1
|
Ke J, Liu F, Yang W, Xu R, Chen L, Yang W, He Y, Liu Z, Hou B, Zhang L, Lin M, Zhang L, Zhang F, Cai F, Xu H, Liu M, Liu Y, Pan Y, He Z, Ke Y. Community prevention and standardized clinical treatment jointly improve cancer outcome: Real-world evidence from an esophageal cancer patient cohort study. Sci Bull (Beijing) 2024; 69:3899-3907. [PMID: 39547907 DOI: 10.1016/j.scib.2024.11.005] [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: 12/11/2023] [Revised: 04/26/2024] [Accepted: 06/12/2024] [Indexed: 11/17/2024]
Abstract
Extensive efforts have been put into reducing the heavy burden of esophageal squamous cell carcinoma (ESCC) in China. However, the joint impact of prevention and treatment on the long-term overall survival (OS) of ESCC patients remains largely unknown. We consecutively recruited 13,255 ESCC patients from two Chinese centers: the Northern center, located in a high-risk area with abundant screening programs; and the Southern center, situated in a non-high-risk area with improved clinical practices. Inter-center comparison, longitudinal intra-center comparison, and a simulation analysis were conducted to investigate the influence of tumor downstaging and high-quality clinical treatment on OS. During a follow-up period of 12.52 years, the Northern center exhibited higher median survival than the Southern center (6.22 vs. 3.15 years; HRadjusted = 0.73, 95% CI: 0.69-0.77). Mediation analysis demonstrated that its OS advantage was largely (77.7%) attributed to earlier TNM stage (stage 0-II: 51.3% vs. 24.6%). In temporal analyses, patient survival in the Southern center gradually improved (median survival during 2015-2018 vs. 2009-2014: 3.58 vs. 2.93 years; HRadjusted = 0.86, 95% CI: 0.79-0.94), coinciding with the progress of treatment-related indices (completeness of TNM staging in discharge diagnosis [from 53.7% to 99.6%], adoption of minimally invasive esophagectomy [from 0.0% to 51.1%] and right thoracic esophagectomy [from 12.4% to 86.4%], etc.). Simulation analysis further demonstrated that integrating both downstaging and high-quality treatment would lead to the best survival. Tumor downstaging and high-quality clinical treatment have a joint impact on ESCC patient survival. Establishing a comprehensive strategy that integrates cancer prevention with optimal clinical treatment is crucial for alleviating the ESCC burden.
Collapse
Affiliation(s)
- Ji Ke
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Fangfang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Wei Yang
- Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Ruiping Xu
- Anyang Cancer Hospital, Anyang 455000, China
| | - Lei Chen
- Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Wenlei Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yu He
- Department of Non-communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Zhen Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Bolin Hou
- Linkdoc AI Research (LAIR), Beijing 100080, China
| | - Liqun Zhang
- Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Miaoping Lin
- Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Lixin Zhang
- Anyang Cancer Hospital, Anyang 455000, China
| | - Fan Zhang
- Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Fen Cai
- Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Huawen Xu
- Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Mengfei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ying Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yaqi Pan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhonghu He
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| | - Yang Ke
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| |
Collapse
|
2
|
Jiang W, Zhang B, Xu J, Xue L, Wang L. Current status and perspectives of esophageal cancer: a comprehensive review. Cancer Commun (Lond) 2024. [PMID: 39723635 DOI: 10.1002/cac2.12645] [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: 07/01/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Esophageal cancer (EC) continues to be a significant global health concern, with two main subtypes: esophageal squamous cell carcinoma and esophageal adenocarcinoma. Prevention and changes in etiology, improvements in early detection, and refinements in the treatment have led to remarkable progress in the outcomes of EC patients in the past two decades. This seminar provides an in-depth analysis of advances in the epidemiology, disease biology, screening, diagnosis, and treatment landscape of esophageal cancer, focusing on the ongoing debate surrounding multimodality therapy. Despite significant advancements, EC remains a deadly disease, underscoring the need for continued research into early detection methods, understanding the molecular mechanisms, and developing effective treatments.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, P. R. China
| | - Bo Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Jiaqi Xu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Liyan Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Luhua Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, P. R. China
| |
Collapse
|
3
|
Tian K, Yao Z, Pan D. Leveraging single-cell and multi-omics approaches to identify MTOR-centered deubiquitination signatures in esophageal cancer therapy. Front Immunol 2024; 15:1490623. [PMID: 39742278 PMCID: PMC11685190 DOI: 10.3389/fimmu.2024.1490623] [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: 09/03/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) remains a significant challenge in oncology due to its aggressive nature and heterogeneity. As one of the deadliest malignancies, ESCC research lags behind other cancer types. The balance between ubiquitination and deubiquitination processes plays a crucial role in cellular functions, with its disruption linked to various diseases, including cancer. Methods Our study utilized diverse analytical approaches, encompassing Cox regression models, single-cell RNA sequencing, intercellular communication analysis, and Gene Ontology enrichment. We also conducted mutation profiling and explored potential immunotherapeutic agents. Furthermore, in vitro cellular experiments and in vivo mouse models were performed to validate findings. These methodologies aimed to establish deubiquitination-related gene signatures (DRGS) for predicting ESCC patient outcomes and response to immunotherapy. Results By integrating datasets from TCGA-ESCC and GSE53624, we developed a DRGS model based on 14 deubiquitination-related genes (DUBGs). This signature effectively forecasts ESCC prognosis, drug responsiveness, and immune cell infiltration patterns. It also influences the mutational landscape of patients. Those classified as high-risk exhibited reduced survival rates, increased genetic alterations, and more complex cellular interactions, potentially explaining their poor outcomes. Notably, in vitro and in vivo experiments identified MTOR, a key component of the signature, as a promising therapeutic target for ESCC treatment. Conclusion Our research highlights the significance of 14 DUBGs in ESCC progression. The risk score derived from this gene set enables clinical stratification of patients into distinct prognostic groups. Moreover, MTOR emerges as a potential target for personalized ESCC therapy, offering new avenues for treatment strategies.
Collapse
Affiliation(s)
- Kang Tian
- Department of Oncology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Ziang Yao
- Department of Traditional Chinese Medicine, Peking University People’s Hospital, Beijing, China
| | - Da Pan
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou, China
| |
Collapse
|
4
|
Tu R, Zhong D, Li P, Li Y, Chen Z, Hu F, Yuan G, Chen Z, Yu S, Song J. Assessment of LINC-PINT genetic polymorphisms and esophageal squamous cell carcinoma risk in the Hainan Han population. Ann Med 2024; 56:2397569. [PMID: 39221756 PMCID: PMC11370687 DOI: 10.1080/07853890.2024.2397569] [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: 01/10/2024] [Revised: 05/04/2024] [Accepted: 05/09/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVES Esophageal squamous cell carcinoma (ESCC) is a malignant tumor with high incidence and mortality rates worldwide. This study aimed to investigate the correlation between LINC-PINT polymorphisms and ESCC risk in the Hainan Han population. METHODS A total of 391 patients with ESCC and 452 healthy controls were enrolled to evaluate the effect of LINC-PINT SNPs (single nucleotide polymorphisms) on ESCC susceptibility. Associations were evaluated by calculating odds ratios (OR) and 95% confidence intervals (CIs). Multifactor dimensionality reduction analysis was performed to explore the association between SNP-SNP interactions and ESCC susceptibility. We further determined the correlation between clinical indicators and SNP in patients with ESCC. RESULTS Our study showed that rs157916 (OR 0.63, p = 0.011) and rs157928 (OR 0.80, p = 0.021) were associated with a decreased risk of ESCC. Stratified analysis indicated that rs157916 could decrease the risk of ESCC in people aged >64 years, in males, and non-drinkers (OR 0.58, p = 0.042; OR 0.58, p = 0.010; OR 0.62, p = 0.025, respectively). Rs16873842 was related to a decreased risk of ESCC in males (OR 0.70, p = 0.015). Rs7801029 was associated with ESCC risk in females (OR 0.39, p = 0.033) and non-drinkers (OR 0.68, p = 0.040). Rs7781295 decreased the ESCC risk in smokers (OR 0.58, p = 0.046) and drinkers (OR 0.58, p = 0.046). In addition, rs157928 played a protective role in ESCC risk in females (OR 0.39, p = 0.033) and non-smokers (OR 0.32, p = 0.006). Additionally, the best predictive model for ESCC was a combination of rs157916, rs16873842, rs7801029, rs7781295, rs28662387, and rs157928. CONCLUSION Our study revealed that LINC-PINT polymorphisms were associated with ESCC risk.
Collapse
Affiliation(s)
- Ruisha Tu
- Department of Gastrointestinal Surgery, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Dunjing Zhong
- Department of Gastroenterology, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Ping Li
- Department of Digestive Endoscopy Center, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Yongyu Li
- Department of Gastroenterology, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Zhuang Chen
- Department of Gastroenterology, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Feixiang Hu
- Department of Gastrointestinal Surgery, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Guihong Yuan
- Department of Gastroenterology, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Zhaowei Chen
- Department of Gastroenterology, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Shuyong Yu
- Department of Gastrointestinal Surgery, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Jian Song
- Department of Gastroenterology, Southern University of Science and Technology Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
5
|
Chen HY, Por CR, Hong YK, Kong EQZ, Subramaniyan V. Molecular mechanisms underlying oesophageal cancer development triggered by chronic alcohol consumption. CLINICAL AND TRANSLATIONAL DISCOVERY 2024; 4. [DOI: 10.1002/ctd2.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/24/2024] [Indexed: 12/27/2024]
Abstract
AbstractThis review explores the mechanisms underlying alcohol‐induced oesophageal carcinogenesis, including DNA damage, oxidative stress, and nutritional deficiencies. Alcohol metabolism primarily involves alcohol dehydrogenase (ADH) converting ethanol to acetaldehyde, which can cause DNA damage, inhibit repair mechanisms, and form DNA adducts thus inhibiting DNA replication. Plus, it delves into the epidemiological evidence, genetic susceptibility, epigenetic modifications, biomarkers, and preventive strategies associated with alcohol‐related oesophageal cancers. Consumption of alcohol increases the risk of gastroesophageal reflux disease thus compromising mucosal integrity of the oesophagus as dysregulation of cytokines such as IL‐18, TNFA, GATA3, TLR4, and CD68 expands the intercellular spaces of epithelial cells. Genetic variants, such as ADH1B rs1229984 and ALDH2 rs671, significantly influence susceptibility to alcohol‐related oesophageal cancers, with these variations affecting acetaldehyde metabolism and cancer risk. Understanding these factors is crucial for early detection, effective treatment, and the development of targeted prevention strategies. Biomarkers, such as miRNA and metabolite markers, offer non‐invasive methods for early detection, while advanced endoscopic techniques provide better diagnostic accuracy. Pharmacological interventions, such as statins and proton pump inhibitors, also show potential for reducing cancer progression in high‐risk individuals. Despite advances, late‐stage oesophageal cancer diagnoses are still common, highlighting the need for better screening and prevention. Further research, including this study, should aim to improve early detection, personalise prevention, and explore new treatments to reduce cases and enhance outcomes in alcohol‐related oesophageal cancers.
Collapse
Affiliation(s)
- Huai Yi Chen
- Jeffrey Cheah School of Medicine and Health Sciences Monash University Jalan Lagoon Selatan Bandar Sunway Subang Jaya Malaysia
| | - Chia Rou Por
- Jeffrey Cheah School of Medicine and Health Sciences Monash University Jalan Lagoon Selatan Bandar Sunway Subang Jaya Malaysia
| | - Yong Kai Hong
- Jeffrey Cheah School of Medicine and Health Sciences Monash University Jalan Lagoon Selatan Bandar Sunway Subang Jaya Malaysia
| | - Eason Qi Zheng Kong
- Jeffrey Cheah School of Medicine and Health Sciences Monash University Jalan Lagoon Selatan Bandar Sunway Subang Jaya Malaysia
| | - Vetriselvan Subramaniyan
- Jeffrey Cheah School of Medicine and Health Sciences Monash University Jalan Lagoon Selatan Bandar Sunway Subang Jaya Malaysia
- School of Medical and Life Sciences Sunway University Jalan Lagoon Selatan Bandar Sunway Petaling Jaya Malaysia
| |
Collapse
|
6
|
Zhu Q, Zhang C, Li Z, Ma T, Wang N, Liu W, He Z, Shen J, Wei T, Zhao S, Feng L, Tian Y. The significance of consolidation chemotherapy after concurrent chemoradiotherapy in esophageal squamous cell carcinoma: a randomized controlled phase III clinical trial. Thorac Cancer 2024; 15:2038-2048. [PMID: 39199003 PMCID: PMC11444924 DOI: 10.1111/1759-7714.15424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND This study explored the significance of consolidation maintenance chemotherapy after concurrent chemoradiotherapy with different regimens in patients with esophageal squamous cell carcinoma. METHOD A prospective randomized controlled phase III clinical trial was designed and registered in the China Clinical Trials Registry (Registration number: ChiCTR-TRC-12002719). Survival data were analyzed in terms of intention-to-treat (ITT) and per-protocol (PP) sets for patients undergoing cisplatin and 5-fluorouracil (PF) (group A), or cisplatin and paclitaxel (TP) (group B). RESULTS The incidence risk of grade III-IV leukopenia in group B was higher than in group A (49.2% vs. 25.5%, p = 0.012). The survival rates at 1, 2, 3, and 5 years were 83.8%, 62.6%, 53.1%, and 41.3%, respectively. Consolidation chemotherapy after concurrent chemoradiation therapy had no benefit on median progression-free survival (PFS) (p = 0.95) and overall survival (OS) (p = 0.809). According to the ITT analysis, the median PFS in group A and group B was 28.6 months and 30.3 months (X2 = 0.242, p = 0.623), while the median OS was 31.0 months and 50.3 months (X2 = 1.25,p = 0.263). For the PP analysis, the median PFS in group A and group B were 28.6 months and 30.3 months (p = 0.584), while the median OS was 31.0 months and 50.3 months (p = 0.259), respectively. Patients receiving consolidation chemotherapy did not show significant OS benefits (46.9 months vs. 38.3 months; X2 = 0.059, p = 0.866). CONCLUSION Similar PFS and OS were found between PF and TP regimens with concurrent chemoradiotherapy. Consolidation chemotherapy did not show any significant OS benefits.
