1
|
Lei C, Sun W, Wang K, Weng R, Kan X, Li R. Artificial intelligence-assisted diagnosis of early gastric cancer: present practice and future prospects. Ann Med 2025; 57:2461679. [PMID: 39928093 PMCID: PMC11812113 DOI: 10.1080/07853890.2025.2461679] [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: 10/08/2024] [Revised: 12/09/2024] [Accepted: 01/23/2025] [Indexed: 02/11/2025] Open
Abstract
Gastric cancer (GC) occupies the first few places in the world among tumors in terms of incidence and mortality, causing serious harm to human health, and at the same time, its treatment greatly consumes the health care resources of all countries in the world. The diagnosis of GC is usually based on histopathologic examination, and it is very important to be able to detect and identify cancerous lesions at an early stage, but some endoscopists' lack of diagnostic experience and fatigue at work lead to a certain rate of under diagnosis. The rapid and striking development of Artificial intelligence (AI) has helped to enhance the ability to extract abnormal information from endoscopic images to some extent, and more and more researchers are applying AI technology to the diagnosis of GC. This initiative has not only improved the detection rate of early gastric cancer (EGC), but also significantly improved the survival rate of patients after treatment. This article reviews the results of various AI-assisted diagnoses of EGC in recent years, including the identification of EGC, the determination of differentiation type and invasion depth, and the identification of borders. Although AI has a better application prospect in the early diagnosis of ECG, there are still major challenges, and the prospects and limitations of AI application need to be further discussed.
Collapse
Affiliation(s)
- Changda Lei
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Wenqiang Sun
- Suzhou Medical College, Soochow University, Suzhou, China
- Department of Neonatology, Children’s Hospital of Soochow University, Suzhou, China
| | - Kun Wang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Ruixia Weng
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Xiuji Kan
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Rui Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
2
|
Lu Z, Lyu Z, Dong P, Liu Y, Huang L. N6-methyladenosine RNA modification in stomach carcinoma: Novel insights into mechanisms and implications for diagnosis and treatment. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167793. [PMID: 40088577 DOI: 10.1016/j.bbadis.2025.167793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/16/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
N6-methyladenosine (m6A) RNA methylation is crucially involved in the genesis and advancement of gastric cancer (GC) by controlling various pathobiological aspects including gene expression, signal transduction, metabolism, cell death, epithelial-mesenchymal transition, angiogenesis, and exosome function. Despite its importance, the exact mechanisms by which m6A modification influences GC biology remain inadequately explored. This review consolidates the latest advances in uncovering the mechanisms and diverse roles of m6A in GC and proposes new research and translational directions. Key regulators (writers, readers, and erasers) of m6A, such as METTL3/14/16 and WTAP, significantly affect cancer progression, anticancer immune response, and treatment outcomes. m6A modification also impacts immune cell infiltration and the tumor microenvironment, highlighting its potential as a diagnostic and prognostic marker. Interactions between m6A methylation and non-coding RNAs offer further novel insights into GC development and therapeutic targets. Targeting m6A regulators could enhance immunotherapy response, overcome treatment resistance, and improve oncological and clinical outcomes. Models based on m6A can precisely predict treatment response and prognosis in GC. Additional investigation is needed to fully understand the mechanisms of m6A methylation and its potential clinical applications and relevance (e.g., as precise markers for early detection, prediction of outcome, and response to therapy and as therapeutic targets) in GC. Future research should focus on in vivo studies, potential clinical trials, and the examination of m6A modification in other types of cancers.
Collapse
Affiliation(s)
- Zhengmao Lu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Zhaojie Lyu
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Peixin Dong
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan.
| | - Yunmei Liu
- School of Cultural Heritage and Information Management, Shanghai University, Shanghai, China.
| | - Lei Huang
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, Shanghai Institute of Pancreatic Diseases, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, Shanghai 200433, China; National Key Laboratory of Immunity and Inflammation, Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
3
|
Lv M, Wang Y, Yuan Z, Zhai L, Iqbal H, Ur-Rehman U, Ning X, Wei H, Xin J, Jin Z, Yi Z, Wang B, Chen W, Xiao R. Decitabine promotes the differentiation of poorly differentiated gastric cancer cells and enhances the sensitivity of NK cell cytotoxicity via TNF-α. Sci Rep 2025; 15:13119. [PMID: 40240368 PMCID: PMC12003911 DOI: 10.1038/s41598-025-95741-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Poorly differentiated gastric cancer (PDGC) is characterized by high invasiveness, rapid progression, and poor prognosis for patients. Differentiation therapy has long been a promising approach by manipulating the differentiation state of tumor cells to inhibit tumor growth, offering fewer side effects. Decitabine (DAC), is known as an inhibitor of DNA methylation, thus reactivating the transcription of previously methylated silenced genes associated with differentiation to induce a more differentiated state. This study used the differentiation-inducing agents DAC to treat two PDGC cell lines, MKN45 and NUGC4, and explored the impact of DAC on cell proliferation and influence of their sensitivity to Natural Killer cells (NK cells) mediated cytotoxicity. The results demonstrated a significant reduction in cell proliferation, migration, and invasion without affecting cell viability after DAC treatment. Additionally, transcriptomic analysis revealed that DAC-treated PDGC cells upregulated multiple immune-related genes, including the gene encoding for tumor necrosis factor alpha (TNF-α). Co-culture study of NK cells and PDGC cells showed that DAC treatment enhanced the sensitivity of these cancer cells to NK cell-mediated cytotoxicity, and TNF-α played a crucial role in promoting NK cell cytotoxicity. Following the subcutaneous implantation of tumors in nude mice, DAC administration significantly inhibited the growth of PDGC tumors and induced the upregulation of differentiation related genes. In summary, DAC effectively reduces the malignant characteristics of the PDGC cells by promoting their transition towards a higher state of differentiation and enhancing their sensitivity to NK cell-mediated killing, providing new insights for the mechanisms of the antitumor effects of DAC.
Collapse
Affiliation(s)
- Man Lv
- School of Life Science, Tianjin University, Tianjin, 300072, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Yue Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Medical College of Tianjin University, Tianjin University, Tianjin, 300072, China
| | - Ziyin Yuan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Lina Zhai
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Haroon Iqbal
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Uzair Ur-Rehman
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Xin Ning
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Huiying Wei
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences (UCAS), Hangzhou, 310024, China
| | - Jun Xin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Medical College of Tianjin University, Tianjin University, Tianjin, 300072, China
| | - Zihui Jin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Zhou Yi
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Baichuan Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Wangkai Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Run Xiao
- School of Life Science, Tianjin University, Tianjin, 300072, China.
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
4
|
Fan Y, Ma M, Liu Q, Wu Y. The impact of refined nursing management on the diagnosis of early gastric cancer under ME-NBI. BMC Gastroenterol 2025; 25:225. [PMID: 40188024 PMCID: PMC11972532 DOI: 10.1186/s12876-025-03792-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 03/17/2025] [Indexed: 04/07/2025] Open
Abstract
OBJECTIVE To explore the impact of magnifying endoscopy with narrow-band imaging (ME-NBI) combined with refined nursing management on the endoscopic diagnosis of early gastric cancer. METHODS Patients who underwent painless gastroscopy at the Affiliated Hospital of Zunyi Medical University from January 1, 2021 to December 31, 2021 were randomly selected as study subjects. They were randomly divided into an experimental group and a control group. The experimental group received ME-NBI examination and refined nursing interventions included psychological support, environmental management, and structured patient preparation to optimize endoscopic conditions. The control group received routine endoscopic examination and nursing. The gastric cancer detection rates, patient compliance, and mucosal visibility were evaluated. The patient compliance scale used in this study evaluates adherence based on medication intake, positional changes, and examination cooperation. RESULTS A total of 998 patients were included, with 499 in each group. The gastric cancer detection rate was significantly higher in the experimental group (4.2%) compared to the control group (0.6%) (χ2 = 13.721, p < 0.0001). Patients were randomly assigned to an experimental group (n = 499) receiving ME-NBI with refined nursing, and a control group (n = 499) receiving routine care. There were no statistically significant differences in general data such as gender, age, family history of gastric cancer, and Helicobacter pylori infection between the two groups (all p > 0.05), indicating comparability. In the experimental group, 334 cases (66.93%) had good compliance scores (9-10 points) and 165 cases (33.07%) had general compliance scores (6-8 points), while in the control group, 31 cases (6.21%) had good compliance scores and 468 cases (93.79%) had general compliance scores. Patient compliance was significantly higher in the experimental group compared to the control group (χ2 = 396.569, p < 0.0001), indicating that refined nursing can improve patient compliance. In addition, the comparison of gastric mucosal visibility scores during endoscopic examination showed that in the experimental group, 384 cases (76.95%) scored 1 point, 115 cases (23.05%) scored 2 points, and 0 cases (0.00%) scored 3 points; while in the control group, 27 cases (5.41%) scored 1 point, 228 cases (45.69%) scored 2 points, and 244 cases (48.90%) scored 3 points. The mucosal visibility was significantly higher in the experimental group compared to the control group (χ2 = 591.322, p < 0.0001), indicating that refined nursing can improve gastric mucosal visibility. The gastric cancer detection rate was significantly higher in the experimental group (4.2%) compared to the control group (0.6%) (χ2 = 13.721, p < 0.0001), indicating that refined care can improve the gastric cancer detection rate. CONCLUSION The application of refined nursing management combined with ME-NBI technology for the diagnosis of early gastric cancer can significantly improve patient compliance, gastric mucosal visibility, and gastric cancer detection rate, which is worthy of clinical promotion and application.
Collapse
Affiliation(s)
- Yi Fan
- Nursing Department, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China.
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China.
| | - Ming Ma
- Nursing Department, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Qing Liu
- Nursing Department, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Yu Wu
- Nursing Department, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| |
Collapse
|
5
|
Huang X, Qin M, Fang M, Wang Z, Hu C, Zhao T, Qin Z, Zhu H, Wu L, Yu G, De Cobelli F, Xie X, Palumbo D, Tian J, Dong D. The application of artificial intelligence in upper gastrointestinal cancers. JOURNAL OF THE NATIONAL CANCER CENTER 2025; 5:113-131. [PMID: 40265096 PMCID: PMC12010392 DOI: 10.1016/j.jncc.2024.12.006] [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: 06/13/2024] [Revised: 09/17/2024] [Accepted: 12/20/2024] [Indexed: 04/24/2025] Open
Abstract
Upper gastrointestinal cancers, mainly comprising esophageal and gastric cancers, are among the most prevalent cancers worldwide. There are many new cases of upper gastrointestinal cancers annually, and the survival rate tends to be low. Therefore, timely screening, precise diagnosis, appropriate treatment strategies, and effective prognosis are crucial for patients with upper gastrointestinal cancers. In recent years, an increasing number of studies suggest that artificial intelligence (AI) technology can effectively address clinical tasks related to upper gastrointestinal cancers. These studies mainly focus on four aspects: screening, diagnosis, treatment, and prognosis. In this review, we focus on the application of AI technology in clinical tasks related to upper gastrointestinal cancers. Firstly, the basic application pipelines of radiomics and deep learning in medical image analysis were introduced. Furthermore, we separately reviewed the application of AI technology in the aforementioned aspects for both esophageal and gastric cancers. Finally, the current limitations and challenges faced in the field of upper gastrointestinal cancers were summarized, and explorations were conducted on the selection of AI algorithms in various scenarios, the popularization of early screening, the clinical applications of AI, and large multimodal models.
