shengli Rd, Xiangcheng District, Zhangzhou, 363000, Fujian, People's Republic of China
| | - Li Lin
- Department of Respiratory and Critical Care Medicine, Zhangzhou Affiliated Hospital of Fujian Medical University, No. 59, Shengli Rd, Xiangcheng District, Zhangzhou, 363000, Fujian, People's Republic of China.
| | - Yuezhen Zhuang
- Department of Gynecology, Zhangzhou Affiliated Hospital of Fujian Medical University, No. 59, Shengli Rd., Xiangcheng District, Zhangzhou, 363000, Fujian, People's Republic of China.
| |
Collapse
|
40
|
Wu Y, Hu W, Jia Z, Zhu Q, Xu J, Peng L, Wang R. Impact of Extracellular Matrix-Related Genes on the Tumor Microenvironment and Prognostic Indicators in Esophageal Cancer: A Comprehensive Analytical Study. Genet Res (Camb) 2024; 2024:3577395. [PMID: 39139739 PMCID: PMC11300105 DOI: 10.1155/2024/3577395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/10/2024] [Accepted: 07/03/2024] [Indexed: 08/15/2024] Open
Abstract
Esophageal cancer is a major global health challenge with a poor prognosis. Recent studies underscore the extracellular matrix (ECM) role in cancer progression, but the full impact of ECM-related genes on patient outcomes remains unclear. Our study utilized next-generation sequencing and clinical data from esophageal cancer patients provided by The Cancer Genome Atlas, employing the R package in RStudio for computational analysis. This analysis identified significant associations between patient survival and various ECM-related genes, including IBSP, LINGO4, COL26A1, MMP12, KLK4, RTBDN, TENM1, GDF15, and RUNX1. Consequently, we developed a prognostic model to predict patient outcomes, which demonstrated clear survival differences between high-risk and low-risk patient groups. Our comprehensive review encompassed clinical correlations, biological pathways, and variations in immune response among these risk categories. We also constructed a nomogram integrating clinical information with risk assessment. Focusing on the TENM1 gene, we found it significantly impacts immune response, showing a positive correlation with T helper cells, NK cells, and CD8+ T cells, but a negative correlation with neutrophils and Th17 cells. Gene Set Enrichment Analysis revealed enhanced pathways related to pancreatic beta cells, spermatogenesis, apical junctions, and muscle formation in patients with high TENM1 expression. This research provides new insights into the role of ECM genes in esophageal cancer and informs future research directions.
Collapse
Affiliation(s)
- Yinghong Wu
- The Second People's Hospital of Jingdezhen, Jingdezhen 333000, Jiangxi, China
| | - Wenjie Hu
- The Second People's Hospital of Jingdezhen, Jingdezhen 333000, Jiangxi, China
| | - Zhihong Jia
- The Second People's Hospital of Jingdezhen, Jingdezhen 333000, Jiangxi, China
| | - Qiying Zhu
- The Second People's Hospital of Jingdezhen, Jingdezhen 333000, Jiangxi, China
| | - Jinghui Xu
- The Second People's Hospital of Jingdezhen, Jingdezhen 333000, Jiangxi, China
| | - Liang Peng
- The Second People's Hospital of Jingdezhen, Jingdezhen 333000, Jiangxi, China
| | - Renjie Wang
- The Second People's Hospital of Jingdezhen, Jingdezhen 333000, Jiangxi, China
| |
Collapse
|
41
|
Pan H, Hong J, Shao A, Zhao Z, Ding G, Fang Z, Chen K, Zhu J. Keratin 17 and Collagen type 1 genes: Esophageal cancer molecular marker discovery and evaluation. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e13793. [PMID: 38979664 PMCID: PMC11231643 DOI: 10.1111/crj.13793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 07/10/2024]
Abstract
One hundred eighty pairs of tissues of esophageal squamous cell carcinoma (ESCC) were tested by the transcriptome sequencing in order to explore etiology factors. The chi-square test and correlation analysis demonstrated that the relative expression levels of keratin 17 (KRT17) and collagen type I α1 chain (COL1A1) were significantly higher in EC with diabetes. Expression of KRT17 was correlated with blood glucose (r = 0.204, p = 0.001) and tumor size (r = -0.177, p = 0.038) in patients. COL1A1 correlated with age (r = -0.170, p = 0.029) and blood glucose levels (r = 0.190, p = 0.015). Experimental results of qRT-PCR: KRT17 and COL1A1 genes were highly expressed in ESCC (p < 0.05). When the two genes were used as a combination test, the positive detection rate of EC was 90.6%, and the ROC curve had greater power. The KRT17 and COL1A1 genes had the potential to be biomarkers for the diagnosis of ESCC.