Collapse
Affiliation(s)
- Qingshan Zhu
- Radiotherapy DepartmentAnyang Cancer Hospital of Henan ProvinceAnyangChina
- Radiotherapy Department, Anyang Cancer Hospital affiliated with Henan University of Science and TechnologyAnyangChina
- Radiotherapy DepartmentHenan Provincial Key Laboratory of Precision Prevention and Treatment of Esophageal CancerAnyangChina
| | - Chi Zhang
- Department of CardiologyThe Second Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
| | - Zhuoqi Li
- Department of Radiotherapy OncologyAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinanChina
| | - Tingwei Ma
- Radiotherapy DepartmentAnyang Cancer Hospital of Henan ProvinceAnyangChina
- Radiotherapy Department, Anyang Cancer Hospital affiliated with Henan University of Science and TechnologyAnyangChina
- Radiotherapy DepartmentHenan Provincial Key Laboratory of Precision Prevention and Treatment of Esophageal CancerAnyangChina
| | - Nengchao Wang
- Radiotherapy DepartmentAnyang Cancer Hospital of Henan ProvinceAnyangChina
- Radiotherapy Department, Anyang Cancer Hospital affiliated with Henan University of Science and TechnologyAnyangChina
- Radiotherapy DepartmentHenan Provincial Key Laboratory of Precision Prevention and Treatment of Esophageal CancerAnyangChina
| | - Weipeng Liu
- Radiotherapy DepartmentAnyang Cancer Hospital of Henan ProvinceAnyangChina
- Radiotherapy Department, Anyang Cancer Hospital affiliated with Henan University of Science and TechnologyAnyangChina
- Radiotherapy DepartmentHenan Provincial Key Laboratory of Precision Prevention and Treatment of Esophageal CancerAnyangChina
| | - Zhijie He
- Radiotherapy DepartmentAnyang Cancer Hospital of Henan ProvinceAnyangChina
- Radiotherapy Department, Anyang Cancer Hospital affiliated with Henan University of Science and TechnologyAnyangChina
- Radiotherapy DepartmentHenan Provincial Key Laboratory of Precision Prevention and Treatment of Esophageal CancerAnyangChina
| | - Jing Shen
- Radiotherapy DepartmentAnyang Cancer Hospital of Henan ProvinceAnyangChina
- Radiotherapy Department, Anyang Cancer Hospital affiliated with Henan University of Science and TechnologyAnyangChina
- Radiotherapy DepartmentHenan Provincial Key Laboratory of Precision Prevention and Treatment of Esophageal CancerAnyangChina
| | - Tao Wei
- Radiotherapy DepartmentAnyang Cancer Hospital of Henan ProvinceAnyangChina
- Radiotherapy Department, Anyang Cancer Hospital affiliated with Henan University of Science and TechnologyAnyangChina
- Radiotherapy DepartmentHenan Provincial Key Laboratory of Precision Prevention and Treatment of Esophageal CancerAnyangChina
| | - Shijie Zhao
- Radiotherapy DepartmentAnyang Cancer Hospital of Henan ProvinceAnyangChina
- Radiotherapy Department, Anyang Cancer Hospital affiliated with Henan University of Science and TechnologyAnyangChina
- Radiotherapy DepartmentHenan Provincial Key Laboratory of Precision Prevention and Treatment of Esophageal CancerAnyangChina
| | - Lianjie Feng
- Radiotherapy DepartmentAnyang Cancer Hospital of Henan ProvinceAnyangChina
- Radiotherapy Department, Anyang Cancer Hospital affiliated with Henan University of Science and TechnologyAnyangChina
- Radiotherapy DepartmentHenan Provincial Key Laboratory of Precision Prevention and Treatment of Esophageal CancerAnyangChina
| | - Yuan Tian
- Department of Radiotherapy OncologyAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinanChina
| |
Collapse
|
7
|
Qiu Y, Tian Z, Miao TY, Shen L, Chen J, Li PF, Zhu ZX, Zhu ZF, Wu WJ, Xu X, Shen WG. The METTL3-m 6A-YTHDC1-AMIGO2 axis contributes to cell proliferation and migration in esophageal squamous cell carcinoma. Gene 2024; 908:148281. [PMID: 38360124 DOI: 10.1016/j.gene.2024.148281] [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: 10/06/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024]
Abstract
The upregulation of methyltransferase-like 3 (METTL3) has been associated with the progression of esophageal cancer. However, METTL3-induced N6-methyladenosine (m6A) alterations on the downstream target mRNAs in esophageal squamous cell carcinoma (ESCC) are not yet fully understood. Our study revealed that silencing METTL3 resulted in a significant decrease in ESCC cell proliferation and metastasis in vitro and in vivo. Additionally, the adhesion molecule with Ig like domain 2 (AMIGO2) was identified as a potential downstream target of both METTL3 and YTH Domain-Containing Protein 1 (YTHDC1) in ESCC cells. Functionally, AMIGO2 augmented the malignant behaviors of ESCC cells in vitro and in vivo, and its overexpression can rescue the inhibition of the proliferation and migration in ESCC cells induced by METTL3 or YTHDC1 knockdown. Furthermore, our findings revealed that knockdown of METTL3 decreased m6A modification in the 5'-untranslated regions (5'UTR) of AMIGO2 precursor mRNA (pre-mRNA), and YTHDC1 interacted with AMIGO2 pre-mRNA to regulate AMIGO2 expression by modulating the splicing process of AMIGO2 pre-mRNA in ESCC cells. These findings highlighted a novel role of the METTL3-m6A-YTHDC1-AMIGO2 axis in regulating ESCC cell proliferation and motility, suggesting its potential as a therapeutic target for ESCC.
Collapse
Affiliation(s)
- Yue Qiu
- Department of Cell Biology, Yangzhou University Medical College, Yangzhou, Jiangsu, China
| | - Zhen Tian
- Department of Cell Biology, Yangzhou University Medical College, Yangzhou, Jiangsu, China
| | - Ting-Yu Miao
- Department of Cell Biology, Yangzhou University Medical College, Yangzhou, Jiangsu, China
| | - Lin Shen
- Department of Cell Biology, Yangzhou University Medical College, Yangzhou, Jiangsu, China
| | - Jing Chen
- Department of Cell Biology, Yangzhou University Medical College, Yangzhou, Jiangsu, China
| | - Pei-Fen Li
- Department of Cell Biology, Yangzhou University Medical College, Yangzhou, Jiangsu, China
| | - Zi-Xuan Zhu
- Department of Cell Biology, Yangzhou University Medical College, Yangzhou, Jiangsu, China
| | - Zi-Fan Zhu
- Department of Cell Biology, Yangzhou University Medical College, Yangzhou, Jiangsu, China
| | - Wen-Juan Wu
- Department of Oncology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiao Xu
- Department of Oncology, Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Jiangsu, China.
| | - Wei-Gan Shen
- Department of Cell Biology, Yangzhou University Medical College, Yangzhou, Jiangsu, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
8
|
Ling CQ, Liao HX, Wen JR, Nie HY, Zhang LY, Xu FR, Cheng YX, Dong X. Investigation of the Inhibitory Effects of Illicium verum Essential Oil Nanoemulsion on Fusarium proliferatum via Combined Transcriptomics and Metabolomics Analysis. Curr Microbiol 2024; 81:182. [PMID: 38769214 DOI: 10.1007/s00284-024-03724-7] [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: 02/01/2024] [Accepted: 04/29/2024] [Indexed: 05/22/2024]
Abstract
Fusarium proliferatum is the main pathogen that causes Panax notoginseng root rot. The shortcomings of strong volatility and poor water solubility of Illicium verum essential oil (EO) limit its utilization. In this study, we prepared traditional emulsion (BDT) and nanoemulsion (Bneo) of I. verum EO by ultrasonic method with Tween-80 and absolute ethanol as solvents. The chemical components of EO, BDT, and Bneo were identified by gas chromatography-mass spectrometry (GC-MS) and the antifungal activity and mechanism were compared. The results show that Bneo has good stability and its particle size is 34.86 nm. The contents of (-) -anethole and estragole in Bneo were significantly higher than those in BDT. The antifungal activity against F. proliferatum was 5.8-fold higher than BDT. In the presence of I. verum EO, the occurrence of P. notoginseng root rot was significantly reduced. By combining transcriptome and metabolomics analysis, I. verum EO was found to be involved in the mutual transformation of pentose and glucuronic acid, galactose metabolism, streptomycin biosynthesis, carbon metabolism, and other metabolic pathways of F. proliferatum, and it interfered with the normal growth of F. proliferatum to exert antifungal effects. This study provide a theoretical basis for expanding the practical application of Bneo.
Collapse
Affiliation(s)
- Cui-Qiong Ling
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, 650000, China
| | - Hong-Xin Liao
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, 650000, China
| | - Jin-Rui Wen
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, 650000, China
| | - Hong-Yan Nie
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, 650000, China
| | - Li-Yan Zhang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, 650000, China
| | - Fu-Rong Xu
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, 650000, China
| | - Yong-Xian Cheng
- School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, 518060, People's Republic of China
| | - Xian Dong
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, 650000, China.
| |
Collapse
|
9
|
Jia L, Wang M, Duan S, Chen J, Zhao M, Ji S, Lv B, Jiang X, He G, Yang J. Genetic history of esophageal cancer group in southwestern China revealed by Y-chromosome STRs and genomic evolutionary connection analysis. Heliyon 2024; 10:e29867. [PMID: 38720733 PMCID: PMC11076658 DOI: 10.1016/j.heliyon.2024.e29867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/09/2024] [Accepted: 04/16/2024] [Indexed: 05/12/2024] Open
Abstract
Genetic and environmental factors play crucial roles in the development of esophageal cancer (EC) and contribute uniquely or cooperatively to human cancer susceptibility. Sichuan is located in the interior of southwestern China, and the northern part of Sichuan is one of the regions with a high occurrence of EC. However, the factors influencing the high incidence rate of EC in the Sichuan Han Chinese population and its corresponding genetic background and origins are still poorly understood. Here, we utilized genome-wide single nucleotide polymorphisms (SNPs) and Y-chromosome short tandem repeats (Y-STRs) to characterize the genetic structure, connection, and origin of cancer groups and general populations. We generated Y-STR-based haplotype data from 214 Sichuan individuals, including the Han Chinese EC population and a control group of Han Chinese individuals. Our results, obtained from Y-STR-based population statistical methods (analysis of molecular variance (AMOVA), principal component analysis (PCA), and phylogenetic analysis), demonstrated that there was a genetic substructure difference between the EC population in the high-incidence area of northern Sichuan Province and the control population. Additionally, there was a strong genetic relationship between the EC population in the northern Sichuan high-incidence area and those at high risk in both the Fujian and Chaoshan areas. In addition, we obtained high-density SNP data from saliva samples of 60 healthy Han Chinese individuals from three high-prevalence areas of EC in China: Sichuan Nanchong, Fujian Quanzhou, and Henan Xinxiang. As inferred from the allele frequency of SNPs and sharing patterns of haplotype segments, the evolutionary history and admixture events suggested that the Han population from Nanchong in northern Sichuan Province shared a close genetic relationship with the Han populations from Xinxiang in Henan Province and Quanzhou in Fujian Province, both of which are regions with a high prevalence of EC. Our study illuminated the genetic profile and connection of the Northern Sichuan Han population and enriched the genomic resources and features of the Han Chinese populations in China, especially for the Y-STR genetic data of the Han Chinese EC population. Populations living in different regions with high incidences of EC may share similar genetic backgrounds, which offers new insights for further exploring the genetic mechanisms underlying EC.
Collapse
Affiliation(s)
- Lihua Jia
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College and Center for Genetics and Prenatal diagnosis, Affiliated Hospital of Northern Sichuan Medical College, Nanchong, Sichuan, 637007, China
| | - Mengge Wang
- Center for Archaeological Science, Sichuan University, Chengdu, 610000, China
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China
| | - Shuhan Duan
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College and Center for Genetics and Prenatal diagnosis, Affiliated Hospital of Northern Sichuan Medical College, Nanchong, Sichuan, 637007, China
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China
| | - Jianghua Chen
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College and Center for Genetics and Prenatal diagnosis, Affiliated Hospital of Northern Sichuan Medical College, Nanchong, Sichuan, 637007, China
| | - Mei Zhao
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College and Center for Genetics and Prenatal diagnosis, Affiliated Hospital of Northern Sichuan Medical College, Nanchong, Sichuan, 637007, China
| | - Simeng Ji
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Bingbing Lv
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Xiucheng Jiang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College and Center for Genetics and Prenatal diagnosis, Affiliated Hospital of Northern Sichuan Medical College, Nanchong, Sichuan, 637007, China
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China
| | - Guanglin He
- Center for Archaeological Science, Sichuan University, Chengdu, 610000, China
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China
| | - Junbao Yang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College and Center for Genetics and Prenatal diagnosis, Affiliated Hospital of Northern Sichuan Medical College, Nanchong, Sichuan, 637007, China
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| |
Collapse
|
10
|
Yan CY, Gu YM, Shi GD, Shang QX, Zhang HL, Yang YS, Wang WP, Yuan Y, Chen LQ. Impact of deep muscle invasion on nodal status and survival in patients with pT2 esophageal squamous cancer. J Surg Oncol 2024; 129:1056-1062. [PMID: 38314575 DOI: 10.1002/jso.27593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 12/24/2023] [Accepted: 01/15/2024] [Indexed: 02/06/2024]
Abstract
BACKGROUND Whether T2 esophageal squamous cell carcinoma should be subclassified remains controversial. We aimed to investigate the impact of the depth of muscularis propria invasion on nodal status and survival outcomes. METHODS We identified patients with pT2 esophageal squamous cell carcinoma who underwent primary surgery from January 2009 to June 2017. Clinical data were extracted from prospectively maintained databases. Tumor muscularis propria invasion was stratified into superficial or deep. Binary logistic regression was used to determine risk factors for lymph node metastases. The impact of the depth of muscularis propria invasion on survival was investigated using Kaplan‒Meier analysis and a Cox proportional hazard regression model. RESULTS A total of 750 patients from three institutes were investigated. The depth of muscularis propria invasion (odds ratio [OR]: 3.95, 95% confidence interval [CI]: 2.46-6.35; p < 0.001) was correlated with lymph node metastases using logistic regression. T substage (hazard ratio [HR]: 1.37, 95% CI: 1.05-1.79; p < 0.001) and N status (HR: 1.51, 95% CI: 1.05-2.17; p < 0.001) were independent risk factors in multivariate Cox regression analysis. The deep muscle invasion was associated with worse overall survival (HR: 1.52, 95% CI: 1.19-1.94; p = 0.001) than superficial, specifically in T2N0 patients (HR: 1.38, 95% CI: 1.08-1.94; p = 0.035). CONCLUSIONS We found that deep muscle invasion was associated with significantly worse outcomes and recommended the substaging of pT2 esophageal squamous cell carcinoma in routine pathological examination.
Collapse
Affiliation(s)
- Cheng-Yi Yan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Cardiothoracic Surgery, Changsha Central Hospital, University of South China, Changsha, Hunan, China
| | - Yi-Min Gu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Gui-Dong Shi
- Department of Cardiothoracic Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Qi-Xin Shang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Han-Lu Zhang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu-Shang Yang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wen-Ping Wang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yong Yuan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Long-Qi Chen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
11
|
Bai G, Mahati S, Tulahong A, Eli M, Mao R. ZNF468 inhibits irradiation-induced G2/M cell cycle arrest and apoptosis by facilitating AURKA transcription in Esophageal Squamous Cell Carcinoma. Biochem Biophys Res Commun 2024; 703:149687. [PMID: 38368674 DOI: 10.1016/j.bbrc.2024.149687] [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: 09/29/2023] [Revised: 02/01/2024] [Accepted: 02/13/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND ZNF468 is a relatively unexplored gene that has been implicated in potential oncogenic properties in various cancer types. However, the exact role of ZNF468 in radiotherapy resistance of esophageal squamous cell carcinomas (ESCCs) is not well understood. METHODS Bioinformatic analysis was performed using the TCGA database to assess ZNF468 expression and prognostic significance in pan-cancer and ESCC. Functional experiments were conducted using ZNF468 overexpressing and knockdown cell lines to assess its impact on cell survival, DNA damage response, cell cycle, and apoptosis upon radiation. A luciferase reporter assay was utilized to validate ZNF468 binding to the AURKA promoter. RESULTS ZNF468 was significantly upregulated in diverse cancer types, including ESCC, and its high expression correlated with adverse prognosis in specific tumors. In the ESCC cohort, ZNF468 exhibited substantial upregulation in post-radiotherapy tissues, indicating its potential role in conferring radiotherapy resistance. Functional experiments revealed that ZNF468 enhances cell viability and facilitates DNA damage repair in radiotherapy-treated ESCC cells, while dampening the G2/M cell cycle arrest and apoptosis induced by radiation. Moreover, ZNF468 facilitated AURKA transcription, resulting in upregulated Aurora A expression, and subsequently inhibited P53 expression, unveiling key molecular mechanisms underlying radiotherapy resistance in ESCC. CONCLUSION ZNF468 plays an oncogenic role in ESCC and contributes to radiotherapy resistance. It enhances cell survival while dampening radiation-induced G2/M cell cycle arrest and apoptosis. By modulating AURKA and P53 expression, ZNF468 represents a promising therapeutic target for enhancing radiotherapy efficacy in ESCC.