Collapse
Affiliation(s)
- Xiaoying Huang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Minghao Qin
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- University of Science and Technology Beijing, Beijing, China
| | - Mengjie Fang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Zipei Wang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Chaoen Hu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Tongyu Zhao
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- University of Science and Technology of China, Hefei, China
| | - Zhuyuan Qin
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | | | - Ling Wu
- KiangWu Hospital, Macau, China
| | | | | | | | - Diego Palumbo
- Department of Radiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Di Dong
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
6
|
Li N, Lin J, Wang C, Ran X, Zhao Z. Artificial intelligence-driven integration of single-cell RNA sequencing and transcriptome analysis to decipher APOE's role in gastric cancer prognosis and therapy. Discov Oncol 2025; 16:314. [PMID: 40082390 PMCID: PMC11906938 DOI: 10.1007/s12672-025-02100-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/10/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) remains a global health challenge due to its high mortality rate and the lack of specific diagnostic methods. Tumor heterogeneity significantly hinders effective treatment, necessitating advanced techniques to dissect its complexity. Artificial intelligence (AI) offers transformative potential in uncovering intricate cellular dynamics and gene regulatory networks. This study leverages single-cell RNA sequencing (scRNA-seq) combined with transcriptome analysis, enhanced by AI-driven analytics, to explore the tumor microenvironment and identify novel prognostic markers and therapeutic targets in GC. METHODS scRNA-seq and transcriptome datasets of GC patients were analyzed using AI-enhanced methodologies to unravel tumor heterogeneity and microenvironmental dynamics. Macrophage subsets were identified as critical components within the GC microenvironment. High-variance gene screening in these subsets pinpointed apolipoprotein E (APOE) as a hub gene. Experimental validation of APOE expression in GC samples and functional studies in GC cell lines were conducted. RESULTS Bioinformatics and AI-enabled analyses confirmed the immunosuppressive role of APOE in GC. An immune-related survival model was developed to predict immunotherapy responses and patient prognoses. Mechanistically, APOE was found to induce immunosuppression through M2 macrophage polarization, promoting tumor progression and leading to poorer outcomes in GC patients. CONCLUSION This study highlights the potential of AI-driven approaches in elucidating the role of APOE in GC progression. APOE's regulatory effects on M2 macrophages underscore its value as a prognostic marker and therapeutic target, paving the way for precision medicine in GC management.
Collapse
Affiliation(s)
- Ni Li
- Health Management Center, People's Hospital of Guilin, Guilin, 541002, Guangxi, China
| | - Jinquan Lin
- Department of Gastrointestinal and Hernia Surgery, People's Hospital of Guilin, Guilin, 541002, Guangxi, China
- Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Chunyu Wang
- Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Xiao Ran
- Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Zhi Zhao
- Department of Gastrointestinal and Hernia Surgery, People's Hospital of Guilin, Guilin, 541002, Guangxi, China.
| |
Collapse
|
7
|
Iwai N, Dohi O, Kotachi T, Tsuda M, Yagi N, Ono S, Seya M, Teshima H, Kubo K, Nakahata Y, Obora A, Oka S, Tanaka S, Itoh Y, Kato M. Linked Color Imaging with Light-Emitting Diode Light Enhances the Visibility of Gastric Neoplasm: A Prospective, Multicenter, Comparative Trial. Dig Dis Sci 2025; 70:794-801. [PMID: 39671065 DOI: 10.1007/s10620-024-08785-z] [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: 10/16/2024] [Accepted: 12/01/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Early detection of gastric cancer can play a key role in improving prognosis. Recently, light-emitting diodes (LED) have been developed as novel endoscopic systems. However, the differences in the visibility of gastric neoplastic lesions between LED and laser endoscopy remains unclear. We conducted a prospective multicenter trial to evaluate the non-inferiority of LED endoscopy in the visibility of gastric neoplastic lesions undergoing endoscopic submucosal dissection (ESD) in comparison to laser endoscopy. METHODS A multicenter, prospective, cross-sectional study was conducted in patients undergoing ESD for gastric neoplastic lesions at five hospitals throughout Japan. Seventy patients with 74 lesions were included in this study. The primary endpoint was the non-inferiority of the difference in the individual scores of linked color imaging (LCI) and white-light imaging (WLI) for LED and laser endoscopy for gastric neoplastic lesions. RESULTS The mean individual score was 2.66 ± 1.02, 3.17 ± 0.83, 2.75 ± 1.05, and 3.21 ± 0.84 in LED-WLI, LED-LCI, laser-WLI, and laser-LCI, respectively. The difference in individual scores of LCI and WLI was 0.51 ± 0.77 and 0.46 ± 0.80 in LED and laser endoscopy, respectively. The mean difference between LED and laser endoscopy was 0.04 (95% confidence interval [CI]: - 0.05 to 0.13, P < 0.001). CONCLUSIONS This study revealed the non-inferiority of the differences in individual scores between LCI and WLI in the comparison of LED and laser endoscopy for gastric neoplastic lesions.
Collapse
Affiliation(s)
- Naoto Iwai
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Osamu Dohi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Takahiro Kotachi
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima, Japan
| | - Momoko Tsuda
- Department of Gastroenterology, National Hospital Organization Hakodate National Hospital, Hakodate, Japan
- Department of Gastroenterology, Sapporo Cancer Screening Center, Public Interest Foundation Hokkaido Cancer Society, Sapporo, Japan
| | - Nobuaki Yagi
- Department of Gastroenterology, Asahi University Hospital, Gifu, Japan
| | - Shoko Ono
- Department of Gastroenterology and Hepatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Mayuko Seya
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hajime Teshima
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima, Japan
| | - Kimitoshi Kubo
- Department of Gastroenterology, National Hospital Organization Hakodate National Hospital, Hakodate, Japan
| | - Yuki Nakahata
- Department of Gastroenterology, Asahi University Hospital, Gifu, Japan
| | - Akihiro Obora
- Department of Gastroenterology, Asahi University Hospital, Gifu, Japan
| | - Shiro Oka
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima, Japan
| | | | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Mototsugu Kato
- Department of Gastroenterology, National Hospital Organization Hakodate National Hospital, Hakodate, Japan
| |
Collapse
|
8
|
Shi X, Li R, Shi X, Yan Y, Gong A. The impact of the age-adjusted Charlson comorbidity index as a prognostic factor in patients with early gastric cancer after endoscopic submucosal dissection. Scand J Gastroenterol 2025; 60:136-142. [PMID: 39773271 DOI: 10.1080/00365521.2024.2449072] [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: 10/26/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND The Charlson Comorbidity Index (CCI) and prognostic nutritional index (PNI) have proven to be valuable tools in predicting prognosis based on comorbidities and nutritional status in the context of surgical procedures and endoscopic resections. The age-Adjusted CCI (ACCI) has also shown utility in surgical settings, but its application to early gastric cancer (EGC) remains unexplored. Consequently, we aimed at clarifying the prognostic factors for EGC treated with endoscopic submucosal dissection (ESD). METHODS Patients who underwent ESD for EGC at the First Affiliated Hospital of Dalian Medical University from January 2015 to February 2023 were included. The overall survival (OS) and prognostic predictive ability were evaluated based on patients and lesion characteristics. RESULTS During a median follow-up period of 50 months, 15 patients died, but none from the gastric cancer. The 5-year survival rate was 90.0%. In univariate and multivariate analyses, a high ACCI (>4.5) was the only significant prognostic factor (Hazard ratio, 27.78; 95% confidence interval, 3.62-213.40; p < 0.01). The 5-year survival rates for patients with low ACCI (<4.5) and high ACCI were 98.9% and 72.9%, respectively (p < 0.01). CONCLUSIONS A high ACCI is a significant prognostic indicator for 5-year survival and the risk of mortality caused by other comorbidities. EGC suitable for ESD is unlikely to serve as a prognostic factor, and ACCI should be considered as an important reference when considering additional surgical procedures in high-ACCI patients after ESD with endoscopic curability (eCura) C-2 for EGC.
Collapse
Affiliation(s)
- Xiao Shi
- Department of Gastroenterology, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Ruibo Li
- Department of Gastroenterology, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiaoyi Shi
- Department of Gastroenterology, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yuxing Yan
- Department of Gastroenterology, the Second People's Hospital of Liaocheng, Liaocheng, China
| | - Aixia Gong
- Department of Gastroenterology, First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
9
|
Ono Y, Hatta W, Tarasawa K, Ogata Y, Abe H, Sato I, Hatayama Y, Saito M, Jin X, Uno K, Koike T, Imatani A, Hamada S, Fujimori K, Fushimi K, Masamune A. Optimal direct oral anticoagulant for upper gastrointestinal endoscopic submucosal dissection. J Gastroenterol 2025; 60:66-76. [PMID: 39601804 PMCID: PMC11717785 DOI: 10.1007/s00535-024-02171-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/03/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND The patients taking direct oral anticoagulants (DOACs) are at high risk for developing ischemic stroke and delayed bleeding in upper gastrointestinal endoscopic submucosal dissection (ESD). We aimed to identify the optimal DOAC based on both adverse events in upper gastrointestinal ESD. METHODS A retrospective population-based cohort study was conducted using the Diagnosis Procedure Combination database in Japan. We included patients on a DOAC undergoing upper gastrointestinal ESD between 2012 and 2021. The primary outcomes were ischemic stroke occurring after upper gastrointestinal ESD and delayed bleeding in gastroduodenal and esophageal ESD. Inverse probability weightings were applied to balance the four DOAC groups (dabigatran, rivaroxaban, apixaban, and edoxaban), and logistic regression analyses were performed to compare the outcomes. RESULTS We analyzed 9729 patients on a DOAC undergoing upper gastrointestinal ESD. Ischemic stroke developed after upper gastrointestinal ESD in 1.4%, 0.7%, 0.6%, and 0.8% of patients taking dabigatran, rivaroxaban, apixaban, and edoxaban, respectively, after weighting. Rivaroxaban and apixaban showed significantly lower risk of ischemic stroke compared with dabigatran (odds ratio, 0.15 and 0.12, respectively) in standard doses. The delayed bleeding developed after gastroduodenal ESD in 7.6%, 14.6%, 19.2%, and 17.3% of patients taking each DOAC, respectively, with the lowest risk in dabigatran, followed by rivaroxaban. A similar pattern was observed in delayed bleeding in esophageal ESD (3.2%, 5.4%, 7.5%, and 5.5% in each DOAC), but with no significant results. CONCLUSIONS Rivaroxaban might be an optimal DOAC for upper gastrointestinal ESD showing a lower risk for both ischemic stroke and delayed bleeding.