Collapse
Affiliation(s)
- Huiwen Pan
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
- The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Jie Hong
- The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Aizhong Shao
- The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Zhiguo Zhao
- The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Guowen Ding
- The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Zhijie Fang
- Department of Otolaryngology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzou, China
| | - Keping Chen
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Jingfeng Zhu
- The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
42
|
Wang Z, Sun X, Li Z, Yu H, Li W, Xu Y. Metabolic reprogramming in esophageal squamous cell carcinoma. Front Pharmacol 2024; 15:1423629. [PMID: 38989149 PMCID: PMC11233760 DOI: 10.3389/fphar.2024.1423629] [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: 04/26/2024] [Accepted: 06/06/2024] [Indexed: 07/12/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a malignancy with high incidence in China. Due to the lack of effective molecular targets, the prognosis of ESCC patients is poor. It is urgent to explore the pathogenesis of ESCC to identify promising therapeutic targets. Metabolic reprogramming is an emerging hallmark of ESCC, providing a novel perspective for revealing the biological features of ESCC. In the hypoxic and nutrient-limited tumor microenvironment, ESCC cells have to reprogram their metabolic phenotypes to fulfill the demands of bioenergetics, biosynthesis and redox homostasis of ESCC cells. In this review, we summarized the metabolic reprogramming of ESCC cells that involves glucose metabolism, lipid metabolism, and amino acid metabolism and explore how reprogrammed metabolism provokes novel opportunities for biomarkers and potential therapeutic targets of ESCC.
Collapse
Affiliation(s)
- Ziyi Wang
- Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Thoracic Surgery, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiangyu Sun
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zehui Li
- Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Huidong Yu
- Department of Thoracic Surgery, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wenya Li
- Department of Thoracic Surgery, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yan Xu
- Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
43
|
Guo J, Zhao Y, Sui H, Liu L, Liu F, Yang L, Gao F, Wang J, Zhu Y, Li L, Song X, Li P, Tian Z, Li P, Zhao X. USP21-mediated G3BP1 stabilization accelerates proliferation and metastasis of esophageal squamous cell carcinoma via activating Wnt/β-Catenin signaling. Oncogenesis 2024; 13:23. [PMID: 38906857 PMCID: PMC11192907 DOI: 10.1038/s41389-024-00524-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 06/23/2024] Open
Abstract
Lacking effective therapeutic targets heavily restricts the improvement of clinical prognosis for patients diagnosed with esophageal squamous cell carcinoma (ESCC). Ubiquitin Specific Peptidase 21 (USP21) is dysregulated in plenty of human cancers, however, its potential function and relevant molecular mechanisms in ESCC malignant progression as well as its value in clinical translation remain largely unknown. Here, in vitro and in vivo experiments revealed that aberrant upregulation of USP21 accelerated the proliferation and metastasis of ESCC in a deubiquitinase-dependent manner. Mechanistically, we found that USP21 binds to, deubiquitinates, and stabilizes the G3BP Stress Granule Assembly Factor 1 (G3BP1) protein, which is required for USP21-mediated ESCC progression. Further molecular studies demonstrated that the USP21/G3BP1 axis played a tumor-promoting role in ESCC progression by activating the Wnt/β-Catenin signaling pathway. Additionally, disulfiram (DSF), an inhibitor against USP21 deubiquitylation activity, markedly abolished the USP21-mediated stability of G3BP1 protein and significantly displayed an anti-tumor effect on USP21-driving ESCC progression. Finally, the regulatory axis of USP21/G3BP1 was demonstrated to be aberrantly activated in ESCC tumor tissues and closely associated with advanced clinical stages and unfavorable prognoses, which provides a promising therapeutic strategy targeting USP21/G3BP1 axis for ESCC patients.