Collapse
Affiliation(s)
- Ge Bai
- Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Ürümqi, Xinjiang Uyghur Autonomous Region, 830011, China
| | - Shaya Mahati
- Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Ürümqi, Xinjiang Uyghur Autonomous Region, 830011, China
| | - Asikeer Tulahong
- Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Ürümqi, Xinjiang Uyghur Autonomous Region, 830011, China
| | - Mayinur Eli
- Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Ürümqi, Xinjiang Uyghur Autonomous Region, 830011, China.
| | - Rui Mao
- Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Ürümqi, Xinjiang Uyghur Autonomous Region, 830011, China.
| |
Collapse
|
12
|
Chen J, Zhang L, Zhu Y, Zhao D, Zhang J, Zhu Y, Pang J, Xiao Y, Wu Q, Wang Y, Zhan Q. AKT2 S128/CCTα S315/319/323-positive cancer-associated fibroblasts (CAFs) mediate focal adhesion kinase (FAK) inhibitors resistance via secreting phosphatidylcholines (PCs). Signal Transduct Target Ther 2024; 9:21. [PMID: 38280862 PMCID: PMC10821909 DOI: 10.1038/s41392-023-01728-6] [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: 07/07/2023] [Revised: 10/26/2023] [Accepted: 12/10/2023] [Indexed: 01/29/2024] Open
Abstract
Abnormal metabolism is regarded as an oncogenic hallmark related to tumor progression and therapeutic resistance. Present study employed multi-omics, including phosphoproteomics, untargeted metabolomics and lipidomics, to demonstrate that the pAKT2 Ser128 and pCCTα Ser315/319/323-positive cancer-associated fibroblasts (CAFs) substantially release phosphatidylcholines (PCs), contributing to the resistance of focal adhesion kinase (FAK) inhibitors in esophageal squamous cell carcinoma (ESCC) treatment. Additionally, we observed extremely low levels of FAK Tyr397 expression in CAFs, potentially offering no available target for FAK inhibitors playing their anti-growth role in CAFs. Consequently, FAK inhibitor increased the intracellular concentration of Ca2+ in CAFs, promoting the formation of AKT2/CCTα complex, leading to phosphorylation of CCTα Ser315/319/323 sites and eventually enhancing stromal PC production. This activation could stimulate the intratumoral Janus kinase 2 (JAK2)/Signal transducer and activator of transcription 3 (STAT3) pathway, triggering resistance to FAK inhibition. Analysis of clinical samples demonstrated that stromal pAKT2 Ser128 and pCCTα Ser315/319/323 are related to the tumor malignancy and reduced patient survival. Pseudo-targeted lipidomics and further validation cohort quantitatively showed that plasma PCs enable to distinguish the malignant extent of ESCC patients. In conclusion, inhibition of stroma-derived PCs and related pathway could be possible therapeutic strategies for tumor therapy.
Collapse
Affiliation(s)
- Jie Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
- Peking University International Cancer Institute, Peking University, 100191, Beijing, China.
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China.
- Soochow University Cancer Institute, Suzhou, 215000, China.
| | - Lingyuan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Peking University International Cancer Institute, Peking University, 100191, Beijing, China
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuheng Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Di Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Peking University International Cancer Institute, Peking University, 100191, Beijing, China
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Peking University International Cancer Institute, Peking University, 100191, Beijing, China
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Yanmeng Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Peking University International Cancer Institute, Peking University, 100191, Beijing, China
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingyuan Pang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Yuanfan Xiao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Peking University International Cancer Institute, Peking University, 100191, Beijing, China
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Qingnan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Peking University International Cancer Institute, Peking University, 100191, Beijing, China
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Yan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- Peking University International Cancer Institute, Peking University, 100191, Beijing, China
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Qimin Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
- Peking University International Cancer Institute, Peking University, 100191, Beijing, China.
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China.
- Soochow University Cancer Institute, Suzhou, 215000, China.
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China.
| |
Collapse
|
13
|
Zhang M, Wang Z, Wu Y, Chen M, Li J, Liu G. Hypoxia-induced factor-1α promotes radioresistance of esophageal cancer cells by transcriptionally activating LINC01116 and suppressing miR-3612 under hypoxia. J Biochem Mol Toxicol 2024; 38:e23551. [PMID: 37983895 DOI: 10.1002/jbt.23551] [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: 12/13/2022] [Revised: 08/02/2023] [Accepted: 09/27/2023] [Indexed: 11/22/2023]
Abstract
Esophageal cancer (EC) is a challenging tumor to treat with radiotherapy, often exhibiting resistance to this treatment modality. To explore the factors influencing radioresistance, we focused on the role of hypoxia-induced factor-1α (HIF-1α), and its interaction with the long noncoding RNA long intergenic nonprotein coding RNA 1116 (LINC01116). We analyzed the LINC01116 expression in EC and EC cell lines/human normal esophageal epithelial cell line (Het-1A). LINC01116 was silenced/overexpressed in EC109/KYSE30 cells under hypoxia, followed by radioresistance assessment. We measured HIF-1α levels in hypoxic EC cells and further validated the binding of HIF-1α with LINC01116, analyzing their interaction in EC cells. We then performed experiments in EC109 cells by transfection them with sh-HIF-1α/oe-LINC01116 to verify the effects. Additonally, we analyzed the localization of LINC01116 and its binding with miR-3612, followed by a combined experiment performed to validate the results. Our findings indicated that LINC01116 was highly expressed in EC and further elevated in hypoxic EC cells. LINC01116 was expressed at a high level in EC, which was further elevated in EC cells under hypoxic conditions. Knockdown of LINC01116 triggered EC cell apoptosis, thus suppressing radioresistance. Further investigation revealed that HIF-1α transcriptionally activated LINC01116 expression under hypoxia, and silencing HIF-1α lowered EC cell radioresistance by downregulating LINC01116. Under hypoxic conditions, LINC01116 could function as a sponge for miR-3612 and inhibit its expression. This interaction between LINC01116 and miR-3612 played a crucial role in mediating radioresistance in EC cells. Briefly, under hypoxic conditions, HIF-1α facilitates radioresistance of EC cells by transcriptionally activating LINC01116 expression and downregulating miR-3612.
Collapse
Affiliation(s)
- Mengyan Zhang
- Oncology Department, Guangzhou No.1 People's Hospital, Guangzhou City, Guangdong Province, P.R. China
- Thoracic Radiotherapy Department, Fujian Medical University Cancer Hospital Fujian Cancer Hospital, Fuzhou City, Fujian Province, P.R. China
| | - Zhiping Wang
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou City, Fujian Province, P.R. China
| | - Yahua Wu
- Thoracic Radiotherapy Department, Fujian Medical University Union Hospital, Fuzhou City, Fujian Province, P.R. China
| | - Mingqiu Chen
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou City, Fujian Province, P.R. China
| | - Jiancheng Li
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou City, Fujian Province, P.R. China
| | - Guolong Liu
- Oncology Department, Guangzhou No.1 People's Hospital, Guangzhou City, Guangdong Province, P.R. China
| |
Collapse
|
14
|
Conway E, Wu H, Tian L. Overview of Risk Factors for Esophageal Squamous Cell Carcinoma in China. Cancers (Basel) 2023; 15:5604. [PMID: 38067307 PMCID: PMC10705141 DOI: 10.3390/cancers15235604] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 05/27/2024] Open
Abstract
(1) Background: China has the highest esophageal squamous cell carcinoma (ESCC) incidence areas in the world, with some areas of incidence over 100 per 100,000. Despite extensive public health efforts, its etiology is still poorly understood. This study aims to review and summarize past research into potential etiologic factors for ESCC in China. (2) Methods: Relevant observational and intervention studies were systematically extracted from four databases using key terms, reviewed using Rayyan software, and summarized into Excel tables. (3) Results: Among the 207 studies included in this review, 129 studies were focused on genetic etiologic factors, followed by 22 studies focused on dietary-related factors, 19 studies focused on HPV-related factors, and 37 studies focused on other factors. (4) Conclusions: ESCC in China involves a variety of factors including genetic variations, gene-environment interactions, dietary factors like alcohol, tobacco use, pickled vegetables, and salted meat, dietary behavior such as hot food/drink consumption, infections like HPV, poor oral health, gastric atrophy, and socioeconomic factors. Public health measures should prioritize genetic screening for relevant polymorphisms, conduct comprehensive investigations into environmental, dietary, and HPV influences, enhance oral health education, and consider socioeconomic factors overall as integral strategies to reduce ESCC in high-risk areas of China.
Collapse
Affiliation(s)
| | | | - Linwei Tian
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 7 Sassoon Road, Hong Kong SAR, China; (E.C.); (H.W.)
| |
Collapse
|
15
|
Du B, Wei L, Wang J, Li Y, Huo J, Wang J, Wang P. KIFC1 promotes proliferation and pseudo-bipolar division of ESCC through the transportation of Aurora B kinase. Aging (Albany NY) 2023; 15:12633-12650. [PMID: 37955677 PMCID: PMC10683620 DOI: 10.18632/aging.205203] [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: 07/04/2023] [Accepted: 10/15/2023] [Indexed: 11/14/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) accounts for over 90% of total in China, and the five-year survival rate for patients is less than 30%. Accordingly, the identification of novel, effective early diagnosis markers and therapeutic targets for ESCC is of paramount importance. KIFC1 has been identified as highly expressed in several types of cancer, although its prognostic value is inconsistent, and no research has been conducted specifically on its effect on ESCC. To investigate the expression and function of KIFC1 in ESCC, we conducted immunohistochemical staining on 30 pairs of para-carcinoma tissue and cancerous tissues, revealing a significant increase in KIFC1 expression in ESCC tissues. Using siRNA to knock down KIFC1 significantly reduced the proliferation of EC109 ESCC cells both in vitro and in vivo. Bioinformatics analysis revealed a highly significant positive correlation between KIFC1 overexpression and signaling pathways associated with tumor proliferation pathways. In EC109 cells, overexpression of KIFC1 significantly increased the rate of centrosome amplification and the likelihood of pseudo-bipolar division. Furthermore, the expression of KIFC1 and the rate of centrosome amplification in ESCC tissues were also positively correlated. In order to explore the underline molecular mechanisms, we identified, through proteomics, that KIFC1 binds to the protein Aurora B. The knockdown of KIFC1 significantly reduced the distribution of Aurora B on the metaphase plate and substantially inhibited the phosphorylation of its classical substrate, Histone H3. In conclusion, these findings indicate the potential utility of KIFC1 as both a tumor marker and a promising target for therapeutic interventions.
Collapse
Affiliation(s)
- Bin Du
- Center of Healthy Aging, Changzhi Medical College, Changzhi 047500, China
| | - Lingyu Wei
- Department of Pathology, Affiliated HePing Hospital of Changzhi Medical College, Changzhi 047500, China
| | - Jia Wang
- Center of Healthy Aging, Changzhi Medical College, Changzhi 047500, China
| | - Yanyan Li
- Center of Healthy Aging, Changzhi Medical College, Changzhi 047500, China
| | - Jing Huo
- Department of Pathology, The First Clinical College of Changzhi Medical College, Changzhi 047500, China
| | - Jinsheng Wang
- Department of Biology, Changzhi Medical College, Changzhi 047500, China
| | - Pu Wang
- Center of Healthy Aging, Changzhi Medical College, Changzhi 047500, China
| |
Collapse
|
16
|
Lu Q, Tang Y, Luo S, Gong Q, Li C. Coptisine, the Characteristic Constituent from Coptis chinensis, Exhibits Significant Therapeutic Potential in Treating Cancers, Metabolic and Inflammatory Diseases. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2023; 51:2121-2156. [PMID: 37930333 DOI: 10.1142/s0192415x2350091x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Naturally derived alkaloids belong to a class of quite significant organic compounds. Coptisine, a benzyl tetrahydroisoquinoline alkaloid, is one of the major bioactive constituents in Coptis chinensis Franch., which is a famous traditional Chinese medicine. C. chinensis possesses many kinds of functions, including the ability to eliminate heat, expel dampness, purge fire, and remove noxious substances. In Asian countries, C. chinensis is traditionally employed to treat carbuncle and furuncle, diabetes, jaundice, stomach and intestinal disorders, red eyes, toothache, and skin disorders. Up to now, there has been plenty of research of coptisine with respect to its pharmacology. Nevertheless, a comprehensive review of coptisine-associated research is urgently needed. This paper was designed to summarize in detail the progress in the research of the pharmacology, pharmacokinetics, safety, and formulation of coptisine. The related studies included in this paper were retrieved from the following academic databases: The Web of Science, PubMed, Google scholar, Elsevier, and CNKI. The cutoff date was January 2023. Coptisine manifests various pharmacological actions, including anticancer, antimetabolic disease, anti-inflammatory disease, and antigastrointestinal disease effects, among others. Based on its pharmacokinetics, the primary metabolic site of coptisine is the liver. Coptisine is poorly absorbed in the gastrointestinal system, and most of it is expelled in the form of its prototype through feces. Regarding safety, coptisine displayed potential hepatotoxicity. Some novel formulations, including the [Formula: see text]-cyclodextrin-based inclusion complex and nanocarriers, could effectively enhance the bioavailability of coptisine. The traditional use of C. chinensis is closely connected with the pharmacological actions of coptisine. Although there are some disadvantages, including poor solubility, low bioavailability, and possible hepatotoxicity, coptisine is still a prospective naturally derived drug candidate, especially in the treatment of tumors as well as metabolic and inflammatory diseases. Further investigation of coptisine is necessary to facilitate the application of coptisine-based drugs in clinical practice.
Collapse
Affiliation(s)
- Qiang Lu
- Department of Pharmaceutical Sciences, Zhuhai Campus, Zhuhai 519041, P. R. China
| | - Ying Tang
- Department of Pharmacology, Zunyi Medical University, Zhuhai Campus, Zhuhai 519041, P. R. China
| | - Shuang Luo
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518005, P. R. China
| | - Qihai Gong
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, P. R. China
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563000, P. R. China
| | - Cailan Li
- Department of Pharmacology, Zunyi Medical University, Zhuhai Campus, Zhuhai 519041, P. R. China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, P. R. China
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563000, P. R. China
| |
Collapse
|
17
|
He L, Zhou X, Liu J, Yao Y, Lin J, Chen J, Qiu S, Liu Z, He Y, Yi Y, Zhou X, Zou F. RAE1 promotes nitrosamine-induced malignant transformation of human esophageal epithelial cells through PPARα-mediated lipid metabolism. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 265:115513. [PMID: 37774541 DOI: 10.1016/j.ecoenv.2023.115513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 09/11/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023]
Abstract
Esophageal cancer (EC) is the sixth cause of cancer-related deaths and still is a significant public health problem globally. Nitrosamines exposure represents a major health concern increasing EC risks. Exploring the mechanisms induced by nitrosamines may contribute to the prevention and early detection of EC. However, the mechanism of nitrosamine carcinogenesis remains unclear. Ribonucleic acid export 1 (RAE1), has an important role in mediating diverse cancer types, but, to date, there has been no study for any functional role of RAE1 in esophageal carcinogenesis. Here, we successfully verified the nitrosamine-induced malignant transformation cell (MNNG-M) by xenograft tumor model, based on which it was found that RAE1 was upregulation in the early stage of nitrosamine-induced esophageal carcinogenesis and EC tissues. RAE1 knockdown led to severe blockade in G2/M phase and significant inhibition of proliferation of MNNG-M cells, whereas RAE1 overexpression had the opposite effect. In addition, peroxisome proliferator-activated receptor-alpha (PPARα), was demonstrated as a downstream target gene of RAE1, and its down-regulation reduced lipid accumulation, resulting in causing cells accumulation in the G2/M phase. Mechanistically, we found that RAE1 regulates the lipid metabolism by maintaining the stability of PPARα mRNA. Taken together, our study reveals that RAE1 promotes malignant transformation of human esophageal epithelial cells (Het-1A) by regulating PPARα-mediated lipid metabolism to affect cell cycle progression, and offers a new explanation of the mechanisms underlying esophageal carcinogenesis.