Collapse
Affiliation(s)
- Yoshitaka Ono
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Waku Hatta
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan.
| | - Kunio Tarasawa
- Department of Health Administration and Policy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan
| | - Yohei Ogata
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Hiroko Abe
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Isao Sato
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Yutaka Hatayama
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Masahiro Saito
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Xiaoyi Jin
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Kaname Uno
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Tomoyuki Koike
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Akira Imatani
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Shin Hamada
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Kenji Fujimori
- Department of Health Administration and Policy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan
| | - Kiyohide Fushimi
- Department of Health Policy and Informatics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, S1560/S1568 M&D Tower 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| |
Collapse
|
10
|
Ma X, Lu T, Yang Y, Qin D, Tang Z, Cui Y, Wang R. DEAD-box helicase family proteins: emerging targets in digestive system cancers and advances in targeted drug development. J Transl Med 2024; 22:1120. [PMID: 39707322 DOI: 10.1186/s12967-024-05930-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/30/2024] [Indexed: 12/23/2024] Open
Abstract
Cancer has become one of the major diseases threatening human health in the twenty-first century due to its incurability. In 2022, new cases of esophageal and gastrointestinal cancers accounted for 17.1% of all newly diagnosed cancer cases worldwide. Despite significant improvements in early cancer screening, clinical diagnostics, and treatments in recent years, the overall prognosis of digestive system cancer patients remains poor. The DEAD-box helicase family, a crucial member of the RNA helicase family, participates in almost every aspect of RNA metabolism, including transcription, splicing, translation, and degradation, and plays a key role in the occurrence and progression of various cancers. This article aims to summarize and discuss the role and potential clinical applications of DEAD-box helicase family proteins in digestive system cancers. The discussion includes the latest progress in the occurrence, development, and treatment of esophageal and gastrointestinal tumors; the main functions of DEAD-box helicase family proteins; their roles in digestive system cancers, including their relationships with clinical factors; effects on cancer proliferation, migration, and invasion; and involved signaling pathways; as well as the existing inhibitory strategies targeting DDX family proteins, are discussed. Additionally, outlooks on future research directions are provided.
Collapse
Affiliation(s)
- Xiaochao Ma
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| | - Tianyu Lu
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| | - Yue Yang
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| | - Da Qin
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| | - Ze Tang
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| | - Youbin Cui
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China.
| | - Rui Wang
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| |
Collapse
|
11
|
Delgado-Guillena P, Jimeno M, López-Nuñez A, Córdova H, Fernández-Esparrach G. The endoscopic model for gastric carcinogenesis and Helicobacter pylori infection: A potential visual mind-map during gastroscopy examination. GASTROENTEROLOGIA Y HEPATOLOGIA 2024; 47:502214. [PMID: 38844201 DOI: 10.1016/j.gastrohep.2024.502214] [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: 03/23/2024] [Revised: 05/04/2024] [Accepted: 05/24/2024] [Indexed: 06/29/2024]
Abstract
Helicobacter pylori (Hp) is the main trigger of chronic gastric atrophy and the main leading cause of gastric cancer. Hp infects the normal gastric mucosa and can lead to chronic inflammation, glandular atrophy, intestinal metaplasia, dysplasia and finally adenocarcinoma. Chronic inflammation and gastric atrophy associated with Hp infection appear initially in the distal part of the stomach (the antrum) before progressing to the proximal part (the corpus-fundus). In recent years, endoscopic developments have allowed for the characterization of various gastric conditions including the normal mucosa (pyloric/fundic gland pattern and regular arrangement of collecting venules), Hp-related gastritis (Kyoto classification), glandular atrophy (Kimura-Takemoto classification), intestinal metaplasia (Endoscopic Grading of Gastric Intestinal Metaplasia), and dysplasia/adenocarcinoma (Vessel plus Surface classification). Despite being independent classifications, all these scales can be integrated into a single model: the endoscopic model for gastric carcinogenesis. This model would assist endoscopists in comprehending the process of gastric carcinogenesis and conducting a systematic examination during gastroscopy. Having this model in mind would enable endoscopists to promptly recognize the implications of Hp infection and the potential patient's risk of developing gastric cancer.
Collapse
Affiliation(s)
| | - Mireya Jimeno
- Department of Pathology, Hospital of Germans Trias i Pujol, Badalona, Spain
| | | | - Henry Córdova
- Department of Gastroenterology, Hospital Clinic of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
| | - Gloria Fernández-Esparrach
- Department of Gastroenterology, Hospital Clinic of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
| |
Collapse
|
12
|
Wang B, Wang SW, Zhou Y, Wang SP, Gao Y, Liu HM, Ji SK, Wang SQ, Zheng YC, Zhang C, Mardinoglu A, Liu HM, Chen XB, Dai XJ. Discovery of 2-Aryl-4-aminoquinazolin-Based LSD1 Inhibitors to Activate Immune Response in Gastric Cancer. J Med Chem 2024; 67:16165-16184. [PMID: 39264726 DOI: 10.1021/acs.jmedchem.4c00972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2024]
Abstract
LSD1 (histone lysine-specific demethylase 1) has been gradually disclosed to act as an immunomodulator to enhance antitumor immune response. Despite the identification of numerous potent LSD1 inhibitors, there remains a lack of LSD1 inhibitors approved for marketing. Novel LSD1 inhibitors with different mechanisms are therefore needed. Herein, we reported a series of novel quinazoline-based LSD1 inhibitors. Among them, compound Z-1 exhibited the best LSD1 inhibitory activity (IC50 = 0.108 μM). Z-1 also acted as a selective and cellular active as an LSD1 inhibitor. Furthermore, Z-1 promoted response of gastric cancer cells to T-cell killing effect by decreasing PD-L1 expression and further attenuated the PD-1/PD-L1 interaction. In vivo, Z-1 exhibited significant suppression effect on the growth of gastric cancer cells without obvious toxicity. Therefore, Z-1 represents a potential novel immunomodulator that targets LSD1, providing a lead compound with new function mechanism for gastric cancer treatment.
Collapse
Affiliation(s)
- Bo Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shu-Wu Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ying Zhou
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shao-Peng Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ya Gao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Hui-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shi-Kun Ji
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Sai-Qi Wang
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer & Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou 450008, China
| | - Yi-Chao Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Cheng Zhang
- Science for Life Laboratory, KTH-Royal Institute of Technology, SE-17121 Stockholm, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH-Royal Institute of Technology, SE-17121 Stockholm, Sweden
- Faculty of Dentistry, Oral & Craniofacial Sciences, Centre for Host-Microbiome Interactions, King's College London, London WC2R 2LS, U.K
| | - Hong-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiao-Bing Chen
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer & Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou 450008, China
| | - Xing-Jie Dai
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
13
|
Liao Z, Xie Z. Construction of a disulfidptosis-related glycolysis gene risk model to predict the prognosis and immune infiltration analysis of gastric adenocarcinoma. Clin Transl Oncol 2024; 26:2309-2322. [PMID: 38587603 DOI: 10.1007/s12094-024-03457-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/11/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND The pattern of cell death known as disulfidptosis was recently discovered. Disulfidptosis, which may affect the growth of tumor cells, represents a potential new approach to treating tumors. Glycolysis affects tumor proliferation, invasion, chemotherapy resistance, the tumor microenvironment (TME), and immune evasion. However, the efficacy and therapeutic significance of disulfidptosis-related glycolysis genes (DRGGs) in stomach adenocarcinoma (STAD) remain uncertain. METHODS STAD clinical data and RNA sequencing data were downloaded from the TCGA database. DRGGs were screened using Cox regression and Lasso regression analysis to construct a prognostic risk model. The accuracy of the model was verified using survival studies, receiver operating characteristic (ROC) curves, column plots, and calibration curves. Additionally, our study investigated the relationships between the risk scores and immune cell infiltration, tumor mutational burden (TMB), and anticancer drug sensitivity. RESULTS We have successfully developed a prognosis risk model with 4 DRGGs (NT5E, ALG1, ANKZF1, and VCAN). The model showed excellent performance in predicting the overall survival of STAD patients. The DRGGs prognostic model significantly correlated with the TME, immune infiltrating cells, and treatment sensitivity. CONCLUSIONS The risk model developed in this work has significant clinical value in predicting the impact of immunotherapy in STAD patients and assisting in the choice of chemotherapeutic medicines. It can correctly estimate the prognosis of STAD patients.
Collapse
Affiliation(s)
- Zhaohui Liao
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, China
| | - Zhengyuan Xie
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, China.
| |
Collapse
|
14
|
Ji XW, Lin J, Wang YT, Ruan JJ, Xu JH, Song K, Mao JS. Endoscopic detection and diagnostic strategies for minute gastric cancer: A real-world observational study. World J Gastrointest Oncol 2024; 16:3529-3538. [PMID: 39171159 PMCID: PMC11334033 DOI: 10.4251/wjgo.v16.i8.3529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/26/2024] [Accepted: 06/18/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Minute gastric cancers (MGCs) have a favorable prognosis, but they are too small to be detected by endoscopy, with a maximum diameter ≤ 5 mm. AIM To explore endoscopic detection and diagnostic strategies for MGCs. METHODS This was a real-world observational study. The endoscopic and clinicopathological parameters of 191 MGCs between January 2015 and December 2022 were retrospectively analyzed. Endoscopic discoverable opportunity and typical neoplastic features were emphatically reviewed. RESULTS All MGCs in our study were of a single pathological type, 97.38% (186/191) of which were differentiated-type tumors. White light endoscopy (WLE) detected 84.29% (161/191) of MGCs, and the most common morphology of MGCs found by WLE was protruding. Narrow-band imaging (NBI) secondary observation detected 14.14% (27/191) of MGCs, and the most common morphology of MGCs found by NBI was flat. Another three MGCs were detected by indigo carmine third observation. If a well-demarcated border lesion exhibited a typical neoplastic color, such as yellowish-red or whitish under WLE and brownish under NBI, MGCs should be diagnosed. The proportion with high diagnostic confidence by magnifying endoscopy with NBI (ME-NBI) was significantly higher than the proportion with low diagnostic confidence and the only visible groups (94.19% > 56.92% > 32.50%, P < 0.001). CONCLUSION WLE combined with NBI and indigo carmine are helpful for detection of MGCs. A clear demarcation line combined with a typical neoplastic color using nonmagnifying observation is sufficient for diagnosis of MGCs. ME-NBI improves the endoscopic diagnostic confidence of MGCs.