Collapse
Affiliation(s)
- Jiazhong Guo
- Department of Critical Care Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yunpeng Zhao
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huacong Sui
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lei Liu
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fanrong Liu
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lingxiao Yang
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fengyuan Gao
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jinfu Wang
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yilin Zhu
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lingbing Li
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiangqing Song
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peng Li
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhongxian Tian
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Chest Cancer, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peichao Li
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Key Laboratory of Chest Cancer, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Xiaogang Zhao
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Key Laboratory of Chest Cancer, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
44
|
Zhang L, Wang Y, Gao J, Zhou X, Huang M, Wang X, He Z. Non‑coding RNA: A promising diagnostic biomarker and therapeutic target for esophageal squamous cell carcinoma (Review). Oncol Lett 2024; 27:255. [PMID: 38646493 PMCID: PMC11027111 DOI: 10.3892/ol.2024.14388] [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: 11/04/2023] [Accepted: 03/22/2024] [Indexed: 04/23/2024] Open
Abstract
Esophageal cancer (EC) is a common form of malignant tumor in the digestive system that is classified into two types: Esophageal squamous cell carcinomas (ESCC) and esophageal adenocarcinoma. ESCC is known for its early onset of symptoms, which can be difficult to identify, as well as its rapid progression and tendency to develop drug resistance to chemotherapy and radiotherapy. These factors contribute to the high incidence of disease and low cure rate. Therefore, a diagnostic biomarker and therapeutic target need to be identified for ESCC. Non-coding RNAs (ncRNAs) are a class of molecules that are transcribed from DNA but do not encode proteins. Initially, ncRNAs were considered to be non-functional segments generated during transcription. However, with advancements in high-throughput sequencing technologies in recent years, ncRNAs have been associated with poor prognosis, drug resistance and progression of ESCC. The present study provides a comprehensive overview of the biogenesis, characteristics and functions of ncRNAs, particularly focusing on microRNA, long ncRNAs and circular RNAs. Furthermore, the ncRNAs that could potentially be used as diagnostic biomarkers and therapeutic targets for ESCC are summarized to highlight their application value and prospects in ESCC.
Collapse
Affiliation(s)
- Longze Zhang
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Yanyang Wang
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Department of Cell Engineering Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jianmei Gao
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Xue Zhou
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Minglei Huang
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Xianyao Wang
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Zhixu He
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Department of Cell Engineering Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
45
|
Zhang P, Wen B, Gong J, Liu Z, Zhang M, Zhou G, Zhang L, Zhang Z. Clinical prognostication and immunotherapy response prediction in esophageal squamous cell carcinoma using the DNA damage repair-associated signature. ENVIRONMENTAL TOXICOLOGY 2024; 39:2803-2816. [PMID: 38287713 DOI: 10.1002/tox.24155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/06/2024] [Accepted: 01/18/2024] [Indexed: 01/31/2024]
Abstract
BACKGROUND The relationship between DNA damage repair (DDR) and cancer is intricately intertwined; however, its specific role in esophageal squamous cell carcinoma (ESCC) remains enigmatic. METHODS Employing single-cell analysis, we delineated the functionality of DDR-related genes within the tumor microenvironment (TME). A diverse array of scoring mechanisms, including AUCell, UCell, singscore, ssgsea, and AddModuleScore, were harnessed to scrutinize the activity of DDR-related genes across different cell types. Differential pathway alterations between high-and low-DDR activity cell clusters were compared. Furthermore, leveraging multiple RNA-seq datasets, we constructed a robust DDR-associated signature (DAS), and through integrative multiomics analysis, we explored differences in prognosis, pathways, mutational landscapes, and immunotherapy predictions among distinct DAS groups. RESULTS Notably, high-DDR activity cell subpopulations exhibited markedly enhanced cellular communication. The DAS demonstrated uniformity across multiple datasets. The low-DAS group exhibited improved prognoses, accompanied by heightened immune infiltration and elevated immune checkpoint expression. SubMap analysis of multiple immunotherapy datasets suggested that low-DAS group may experience enhanced immunotherapy responses. The "oncopredict" R package analyzed and screened sensitive drugs for different DAS groups. CONCLUSION Through the integration of single-cell and bulk RNA-seq data, we have developed a DAS associated with prognosis and immunotherapy response. This signature holds promise for the future stratification and personalized treatment of ESCC patients in clinical settings.