Collapse
Affiliation(s)
- Ling He
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Xiangjun Zhou
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Jia Liu
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Yina Yao
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Junyuan Lin
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Jialong Chen
- Department of Preventive Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Shizhen Qiu
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Zeyu Liu
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Yingzheng He
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Yujie Yi
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Xueqiong Zhou
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China.
| | - Fei Zou
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China.
| |
Collapse
|
18
|
Yin LB, Li ZW, Wang JL, Wang L, Hou L, Hu SY, Chen H, Luo P, Cui XB, Zhu JL. Sulfasalazine inhibits esophageal cancer cell proliferation by mediating ferroptosis. Chem Biol Drug Des 2023; 102:730-737. [PMID: 37291716 DOI: 10.1111/cbdd.14281] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/19/2023] [Accepted: 05/26/2023] [Indexed: 06/10/2023]
Abstract
This study aimed to explore the potential mechanism by which sulfasalazine (SAS) inhibits esophageal cancer cell proliferation. A cell counting kit-8 (CCK-8) assay was used to detect the effect of SAS (0, 1, 2, and 4 mM) on the proliferation of TE-1 cells. Subsequently, TE-1 cells were divided into control group, SAS group, SAS + ferrostatin-1 (ferroptosis inhibitor) group, and SAS + Z-VAD (OH)-FMK (apoptosis inhibitor) group, and cell proliferation was measured using a CCK-8 assay. Real-time quantitative polymerase chain reaction and western blotting were used to determine the expression of solute carrier family member 7 11 (SLC7A11, also called xCT), glutathione peroxidase 4 (GPX4), and acyl-CoA synthase long-chain family member 4 (ACSL4) in TE-1 cells. Measurement of ferroptosis in TE-1 cells was achieved by flow cytometry. Compared with the control group (0 mM SAS), the proliferation of TE-1 cells was significantly inhibited by different concentrations of SAS for different time lengths, and 4 mM SAS treatment for 48 h could obtain the maximum inhibition rate (53.9%). In addition, SAS treatment caused a significant decrease in the mRNA and protein expression of xCT and GPX4, and a significant increase in ACSL4 expression in TE-1 cells treated with SAS. Flow cytometry results showed that the ferroptosis level was significantly increased after SAS treatment. However, the activation of ferroptosis by SAS was partially eliminated by treatment with ferrostatin-1 or Z-VAD (OH)-FMK. In conclusion, SAS inhibits the proliferation of esophageal carcinoma cells by activating the ferroptosis pathway.
Collapse
Affiliation(s)
- Lai-Bo Yin
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Zhi-Wei Li
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Jing-Ling Wang
- The Second Department of Traditional Chinese Medicine, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Lei Wang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Liang Hou
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Si-Yuan Hu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Huan Chen
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Pan Luo
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Xiao-Bin Cui
- Department of Pathology, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Jia-Long Zhu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| |
Collapse
|
19
|
Mokhlesi A, Sharifi Z, Berimipour A, Taleahmad S, Talkhabi M. Identification of hub genes and microRNAs with prognostic values in esophageal cancer by integrated analysis. Noncoding RNA Res 2023; 8:459-470. [PMID: 37416747 PMCID: PMC10319852 DOI: 10.1016/j.ncrna.2023.05.009] [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: 02/12/2023] [Revised: 05/07/2023] [Accepted: 05/31/2023] [Indexed: 07/08/2023] Open
Abstract
Esophageal cancer (EC) is the eighth most common cancer in the world, and the sixth most common cause of cancer-related mortality. The aim of the present study was to identify cell and molecular mechanisms involved in EC, and to provide the potential targets for diagnosis and treatment. Here, a microarray dataset (GSE20347) was screened to find differentially expressed genes (DEGs). Different bioinformatic methods were used to analyze the identified DEGs. The up-regulated DEGs were significantly involved in different biological processes and pathways including extracellular matrix organization and ECM-receptor interaction. FN1, CDK1, AURKA, TOP2A, FOXM1, BIRC5, CDC6, UBE2C, TTK, and TPX2 were identified as the most important genes among the up-regulated DEGs. Our analysis showed that has-miR-29a-3p, has-miR-29b-3p, has-miR-29c-3p, and has-miR-767-5p had the largest number of common targets among the up-regulated DEGs. These findings strengthen the understanding of EC development and progression, as well as representing potential markers for EC diagnosis and treatment.
Collapse
Affiliation(s)
- Amir Mokhlesi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Zahra Sharifi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Ahmad Berimipour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sara Taleahmad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahmood Talkhabi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
20
|
Li C, Zhang J, Bi Y. Unveiling the prognostic significance of SOX5 in esophageal squamous cell carcinoma: a comprehensive bioinformatic and experimental analysis. Aging (Albany NY) 2023; 15:7565-7582. [PMID: 37531195 PMCID: PMC10457070 DOI: 10.18632/aging.204924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/10/2023] [Indexed: 08/03/2023]
Abstract
BACKGROUND This study aimed to investigate the expression and prognostic significance of SOX5 in esophageal squamous cell carcinoma (ESCC). METHODS Gene Expression Omnibus (GEO) data were analyzed to assess SOX5 expression in ESCC and normal tissues. Survival analysis was performed to evaluate its prognostic significance. Pathway enrichment analysis was conducted to identify pathways associated with low SOX5 expression. Methylation status of CpG sites in ESCC cases was examined, and SOX5 expression was evaluated. Differential expression and ChIP-seq data analyses were used to identify genes significantly correlated with SOX5 and to obtain target genes. A protein-protein interaction (PPI) network was constructed using hub genes, and their association with immune cell infiltration was determined. In vitro ESCC cell experiments validated the findings. RESULTS SOX5 was significantly downregulated in ESCC samples compared to normal samples. Its downregulation was associated with shorter survival in ESCC patients. Pathway enrichment analysis revealed enrichment in regulated necrosis, NLRP3 inflammasome, formation of the cornified envelope, and PD-1 signaling. Methylation status of two CpG sites negatively correlated with SOX5 expression. Differential expression analysis identified 122 genes significantly correlated with SOX5, and 28 target genes were obtained from ChIP-seq analysis. Target genes were enriched in DNA replication, cell cycle, spindle, and ATPase activity. Five hub genes were identified, and the PPI network showed significant associations with immune cell infiltration. In vitro experiments confirmed SOX5 downregulation, upregulation of hub genes, and their functional effects on ESCC cell apoptosis and proliferation. CONCLUSIONS These findings enhance understanding of SOX5 in ESCC and potential therapeutic strategies.
Collapse
Affiliation(s)
- Chenglin Li
- Department of Cardiothoracic Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
- Department of Cardiothoracic Surgery, The Affiliated Huaian No.1 People’s Hospital of Nanjing Medical University, Huaian 223300, Jiangsu, China
| | - Jialing Zhang
- Department of Gastroenterology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian 223300, Jiangsu, China
| | - Yanwen Bi
- Department of Cardiothoracic Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| |
Collapse
|
21
|
Zeng X, Ye L, Luo M, Zeng D, Chen Y. Prognostic value of pretreatment systemic immune-inflammation index in Chinese esophageal squamous cell carcinoma patients receiving radical radiotherapy: A meta-analysis. Medicine (Baltimore) 2023; 102:e34117. [PMID: 37352061 PMCID: PMC10289742 DOI: 10.1097/md.0000000000034117] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND The association between pretreatment systemic immune-inflammation index (SII) and long-term survival among Chinese esophageal squamous cell carcinoma (ESCC) patients who received radical radiotherapy remains unclear. The aim of this study was to identify the prognostic role of pretreatment SII in Chinese ESCC patients receiving radical radiotherapy based on current evidence. METHODS The PubMed, EMBASE, Web of Science and CNKI databases were searched up to March 18, 2023. Primary and secondary outcomes were overall survival (OS) and progression-free survival (PFS), respectively. The hazard ratios (HRs) with corresponding 95% confidence intervals (CIs) were combined to assess the predictive role of pretreatment SII for long-term survival of Chinese ESCC patients receiving radiotherapy. All statistical analyses were conducted by STATA 15.0 software. RESULTS A total of 8 eligibility studies involving 2101 cases were included in this meta-analysis. The pooled results demonstrated that elevated pretreatment SII was significantly related to worse OS (HR = 1.59, 95% CI: 1.24-2.02, P < .001) and PFS (HR = 1.33, 95% CI: 1.13-1.57, P < .001). Besides, subgroup based on TNM stage showed similar results. CONCLUSION Pretreatment SII could serve as a novel prognostic factor in Chinese ESCC patients receiving definitive radiotherapy and patients with an elevated SII may experience poorer survival.
Collapse
Affiliation(s)
- Xiaoxiao Zeng
- Department of Oncology, The People’s Hospital of JianYang City, Jian Yang, China
| | - Ling Ye
- Department of Oncology, The People’s Hospital of JianYang City, Jian Yang, China
| | - Mingying Luo
- Department of Oncology, The People’s Hospital of JianYang City, Jian Yang, China
| | - Danli Zeng
- Department of Oncology, The People’s Hospital of JianYang City, Jian Yang, China
| | - Yang Chen
- Department of Oncology, The People’s Hospital of JianYang City, Jian Yang, China
| |
Collapse
|
22
|
Huang YY, Zheng Y, Liang SH, Wu LL, Liu X, Xing WQ, Ma GW. Establishment and validation of a prognostic risk classification for patients with stage T1-3N0M0 esophageal squamous cell carcinoma. J Cardiothorac Surg 2023; 18:192. [PMID: 37316912 DOI: 10.1186/s13019-023-02294-2] [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: 12/29/2021] [Accepted: 04/15/2023] [Indexed: 06/16/2023] Open
Abstract
INTRODUCTION At present, clinical factors and hematological indicators have been proved to have great potential in predicting the prognosis of cancer patients, and no one has combined these two valuable indicators to establish a prognostic model for esophageal squamous cell carcinoma (ESCC) patients with stage T1-3N0M0 after R0 resection. To verify, we aimed to combine these potential indicators to establish a prognostic model. METHODS Stage T1-3N0M0 ESCC patients from two cancer centers (including training cohort: N = 819, and an external validation cohort: N = 177)-who had undergone esophagectomy in 1995-2015 were included. We integrated significant risk factors for death events by multivariable logistic regression methods and applied them to the training cohort to build Esorisk. The parsimonious aggregate Esorisk score was calculated for each patient; the training set was divided into three prognostic risk classes according to the 33rd and 66th percentiles of the Esorisk score. The association of Esorisk with cancer-specific survival (CSS) was assessed using Cox regression analyses. RESULTS The Esorisk model was: [10 + 0.023 × age + 0.517 × drinking history - 0.012 × hemoglobin-0.042 × albumin - 0.032 × lymph nodes]. Patients were grouped into three classes-Class A (5.14-7.26, low risk), Class B (7.27-7.70, middle risk), and Class C (7.71-9.29, high risk). In the training group, five-year CSS decreased across the categories (A: 63%; B: 52%; C: 30%, Log-rank P < 0.001). Similar findings were observed in the validation group. Additionally, Cox regression analysis showed that Esorisk aggregate score remained significantly associated with CSS in the training cohort and validation cohort after adjusting for other confounders. CONCLUSIONS We combined the data of two large clinical centers, and comprehensively considered their valuable clinical factors and hematological indicators, established and verified a new prognostic risk classification that can predict CSS of stage T1-3N0M0 ESCC patients.
Collapse
Affiliation(s)
- Yang-Yu Huang
- The Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University, No. 651 Dongfengdong Road, Guangzhou, 510060, People's Republic of China
- Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Yan Zheng
- The Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, No. 1 Jianshedong Road, Zhengzhou, 45000, People's Republic of China
| | - Shen-Hua Liang
- The Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University, No. 651 Dongfengdong Road, Guangzhou, 510060, People's Republic of China
| | - Lei-Lei Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xuan Liu
- The Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University, No. 651 Dongfengdong Road, Guangzhou, 510060, People's Republic of China
| | - Wen-Qun Xing
- The Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, No. 1 Jianshedong Road, Zhengzhou, 45000, People's Republic of China
| | - Guo-Wei Ma
- The Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University, No. 651 Dongfengdong Road, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
23
|
Shah MA, Altorki N, Patel P, Harrison S, Bass A, Abrams JA. Improving outcomes in patients with oesophageal cancer. Nat Rev Clin Oncol 2023; 20:390-407. [PMID: 37085570 DOI: 10.1038/s41571-023-00757-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 04/23/2023]
Abstract
The care of patients with oesophageal cancer or of individuals who have an elevated risk of oesophageal cancer has changed dramatically. The epidemiology of squamous cell and adenocarcinoma of the oesophagus has diverged over the past several decades, with a marked increase in incidence only for oesophageal adenocarcinoma. Only in the past decade, however, have molecular features that distinguish these two forms of the disease been identified. This advance has the potential to improve screening for oesophageal cancers through the development of novel minimally invasive diagnostic technologies predicated on cancer-specific genomic or epigenetic alterations. Surgical techniques have also evolved towards less invasive approaches associated with less morbidity, without compromising oncological outcomes. With improvements in multidisciplinary care, advances in radiotherapy and new tools to detect minimal residual disease, certain patients may no longer even require surgical tumour resection. However, perhaps the most anticipated advance in the treatment of patients with oesophageal cancer is the advent of immune-checkpoint inhibitors, which harness and enhance the host immune response against cancer. In this Review, we discuss all these advances in the management of oesophageal cancer, representing only the beginning of a transformation in our quest to improve patient outcomes.
Collapse
Affiliation(s)
- Manish A Shah
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - Nasser Altorki
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Pretish Patel
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sebron Harrison
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Adam Bass
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Julian A Abrams
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
24
|
Xu W, Jiang H, Liu Y, Liu X, Jiang Y. Retrospective cohort study on treatment modalities and survival time after oesophageal fistula in patients with oesophageal cancer in a regional cancer care centre in China. BMJ Open 2023; 13:e069703. [PMID: 37045573 PMCID: PMC10106048 DOI: 10.1136/bmjopen-2022-069703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND Oesophageal fistula (perforation) is a devastating complication in patients with oesophageal cancer . The optimal treatment remains uncertain. OBJECTIVE We sought to present real-world evidence on treatment modalities and survival postfistula in patients with oesophageal cancer. DESIGN, SETTINGS AND MAIN OUTCOMES This was a retrospective cohort study of patients with oesophageal cancer with oesophageal fistulae diagnosed between June 2010 and June 2020 in a regional cancer care centre in Zhengzhou, China (n=352). The treatment options included surgical resection, oesophageal stent grafting, gastrostomy, nasogastric tube and conservative care. The primary outcome was survival time (months) postfistula. Inverse probability of treatment weighting (IPTW) life regression was used to estimate the differences in survival time accounting for potential confounders. RESULTS The median survival time was 2.3 months (IQR: 0.7-6.0 months). Survival times were shorter in patients of male sex, T4 stage and oesophagotracheal versus oesophageal-mediastinal fistulae, and longer for any treatment option versus conservative care. The IPTW life regression analyses showed that in patients with oesophagotracheal fistulae, survival times were longer for stent grafting (+0.90 (95% CI 0.60 to 1.19) months) or gastrostomy (+0.81 (95% CI 0.47 to 1.13) months) versus nasogastric tube. In patients with oesophageal-mediastinal fistulae, survival times were shorter for stent grafting versus nasogastric tube (-0.36 (95% CI -0.63 to -0.09) months) and gastric tube (-0.29 (95% CI -0.50 to -0.08) months). Surgical resection was recorded in nine patients with oesophageal-mediastinal fistulae, and it was associated with the longest survival time in these patients. CONCLUSIONS Stent grafting or gastrostomy may be preferable to nasogastric tube in survival prognosis for patients with oesophageal cancer with oesophagotracheal fistulae. In contrast, stent grafting may be not preferable to nasogastric tube or gastrostomy in survival prognosis for patients with oesophageal-mediastinal fistulae.