Collapse
Affiliation(s)
- Xiao-Wei Ji
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Jie Lin
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Yan-Ting Wang
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Jing-Jing Ruan
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Jing-Hong Xu
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Kai Song
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Jian-Shan Mao
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| |
Collapse
|
15
|
Sung YN, Lee H, Kim E, Jung WY, Sohn JH, Lee YJ, Keum B, Ahn S, Lee SH. Interpretable deep learning model to predict lymph node metastasis in early gastric cancer using whole slide images. Am J Cancer Res 2024; 14:3513-3522. [PMID: 39113867 PMCID: PMC11301296 DOI: 10.62347/rjbh6076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/24/2024] [Indexed: 08/10/2024] Open
Abstract
In early gastric cancer (EGC), the presence of lymph node metastasis (LNM) is a crucial factor for determining the treatment options. Endoscopic resection is used for treatment of EGC with minimal risk of LNM. However, owing to the lack of definitive criteria for identifying patients who require additional surgery, some patients undergo unnecessary additional surgery. Considering that histopathologic patterns are significant factor for predicting lymph node metastasis in gastric cancer, we aimed to develop a machine learning algorithm which can predict LNM status using hematoxylin and eosin (H&E)-stained images. The images were obtained from several institutions. Our pipeline comprised two sequential approaches including a feature extractor and a risk classifier. For the feature extractor, a segmentation network (DeepLabV3+) was trained on 243 WSIs across three datasets to differentiate each histological subtype. The risk classifier was trained with XGBoost using 70 morphological features inferred from the trained feature extractor. The trained segmentation network, the feature extractor, achieved high performance, with pixel accuracies of 0.9348 and 0.8939 for the internal and external datasets in patch level, respectively. The risk classifier achieved an overall AUC of 0.75 in predicting LNM status. Remarkably, one of the datasets also showed a promising result with an AUC of 0.92. This is the first multi-institution study to develop machine learning algorithm for predicting LNM status in patients with EGC using H&E-stained histopathology images. Our findings have the potential to improve the selection of patients who require surgery among those with EGC showing high-risk histological features.
Collapse
Affiliation(s)
- You-Na Sung
- Department of Pathology, Korea University Anam Hospital, College of Medicine, Korea UniversitySeoul, South Korea
| | - Hyeseong Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic UniversitySeoul, South Korea
| | - Eunsu Kim
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic UniversitySeoul, South Korea
| | - Woon Yong Jung
- Department of Pathology, Hanyang University Guri Hospital, College of Medicine, Hanyang UniversityGuri, South Korea
| | - Jin-Hee Sohn
- Department of Pathology, Samkwang Medical LaboratoriesSeoul, South Korea
| | - Yoo Jin Lee
- Department of Pathology, Korea University Anam Hospital, College of Medicine, Korea UniversitySeoul, South Korea
| | - Bora Keum
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University Anam Hospital, College of Medicine, Korea UniversitySeoul, South Korea
| | - Sangjeong Ahn
- Department of Pathology, Korea University Anam Hospital, College of Medicine, Korea UniversitySeoul, South Korea
- Artificial Intelligence Center, Korea University Anam Hospital, College of Medicine, Korea UniversitySeoul, South Korea
- Department of Medical Informatics, College of Medicine, Korea UniversitySeoul, South Korea
| | - Sung Hak Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic UniversitySeoul, South Korea
| |
Collapse
|
16
|
Ichikawa H, Usui K, Aizawa M, Shimada Y, Muneoka Y, Kano Y, Sugai M, Moro K, Hirose Y, Miura K, Sakata J, Yabusaki H, Nakagawa S, Kawasaki T, Umezu H, Okuda S, Wakai T. Clinical application of targeted tumour sequencing tests for detecting ERBB2 amplification and optimizing anti-HER2 therapy in gastric cancer. BMC Cancer 2024; 24:719. [PMID: 38862927 PMCID: PMC11167924 DOI: 10.1186/s12885-024-12482-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Evaluation of human epidermal growth factor receptor 2 (HER2) overexpression caused by erb-b2 receptor tyrosine kinase 2 (ERBB2) amplification (AMP) by immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) is essential for treating unresectable metastatic gastric cancer (GC). A targeted tumour sequencing test enables comprehensive assessment of alterations in cancer-related genes, including ERBB2. This study aimed to evaluate the concordance between the targeted tumour sequencing test and IHC/FISH for detecting HER2-positive GC and to clarify the significance of ERBB2 AMP and concomitant genetic alterations in HER2 downstream pathways (DPs) in anti-HER2 therapy for unresectable metastatic GC patients. METHODS ERBB2 copy number alteration (CNA) was examined via a targeted tumour sequencing test in 152 formalin-fixed paraffin-embedded (FFPE) GC tissues. ERBB2 CNA was compared to HER2 status evaluated by IHC/FISH in FFPE block sections, which were identical to those subjected to the targeted tumour sequencing test. Treatment outcomes of anti-HER2 therapy in 11 patients with unresectable metastatic GC was evaluated. RESULTS ERBB2 AMP (≥ 2.5-fold change) was detected by the targeted tumour sequencing test in 15 patients (9.9%), and HER2 positivity (IHC 3 + or IHC 2+/FISH positive) was detected in 21 patients (13.8%). The overall percent agreement, positive percent agreement, negative percent agreement and Cohen's kappa between ERBB2 CNA and HER2 status were 94.7%, 66.7%, 99.2% and 0.75, respectively. Progression-free survival for trastuzumab therapy in patients with ERBB2 AMP was significantly longer than that in patients with no ERBB2 AMP detected by the targeted tumour sequencing test (median 14 months vs. 4 months, P = 0.007). Treatment response to trastuzumab therapy was reduced in patients with ERBB2 AMP and concomitant CNAs of genes in HER2 DPs. One patient with ERBB2 AMP and concomitant CNAs of genes in HER2 DPs achieved a durable response to trastuzumab deruxtecan as fourth-line therapy. CONCLUSIONS A targeted tumour sequencing test is a reliable modality for identifying HER2-positive GC. ERBB2 AMP and concomitant genetic alterations detected through the targeted tumour sequencing test are potential indicators of treatment response to trastuzumab therapy. The targeted tumour sequencing test has emerged as a plausible candidate for companion diagnostics to determine indications for anti-HER2 therapy in the era of precision medicine for GC.
Collapse
Affiliation(s)
- Hiroshi Ichikawa
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan.
| | - Kenji Usui
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Masaki Aizawa
- Department of Gastroenterological Surgery, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Chuo-ku, Niigata City, Niigata, 951-8566, Japan
| | - Yoshifumi Shimada
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Yusuke Muneoka
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Yosuke Kano
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Mika Sugai
- Division of Medical Technology, Niigata University Graduate School of Health Sciences, 2-746 Asahimachi-Dori, Chuo-ku, Niigata City, Niigata, 951-8518, Japan
| | - Kazuki Moro
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Yuki Hirose
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Kohei Miura
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Jun Sakata
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Hiroshi Yabusaki
- Department of Gastroenterological Surgery, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Chuo-ku, Niigata City, Niigata, 951-8566, Japan
| | - Satoru Nakagawa
- Department of Gastroenterological Surgery, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Chuo-ku, Niigata City, Niigata, 951-8566, Japan
| | - Takashi Kawasaki
- Department of Pathology, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Chuo-ku, Niigata City, Niigata, 951-8566, Japan
| | - Hajime Umezu
- Division of Pathology, Niigata University Medical and Dental Hospital, 1-754 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8520, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, 2-5274, Gakkocho-dori, Chuo-ku, Niigata City, Niigata, 951-8514, Japan
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| |
Collapse
|
17
|
Shi H, Kong S. A comprehensive evaluation of serum circCSPP1 as a novel diagnostic and prognostic biomarker for gastric cancer. Clin Res Hepatol Gastroenterol 2024; 48:102367. [PMID: 38744073 DOI: 10.1016/j.clinre.2024.102367] [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: 02/13/2024] [Revised: 04/05/2024] [Accepted: 05/01/2024] [Indexed: 05/16/2024]
Abstract
PURPOSE Gastric cancer (GC) has high incidence and mortality due to its low early screening efficiency. Circular RNAs (CircRNAs) are a new class of non-coding RNAs which is closely related to GC. Nevertheless, the clinical application value of circRNAs in GC are largely unknown. Therefore, we studied the role of a novel circRNA named circCSPP1 in patients with GC. METHODS CircRNA sequencing was performed to screen out the target molecule. Real-time fluorescent quantitative PCR (RT-qPCR) was utilized to detect the expression level of circCSPP1 in GC tissues, cells, and serum. Gel and Sanger sequencing were utilized to verify the ring structure of circCSPP1. RNase R enzyme digestion experiment and actinomycin D experiment were verifed the advantage of circCSPP1 as a diagnostic biomarker in patients with GC when that compared with linear RNA. The correlation between the expression level of serum circCSPP1 and clinicopathological data of GC patients was further analyzed. Receiver operating characteristic curve (ROC) and the area under ROC curve (AUC) were utilied to evaluate the diagnostic performance. RESULTS CircCSPP1 has a circular structure which with resistance to RNA exonuclease digestion and long half-life compared with linear RNA. In our study, circCSPP1 was first found up-regulated in patients with GC. Serum circCSPP1 level was decreased significantly after surgical resection whereas increased after recurrence. High expression of circCSPP1 was associated with poor survival rates. The expression level of circCSPP1 was significantly correlated to tumor size, T stage, lymph node metastasis, and TNM stage. The AUC of serum circCSPP1 was 0.834, with high sensitivity and specificity in discriminating patients with GC from healthy donors. More importantly, the combined diagnosis of circCSPP1, CEA, and CA19-9 achieved the superior AUC of 0.882, with the highest specificity. CONCLUSION Serum circCSPP1 may prove to be a potential non-invasive auxiliary diagnostic biomarker for patients with GC.
Collapse
Affiliation(s)
- Hengchuan Shi
- Department of Laboratory Medicine, Geriatric Hospital of Nanjing Medical University: Jiangsu Province Geriatric Hospital, Nanjing 210009 Jiangsu, China
| | - Shan Kong
- Department of Laboratory Medicine, Geriatric Hospital of Nanjing Medical University: Jiangsu Province Geriatric Hospital, Nanjing 210009 Jiangsu, China.
| |
Collapse
|
18
|
Jiang Z, Liang Y, Huang P, Ning J, Qi J. Value of quantitative microsurface structure analysis for evaluating the invasion depth of type 0-II early gastric cancer. JGH Open 2024; 8:e13055. [PMID: 38628386 PMCID: PMC11019524 DOI: 10.1002/jgh3.13055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/31/2024] [Accepted: 03/02/2024] [Indexed: 04/19/2024]
Abstract
Background and Aim The microsurface structure reflects the degree of damage to the glands, which is related to the invasion depth of early gastric cancer. To evaluate the diagnostic value of quantitative microsurface structure analysis for estimating the invasion depth of early gastric cancer. Methods White-light imaging and narrow-band imaging (NBI) endoscopy were used to visualize the lesions of the included patients. The area ratio and depth-predicting score (DPS) of each patient were calculated; meanwhile, each lesion was examined by endoscopic ultrasonography (EUS). Results Ninety-three patients were included between 2016 and 2019. Microsurface structure is related to the histological differentiation and progression of early gastric cancer. The receiver operating characteristic curve showed that when an area ratio of 80.3% was used as a cut-off value for distinguishing mucosal (M) and submucosal (SM) type 0-II gastric cancers, the sensitivity, specificity, and accuracy were 82.9%, 80.2%, and 91.6%, respectively. The accuracies for distinguishing M/SM differentiated and undifferentiated early gastric cancers were 87.4% and 84.8%, respectively. The accuracy of EUS for distinguishing M/SM early gastric cancer was 74.9%. DPS can only distinguish M-SM1 (SM infiltration <500 μm)/SM (SM infiltration ≥500 μm) with an accuracy of 83.8%. The accuracy of using area ratio for distinguishing 0-II early gastric cancers was better than those of using DPS and EUS (P < 0.05). Conclusion Quantitative analysis of microsurface structure can be performed to assess M/SM type 0-II gastric cancer and is expected to be effective for judging the invasion depth of gastric cancer.