Collapse
Affiliation(s)
- Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Bing Wen
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Cardiothoracic Surgery, The Second People's Hospital of Yibin, Yibin, Sichuan, China
| | - Jialin Gong
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zuo Liu
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Mengzhe Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Guangyao Zhou
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lianmin Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhenfa Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
46
|
Luan M, Feng Z, Zhu W, Xing Y, Ma X, Zhu J, Wang Y, Jia Y. Mechanism of metal ion-induced cell death in gastrointestinal cancer. Biomed Pharmacother 2024; 174:116574. [PMID: 38593706 DOI: 10.1016/j.biopha.2024.116574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/26/2024] [Accepted: 04/05/2024] [Indexed: 04/11/2024] Open
Abstract
Gastrointestinal (GI) cancer is one of the most severe types of cancer, with a significant impact on human health worldwide. Due to the urgent demand for more effective therapeutic strategies against GI cancers, novel research on metal ions for treating GI cancers has attracted increasing attention. Currently, with accumulating research on the relationship between metal ions and cancer therapy, several metal ions have been discovered to induce cell death. In particular, the three novel modes of cell death, including ferroptosis, cuproptosis, and calcicoptosis, have become focal points of research in the field of cancer. Meanwhile, other metal ions have also been found to trigger cell death through various mechanisms. Accordingly, this review focuses on the mechanisms of metal ion-induced cell death in GI cancers, hoping to provide theoretical support for further GI cancer therapies.
Collapse
Affiliation(s)
- Muhua Luan
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan 250013, People's Republic of China; Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Zhaotian Feng
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China; Department of Medical Laboratory, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Wenshuai Zhu
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Yuanxin Xing
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Xiaoli Ma
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Jingyu Zhu
- Department of Gastroenterology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Yunshan Wang
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan 250013, People's Republic of China; Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Yanfei Jia
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan 250013, People's Republic of China; Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China; Department of Medical Laboratory, Weifang Medical University, Weifang 261053, People's Republic of China.