Collapse
Affiliation(s)
- Wencai Xu
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Hui Jiang
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Yang Liu
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Xiao Liu
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Yue Jiang
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| |
Collapse
|
25
|
Liang F, Luo Q, Han H, Zhang J, Yang Y, Chen J. Long noncoding RNA LINC01088 inhibits esophageal squamous cell carcinoma progression by targeting the NPM1-HDM2-p53 axis. Acta Biochim Biophys Sin (Shanghai) 2023; 55:367-381. [PMID: 36942988 PMCID: PMC10160232 DOI: 10.3724/abbs.2023021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is characterized by extensive metastasis and poor prognosis. Long noncoding RNAs (lncRNAs) have been shown to play important roles in ESCC. However, the specific roles of lncRNAs in ESCC tumorigenesis and metastasis remain largely unknown. Here, we investigate LINC01088 in ESCC. Differentially expressed LINC01088 levels are screened from the GEO database. We find that LINC01088 is expressed at low level in collected clinical samples and is correlated with vascular tumor emboli and poor overall survival time of patients after surgery. LINC01088 inhibits not only ESCC cell migration and invasion in vitro, but also tumorigenesis and metastasis in vivo. Mechanistically, LINC01088 directly interacts with nucleophosmin (NPM1) and increases the expression of NPM1 in the nucleoplasm compared to that in the nucleolar region. LINC01088 decreases mutant p53 (mut-p53) expression and rescues the transcriptional activity of p53 by targeting the NPM1-HDM2-p53 axis. LINC01088 may also interfere with the DNA repair function of NPM1 by affecting its translocation. Our results highlight the potential of LINC01088 as a prognostic biomarker and therapeutic target of ESCC.
Collapse
Affiliation(s)
- Fan Liang
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Qiuli Luo
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100020, China
| | - Haibo Han
- Department of Clinical Laboratory, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jianzhi Zhang
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yue Yang
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jinfeng Chen
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
26
|
Han Y, Zhu X, Hu Y, Yu C, Guo Y, Hang D, Pang Y, Pei P, Ma H, Sun D, Yang L, Chen Y, Du H, Yu M, Chen J, Chen Z, Huo D, Jin G, Lv J, Hu Z, Shen H, Li L. Electronic Health Record-Based Absolute Risk Prediction Model for Esophageal Cancer in the Chinese Population: Model Development and External Validation. JMIR Public Health Surveill 2023; 9:e43725. [PMID: 36781293 PMCID: PMC10132027 DOI: 10.2196/43725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/09/2023] [Accepted: 02/03/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND China has the largest burden of esophageal cancer (EC). Prediction models can be used to identify high-risk individuals for intensive lifestyle interventions and endoscopy screening. However, the current prediction models are limited by small sample size and a lack of external validation, and none of them can be embedded into the booming electronic health records (EHRs) in China. OBJECTIVE This study aims to develop and validate absolute risk prediction models for EC in the Chinese population. In particular, we assessed whether models that contain only EHR-available predictors performed well. METHODS A prospective cohort recruiting 510,145 participants free of cancer from both high EC-risk and low EC-risk areas in China was used to develop EC models. Another prospective cohort of 18,441 participants was used for validation. A flexible parametric model was used to develop a 10-year absolute risk model by considering the competing risks (full model). The full model was then abbreviated by keeping only EHR-available predictors. We internally and externally validated the models by using the area under the receiver operating characteristic curve (AUC) and calibration plots and compared them based on classification measures. RESULTS During a median of 11.1 years of follow-up, we observed 2550 EC incident cases. The models consisted of age, sex, regional EC-risk level (high-risk areas: 2 study regions; low-risk areas: 8 regions), education, family history of cancer (simple model), smoking, alcohol use, BMI (intermediate model), physical activity, hot tea consumption, and fresh fruit consumption (full model). The performance was only slightly compromised after the abbreviation. The simple and intermediate models showed good calibration and excellent discriminating ability with AUCs (95% CIs) of 0.822 (0.783-0.861) and 0.830 (0.792-0.867) in the external validation and 0.871 (0.858-0.884) and 0.879 (0.867-0.892) in the internal validation, respectively. CONCLUSIONS Three nested 10-year EC absolute risk prediction models for Chinese adults aged 30-79 years were developed and validated, which may be particularly useful for populations in low EC-risk areas. Even the simple model with only 5 predictors available from EHRs had excellent discrimination and good calibration, indicating its potential for broader use in tailored EC prevention. The simple and intermediate models have the potential to be widely used for both primary and secondary prevention of EC.
Collapse
Affiliation(s)
- Yuting Han
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Xia Zhu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Yizhen Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
| | - Yu Guo
- Chinese Academy of Medical Sciences, Beijing, China
| | - Dong Hang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Yuanjie Pang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Pei Pei
- Chinese Academy of Medical Sciences, Beijing, China
| | - Hongxia Ma
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Dianjianyi Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
| | - Ling Yang
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, United Kingdom
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Yiping Chen
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, United Kingdom
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Huaidong Du
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, United Kingdom
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Min Yu
- Zhejiang Center for Disease Control and Prevention, Hangzhou, China
| | - Junshi Chen
- China National Center for Food Safety Risk Assessment, Beijing, China
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Dezheng Huo
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, United States
| | - Guangfu Jin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
| | - Zhibin Hu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Hongbing Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
| |
Collapse
|
27
|
Meng L, Hu YT, Xu AM. F-box and leucine-rich repeat 6 promotes gastric cancer progression via the promotion of epithelial-mesenchymal transition. World J Gastrointest Oncol 2023; 15:490-503. [PMID: 37009323 PMCID: PMC10052668 DOI: 10.4251/wjgo.v15.i3.490] [Citation(s) in RCA: 2] [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: 12/04/2022] [Revised: 01/06/2023] [Accepted: 02/15/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND F-box and leucine-rich repeat 6 (FBXL6) have reportedly been associated with several cancer types. However, the role and mechanisms of FBXL6 in gastric cancer (GC) require further elucidation.
AIM To investigate the effect of FBXL6 in GC tissues and cells and the underlying mechanisms.
METHODS TCGA and GEO database analysis was performed to evaluate the expression of FBXL6 in GC tissues and adjacent normal tissues. Reverse transcription-quantitative polymerase chain reaction, immunofluorescence, and western blotting were used to detect the expression of FBXL6 in GC tissue and cell lines. Cell clone formation, 5-ethynyl-2’-deoxyuridine (EdU) assays, CCK-8, transwell migration assay, and wound healing assays were performed to evaluate the malignant biological behavior in GC cell lines after transfection with FBXL6-shRNA and the overexpression of FBXL6 plasmids. Furthermore, in vivo tumor assays were performed to prove whether FBXL6 promoted cell proliferation in vivo.
RESULTS FBXL6 expression was upregulated more in tumor tissues than in adjacent normal tissues and positively associated with clinicopathological characteristics. The outcomes of CCK-8, clone formation, and Edu assays demonstrated that FBXL6 knockdown inhibited cell proliferation, whereas upregulation of FBXL6 promoted proliferation in GC cells. Additionally, the transwell migration assay revealed that FBXL6 knockdown suppressed migration and invasion, whereas the overexpression of FBXL6 showed the opposite results. Through the subcutaneous tumor implantation assay, it was evident that the knockdown of FBXL6 inhibited GC graft tumor growth in vivo. Western blotting showed that the effects of FBXL6 on the expression of the proteins associated with the epithelial-mesenchymal transition-associated proteins in GC cells.
CONCLUSION Silencing of FBXL6 inactivated the EMT pathway to suppress GC malignancy in vitro. FBXL6 can potentially be used for the diagnosis and targeted therapy of patients with GC.
Collapse
Affiliation(s)
- Lei Meng
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Yu-Ting Hu
- Department of Immunology, College of Basic Medicine, Anhui Medical University, Hefei 230022, Anhui Province, China
| | - A-Man Xu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| |
Collapse
|
28
|
Meng L, Hu P, Xu A. PGAM5 promotes tumorigenesis of gastric cancer cells through PI3K/AKT pathway. Pathol Res Pract 2023; 244:154405. [PMID: 36889176 DOI: 10.1016/j.prp.2023.154405] [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: 10/24/2022] [Revised: 02/22/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
PGAM5 has been associated with the development of tumours, however, its function in gastric cancer (GC) remains unexplored. Here, we investigated the role and mechanism of PGAM5 in regulating GC. The results revealed that PGAM5 was upregulated in GC tissues and cell lines, which was correlated with tumour size and TNM stage. Moreover, PGAM5 knockdown inhibited proliferation, migration, and invasion progression, whereas PGAM5 overexpression promoted the function of GC cells in vitro. PGAM5 also promoted the activation of the PI3K/AKT signalling pathway. Furthermore, MK-2206, an AKT inhibitor, reversed the proliferation and activation of the PI3K/AKT signalling pathway induced by PGAM5 knockdown in GC cells. In conclusion, PGAM5 promotes the proliferation of GC by positively regulating the activation of the PI3K/AKT signalling pathway in GC cells.
Collapse
Affiliation(s)
- Lei Meng
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Pibo Hu
- Department of General Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No. 1558, Sanhuan North Road, Huzhou, Zhejiang, China
| | - Aman Xu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| |
Collapse
|
29
|
Luo Y, Yao Y, Wu P, Zi X, Sun N, He J. Profile of treatment-related adverse events of PD-1 blockade-based therapies in advanced esophageal cancer: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2023; 183:103922. [PMID: 36696933 DOI: 10.1016/j.critrevonc.2023.103922] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/29/2022] [Accepted: 01/20/2023] [Indexed: 01/23/2023] Open
Abstract
PD-1 blockade-based therapies are the most promising treatment for advanced esophageal cancer (EC). It is crucial to investigate the corresponding toxicity profiles of treatment-related adverse events (TRAEs). We conducted a systematic review and meta-analysis to explore toxicity profiles across different PD-1 blockade-based treatments in EC. A total of 5595 patients from 10 clinical trials were included. The overall rates of TRAEs were 88 % (95 % CI 72.0-95.0), 98.0 % (97.0-99.0), and 79.5 % (74.6-83.7) for all grade TRAEs, 24.0 % (15.0-36.0), 64.0 % (56.0-71.0), and 34.2 % (29.1-39.7) for grade 3 or higher TRAEs in PD-1 blockade alone, PD-1 blockade plus chemotherapy, and dual blockade group, respectively. Compared to chemotherapy, RRs for patients receiving PD-1 blockade-based treatments for all grade TRAEs were 0.96 (93.0-100.0) and 0.75 (60.0-94.0) for grade 3 or higher TRAEs. We exhibited comprehensive statistics on the toxicity of the PD-1 blockade-based regimens, providing useful references for clinicians.
Collapse
Affiliation(s)
- Yuejun Luo
- 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 100021, China; State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuxin Yao
- 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 100021, China
| | - Peng Wu
- 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 100021, China; State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohui Zi
- 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 100021, China; State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Sun
- 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 100021, China; State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jie He
- 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 100021, China; State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
30
|
Ding N, Cheng Y, Liu H, Wu Y, Weng Y, Cui H, Cheng C, Zhang W, Cui Y. Fusobacterium nucleatum Infection Induces Malignant Proliferation of Esophageal Squamous Cell Carcinoma Cell by Putrescine Production. Microbiol Spectr 2023; 11:e0275922. [PMID: 36840590 PMCID: PMC10101128 DOI: 10.1128/spectrum.02759-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 01/13/2023] [Indexed: 02/24/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a malignant upper digestive tract cancer, and its pathogenesis and etiology are poorly understood. Because gut microbes commonly impact progression, metastasis, and immunotherapy responses in colorectal cancer (CRC), the roles of the esophageal microbiota in ESCC have gradually drawn attention. As reported previously, Fusobacterium nucleatum (Fn), the notable "culprit" of CRC, can also influence the prognosis of ESCC in clinical studies. However, thus far, the underlying mechanism is unclear. In this study, 73 Chinese ESCC samples were collected. In those clinical samples, the abundance of Fn was found to be higher in tumors than in adjacent normal tissues, and a high abundance of Fn was correlated with shorter survival. Furthermore, using in vitro experiments, we demonstrated that Fn can invade ESCC cells, enhancing their proliferation capacity. The mechanism study revealed that Fn can produce high levels of putrescine after invasion, which disturbs polyamine metabolism and promotes the malignant proliferation of ESCC cells. In conclusion, Fn infection was found in Chinese ESCC tumor tissue samples and may promote ESCC progression by disturbing the polyamine metabolism pathway. IMPORTANCE Nowadays, the complex and varied interactions between microbes and human body are known to be crucial for maintaining the health of the human body. However, knowledge concerning the influence of esophageal microbes on the progression of esophageal squamous cell carcinoma is limited. Here, in our study, we confirmed that F. nucleatum can invade ESCC cells and consequently promote their proliferation, suggesting that esophageal microbes likely influence the progression of ESCC in clinical settings. Because the esophagus connects the oral cavity and stomach, acting as a canal for transporting foods, its special physical location makes it easily exposed to microorganisms. Thus, it is necessary to explore the interaction between esophageal microbes and ESCC.
Collapse
Affiliation(s)
- Ning Ding
- Cancer Institute, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Peking University Shenzhen Hospital, Shenzhen Peking University–Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, People’s Republic of China
| | - Yikun Cheng
- College of Letters & Science, University of California Berkeley, Berkeley, California, USA
| | - Huijuan Liu
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Yueguang Wu
- Cancer Institute, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Peking University Shenzhen Hospital, Shenzhen Peking University–Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, People’s Republic of China
| | - Yongjia Weng
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Heyang Cui
- Cancer Institute, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Peking University Shenzhen Hospital, Shenzhen Peking University–Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, People’s Republic of China
| | - Chen Cheng
- Cancer Institute, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Peking University Shenzhen Hospital, Shenzhen Peking University–Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, People’s Republic of China
| | - Weimin Zhang
- Cancer Institute, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Peking University Shenzhen Hospital, Shenzhen Peking University–Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, People’s Republic of China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Molecular Oncology, Peking University Cancer Hospital and Institute, Beijing, People’s Republic of China
| | - Yongping Cui
- Cancer Institute, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Peking University Shenzhen Hospital, Shenzhen Peking University–Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, People’s Republic of China
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| |
Collapse
|
31
|
Hu JF, Song X, Zhong K, Zhao XK, Zhou FY, Xu RH, Li JL, Wang XZ, Li XM, Wang PP, Lei LL, Wei MX, Wang R, Fan ZM, Han XN, Chen Y, Li LY, Ji JJ, Yang YZ, Li B, Yang MM, Yang HJ, Chang FB, Ren JL, Zhou SL, Wang LD. Increases prognostic value of clinical-pathological nomogram in patients with esophageal squamous cell carcinoma. Front Oncol 2023; 13:997776. [PMID: 36865805 PMCID: PMC9973522 DOI: 10.3389/fonc.2023.997776] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 01/04/2023] [Indexed: 02/16/2023] Open
Abstract
Background This study was intended to construct a brand new prognostic nomogram after combine clinical and pathological characteristics to increases prognostic value in patients with esophageal squamous cell carcinoma. Methods A total of 1,634 patients were included. Subsequently, the tumor tissues of all patients were prepared into tissue microarrays. AIPATHWELL software was employed to explore tissue microarrays and calculate the tumor-stroma ratio. X-tile was adopted to find the optimal cut-off value. Univariate and multivariate Cox analyses were used to screen out remarkable characteristics for constructing the nomogram in the total populations. A novel prognostic nomogram with clinical and pathological characteristics was constructed on the basis of the training cohort (n=1,144). What's more performance was validated in the validation cohort (n=490). Clinical-pathological nomogram were assessed by concordance index, time-dependent receiver operating characteristic, calibration curve and decision curve analysis. Results The patients can divide into two groups with cut-off value of 69.78 for the tumor-stroma ratio. It is noteworthy that the survival difference was noticeable (P<0.001). A clinical-pathological nomogram was constructed by combining clinical and pathological characteristics to predict the overall survival. In comparison with TNM stage, the concordance index and time-dependent receiver operating characteristic of the clinical-pathological nomogram showed better predictive value (P<0.001). High quality of calibration plots in overall survival was noticed. As demonstrated by the decision curve analysis, the nomogram has better value than the TNM stage. Conclusions As evidently revealed by the research findings, tumor-stroma ratio is an independent prognostic factor in patients with esophageal squamous cell carcinoma. The clinical-pathological nomogram has an incremental value compared TNM stage in predicting overall survival.