Collapse
Affiliation(s)
- Zhang‐Xiu Jiang
- Division of Gastroenterology, Guangxi Hospital Division of The First Affiliated HospitalSun Yat‐sen University, People's Hospital of Guangxi Zhuang Autonomous RegionNanningGuangxiChina
| | - Yun‐Xiao Liang
- Department of Digestion (Division of Gastroenterology)People's Hospital of Guangxi Zhuang Autonomous RegionNanningChina
| | - Peng‐Yu Huang
- Department of Digestion (Division of Gastroenterology)People's Hospital of Guangxi Zhuang Autonomous RegionNanningChina
| | - Jia‐Juan Ning
- Department of Digestion (Division of Gastroenterology)People's Hospital of Guangxi Zhuang Autonomous RegionNanningChina
| | - Jing‐Jing Qi
- Department of PathologyPeople's Hospital of Guangxi Zhuang Autonomous RegionNanningChina
| |
Collapse
|
19
|
Xu WS, Zhang HY, Jin S, Zhang Q, Liu HD, Wang MT, Zhang B. Efficacy and safety of endoscopic submucosal dissection for early gastric cancer and precancerous lesions in elderly patients. World J Gastrointest Surg 2024; 16:511-517. [PMID: 38463378 PMCID: PMC10921217 DOI: 10.4240/wjgs.v16.i2.511] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/05/2024] [Accepted: 01/17/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND With advancements in the development of endoscopic technologies, the endoscopic submucosal dissection (ESD) has been one of the gold-standard therapies for early gastric cancer. AIM To investigate the efficacy and safety ESD in the treatment of early gastric cancer and precancerous lesions in the elderly patients. METHODS Seventy-eight elderly patients with early gastric cancer and precancerous lesions admitted to the Third Affiliated Hospital of Qiqihar Medical University were selected and classified into two groups according to the different surgical therapies they received between January 2021 and June 2022. Among them, 39 patients treated with ESD were included in an experimental group, and 39 patients treated with endoscopic mucosal resection (EMR) were included in a control group. We compared the basic intraoperative conditions, postoperative short-term recovery, long-term recovery effects and functional status of gastric mucosa between the two groups; the basic intraoperative conditions included lesion resection, intraoperative bleeding and operation time; the postoperative short-term recovery assessment indexes were length of hospital stay and incidence of surgical complications; and the long-term recovery assessment indexes were the recurrence rate at 1 year postoperatively and the survival situation at 1 year and 3 years postoperatively; and we compared the preoperative and predischarge serum pepsinogen I (PG I) and PG II levels and PG I/PG II ratio in the two groups before surgery and discharge. RESULTS The curative resection rate and the rate of en bloc resection were higher in the experimental group than in the control group. The intraoperative bleeding volume was higher in the experimental group than in the control group. The operation time was longer in the experimental group than that in the control group, and the rate for base residual focus was lower in the experimental group than that of the control group, and the differences were all statistically significant (all P < 0.05). The length of hospital stay was longer in the experimental group than in the control group, and the incidence of surgical complications, 1-year postoperative recurrence rate and 3-year postoperative survival rate were lower in the experimental group than in the control group, and the differences were statistically significant (all P < 0.05). However, the difference in the 1-year postoperative survival rate was not statistically significant between the two groups (P > 0.05). Before discharge, PG I and PG I/PG II ratio were elevated in both groups compared with the preoperative period, and the above indexes were higher in the experimental group than those in the control group, and the differences were statistically significant (both P < 0.05). Moreover, before discharge, PG II level was lower in both groups compared with the preoperative period, and the level was lower in the experimental group than in the control group, and the differences were all statistically significant (all P < 0.05). CONCLUSION Compared with EMR, ESD surgery is more thorough. It reduces the rate of base residual focus, recurrence rate, surgical complications, and promotes the recovery of gastric cells and glandular function. It is safe and suitable for clinical application.
Collapse
Affiliation(s)
- Wen-Si Xu
- Department of Gastroenterology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161099, Heilongjiang Province, China
| | - Hui-Yu Zhang
- Department of Gastroenterology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161099, Heilongjiang Province, China
| | - Shuang Jin
- Department of Gastroenterology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161099, Heilongjiang Province, China
| | - Qi Zhang
- Department of Gastroenterology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161099, Heilongjiang Province, China
| | - Hong-Dan Liu
- Department of Gastroenterology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161099, Heilongjiang Province, China
| | - Ming-Tao Wang
- Department of Gastroenterology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161099, Heilongjiang Province, China
| | - Bo Zhang
- Department of Gastroenterology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161099, Heilongjiang Province, China
| |
Collapse
|
20
|
Xin Y, Zhang Q, Liu X, Li B, Mao T, Li X. Application of artificial intelligence in endoscopic gastrointestinal tumors. Front Oncol 2023; 13:1239788. [PMID: 38144533 PMCID: PMC10747923 DOI: 10.3389/fonc.2023.1239788] [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: 06/14/2023] [Accepted: 11/17/2023] [Indexed: 12/26/2023] Open
Abstract
With an increasing number of patients with gastrointestinal cancer, effective and accurate early diagnostic clinical tools are required provide better health care for patients with gastrointestinal cancer. Recent studies have shown that artificial intelligence (AI) plays an important role in the diagnosis and treatment of patients with gastrointestinal tumors, which not only improves the efficiency of early tumor screening, but also significantly improves the survival rate of patients after treatment. With the aid of efficient learning and judgment abilities of AI, endoscopists can improve the accuracy of diagnosis and treatment through endoscopy and avoid incorrect descriptions or judgments of gastrointestinal lesions. The present article provides an overview of the application status of various artificial intelligence in gastric and colorectal cancers in recent years, and the direction of future research and clinical practice is clarified from a clinical perspective to provide a comprehensive theoretical basis for AI as a promising diagnostic and therapeutic tool for gastrointestinal cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaoyu Li
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
21
|
dos Santos SN, Junior DSG, Pereira JPM, Iadocicco NM, Silva AH, do Nascimento T, Dias LAP, de Oliveira Silva FR, Ricci-Junior E, Santos-Oliveira R, Bernardes ES. Development of glycan-targeted nanoparticles as a novel therapeutic opportunity for gastric cancer treatment. Cancer Nanotechnol 2023. [DOI: 10.1186/s12645-023-00161-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
AbstractChemotherapy resistance remains a major cause of therapeutic failure in gastric cancer. The combination of genetic material such as interference RNAs (iRNAs) to silence cancer-associated genes with chemotherapeutics has become a novel approach for cancer treatment. However, finding the right target genes and developing non-toxic, highly selective nanocarrier systems remains a challenge. Here we developed a novel sialyl-Tn-targeted polylactic acid—didodecyldimethylammonium bromide nanoparticle (PLA-DDAB) nanoparticles (NPs) loaded with dsRNA targeting ST6GalNac-I and/or galectin-3 genes. Using single photon emission computed tomography (SPECT), we have demonstrated that 99mtechnetium radiolabeled sialyl-Tn-targeted nanoparticles can reach the tumor site and downregulate ST6GalNAc-I and galectin-3 RNA expression levels when injected intravenously. Furthermore, using an in vivo gastric tumor model, these nanoparticles increased the effectiveness of 5-FU in reducing tumor growth. Our findings indicate that cancer-associated glycan-targeted NPs loaded with dsRNA targeting ST6GalNAc-I and/or galectin-3 in combination with standard chemotherapy, have the potential to become a novel therapeutic tool for gastric cancer.
Collapse
|
22
|
Dong ZB, Xiang HT, Wu HM, Cai XL, Chen ZW, Chen SS, He YC, Li H, Yu WM, Liang C. LncRNA expression signature identified using genome-wide transcriptomic profiling to predict lymph node metastasis in patients with stage T1 and T2 gastric cancer. Gastric Cancer 2023; 26:947-957. [PMID: 37691031 PMCID: PMC10640531 DOI: 10.1007/s10120-023-01428-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/28/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND Lymph node (LN) status is vital to evaluate the curative potential of relatively early gastric cancer (GC; T1-T2) treatment (endoscopic or surgery). Currently, there is a lack of robust and convenient methods to identify LN metastasis before therapeutic decision-making. METHODS Genome-wide expression profiles of long noncoding RNA (lncRNA) in primary T1 gastric cancer data from The Cancer Genome Atlas (TCGA) was used to identify lncRNA expression signature capable of detecting LN metastasis of GC and establish a 10-lncRNA risk-prediction model based on deep learning. The performance of the lncRNA panel in diagnosing LN metastasis was evaluated both in silico and clinical validation methods. In silico validation was conducted using TCGA and Asian Cancer Research Group (ACRG) datasets. Clinical validation was performed on T1 and T2 patients, and the panel's efficacy was compared with that of traditional tumor markers and computed tomography (CT) scans. RESULTS Profiling of genome-wide RNA expression identified a panel of lncRNA to predict LN metastasis in T1 stage gastric cancer (AUC = 0.961). A 10-lncRNA risk-prediction model was then constructed, which was validated successfully in T1 and T2 datasets (TCGA, AUC = 0.852; ACRG, AUC = 0.834). Thereafter, the clinical performance of the lncRNA panel was validated in clinical cohorts (T1, AUC = 0.812; T2, AUC = 0.805; T1 + T2, AUC = 0.764). Notably, the panel demonstrated significantly better performance compared with CT and traditional tumor markers. CONCLUSIONS The novel 10-lncRNA could diagnose LN metastasis robustly in relatively early gastric cancer (T1-T2), with promising clinical potential.