| |
Collapse
|
47
|
Liu YQ, Xu YW, Zheng ZT, Li D, Hong CQ, Dai HQ, Wang JH, Chu LY, Liao LD, Zou HY, Li EM, Xie JJ, Fang WK. Serine/threonine-protein kinase D2-mediated phosphorylation of DSG2 threonine 730 promotes esophageal squamous cell carcinoma progression. J Pathol 2024; 263:99-112. [PMID: 38411280 DOI: 10.1002/path.6264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 12/23/2023] [Accepted: 01/16/2024] [Indexed: 02/28/2024]
Abstract
Desmoglein-2 (DSG2) is a transmembrane glycoprotein belonging to the desmosomal cadherin family, which mediates cell-cell junctions; regulates cell proliferation, migration, and invasion; and promotes tumor development and metastasis. We previously showed serum DSG2 to be a potential biomarker for the diagnosis of esophageal squamous cell carcinoma (ESCC), although the significance and underlying molecular mechanisms were not identified. Here, we found that DSG2 was increased in ESCC tissues compared with adjacent tissues. In addition, we demonstrated that DSG2 promoted ESCC cell migration and invasion. Furthermore, using interactome analysis, we identified serine/threonine-protein kinase D2 (PRKD2) as a novel DSG2 kinase that mediates the phosphorylation of DSG2 at threonine 730 (T730). Functionally, DSG2 promoted ESCC cell migration and invasion dependent on DSG2-T730 phosphorylation. Mechanistically, DSG2 T730 phosphorylation activated EGFR, Src, AKT, and ERK signaling pathways. In addition, DSG2 and PRKD2 were positively correlated with each other, and the overall survival time of ESCC patients with high DSG2 and PRKD2 was shorter than that of patients with low DSG2 and PRKD2 levels. In summary, PRKD2 is a novel DSG2 kinase, and PRKD2-mediated DSG2 T730 phosphorylation promotes ESCC progression. These findings may facilitate the development of future therapeutic agents that target DSG2 and DSG2 phosphorylation. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Yin-Qiao Liu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, PR China
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, PR China
| | - Yi-Wei Xu
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, PR China
| | - Zheng-Tan Zheng
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, PR China
| | - Die Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, PR China
| | - Chao-Qun Hong
- Department of Oncological Laboratory Research, The Cancer Hospital of Shantou University Medical College, Shantou, PR China
| | - Hao-Qiang Dai
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, PR China
| | - Jun-Hao Wang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, PR China
| | - Ling-Yu Chu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, PR China
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, PR China
| | - Lian-Di Liao
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, PR China
| | - Hai-Ying Zou
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, PR China
| | - En-Min Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, PR China
- Shantou Academy Medical Sciences, Shantou, PR China
| | - Jian-Jun Xie
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, PR China
| | - Wang-Kai Fang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, PR China
| |
Collapse
|
48
|
Liang J, Lei K, Liang R, Huang J, Tan B, Lin H, Wang M. Single-cell RNA sequencing reveals the MIF-ACKR3 receptor-ligand interaction between iCAFs and tumor cells in esophageal squamous cell carcinoma. Cell Signal 2024; 117:111093. [PMID: 38336189 DOI: 10.1016/j.cellsig.2024.111093] [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: 12/01/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a malignant tumor of the gastrointestinal tract with a high morbidity and mortality rate. The heterogeneity of ESCC poses challenges in treatment and contributes to the poor prognosis of patients. Therefore, it is crucial to gain a better understanding of the tumor microenvironment (TME) heterogeneity and identify novel therapeutic targets. METHODS To solve this problem, we performed a single-cell RNA sequencing (scRNA-seq) analysis of ESCC samples obtained from the GEO database. RESULTS A total of 31,283 single cells were categorized into nine cell types, which included four non-immune cells (epithelial cells, endothelial cells, fibroblasts, schwann cells) and five immune cells (T cells, macrophages, mast cells, neutrophils, B cells). Our study revealed the presence of immunosuppressive tumor microenvironments in ESCC. We have also identified not only inflammatory cancer-associated fibroblast (iCAFs) and myofibroblastic cancer-associated fibroblasts (myCAFs) but also a subset of antigen presenting cancer-associated fibroblasts (apCAFs) which express high levels of HLA class II molecules in ESCC. Furthermore, our analysis of cell communication showed up-regulation of MIF-ACKR3 interaction between iCAFs and tumor cells in tumors compared to normal tissues. Finally, it was demonstrated that macrophage migration inhibitory factor (MIF) facilitates tumor cell migration and invasion through interacting with ACKR3 in vitro. CONCLUSIONS This study exposes the features of the tumor microenvironment of ESCC via scRNA-seq and examines the dynamics of various cellular subpopulations, thus facilitating the identification of future therapeutic targets for ESCC.