Collapse
Affiliation(s)
- Jing Feng Hu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xin Song
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Kan Zhong
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xue Ke Zhao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Fu You Zhou
- Department of Thoracic Surgery, Anyang Tumor Hospital, Anyang, Henan, China
| | - Rui Hua Xu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ji Lin Li
- Department of Pathology, Linzhou Esophageal Cancer Hospital, Linzhou, Henan, China
| | - Xian Zeng Wang
- Department of Thoracic Surgery, Linzhou People’s Hospital, Linzhou, Henan, China
| | - Xue Min Li
- Department of Pathology, Hebei Provincial Cixian People’s Hospital, Cixian, Hebei, China
| | - Pan Pan Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ling Ling Lei
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Meng Xia Wei
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ran Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Zong Min Fan
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xue Na Han
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yao Chen
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Liu Yu Li
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Jia Jia Ji
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuan Ze Yang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Bei Li
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Miao Miao Yang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Hai Jun Yang
- Department of Thoracic Surgery, Anyang Tumor Hospital, Anyang, Henan, China
| | - Fu Bao Chang
- Department of Surgery, Central Hospital of Linzhou City, Linzhou, Henan, China
| | - Jing Li Ren
- Department of Pathology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Sheng Li Zhou
- Department of Pathology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, Henan, China
| | - Li Dong Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China,*Correspondence: Li Dong Wang,
| |
Collapse
|
32
|
Zhang R, Li C, Wan Z, Qin J, Li Y, Wang Z, Zheng Q, Kang X, Chen X, Li Y, He J, Li Y. Comparative genomic analysis of esophageal squamous cell carcinoma among different geographic regions. Front Oncol 2023; 12:999424. [PMID: 36741715 PMCID: PMC9889985 DOI: 10.3389/fonc.2022.999424] [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: 07/21/2022] [Accepted: 12/30/2022] [Indexed: 01/19/2023] Open
Abstract
Introduction Esophageal squamous cell carcinoma (ESCC) shows remarkable variation in incidence, survival, and risk factors. Although the genomic characteristics of ESCC have been extensively characterized, the genomic differences between different geographic regions remain unclear. Methods In this study, we sequenced 111 patients with ESCC from northern (NC) and southern (SC) China, combined their data with those of 1081 cases from previous reports, and performed a comparative analysis among different regions. In total, 644 ESCC cases were collected from six geographic regions (NC, SC, Xinjiang, China [XJC], Japan [JP], Vietnam [VN], and Europe & America [EA]) as the discovery cohort. Validation cohort 1 included 437 patients with ESCC from the NC region. Validation cohort 2 included 54 and 57 patients from the NC and SC regions, respectively. Results Patients with ESCC in different regions had different genomic characteristics, including DNA signatures, tumor mutation burdens, significantly mutated genes (SMGs), altered signaling pathways, and genes associated with clinical features. Based on both the DNA mutation signature and the mutation profile of the most common genes, the NC and SC groups were clustered close together, followed by the JP, XJC, EA, and VN groups. Compared to patients with ESCC from SC, SMGs, including KMT2D, FAT1, and NOTCH1 were more frequently identified in patients with ESCC from NC. Furthermore, some genes (TDG and DNAH8) correlated with overall survival in completely opposite ways in patients with ESCC from different geographical regions. Conclusions Our study provides insights into genomic differences in ESCC among different regions. These differences may be related to differences in environmental carcinogens, incidence, and survival.
Collapse
Affiliation(s)
- Ruixiang Zhang
- 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
| | - Canjun Li
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiyi Wan
- Department of Medicine, Genecast Biotechnology Co., Ltd, Wuxi, China
| | - Jianjun Qin
- 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
| | - Yong Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhen Wang
- 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
| | - Qingfeng Zheng
- 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
| | - Xiaozheng Kang
- 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
| | - Xiankai Chen
- 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
| | - Yun Li
- Department of Medicine, Genecast Biotechnology Co., Ltd, Wuxi, China
| | - Jie He
- 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,*Correspondence: Jie He, ; Yin Li,
| | - Yin Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China,*Correspondence: Jie He, ; Yin Li,
| |
Collapse
|
33
|
Yang W, Liu F, Xu R, Yang W, He Y, Liu Z, Zhou F, Heng F, Hou B, Zhang L, Chen L, Zhang F, Cai F, Xu H, Lin M, Liu M, Pan Y, Liu Y, Hu Z, Chen H, He Z, Ke Y. Is Adjuvant Therapy a Better Option for Esophageal Squamous Cell Carcinoma Patients Treated With Esophagectomy? A Prognosis Prediction Model Based on Multicenter Real-World Data. Ann Surg 2023; 277:e61-e69. [PMID: 34091512 DOI: 10.1097/sla.0000000000004958] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To construct a prediction model for more precise evaluation of prognosis which will allow personalized treatment recommendations for adjuvant therapy in patients following resection of ESCC. BACKGROUND Marked heterogeneity of patient prognosis and limited evidence regarding survival benefit of various adjuvant therapy regimens pose challenges in the clinical treatment of ESCC. METHODS Based on comprehensive clinical data obtained from 4129 consecutive patients with resected ESCC in a high-risk region in China, we identified predictors for overall survival through a 2-phase selection based on Cox proportional hazard regression and minimization of Akaike information criterion. The model was internally validated using bootstrapping and externally validated in 1815 patients from a non-high-risk region in China. RESULTS The final model incorporates 9 variables: age, sex, primary site, T stage, N stage, number of lymph nodes harvested, tumor size, adjuvant treatment, and hemoglobin level. A significant interaction was also observed between N stage and adjuvant treatment. N1+ stage patients were likely to benefit from addition of adjuvant therapy as opposed to surgery alone, but adjuvant therapy did not improve overall survival for N0 stage patients. The C -index of the model was 0.729 in the training cohort, 0.723 after bootstrapping, and 0.695 in the external validation cohort. This model outperformed the seventh edition American Joint Committee on Cancer staging system in prognostic prediction and risk stratification. CONCLUSIONS The prediction model constructed in this study may facilitate precise prediction of survival and inform decision-making about adjuvant therapy according to N stage.
Collapse
Affiliation(s)
- Wenlei Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Fangfang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Ruiping Xu
- Anyang Cancer Hospital, Anyang, Henan Province, People's Republic of China
| | - Wei Yang
- Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Yu He
- Department of Non-communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Zhen Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Fuyou Zhou
- Anyang Cancer Hospital, Anyang, Henan Province, People's Republic of China
| | - Fanxiu Heng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of information Technology Service, Peking University Cancer Hospital & institute, Beijing, People's Republic of China
| | - Bolin Hou
- Linkdoc AI Research (LAIR), Beijing, People's Republic of China
| | - Lixin Zhang
- Anyang Cancer Hospital, Anyang, Henan Province, People's Republic of China
| | - Lei Chen
- Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Fan Zhang
- Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Fen Cai
- Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Huawen Xu
- Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Miaoping Lin
- Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Mengfei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Yaqi Pan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Ying Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Zhe Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Huanyu Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Zhonghu He
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Yang Ke
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| |
Collapse
|
34
|
Chen J, Luo AL, Yang L, Wang W, Zhou X, Yang M. Nutrition management by a multidisciplinary team for prevention of nutritional deficits and morbidity following esophagectomy. Braz J Med Biol Res 2023; 56:e12421. [PMID: 37075345 PMCID: PMC10125802 DOI: 10.1590/1414-431x2023e12421] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/15/2023] [Indexed: 04/21/2023] Open
Abstract
This study evaluated the effects of perioperative nutrition management by a multidisciplinary team on nutrition and postoperative complications of patients with esophageal cancer. A total of 239 patients with esophageal cancer who underwent esophagectomy and gastric conduit reconstruction for esophageal or esophagogastric junction cancer between February 2019 and February 2020 were included in the study. They were divided into the experimental group (120 patients) and the control group (119 patients) using the random number table method. Control group patients received routine diet management and experimental group patients received perioperative nutrition management by a multidisciplinary team. The differences of nutriture and postoperative complications between the two groups were compared. At 3 and 7 days after surgery, the experimental group patients had higher total protein and albumin levels (P<0.05), shorter postoperative anal exhaust time (P<0.05), lower incidence of postoperative gastrointestinal adverse reactions, pneumonia, anastomotic fistula, hypoproteinemia (P<0.05), and lower hospitalization costs (P<0.05) than the control group. Nutrition management by a multidisciplinary team effectively improved the nutriture of patients, promoted the rapid recovery of postoperative gastrointestinal function, reduced postoperative complications, and reduced hospitalization costs.
Collapse
Affiliation(s)
- Juan Chen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ai-Lin Luo
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Yang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Wang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xian Zhou
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Mei Yang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
35
|
Wang M, Yue M, Zhao X, He X, Zhang H, Jin J, Wang H. Effect of extracapsular lymph node involvement on the prognosis of patients with esophageal squamous cell carcinoma. Technol Health Care 2023; 31:1771-1786. [PMID: 37125578 DOI: 10.3233/thc-220645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
BACKGROUND According to the eighth edition of the tumor node metastasis (TNM) staging system for esophageal cancer, it is recommended that extracapsular lymph node involvement (EC-LNI) is included as a registered independent variable for the disease. However, its role in the prognosis has not been clearly explained. OBJECTIVE To study the value of EC-LNI in the prognosis of esophageal cancer and attempt to explore its molecular mechanism via an enrichment analysis. METHODS A retrospective analysis was performed on 544 patients with esophageal squamous cell carcinoma (ESCC) who underwent radical surgery in the department of thoracic surgery of our hospital, focusing on the relationship between EC-LNI and clinicopathological characteristics and its effect on prognosis. Additionally, the mechanism of EC-LNI in esophageal cancer was explored. RESULTS Among the 271 patients with lymph node metastasis, 125 were EC-LNI (+). The degrees of tumor differentiation, location, TNM stage, vascular tumor thrombus, and nerve invasion were related to the occurrence of EC-LNI. The stage of TNM was considered an independent risk factor for the development of EC-LNI. A significant difference was found in terms of overall survival (OS) and disease-free survival (DFS) between the EC-LNI (+) and EC-LNI (-) groups. A univariate analysis showed that the degrees of tumor differentiation, T stage, N stage, TNM stage, EC-LNI, EC-LNI number, and EC-LNI distance were significantly correlated with prognosis. A multivariate survival analysis showed that tumor differentiation, TNM stage, and EC-LNI were independent prognostic factors for OS, while TNM stage and EC-LNI were independent prognostic factors for DFS. The enrichment analysis identified the molecular targets and signaling pathways that can regulate cell proliferation, differentiation, and apoptosis. CONCLUSION Extracapsular LNI has a high prognostic value in patients with esophageal cancer and is closely related to the stage of tumors. Our preliminary molecular mechanism research indicated that the molecular targets of EC-LNI are expected to become a new direction for the treatment of esophageal cancer.
Collapse
Affiliation(s)
- Miao Wang
- Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Meng Yue
- Department of Pathology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaopeng Zhao
- Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xu He
- Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haoran Zhang
- Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jing Jin
- Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hongyan Wang
- Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
36
|
Liu W, Zeng C, Wang S, Zhan Y, Huang R, Luo T, Peng G, Wu Y, Qiu Z, Li D, Wu F, Chen C. A combined predicting model for benign esophageal stenosis after simultaneous integrated boost in esophageal squamous cell carcinoma patients (GASTO1072). Front Oncol 2022; 12:1026305. [PMID: 37078004 PMCID: PMC10107369 DOI: 10.3389/fonc.2022.1026305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
PurposeWe aimed to develop a combined predicting model for benign esophageal stenosis (BES) after simultaneous integrated boost (SIB) with concurrent chemotherapy in patients with esophageal squamous cell carcinoma (ESCC).MethodsThis study included 65 patients with EC who underwent SIB with chemotherapy. Esophageal stenosis was evaluated using esophagograms and the severity of eating disorders. Risk factors were investigated using univariate and multivariate analyses. Radiomics features were extracted based on contrast-enhanced CT (CE-CT) before treatment. The least absolute shrinkage and selection operator (LASSO) regression analysis was used for feature selection and radiomics signature construction. The model’s performance was evaluated using Harrell’s concordance index and receiver operating characteristic curves.ResultsThe patients were stratified into low- and high-risk groups according to BES after SIB. The area under the curves of the clinical model, Rad-score, and the combined model were 0.751, 0.820 and 0.864, respectively. In the validation cohort, the AUCs of these three models were 0.854, 0.883 and 0.917, respectively. The Hosmer-Lemeshow test showed that there was no deviation from model fitting for the training cohort (p=0.451) and validation cohort (p=0.481). The C-indexes of the nomogram were 0.864 and 0.958 for the training and validation cohort, respectively. The model combined with Rad-score and clinical factors achieved favorable prediction ability.ConclusionDefinitive chemoradiotherapy could alleviate tumor-inducing esophageal stenosis but result in benign stenosis. We constructed and tested a combined predicting model for benign esophageal stenosis after SIB. The nomogram incorporating both radiomics signature and clinical prognostic factors showed favorable predictive accuracy for BES in ESCC patients who received SIB with chemotherapy.Trial registration number and date of registrationRegistered in www.Clinicaltrial.gov, ID: NCT01670409, August 12, 2012
Collapse
Affiliation(s)
- Weitong Liu
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
- Department of Radiation Oncology, Jieyang People’s Hospital, Jeiyang, China
| | - Chengbing Zeng
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Siyan Wang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yizhou Zhan
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Ruihong Huang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Ting Luo
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
- Department of Radiation Oncology, Shenshan Central Hospital, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Shanwei, China
| | - Guobo Peng
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yanxuan Wu
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Zihan Qiu
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Derui Li
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Fangcai Wu
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
- *Correspondence: Chuangzhen Chen, ; Fangcai Wu,
| | - Chuangzhen Chen
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
- *Correspondence: Chuangzhen Chen, ; Fangcai Wu,
| |
Collapse
|
37
|
Liu Y, Xiong R, Xiao T, Xiong L, Wu J, Li J, Feng G, Song G, Liu K. SCARA5 induced ferroptosis to effect ESCC proliferation and metastasis by combining with Ferritin light chain. BMC Cancer 2022; 22:1304. [PMID: 36513999 PMCID: PMC9746006 DOI: 10.1186/s12885-022-10414-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) remains one of the most lethal cancers worldwide accompany with an extremely poor prognosis. Therefore, this study aims to screen for new molecules affecting ESCC and explore their mechanisms of action to provide ideas for targeted therapies for ESCC. METHODS Firstly, we screened out the membrane protein SCARA5 by high-throughput sequencing of the ESCC patient tissues, and RT-qPCR and WB were used to verify the differential expression of SCARA5 in esophageal cell lines, and IHC analyzed the expression localization of SCARA5 in ESCC tissue. Then, flow cytometry, wound healing assay, Transwell assay and CCK-8 assay were used to explore the effects of SCARA5 on cell cycle, migration and invasion as well as cell proliferation activity of esophageal squamous carcinoma cells. Meanwhile, transmission electron microscopy was used to detect changes in cellular mitochondrial morphology, and flow cytometry were used to detect changes in intracellular reactive oxygen metabolism, and immunofluorescence and flow cytometry were used to detect changes in intracellular Fe2+. Mechanistically, co-immunoprecipitation was used to detect whether SCARA5 binds to ferritin light chain, and ferroptosis-related protein expression was detected by WB. Finally, the tumor xenograft model was applied to validation the role of SCARA5 tumor growth inhibition in vivo. RESULTS We found that SCARA5 was aberrantly decreased in ESCC tissues and cell lines. Furthermore, we confirmed that SCARA5 suppressed the cell cycle, metastasis and invasion of ESCC cells. Meanwhile, we also found that overexpression of SCARA5 caused changes in mitochondrial morphology, accumulation of intracellular reactive oxygen species and increased intracellular Fe2+ in ESCC cells, which induced ferroptosis in ESCC cells. Mechanically, we validated that SCARA5 combined with ferritin light chain and increased intracellular Fe2+. As well as, overexpression SCARA5 induced ferroptosis by increasing ferritin light chain in nude mice subcutaneous tumors and inhibited the growth of nude mice subcutaneous tumors. CONCLUSION Collectively, our findings demonstrated that SCARA5 suppressed the proliferation and metastasis of ESCC by triggering ferroptosis through combining with ferritin light chain.