Collapse
Affiliation(s)
- Zhe-Bin Dong
- Department of General Surgery, The Affiliated Lihuili Hospital, Ningbo University, 57 Xingning Road, Ningbo, 315000, People's Republic of China
| | - Han-Ting Xiang
- Department of General Surgery, The Affiliated Lihuili Hospital, Ningbo University, 57 Xingning Road, Ningbo, 315000, People's Republic of China
| | - Heng-Miao Wu
- Department of General Surgery, The Affiliated Lihuili Hospital, Ningbo University, 57 Xingning Road, Ningbo, 315000, People's Republic of China
| | - Xian-Lei Cai
- Department of General Surgery, The Affiliated Lihuili Hospital, Ningbo University, 57 Xingning Road, Ningbo, 315000, People's Republic of China
| | - Zheng-Wei Chen
- Department of General Surgery, The Affiliated Lihuili Hospital, Ningbo University, 57 Xingning Road, Ningbo, 315000, People's Republic of China
| | - Sang-Sang Chen
- Department of General Surgery, The Affiliated Lihuili Hospital, Ningbo University, 57 Xingning Road, Ningbo, 315000, People's Republic of China
| | - Yi-Chen He
- Department of General Surgery, The Affiliated Lihuili Hospital, Ningbo University, 57 Xingning Road, Ningbo, 315000, People's Republic of China
| | - Hong Li
- Department of General Surgery, The Affiliated Lihuili Hospital, Ningbo University, 57 Xingning Road, Ningbo, 315000, People's Republic of China
| | - Wei-Ming Yu
- Department of General Surgery, The Affiliated Lihuili Hospital, Ningbo University, 57 Xingning Road, Ningbo, 315000, People's Republic of China
| | - Chao Liang
- Department of General Surgery, The Affiliated Lihuili Hospital, Ningbo University, 57 Xingning Road, Ningbo, 315000, People's Republic of China.
| |
Collapse
|
23
|
Wu H, Liu W, Yin M, Liu L, Qu S, Xu W, Xu C. A nomogram based on platelet-to-lymphocyte ratio for predicting lymph node metastasis in patients with early gastric cancer. Front Oncol 2023; 13:1201499. [PMID: 37719022 PMCID: PMC10502215 DOI: 10.3389/fonc.2023.1201499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/10/2023] [Indexed: 09/19/2023] Open
Abstract
Background Preoperative assessment of the presence of lymph node metastasis (LNM) in patients with early gastric cancer (EGC) remains difficult. We aimed to develop a practical prediction model based on preoperative pathological data and inflammatory or nutrition-related indicators. Methods This study retrospectively analyzed the clinicopathological characteristics of 1,061 patients with EGC who were randomly divided into the training set and validation set at a ratio of 7:3. In the training set, we introduced the least absolute selection and shrinkage operator (LASSO) algorithm and multivariate logistic regression to identify independent risk factors and construct the nomogram. Both internal validation and external validation were performed by the area under the receiver operating characteristic curve (AUC), C-index, calibration curve, and decision curve analysis (DCA). Results LNM occurred in 162 of 1,061 patients, and the rate of LNM was 15.27%. In the training set, four variables proved to be independent risk factors (p < 0.05) and were incorporated into the final model, including depth of invasion, tumor size, degree of differentiation, and platelet-to-lymphocyte ratio (PLR). The AUC values were 0.775 and 0.792 for the training and validation groups, respectively. Both calibration curves showed great consistency in the predictive and actual values. The Hosmer-Lemeshow (H-L) test was carried out in two cohorts, showing excellent performance with p-value >0.05 (0.684422, 0.7403046). Decision curve analysis demonstrated a good clinical benefit in the respective set. Conclusion We established a preoperative nomogram including depth of invasion, tumor size, degree of differentiation, and PLR to predict LNM in EGC patients and achieved a good performance.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Chunfang Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
24
|
Ding SQ, Zhang XP, Pei JP, Bai X, Ma JJ, Zhang CD, Dai DQ. Role of N6-methyladenosine RNA modification in gastric cancer. Cell Death Discov 2023; 9:241. [PMID: 37443100 DOI: 10.1038/s41420-023-01485-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 06/02/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023] Open
Abstract
N6-methyladenosine (m6A) RNA methylation is the most prevalent internal modification of mammalian messenger RNA. The m6A modification affects multiple aspects of RNA metabolism, including processing, splicing, export, stability, and translation through the reversible regulation of methyltransferases (Writers), demethylases (Erasers), and recognition binding proteins (Readers). Accumulating evidence indicates that altered m6A levels are associated with a variety of human cancers. Recently, dysregulation of m6A methylation was shown to be involved in the occurrence and development of gastric cancer (GC) through various pathways. Thus, elucidating the relationship between m6A and the pathogenesis of GC has important clinical implications for the diagnosis, treatment, and prognosis of GC patients. In this review, we evaluate the potential role and clinical significance of m6A-related proteins which function in GC in an m6A-dependent manner. We discuss current issues regarding m6A-targeted inhibition of GC, explore new methods for GC diagnosis and prognosis, consider new targets for GC treatment, and provide a reasonable outlook for the future of GC research.
Collapse
Affiliation(s)
- Si-Qi Ding
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, 110032, Shenyang, China
| | - Xue-Ping Zhang
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, 110032, Shenyang, China
| | - Jun-Peng Pei
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, 110032, Shenyang, China
| | - Xiao Bai
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, 110032, Shenyang, China
| | - Jin-Jie Ma
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, 110032, Shenyang, China
| | - Chun-Dong Zhang
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, 110032, Shenyang, China
| | - Dong-Qiu Dai
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, 110032, Shenyang, China.
- Cancer Center, The Fourth Affiliated Hospital of China Medical University, 110032, Shenyang, China.
| |
Collapse
|
25
|
Cheng R, Peng Y, Sun X, Zhang S, Li P. Circulating Tumor Cells as Diagnostic Markers of Early Gastric Cancer and Gastric Precancerous Lesions. Oncology 2023; 101:512-519. [PMID: 37263263 DOI: 10.1159/000531323] [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: 11/28/2022] [Accepted: 04/21/2023] [Indexed: 06/03/2023]
Abstract
INTRODUCTION Circulating tumor cells (CTCs) may be potential diagnostic biomarkers of various malignancies including gastric cancer. This study aimed to evaluate whether CTCs could be used to facilitate the diagnosis of early gastric cancer (EGC) or precancerous gastric lesions. METHODS The diagnostic study included consecutive patients with EGC, gastric precancerous lesions, or fundic gland polyps admitted to the Gastroenterology Department, Beijing Friendship Hospital Affiliated to Capital Medical University (National Center for Digestive Diseases) between October 2016 and January 2018. RESULTS A total of 92 patients were enrolled, including 57 patients with EGC, 14 patients with gastric precancerous lesions, and 21 patients with fundic gland polyps (control group). CTCs were detected in 47.89% (34/71) of patients with EGC/gastric precancerous lesions and 4.76% (1/21) of patients with fundic gland polyps (p < 0.001). CTC detection distinguished EGC/precancerous lesions from fundic gland polyps with an area under the receiver operating characteristic curve of 0.740 (95% confidence interval, 0.640-0.840; p = 0.001), a sensitivity of 49.10%, a specificity of 95.00%, a positive predictive value of 97.00%, and a negative predictive value of 64.90%. CONCLUSIONS Peripheral blood CTCs are more common in patients with EGC or gastric precancerous lesions than in patients with fundic gland polyps. Measurement of CTCs may be a useful tool to aid in the diagnosis of EGC and precancerous lesions.
Collapse
Affiliation(s)
- Rui Cheng
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yang Peng
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Gastroenterology, Suining First People's Hospital, Suining, China
| | - Xiujing Sun
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shutian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Peng Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
26
|
Chen M, Chen K, Hou H, Li W, Wang X, Dao Q, Wang Z. Incidence and mortality trends in gastric cancer in the United States, 1992-2019. Int J Cancer 2023; 152:1827-1836. [PMID: 36562305 DOI: 10.1002/ijc.34415] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/12/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022]
Abstract
Our study aimed to estimate the epidemiological trends of gastric cancer in the United States from 1992 to 2019. This population-based study used the US Surveillance, Epidemiology and End Results-12 database as a fundamental cohort to analyze gastric cancer incidence, incidence-based mortality (IBM), overall survival (OS) and cancer-specific survival (CSS) probabilities from 1992 to 2019. The Global Burden of Disease study (1990-2018) was used as a likely validation cohort. Age-period-cohort analyses were performed to explore the underlying causes of trend changes. We found that the incidence rate of gastric cancer decreased from 1992 to 2019. IBM also decreased significantly from 1997 to 2019. The 3-year OS and CSS of gastric cancer increased from 22.3% to 28.7% and 25.7% to 33.5%, respectively. However, the proportion of distant gastric cancer cases had unexpectedly increased rapidly from 33.1% in 1992 to 44.7% in 2019. Age-period-cohort modeling found that the incidence and IBM rates remained stable in the groups aged below 50 years, while that in all age groups older than 50 years showed a significant downward trend. High incidence and mortality risks were observed in the younger birth cohorts (birth year after 1990). To conclude, we observed a decline in incidence and mortality rates of gastric cancer in the United States in the past decades. We determined that progression of primary and tertiary preventive measures is the main reason for the reduction in the disease burden of gastric cancer. However, secondary preventive measures for gastric cancer still need to be strengthened.
Collapse
Affiliation(s)
- Mengding Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ke Chen
- Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Huanan Hou
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wanjing Li
- Department of Geriatrics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaoshan Wang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qianze Dao
- Department of General Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhengguang Wang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
27
|
Paredes O, Baca C, Cruz R, Paredes K, Luque-Vasquez C, Chavez I, Taxa L, Ruiz E, Berrospi F, Payet E. Predictive factors of lymphatic metastasis and evaluation of the Japanese treatment guidelines for endoscopic resection of early gastric cancer in a high-volume center in Perú. Heliyon 2023; 9:e16293. [PMID: 37251889 PMCID: PMC10209413 DOI: 10.1016/j.heliyon.2023.e16293] [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: 06/18/2022] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/31/2023] Open
Abstract
Purpose This study aimed to identify the predictive factors of lymph node metastasis (LNM) in patients with early gastric cancer (EGC) and to evaluate the applicability of the Japanese treatment guidelines for endoscopic resection in the western population. Methods Five hundred-one patients with pathological diagnoses of EGC were included. Univariate and multivariate analyses were conducted to identify the predictive factors of LNM. EGC patients were distributed according to the indications for endoscopic resection of the Eastern guidelines. The incidence of LNM was evaluated in each group. Results From 501 patients with EGC, 96 (19.2%) presented LNM. In 279 patients with tumors with submucosal infiltration (T1b), 83 (30%) patients had LNM. Among 219 patients who presented tumors > 3 cm, 63 (29%) patients had LNM. Thirty-one percent of patients with ulcerated tumors presented LMN (33 out of 105). In 76 patients and 24 patients with lymphovascular and perineural invasion, the percentage of LMN was 84% and 87%, respectively. In the multivariate analysis, a tumor diameter >3 cm, submucosal invasion, lymphovascular, and perineural invasion were independent predictors of LMN in EGC. No patient with differentiated, non-ulcerated mucosal tumors presented LNM regardless of tumor size. Three of 17 patients (18%) with differentiated, ulcerated mucosal tumors and ≤ 3 cm presented LNM. No LNM was evidenced in patients with undifferentiated mucosal tumors and ≤ 2 cm. Conclusions The presence of LNM in Western EGC patients was independently related to larger tumors (>3 cm), submucosal invasion, lymphovascular and perineural invasion. The Japanese absolute indications for EMR are safe in the Western population. Likewise, Western patients with differentiated, non-ulcerated mucosal tumors, and larger than 2 cm are susceptible to endoscopic resection. Patients with undifferentiated mucosal tumors smaller than 2 cm presented encouraging results and ESD could be recommended only for selected cases.