Collapse
Affiliation(s)
- Jialu Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kai Lei
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruihao Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Binhua Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huayue Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Minghui Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
49
|
Liu K, Yang H, Xiong R, Shen Y, Song G, Yang J, Wang Z. Generation and characterization of mAb 61H9 against junctional adhesion molecule-a with potent antitumor activity. PeerJ 2024; 12:e17088. [PMID: 38495763 PMCID: PMC10944630 DOI: 10.7717/peerj.17088] [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: 09/19/2023] [Accepted: 02/20/2024] [Indexed: 03/19/2024] Open
Abstract
Junctional adhesion molecule-A (JAM-A) is an adhesion molecule that exists on the surface of certain types of cells, including white blood cells, endothelial cells, and dendritic cells. In this study, the cDNA sequences of JAM-A-Fc were chemically synthesized with optimization for mammalian expression. Afterward, we analyzed JAM-A protein expression through transient transfection in HEK293 cell lines. Mice were immunized with JAM-A-Fc protein, and hybridoma was prepared by fusing myeloma cells and mouse spleen cells. Antibodies were purified from the hybridoma supernatant and four monoclonal strains were obtained and numbered 61H9, 70E5, 71A8, and 74H3 via enzyme-linked immunosorbent assay screening. Immunofluorescence staining assay showed 61H9 was the most suitable cell line for mAb production due to its fluorescence signal being the strongest. Flow cytometric analysis proved that 61H9 possessed high affinity. Moreover, antagonism of JAM-A mAb could attenuate the proliferative, migrative, and invasive abilities of ESCC cells and significantly inhibit tumor growth in mice. By examining hematoxylin-eosin staining mice tumor tissues, we found inflammatory cells infiltrated lightly in the anti-JAM-A group. The expression of BCL-2 and IκBα in the anti-JAM-A group were decreased in mice tumor tissues compared to the control group. Ultimately, a method for preparing high-yield JAM-A-Fc protein was created and a high affinity mAb against JAM-A with an antitumor effect was prepared.
Collapse
Affiliation(s)
- Kang Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, China
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hang Yang
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Rong Xiong
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yunlong Shen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Guiqin Song
- School of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Zhenling Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
50
|
Wang Z, Zhang Y, Yang X, Zhang T, Li Z, Zhong Y, Fang Y, Chong W, Chen H, Lu M. Genetic and molecular characterization of metabolic pathway-based clusters in esophageal squamous cell carcinoma. Sci Rep 2024; 14:6200. [PMID: 38486026 PMCID: PMC10940668 DOI: 10.1038/s41598-024-56391-w] [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: 12/29/2023] [Accepted: 03/06/2024] [Indexed: 03/18/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most aggressive types of squamous cell carcinoma and represents a significant proportion of esophageal cancer. Metabolic reprogramming plays a key role in the occurrence and development of ESCC. Unsupervised clustering analysis was employed to stratify ESCC samples into three clusters: MPC1-lipid type, MPC2-amino acid type, and MPC3-energy type, based on the enrichment scores of metabolic pathways extracted from the Reactome database. The MPC3 cluster exhibited characteristics of energy metabolism, with heightened glycolysis, cofactors, and nucleotide metabolism, showing a trend toward increased aggressiveness and poorer survival rates. On the other hand, MPC1 and MPC2 primarily involved lipid and amino acid metabolism, respectively. In addition, liquid chromatography‒mass spectrometry-based metabolite profiles and potential therapeutic agents were explored and compared among ESCC cell lines with different MPCs. MPC3 amplified energy metabolism markers, especially carnitines. In contrast, MPC1 and MPC2 predominantly had elevated levels of lipids (primarily triacylglycerol) and amino acids, respectively. Furthermore, MPC3 demonstrated a suboptimal clinical response to PD-L1 immunotherapy but showed increased sensitivity to the doramapimod chemotherapy regimen, as evident from drug sensitivity evaluations. These insights pave the way for a more personalized therapeutic approach, potentially enhancing treatment precision for ESCC patients.
Collapse
Affiliation(s)
- Ze Wang
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yuan Zhang
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Xiaorong Yang
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Tongchao Zhang
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Zhen Li
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yang Zhong
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yuan Fang
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Wei Chong
- Department of Gastrointestinal Surgery, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Hao Chen
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| | - Ming Lu
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|