Collapse
Affiliation(s)
- Yanqun Liu
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, 637000, China
- Department of Cell Biology and Genetics, North Sichuan Medical College, Nanchong, 637100, China
- Department of Laboratory Medicine, Sichuan Chengdu Chengfei Hospital, Chengdu, 610092, China
| | - Rong Xiong
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, 637000, China
| | - Ting Xiao
- Department of Cell Biology and Genetics, North Sichuan Medical College, Nanchong, 637100, China
| | - Li Xiong
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, 637000, China
| | - Jialin Wu
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, 637000, China
- Department of Cell Biology and Genetics, North Sichuan Medical College, Nanchong, 637100, China
| | - Junfeng Li
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, 637000, China
| | - Gang Feng
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, 637000, China
| | - Guiqin Song
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, 637000, China.
- Department of Cell Biology and Genetics, North Sichuan Medical College, Nanchong, 637100, China.
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, 637000, China.
| |
Collapse
|
38
|
Wu Y, Guo Y, Wang Q. USP21 accelerates the proliferation and glycolysis of esophageal cancer cells by regulating the STAT3/FOXO1 pathway. Tissue Cell 2022; 79:101916. [DOI: 10.1016/j.tice.2022.101916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/31/2022] [Accepted: 08/31/2022] [Indexed: 11/28/2022]
|
39
|
Wu P, Zhang Z, Yuan Y, Zhang C, Zhang G, Xue L, Yang H, Wang L, Zheng X, Zhang Y, Yuan Y, Lei R, Yang Z, Zheng B, Xue Q, Sun N, He J. A tumor immune microenvironment-related integrated signature can predict the pathological response and prognosis of esophageal squamous cell carcinoma following neoadjuvant chemoradiotherapy: A multicenter study in China. Int J Surg 2022; 107:106960. [PMID: 36257585 DOI: 10.1016/j.ijsu.2022.106960] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/25/2022] [Accepted: 10/11/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Currently, there are insufficient indicators for the reliable assessment of treatment response following neoadjuvant chemoradiotherapy (nCRT) in patients with esophageal squamous cell carcinoma (ESCC). Considering the essential role of protein-coding and non-coding RNAs in gene regulation and cellular processes, we systematically explored the molecular features and clinical significance of mRNA and lncRNA in 249 pretreatment biopsies from four hospitals in three districts with a high incidence of ESCC patients in China. METHODS During the discovery phrase, 13 differentially expressed genes were identified and validated between samples with a complete pathological response (pCR) and those with an incomplete pathological response (<pCR). Subsequently, we constructed a predictive mRNA and lncRNA signature (SERPINE1, LINC00592, and PRKAG2-AS1) using Fisher's linear discriminant analysis (FLDA) with stepwise variant-selection, followed by validation of its predictive ability in both internal and external cohorts. RESULTS Our signature showed great value in predicting the response to nCRT among ESCC samples and acted as an independent predictive indicator, in addition to demonstrating great potential in estimating patient prognosis. Interestingly, we found that patients with a high signature score had lower PD-L1 and IDO1 expression levels but higher CD8+ T cells infiltration, suggesting that PD-L1 and IDO1 are negatively correlated with a high signature score and further associated with pCR and a better prognosis. CONCLUSION The present study identified a promising three-gene-based predictive signature that has powerful clinical implications for the identification of pCR and a good prognosis among patients with ESCC. Further immune-related exploration may provide an opportunity for future therapeutic combination.
Collapse
Affiliation(s)
- Peng Wu
- 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, 100021, China Department of Pharmacology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China Department of Pathology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China Department of Pathology, Anyang Cancer Hospital, The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang, Henan, 455000, China Department of Otology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, 450008, China Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, 450008, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
He Y, Quaresma M, dos-Santos-Silva I. Stage-Specific Survival From Esophageal Cancer in China and Implications for Control Strategies: A Systematic Review and Meta-Analyses. GASTRO HEP ADVANCES 2022; 2:426-437. [PMID: 39132661 PMCID: PMC11307838 DOI: 10.1016/j.gastha.2022.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/25/2022] [Indexed: 08/13/2024]
Abstract
Background and Aims Esophageal cancer claims more than 500,000 deaths worldwide, with half occurring in China. We aimed to synthesize existing evidence on stage-specific survival from this cancer in China to inform cancer control strategies. Methods English and Chinese literature databases were systematically searched to identify original research published up to May 31, 2019 that reported stage-specific survival from esophageal cancer in China. Two meta-analyses were performed using random-effects models to summarize stage-specific survival differences on relative and absolute scales. The number of esophageal cancer deaths that might have been prevented by early detection in China, in 2018, was estimated assuming 2 different downstaging scenarios. Results One hundred fifty eligible studies were identified, 97 had non-overlapping study populations (83,063 participants), 47 were included in the meta-analysis of hazard ratios, and 26 in the meta-analysis of survival probabilities. Late-stage (III-IV) was associated with 92% higher hazard of death compared with early-stage (0-II) (95% confidence interval 1.62-2.28), corresponding to an absolute 5-year survival difference of 31.2% (29.9%-32.4%). In all, 5.2% esophageal cancer deaths could have been prevented in China, in 2018, if the observed stage distribution at diagnosis (∼50% early-stage) was shifted to the real-life conditions of a population-based endoscopic screening program (∼60% early-stage) and 26.9% if shifted to that observed in the controlled setting of a randomized trial (∼90% early-stage). Conclusion Shifting downwards the stage distribution of esophageal cancer through screening would bring moderate reductions in mortality from the disease. Treatment improvements for early-stage patients are needed to reduce further mortality from this cancer.
Collapse
Affiliation(s)
- Yu He
- Department of Non-Communicable Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Manuela Quaresma
- Department of Non-Communicable Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Isabel dos-Santos-Silva
- Department of Non-Communicable Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
41
|
Zheng Y, Niu X, Wei Q, Li Y, Li L, Zhao J. Familial Esophageal Cancer in Taihang Mountain, China: An Era of Personalized Medicine Based on Family and Population Perspective. Cell Transplant 2022; 31:9636897221129174. [PMID: 36300368 PMCID: PMC9618747 DOI: 10.1177/09636897221129174] [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] [Indexed: 11/15/2022] Open
Abstract
In the Taihang Mountain areas, known as the “esophageal cancer zone” in China, the incidence of esophageal cancer (ESCA) ranks the first in the country and shows a familial and regional clustering trend. Taihang Mountain areas are located in a mountainous area, with inconvenient transportation, limited living conditions, unbalanced diet, and poor nutrition. Ninety percent of the pathological types of ESCA in Taihang Mountain areas are squamous cell carcinoma, among which the risk factors have not been well understood. These areas are usually remote villages and mountains with low population mobility, large family members, similar environmental factors, and a clear and stable genetic background. Therefore, according to the current situation, second-generation sequencing and multigroup analysis technology are used to analyze the familial ESCA patients; disease-related genetic variation are located; and then disease-related susceptibility genes associated with ESCA are screened and analyzed. Health education, tobacco control, endoscopic screening, and other health management projects for suspected and high-risk patients in areas with a high incidence of ESCA can be carried out for screening and early diagnosis, and the incidence of ESCA in Taihang Mountain areas can be reduced. A comprehensive continuous care pattern based on traditional medical nursing to track, monitor, evaluate, and intervene with patients diagnosed with ESCA to facilitate them with medications guidance, dietary guidance, and timely health problem-solving is established. Furthermore, statistical analysis of epidemiology, gene sequencing, and family genetics information can be performed on patients with ESCA in the Taihang Mountains areas to clarify the relationship between genetic phenotype and genotype during the occurrence of ESCA.
Collapse
Affiliation(s)
- Yuanyuan Zheng
- National Engineering Laboratory for Internet Medical Systems and Applications, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyu Niu
- Department of Anesthesiology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Qian Wei
- National Engineering Laboratory for Internet Medical Systems and Applications, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yijing Li
- National Engineering Laboratory for Internet Medical Systems and Applications, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lifeng Li
- National Engineering Laboratory for Internet Medical Systems and Applications, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,Biological Cell Therapy Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jie Zhao
- National Engineering Laboratory for Internet Medical Systems and Applications, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,Department of Pharmacy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,Jie Zhao, National Engineering Laboratory for Internet Medical Systems and Applications, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| |
Collapse
|
42
|
Wang L, Ge S, Liang W, Liao W, Li W, Jiao G, Wei X, Shao G, Xie L, Sheng Z, Hu S, Tang S, Hu P. Genome-Wide Characterization Reveals Variation Potentially Involved in Pathogenicity and Mycotoxins Biosynthesis of Fusarium proliferatum Causing Spikelet Rot Disease in Rice. Toxins (Basel) 2022; 14:toxins14080568. [PMID: 36006230 PMCID: PMC9414198 DOI: 10.3390/toxins14080568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/04/2022] [Accepted: 08/17/2022] [Indexed: 12/12/2022] Open
Abstract
Fusarium proliferatum is the primary cause of spikelet rot disease in rice (Oryza sativa L.) in China. The pathogen not only infects a wide range of cereals, causing severe yield losses but also contaminates grains by producing various mycotoxins that are hazardous to humans and animals. Here, we firstly reported the whole-genome sequence of F. proliferatum strain Fp9 isolated from the rice spikelet. The genome was approximately 43.9 Mb with an average GC content of 48.28%, and it was assembled into 12 scaffolds with an N50 length of 4,402,342 bp. There is a close phylogenetic relationship between F. proliferatum and Fusarium fujikuroi, the causal agent of the bakanae disease of rice. The expansion of genes encoding cell wall-degrading enzymes and major facilitator superfamily (MFS) transporters was observed in F. proliferatum relative to other fungi with different nutritional lifestyles. Species-specific genes responsible for mycotoxins biosynthesis were identified among F. proliferatum and other Fusarium species. The expanded and unique genes were supposed to promote F. proliferatum adaptation and the rapid response to the host's infection. The high-quality genome of F. proliferatum strain Fp9 provides a valuable resource for deciphering the mechanisms of pathogenicity and secondary metabolism, and therefore shed light on development of the disease management strategies and detoxification of mycotoxins contamination for spikelet rot disease in rice.
Collapse
|
43
|
Zhang L, Wan X, Shi R, Gong P, Si Y. Comparing spatial patterns of 11 common cancers in Mainland China. BMC Public Health 2022; 22:1551. [PMID: 35971087 PMCID: PMC9377081 DOI: 10.1186/s12889-022-13926-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: 05/19/2022] [Accepted: 07/31/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND A stronger spatial clustering of cancer burden indicates stronger environmental and human behavioral effects. However, which common cancers in China have stronger spatial clustering and knowledge gaps regarding the environmental and human behavioral effects have yet to be investigated. This study aimed to compare the spatial clustering degree and hotspot patterns of 11 common cancers in mainland China and discuss the potential environmental and behavioral risks underlying the patterns. METHODS Cancer incidence data recorded at 339 registries in 2014 was obtained from the "China Cancer Registry Annual Report 2017". We calculated the spatial clustering degree of the common cancers using the global Moran's Index and identified the hotspot patterns using the hotspot analysis. RESULTS We found that esophagus, stomach and liver cancer have a significantly higher spatial clustering degree ([Formula: see text]) than others. When by sex, female esophagus, male stomach, male esophagus, male liver and female lung cancer had significantly higher spatial clustering degree ([Formula: see text]). The spatial clustering degree of male liver was significantly higher than that of female liver cancer ([Formula: see text]), whereas the spatial clustering degree of female lung was significantly higher than that of male lung cancer ([Formula: see text]). The high-risk areas of esophagus and stomach cancer were mainly in North China, Huai River Basin, Yangtze River Delta and Shaanxi Province. The hotspots for liver and male liver cancer were mainly in Southeast China and south Hunan. Hotspots of female lung cancer were mainly located in the Pearl River Delta, Shandong, North and Northeast China. The Yangtze River Delta and the Pearl River Delta were high-risk areas for multiple cancers. CONCLUSIONS The top highly clustered cancer types in mainland China included esophagus, stomach and liver cancer and, by sex, female esophagus, male stomach, male esophagus, male liver and female lung cancer. Among them, knowledge of their spatial patterns and environmental and behavioral risk factors is generally limited. Potential factors such as unhealthy diets, water pollution and climate factors have been suggested, and further investigation and validation are urgently needed, particularly for male liver cancer. This study identified the knowledge gap in understanding the spatial pattern of cancer burdens in China and offered insights into targeted cancer monitoring and control.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Earth System Science, Ministry of Education Key Laboratory for Earth System Modeling, Institute for Global Change Studies, Tsinghua University, Beijing, 100084, China.
| | - Xia Wan
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Runhe Shi
- Key Laboratory of Geographic Information Science, Ministry of Education, East China Normal University, Shanghai, 200241, China
| | - Peng Gong
- Department of Geography and Department of Earth Sciences, University of Hongkong, Hongkong, 999077, China
| | - Yali Si
- Institute of Environmental Sciences CML, Leiden University, Leiden, 2333 CC, The Netherlands.
| |
Collapse
|
44
|
Feng J, Wang L, Yang X, Chen Q, Cheng X. Pathologic Complete Response Prediction to Neoadjuvant Immunotherapy Combined with Chemotherapy in Resectable Locally Advanced Esophageal Squamous Cell Carcinoma: Real-World Evidence from Integrative Inflammatory and Nutritional Scores. J Inflamm Res 2022; 15:3783-3796. [PMID: 35832830 PMCID: PMC9271687 DOI: 10.2147/jir.s367964] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/21/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Neoadjuvant immunotherapy and chemotherapy (nICT) is an emerging hotspot that has been shown to be safe and feasible for locally advanced esophageal squamous cell carcinoma (LA-ESCC). This real-world study aimed to develop and validate a novel predictive model [integrative inflammatory and nutritional score (IINS)] in LA-ESCC patients receiving nICT to predict the pathologic complete response (pCR). Patients and Methods Patients with LA-ESCC who received nICT followed by surgery from Jun 2019 to Dec 2021 were enrolled and randomly divided into two sets (7:3). Using least absolute shrinkage and selection operator (LASSO) logistic regression analysis, the IINS was constructed in LA-ESCC patients received nICT to predict pCR. A nomogram based on IINS for pCR prediction was generated in the training cohort and verified in the validation cohort. Results Of the 285 enrolled LA-ESCC patients received nICT followed by radical resection, 84 (29.5%) patients achieved pCR. A predictive index of IINS based on 8 inflammatory and nutritional indicators was constructed using the LASSO model. According to the cutoff finder, patients were then stratified into two groups (high and low). The pCR rates were significantly higher in high-IINS group than in low-IINS group in both the training cohort (44.7% vs 17.4%, P < 0.001) and validation cohort (50.0% vs 13.3%, P < 0.001). The IINS [odds ratio (OR) = 0.237, 95% confidence interval (CI) = 0.117–0.480, P < 0.001] was an independent significant predictor for pCR in multivariate logistic analyses. The IINS-based nomogram showed an excellent discrimination for pCR prediction (C-indexes = 0.759 and 0.812 for training and validation cohorts, respectively). Conclusion Pretreatment IINS is an independent predictor for pCR in LA-ESCC patients who are treated with nICT. To our knowledge, the IINS-based nomogram is the first model for pCR prediction and may serve as a simple and potential risk stratification model in LA-ESCC who are treated with nICT.