Collapse
Affiliation(s)
- Oscar Paredes
- Department of Abdominal Surgery, National Institute of Neoplastic Diseases INEN, Lima, Peru
| | - Carlos Baca
- Department of Abdominal Surgery, National Institute of Neoplastic Diseases INEN, Lima, Peru
| | - Renier Cruz
- Department of Pathology, National Institute of Neoplastic Disease INEN, Lima, Peru
| | - Kori Paredes
- Department of Abdominal Surgery, National Institute of Neoplastic Diseases INEN, Lima, Peru
| | - Carlos Luque-Vasquez
- Department of Abdominal Surgery, National Institute of Neoplastic Diseases INEN, Lima, Peru
| | - Iván Chavez
- Department of Abdominal Surgery, National Institute of Neoplastic Diseases INEN, Lima, Peru
| | - Luis Taxa
- Department of Pathology, National Institute of Neoplastic Disease INEN, Lima, Peru
| | - Eloy Ruiz
- Department of Abdominal Surgery, National Institute of Neoplastic Diseases INEN, Lima, Peru
| | - Francisco Berrospi
- Department of Abdominal Surgery, National Institute of Neoplastic Diseases INEN, Lima, Peru
| | - Eduardo Payet
- Department of Abdominal Surgery, National Institute of Neoplastic Diseases INEN, Lima, Peru
| |
Collapse
|
28
|
Komai Y, Nakajima K, Saito K, Tomioka Y, Masuda H, Ogawa A, Yonese J, Kobayashi E, Ito M. Development of a New Two-Arm Transurethral Surgical System for En Bloc Resection of Bladder Tumor: A Preclinical Study. J Endourol 2023; 37:165-170. [PMID: 36322793 DOI: 10.1089/end.2021.0917] [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: 02/04/2023] Open
Abstract
Backgrounds and Objectives: To overcome the piecemeal nature of bladder tumor resection, en bloc resection of bladder tumor (ERBT) has been introduced. ERBT is difficult for surgeons to perform using the currently available system because it has only one arm. Herein, we aimed to develop a new transurethral surgical system to facilitate two-arm ERBT and to report the results of preclinical experiments using tumor phantoms. Methods: Initially, we aimed to develop a brand-new surgical system for ERBT but, after trial and error, we redirected our development to the creation of three elements: the left arm to grasp the tumor; the right arm to cut the tumor; and the system to operate the arms that can be attached to the existing surgical system (UES-40 SurgMaster® [Olympus Co. Ltd., Tokyo, Japan]). The current system was evaluated by performing simulated ERBTs using tumor phantoms made from konjac jelly. Results: Following the assembly of developed arms into the UES-40 SurgMaster, we conducted preliminary ERBTs. After performing several resections, we adopted a basket-shaped forceps as the left arm instead of grasping forceps and an arched electrode as the right arm. The two arms and single endoscope were placed in an equilateral triangle. We performed ERBT for the tumor phantoms that ranged from 0.5 to 2.0 cm without major redo. Conclusion: Herein, we introduced our development for two-arm ERBT. The current concept of "two-hand transurethral surgery" has the potential to be developed in future in vivo and clinical trials.
Collapse
Affiliation(s)
- Yoshinobu Komai
- Department of Urology, Canter Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan.,Surgical Device Innovation Office, National Cancer Center Hospital East, Kashiwa, Japan
| | | | | | - Yutaka Tomioka
- Surgical Device Innovation Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hitoshi Masuda
- Department of Urology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Akira Ogawa
- R&D Department, Actment Co., Ltd., Kasukabe, Japan
| | - Junji Yonese
- Department of Urology, Canter Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Etsuko Kobayashi
- Department of Precision Machinery Engineering, Faculty of Engineering, The University of Tokyo, Tokyo, Japan
| | - Masaaki Ito
- Surgical Device Innovation Office, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
29
|
Ito N, Funasaka K, Fujiyoshi T, Nishida K, Furukawa K, Kakushima N, Furune S, Ohno E, Nakamura M, Horiguchi N, Shibata T, Miyahara R, Haruta JI, Hirooka Y, Fujishiro M, Kawashima H. Scoring system for predicting the prognosis of elderly gastric cancer patients after endoscopic submucosal dissection. Dig Endosc 2023; 35:67-76. [PMID: 36052429 DOI: 10.1111/den.14416] [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: 04/30/2022] [Accepted: 07/29/2022] [Indexed: 01/17/2023]
Abstract
OBJECTIVES Comprehensive assessments of the long-term outcomes of endoscopic submucosal dissection (ESD) for early gastric cancer (EGC) in the elderly are unavailable. We aimed to create a scoring system to predict the long-term prognosis after ESD for EGC among patients aged ≥75 years. METHODS We conducted retrospective studies of two cohorts: a single-center cohort (2006-2011) for developing the scoring system, and a multicenter cohort for validating the developed system (2012-2016). In the development cohort, factors related to death after ESD were identified using multivariable Cox regression analysis, and a predictive scoring system was developed. In the validation cohort, the scoring system was validated in 295 patients. RESULTS In the development cohort, Charlson comorbidity index (CCI) ≥3 (hazard ratio [HR] 3.017), high psoas muscle index (PMI) (HR 2.206), and age ≥80 years (HR 1.978) were significantly related to overall survival after ESD. Therefore, high CCI, low PMI, and age ≥80 years were assigned 1 point each. The patients were categorized into low (≤1 point) and high (≥2 points) score groups based on their total scores. In the validation cohort, 184 and 111 patients were assigned to the low- and high-score groups, respectively. In comparisons based on Kaplan-Meier curves, the 5-year survival rate was 91.5% in the low-score group and 57.8% in the high-score group (log-rank test; P < 0.001). CONCLUSION Our scoring system including high CCI, low PMI, and age ≥80 years could stratify the long-term prognosis of elderly patients aged ≥75 years after ESD for EGC.
Collapse
Affiliation(s)
- Nobuhito Ito
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Kohei Funasaka
- Department of Gastroenterology and Hepatology, Fujita Health University School of Medicine, Aichi, Japan
| | - Toshihisa Fujiyoshi
- Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Aichi, Japan
| | - Kazuki Nishida
- Department of Biostatistics Section, Center for Advanced Medicine and Clinical Research, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Kazuhiro Furukawa
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Naomi Kakushima
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Aichi, Japan
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Satoshi Furune
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Eizaburo Ohno
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Masanao Nakamura
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Noriyuki Horiguchi
- Department of Gastroenterology and Hepatology, Fujita Health University School of Medicine, Aichi, Japan
| | - Tomoyuki Shibata
- Department of Gastroenterology and Hepatology, Fujita Health University School of Medicine, Aichi, Japan
| | - Ryoji Miyahara
- Department of Gastroenterology and Hepatology, Fujita Health University School of Medicine, Aichi, Japan
| | - Jun-Ichi Haruta
- Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Aichi, Japan
| | - Yoshiki Hirooka
- Department of Gastroenterology and Hepatology, Fujita Health University School of Medicine, Aichi, Japan
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Aichi, Japan
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Aichi, Japan
| |
Collapse
|
30
|
Wei Y, Jiang C, Han Y, Song W, Li X, Yin X. Characteristics and background mucosa status of early gastric cancer after Helicobacter pylori eradication: A narrative review. Medicine (Baltimore) 2022; 101:e31968. [PMID: 36482539 PMCID: PMC9726367 DOI: 10.1097/md.0000000000031968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/01/2022] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori (H pylori) eradication treatment can reduce the risk of gastric cancer. However, early gastric cancer (EGC) can still be detected after eradication. Meanwhile, EGC after eradication is challenging to diagnose by an endoscopist in some cases due to the lack of apparent characteristics and the complex mucosal status. This review aims to summarize the endoscopic and histological characteristics and the mucosal risk factors for gastric cancer after H pylori eradication. The literature was searched for possible reported gastric cancer after eradication in "PubMed." These included related clinical studies and reviews, and unrelated or non-English articles were excluded. Endoscopically, EGC displays a small, reddish and depressed lesion, indistinct border, "gastritis-like" appearance and submucosal invasion. Histologically, it is divided into surface differentiation, nontumorous epithelium, and intestinal type. The risk factors include severe gastric atrophy, intestinal metaplasia in the corpus, and map-like redness. In conclusion, these studies on the characteristics and risk mucosal factors of patients with gastric cancer after H pylori eradication will drive the establishment of a novel endoscopic surveillance and diagnosis system for H pylori-eradicated patients.
Collapse
Affiliation(s)
- Yali Wei
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Chen Jiang
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Yiping Han
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Wen Song
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xiaoyu Li
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xiaoyan Yin
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
31
|
Yasuda T, Dohi O, Yamada S, Ishida T, Iwai N, Hongo H, Terasaki K, Tanaka M, Yamada N, Kamada K, Horie R, Harusato A, Horii Y, Takayama S, Zen K, Majima A, Mizuno N, Motoyoshi T, Yagi N, Naito Y, Itoh Y. Risk and prognostic factors of invasive gastric cancer detection during surveillance endoscopy: Multi-institutional cross-sectional study. Dig Endosc 2022. [PMID: 36461634 DOI: 10.1111/den.14492] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/01/2022] [Indexed: 12/04/2022]
Abstract
OBJECTIVES Esophagogastroduodenoscopy (EGD) is important for the detection of curable gastric cancer (GC). However, there are no appropriate surveillance data during routine endoscopic inspections. This study aimed to clarify the risk factors of pT1b or deeper GC detection during surveillance endoscopy. METHODS This was a retrospective, multicenter, cross-sectional study conducted in 15 Japanese hospitals. We retrospectively analyzed patients with GC who had previously undergone surveillance endoscopy at each institution from January 2014 to March 2020. Patients who had undergone gastrectomy, non-infection of Helicobacter pylori (Hp), and those with intervals <3 months or >10 years from a previous endoscopy were excluded. RESULTS In total, 1085 patients with GCs detected during surveillance endoscopy were enrolled. The multivariate logistic analysis revealed that current Hp infection (odds ratio [OR] 2.18; 95% confidence interval [CI] 1.50-3.16) and a surveillance interval of >1.5 years (OR 1.96; 95% CI 1.35-2.84) were independent risk factors for pT1b or deeper GC. The 5-year disease-specific survival (5y-DSS) rate of GC was significantly lower in patients with surveillance interval of >1.5 years than in those with surveillance interval of ≤1.5 years (93.7% vs. 98.3%, P < 0.001). Similarly, the 5y-DSS rate of GC was significantly lower in patients with active Hp infection than in those without (93.7% vs. 99.4%, P < 0.001). CONCLUSION In this study, a surveillance interval of >1.5 years and current Hp infection were independent risk factors for detecting pT1b or deeper GC. Additionally, these factors were poor prognostic factors of the detected GC during surveillance endoscopy.