Collapse
Affiliation(s)
- Jifeng Feng
- Department of Thoracic Oncological Surgery, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China.,Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China.,The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Liang Wang
- Department of Thoracic Oncological Surgery, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| | - Xun Yang
- Department of Thoracic Oncological Surgery, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| | - Qixun Chen
- Department of Thoracic Oncological Surgery, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| | - Xiangdong Cheng
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| |
Collapse
|
45
|
Yang H, Wang XK, Wang JB, Zhao FH, Fan JH, Qiao YL, Taylor PR, Abnet CC. Combined risk factors and risk of upper gastrointestinal cancer mortality in the Linxian general population. Int J Cancer 2022; 151:1462-1473. [PMID: 35689438 DOI: 10.1002/ijc.34160] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 12/17/2022]
Abstract
We aimed to explore the association of combined risk factors with risk of death from upper gastrointestinal (UGI) cancer, including esophageal squamous cell carcinoma (ESCC), gastric cardia carcinoma (GCC) and gastric noncardia carcinoma (GNCC) in the Linxian Nutrition Intervention Trial (NIT) cohort. The NIT cohort included 29 584 healthy adults. A combined risk score (CRS) was calculated using a point system method based on 10 risk factors collected at baseline, including gender, smoking, alcohol drinking, body mass index, family history of UGI cancer, drinking tap water, tooth loss and consumption of fresh fruit, eggs and meat. Possible score ranged from 0 to 31, and higher score indicated as poorer health status. Subjects were divided into three groups by the CRS (<12 points, 12 to 20 points and >20 points). The group of CRS <12 points was considered as the reference. During the 30-year follow-up, we identified 4553 UGI cancer deaths. Compared to subjects with a CRS <12 points, the adjusted HRs for CRS of 12 to 20 points and >20 points were 1.69 (95% CI: 1.56-1.83) and 3.06 (95% CI: 2.82-3.33) for UGI cancer mortality, respectively (Ptrend < .001). Comparable associations were also observed for ESCC, GCC and GNCC mortality. Results remained similar across different age groups (Pinteraction > .05). All HRs observed in the second half follow-up period were stronger than that observed in the first half follow-up period. Our study indicated that higher CRS was associated with increased risk of UGI cancer mortality. Appropriate measures should be taken to reduce unhealthy lifestyles.
Collapse
Affiliation(s)
- Huan Yang
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao-Kun Wang
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Bing Wang
- Department of Epidemiology and Biostatistics, The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Fang-Hui Zhao
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jin-Hu Fan
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - You-Lin Qiao
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Philip R Taylor
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Christian C Abnet
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| |
Collapse
|
46
|
Zhao R, Li X, Yang X, Zhang T, Lu M, Ye W, Jin L, Suo C, Chen X. Association of Esophageal Squamous Cell Carcinoma With the Interaction Between Poor Oral Health and Single Nucleotide Polymorphisms in Regulating Cell Cycles and Angiogenesis: A Case-Control Study in High-Incidence Chinese. Cancer Control 2022. [PMCID: PMC9179002 DOI: 10.1177/10732748221075811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Introduction Oral health and genetic factors can independently influence the risk of developing esophageal squamous cell carcinoma (ESCC). Objectives The primary objective of this study was to investigate the interactive effects of oral health and genetic factors on ESCC risk. Methods This was a matched case-control study with 927 ESCC patients and 1701 matched controls. We selected 101 candidate single nucleotide polymorphisms (SNPs) from 59 genes that were associated with ESCC. Oral health was assessed based on tooth-brushing frequency, tooth loss, and age at the time of first tooth loss. An unconditional logistic regression model was employed in which SNP–oral health interactions were assessed as risk factors for ESCC, after adjusting for age and sex. A genetic risk score (GRS) analysis was conducted. Results The association between GRS and ESCC and the synergistic effect of GRS and oral health on ESCC were examined. Daily frequency of tooth-brushing was found to interact with 5 SNPs, rs3765524, rs753724, rs994771, rs3781264, and rs11187842, to increase the risk of ESCC. In particular, individuals with genotype TT of rs3765524 who brushed their teeth less than twice a day had a 5.13-times higher risk of ESCC than those with genotype CC who brushed their teeth at least twice a day. Furthermore, tooth loss interacted with two SNPs: rs1159918 from ADH1B and rs3813867 from CYP2E1. Conclusion Oral health may interact with genetic factors increasing ESCC risk, which provides new insights into the relationship between ESCC and gene–lifestyle interactions which can be used for disease prevention.
Collapse
Affiliation(s)
- Renjia Zhao
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Xiaoxiao Li
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Institute of Health Sciences, Fudan University Taizhou, Taizhou, China
| | - Xiaorong Yang
- Clinical Epidemiology Unit, Qilu Hospital, Shandong University, Jinan, China
| | - Tiejun Zhang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Ming Lu
- Institute of Health Sciences, Fudan University Taizhou, Taizhou, China
- Clinical Epidemiology Unit, Qilu Hospital, Shandong University, Jinan, China
| | - Weimin Ye
- Clinical Epidemiology Unit, Qilu Hospital, Shandong University, Jinan, China
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Li Jin
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Institute of Health Sciences, Fudan University Taizhou, Taizhou, China
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Chen Suo
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
- Institute of Health Sciences, Fudan University Taizhou, Taizhou, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Institute of Health Sciences, Fudan University Taizhou, Taizhou, China
- Human Phenome Institute, Fudan University, Shanghai, China
| |
Collapse
|
47
|
You H, Zhang J, Xia S, Wu S. Farmland transfer and esophageal cancer incidence rate: mediation of pollution-related agricultural input intensity. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:43826-43844. [PMID: 35119636 DOI: 10.1007/s11356-022-18921-9] [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: 10/05/2021] [Accepted: 01/24/2022] [Indexed: 06/14/2023]
Abstract
Cancer is a growing global health threat. Examining the determinants of cancer incidence can benefit for cancer treatment and prevention. Farmland transfer relates to the risk factors of esophageal cancer including environmental pollution, services access, and habits. This study characterizes the associations between farmland transfer and esophageal cancer incidence rate (ECI) that integrate mediated effect of pollution-related agricultural input intensity in Xiaoshan District, China. The state-space model is employed to quantify the relationships among farmland transfer, pollution-related agricultural input intensity, and ECI. The results showed that (1) Total effects of the proportion of transferred farmland (TFA) area cause a reduction in the ECI. Besides, the total positive effects of the proportion of transferred farmland cultivated non-grain crop (NGC) and proportion of farmland transferred to non-farmer users (NFU) show a downward trend. (2) The raise of TFA can result in the reduction of chemical fertilizer use intensity. Meanwhile, the raise of NGC and NFU can result in the growth of pollution-related agricultural input intensity. But these increasing effects generally show a downward trend. (3) Increasing chemical fertilizer use intensity and pesticide use intensity results in the rise of esophageal cancer incidence rate as a whole. (4) In general, farmland transfer has positive direct effects on esophageal cancer incidence rate. (5) The average proportions of mediated effects in all state-space models are larger than 10%. These findings can raise land reform policy designers' awareness of the risk of public health since the land transfer markets are emerging rapidly in land reform in many developing countries to improve agricultural production.
Collapse
Affiliation(s)
- Heyuan You
- School of Public Administration, Zhejiang University of Finance and Economics, Hangzhou, China.
- Department of City and Regional Planning, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Jinrong Zhang
- School of Public Administration, Zhejiang University of Finance and Economics, Hangzhou, China
| | - Shuyi Xia
- School of Public Administration, Zhejiang University of Finance and Economics, Hangzhou, China
| | - Shenyan Wu
- School of Public Administration, Zhejiang University of Finance and Economics, Hangzhou, China
| |
Collapse
|
48
|
Sun J, Huang W, Chen J, Zhang Y. Association of 3D-CRT and IMRT accelerated hyperfractionated radiotherapy with local control rate and 5-year survival in esophageal squamous cell carcinoma patients. Br J Radiol 2022; 95:20211195. [PMID: 35119916 PMCID: PMC10993959 DOI: 10.1259/bjr.20211195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/13/2022] [Accepted: 01/20/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES This retrospective study examined the relevance and prognostic factors of whole-course conformal radiotherapy (CRT) and late-course accelerated hyperfractionation radiotherapy (LCAFRT) for esophageal squamous cell carcinoma (ESCC). METHODS A total of 110 patients with ESCC received whole-course CRT and LCAFRT between May 2004 and January 2015. All patients received conventional CRT of 2 Gy per day, up to 30-40 Gy, followed by LCAFRT using reduced fields at 1.5 Gy/fraction twice a day, up to 24-39 Gy, for a total dose of 60-69 Gy. RESULTS The median follow-up was 85 months. The whole groups 1-, 3-, and 5-year survival rates were 81.8%, 46.4%, and 41.8%, respectively. The local control rates for the whole group at 1, 3, and 5 years were 82.7%, 70.0%, and 68.2%, respectively. There were no significant differences among survival rates and local control rates between the 3D-CRT and intensity-modulated radiotherapy (IMRT) groups. The main reactions to acute radiotherapy were acute radiation tracheitis, esophagitis, and pneumonia. The tumor location and TNM stage were independent prognostic factors for overall survival. CONCLUSION The results showed that whole-course CRT and LCAFRT for ESCC can improve survival and local control with a tolerable acute reaction compared to previous studies. Local recurrence and distant metastasis are the main failure modes of treatment. ADVANCES IN KNOWLEDGE Whole-course CRT and LCAFRT for ESCC can improve the survival and local control rate compared with previous studies from the 2DRT era. It might provide another treatment for patients with inoperable ESCC or refusing surgery.
Collapse
Affiliation(s)
- Jianyong Sun
- Oncology Department Chaozhou City People’s
Hospital, Guangdong,
China
| | - Weiju Huang
- Oncology Department Chaozhou City People’s
Hospital, Guangdong,
China
| | - Jingbin Chen
- Oncology Department Chaozhou City People’s
Hospital, Guangdong,
China
| | - Yaohong Zhang
- Oncology Department Chaozhou City People’s
Hospital, Guangdong,
China
| |
Collapse
|
49
|
Yu Y, Wu H, Qiu J, Ke D, Wu Y, Lin M, Zheng Q, Zheng H, Wang Z, Li H, Liu L, Li J, Yao Q. The novel pretreatment immune prognostic index discriminates survival outcomes in locally advanced non-operative esophageal squamous cell carcinoma patients treated with definitive chemoradiotherapy: a 6-year retrospective study. Transl Oncol 2022; 21:101430. [PMID: 35452997 PMCID: PMC9046998 DOI: 10.1016/j.tranon.2022.101430] [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: 01/22/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 12/24/2022] Open
Abstract
dNLR and LDH were correlated with OS and PFS in locally advanced ESCC patients received dCRT. The EIPI, a novel inflammatory-based and immune-related prognostic score, was an independent prognostic indicator in locally advanced ESCC. The risk stratification of EIPI can help set individualized treatment strategies and intensities for different subgroups of ESCC patients.
Objective We aimed to construct risk stratification to help set individualized treatment strategies and intensities for different subgroups of patients. Methods The Esophagus Immune Prognostic Index (EIPI) scores were constructed according to the levels of derived neutrophil-to-lymphocyte ratio (dNLR) and lactate dehydrogenase (LDH) before treatment, and the patients were divided into low-, medium-, and high-risk groups. Finally, restricted cubic splines (RCS) were used to explore the relationship between dNLR, LDH, and survival outcomes. Results The median follow-up period of overall survival (OS) and progression-free survival (PFS) were 25.2 and 17.6 months, respectively. Multivariate Cox regression analysis showed dNLR were the independent prognostic factors that were associated with OS and PFS. The 3-year OS and PFS rates in the low-, medium-, and high-risk groups were 44.4% and 38.2%, 26.1% and 23.6%, and 10.5% and 5.3%, respectively. Patients who received chemotherapy had better OS and PFS than those who did not receive chemotherapy in low-risk and medium/high-risk groups (all p < 0.05). Besides, the results also revealed significant differences for patients with clinical T, N, and TNM stage groups of the OS and PFS in different risk groups. Finally, RCS analysis indicated a nonlinear relationship between the dNLR, LDH, and survival for esophageal squamous cell carcinoma (ESCC) patients. The death hazard ratios of dNLR and LDH sharply increased at 1.97 and 191, respectively. Conclusions In summary, the EIPI, a novel inflammatory-based and immune-related prognostic score, is an independent prognostic indicator in locally advanced ESCC patients undergoing definitive chemoradiotherapy (dCRT).
Collapse
Affiliation(s)
- Yilin Yu
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Haishan Wu
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Jianjian Qiu
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Dongmei Ke
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Yahua Wu
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Mingqiang Lin
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Qunhao Zheng
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Hongying Zheng
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Zhiping Wang
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Hui Li
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Lingyun Liu
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Jiancheng Li
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China.
| | - Qiwei Yao
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China.
| |
Collapse
|
50
|
Jiang J, Liu J, Gao P, Liu J. Effect of taking aspirin before diagnosis on the prognosis of esophageal squamous cell carcinoma and analysis of prognostic factors. J Int Med Res 2022; 50:3000605221089799. [PMID: 35400214 PMCID: PMC9006383 DOI: 10.1177/03000605221089799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Objective The 5-year survival rate of patients with esophageal squamous cell cancer (ESCC) is very low. However, long-term aspirin use has been suggested to have an adjuvant therapeutic effect. We therefore investigated the effect of long-term aspirin use before ESCC diagnosis on postoperative patient survival. Methods We carried out a retrospective cohort study of patients who underwent esophageal cancer resection in our hospital from 2008 to 2018. Patients were divided into an aspirin group (n = 79) and control group (n = 79), and were followed up until December 2019. We analyzed the clinicopathological and follow-up data of the patients during hospitalization, and the cyclooxygenase-2 (COX-2) protein expression levels by immunohistochemistry, and related these to postoperative survival. Results Patients who took aspirin had significantly lower survival rates than those who did not. COX-2-negative patients had better survival than patients with either low or high COX-2 expression levels. T stage was the only independent predictor of survival in patients who took aspirin. Conclusions Long-term regular use of aspirin before diagnosis had an adverse effect on postoperative survival in patients with ESCC. Different COX-2 protein expression levels were associated with significantly different postoperative survival rates, with COX-2-positive patients having the poorest survival.
Collapse
Affiliation(s)
- Jiang Jiang
- Hebei Medical University Third Affiliated Hospital, 139 Ziqiang Road, Shijiazhuang 050000, Hebei Province, China
| | - Junfeng Liu
- Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, 12 Jiankang Road, Shijiazhuang 050000, Hebei Province, China
| | - Ping Gao
- Hebei Medical University, 361 East Zhongshan Road 050011, Hebei Province, China
| | - Junying Liu
- Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, 12 Jiankang Road, Shijiazhuang 050000, Hebei Province, China
| |
Collapse
|