Collapse
Affiliation(s)
- Takeshi Yasuda
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto, Japan
| | - Osamu Dohi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto, Japan
| | - Shinya Yamada
- Department of Gastroenterology, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto, Japan
| | - Tsugitaka Ishida
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto, Japan
| | - Naoto Iwai
- Department of Gastroenterology, Fukuchiyama City Hospital, Kyoto, Japan
| | - Hitoshi Hongo
- Department of Gastroenterology, Fujita Gastroenterological Hospital, Takatsuki, Osaka, Japan
| | - Kei Terasaki
- Department of Gastroenterology, Saiseikai Suita Hospital, Osaka, Japan
| | - Makoto Tanaka
- Department of Gastroenterology, Saiseikai Shiga Hospital, Shiga, Japan
| | - Nobuhisa Yamada
- Department of Gastroenterology, Matsushita Memorial Hospital, Osaka, Japan
| | - Kazuhiro Kamada
- Department of Gastroenterology, Matsushita Memorial Hospital, Osaka, Japan
| | - Ryusuke Horie
- Department of Gastroenterology, Kyoto Kuramaguchi Medical Center, Kyoto, Japan
| | - Akihito Harusato
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto, Japan.,Department of Gastroenterology, North Medical Center Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yusuke Horii
- Department of Gastroenterology, Kyoto Kizugawa Hospital, Kyoto, Japan
| | - Shun Takayama
- Department of Gastroenterology, Maizuru Medical Center, Kyoto, Japan
| | - Keika Zen
- Department of Gastroenterology, Ōtsu Municipal Hospital, Shiga, Japan
| | - Atsushi Majima
- Department of Gastroenterology, Omihachiman Community Medical Center, Shiga, Japan
| | - Naoki Mizuno
- Department of Gastroenterology, Kyoto City Hospital, Kyoto, Japan
| | | | - Nobuaki Yagi
- Department of Gastroenterology, Asahi University Hospital, Gifu, Japan
| | - Yuji Naito
- Department of Human Immunology and Nutrition Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto, Japan
| |
Collapse
|
32
|
Zeng Q, Feng Z, Zhu Y, Zhang Y, Shu X, Wu A, Luo L, Cao Y, Xiong J, Li H, Zhou F, Jie Z, Tu Y, Li Z. Deep learning model for diagnosing early gastric cancer using preoperative computed tomography images. Front Oncol 2022; 12:1065934. [PMID: 36531076 PMCID: PMC9748811 DOI: 10.3389/fonc.2022.1065934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/07/2022] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Early gastric cancer (EGC) is defined as a lesion restricted to the mucosa or submucosa, independent of size or evidence of regional lymph node metastases. Although computed tomography (CT) is the main technique for determining the stage of gastric cancer (GC), the accuracy of CT for determining tumor invasion of EGC was still unsatisfactory by radiologists. In this research, we attempted to construct an AI model to discriminate EGC in portal venous phase CT images. METHODS We retrospectively collected 658 GC patients from the first affiliated hospital of Nanchang university, and divided them into training and internal validation cohorts with a ratio of 8:2. As the external validation cohort, 93 GC patients were recruited from the second affiliated hospital of Soochow university. We developed several prediction models based on various convolutional neural networks, and compared their predictive performance. RESULTS The deep learning model based on the ResNet101 neural network represented sufficient discrimination of EGC. In two validation cohorts, the areas under the curves (AUCs) for the receiver operating characteristic (ROC) curves were 0.993 (95% CI: 0.984-1.000) and 0.968 (95% CI: 0.935-1.000), respectively, and the accuracy was 0.946 and 0.914. Additionally, the deep learning model can also differentiate between mucosa and submucosa tumors of EGC. CONCLUSIONS These results suggested that deep learning classifiers have the potential to be used as a screening tool for EGC, which is crucial in the individualized treatment of EGC patients.
Collapse
Affiliation(s)
- Qingwen Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
- Institute of Digestive Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zongfeng Feng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
- Institute of Digestive Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yanyan Zhu
- Department of Radiology, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Yang Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
- Institute of Digestive Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xufeng Shu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Ahao Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Lianghua Luo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Yi Cao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Jianbo Xiong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Hong Li
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fuqing Zhou
- Department of Radiology, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Zhigang Jie
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
- Institute of Digestive Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yi Tu
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhengrong Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
- Institute of Digestive Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
33
|
Prognostic Benefit of Additional Treatment After Endoscopic Submucosal Dissection for Esophageal Squamous Cell Carcinoma. Dig Dis Sci 2022; 68:2050-2060. [PMID: 36445654 DOI: 10.1007/s10620-022-07746-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/10/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Although additional treatment is considered for patients with esophageal squamous cell carcinoma (ESCC) invading into the muscularis mucosa (pT1a-MM) or submucosa (pT1b-SM) after endoscopic submucosal dissection (ESD), the actual benefits of this method remain to be elucidated. AIMS We aimed to evaluate the prognostic benefits of additional treatment in such patients. METHODS Between 2006 and 2017, we enrolled patients with pT1a-MM/pT1b-SM ESCC after ESD at 21 institutions in Japan. Overall survival (OS) and disease-specific survival (DSS) were compared between the additional treatment and follow-up groups after propensity score matching, to reduce the bias of baseline characteristics. A subgroup analysis was performed according to the pathological findings: category A, pT1a-MM but negative for lymphovascular invasion (LVI) and vertical margin (VM); category B, tumor invasion into the submucosa ≤ 200 μm but negative for LVI and VM; category C, others. RESULTS Of 593 patients with pT1a-MM/pT1b-SM ESCC after ESD, 101 matched pairs were extracted after propensity score matching. The OSs were similar between the additional treatment and follow-up groups (80.6% vs. 78.6% in 5 years; P = 0.972). In a subgroup analysis, the OS in the additional treatment group was significantly lower than that in the follow-up group (65.7% vs. 95.2% in 5 years; P = 0.037) in category A, whereas OS did not significantly differ in category C (76.8% vs. 69.5% in 5 years; P = 0.360). CONCLUSIONS Additional treatment after ESD in patients with pT1a-MM/pT1b-SM ESCC was not associated with an improved prognosis.
Collapse
|
34
|
Hu RH, Zhang HB, Yuan B, Zhang KH, Xu JY, Cui XM, Du T, Song C, Zhang S, Jiang XH. Quality and accuracy of gastric cancer related videos in social media videos platforms. BMC Public Health 2022; 22:2025. [PMID: 36335401 PMCID: PMC9636631 DOI: 10.1186/s12889-022-14417-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 10/10/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Gastric cancer is a major public health problem worldwide. Social media has affected public's daily lives in ways no one ever thought possible. Both TikoTok and its Chinese version Douyin are the most popular short video posting platform. This study aimed to evaluate the quality, accuracy, and completeness of videos for gastric cancer on TikTok and Douyin. METHODS The terms "gastric cancer" was searched on TikTok in both English and Japanese, and on Douyin in Chinese. The first 100 videos in three languages (website's default setting) were checked. QUality Evaluation Scoring Tool (QUEST) and DISCERN as the instrument for assessing the quality of the information in each video. Content was analysed under six categories (aetiology, anatomy, symptoms, preventions, treatments, and prognosis). The educational value and completeness were evaluated with a checklist developed by the researchers. RESULTS A total of 78 videos in English, 63 in Japanese, and 99 in Chinese were analyzed. The types of sources were as follows: 6.4% in English, 4.8% in Japanese, and 57.6% in Chinese for health professionals; 93.6% in English, 95.2% in Japanese, and 3.0% in Chinese for private users; none in English and Japanese, but 39.4% in Chinese for other sources. In all, 20.5% in English, 17.5% in Japanese, and 93.9% in Chinese of videos had useful information about gastric cancer. Among the useful videos, the videos published in Chinese had the highest QUEST(p < 0.05) and DISCERN scores(p < 0.05), followed by those published in Japanese. Among the educational videos, prognosis in English (37.5%), symptoms in Japanese (54.5%), and prevention in Chinese (47.3%) were the most frequently covered topic. CONCLUSIONS TikTok in English and Japanese might not fully meet the gastric cancer information needs of public, but Douyin in Chinese was the opposite.
Collapse
Affiliation(s)
- Ren-Hao Hu
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Hai-Bin Zhang
- Center of Digestive Endoscopy, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Biao Yuan
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Ke-Hui Zhang
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Jia-Yi Xu
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Xi-Mao Cui
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Tao Du
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Chun Song
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Shun Zhang
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University, Shanghai, China.
| | - Xiao-Hua Jiang
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University, Shanghai, China.
| |
Collapse
|
35
|
Hu B, Meng Y, Qu C, Wang BY, Xiu DR. Combining single-cell sequencing data to construct a prognostic signature to predict survival, immune microenvironment, and immunotherapy response in gastric cancer patients. Front Immunol 2022; 13:1018413. [PMID: 36300104 PMCID: PMC9589350 DOI: 10.3389/fimmu.2022.1018413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/20/2022] [Indexed: 11/22/2022] Open
Abstract
Background and objective Gastric cancer (GC) represents a major factor inducing global cancer-associated deaths, but specific biomarkers and therapeutic targets for GC are lacking at present. Therefore, the present work focused on developing an immune-related genetic signature at the single-cell level for categorizing GC cases and predicting patient prognostic outcome, immune status as well as treatment response. Methods Single-cell RNA-sequencing (scRNA-seq) data were combined with bulk RNA-seq data in GC patients for subsequent analyses. Differences in overall survival (OS), genomic alterations, immune status, together with estimated immunotherapeutic outcomes were measured between different groups. Results Nine cell types were identified by analyzing scRNA-seq data from GC patients, and marker genes of immune cells were also selected for subsequent analysis. In addition, an immune-related signature was established to predict OS while validating the prediction power for GC patients. Afterwards, a nomogram with high accuracy was constructed for improving our constructed signature’s clinical utility. The low-risk group was featured by high tumor mutation burden (TMB), increased immune activation, and microsatellite instability-high (MSI-H), which were related to the prolonged OS and used in immunotherapy. By contrast, high-risk group was associated with microsatellite stability (MSS), low TMB and immunosuppression, which might be more suitable for targeted therapy. Meanwhile, the risk score generated by our signature was markedly related to the cancer stem cell (CSC) index. In addition, the immunotherapeutic response prediction accuracy of our signature was validated in an external dataset IMvigor210 cohort. Conclusion A signature was constructed according to scRNA-seq data analysis. The signature-screened low- and high-risk patients had different prognoses, immune statuses and enriched functions and pathways. Such results shed more lights on immune status of GC, prognosis assessment, and development of efficient immunotherapeutic treatments.
Collapse
|
36
|
Kagawa Y, Fukuzawa M, Sugimoto M, Nemoto D, Muramatsu T, Shinohara H, Matsumoto T, Madarame A, Yamaguchi H, Uchida K, Morise T, Koyama Y, Sugimoto A, Yamauchi Y, Kono S, Naito S, Yamamoto K, Kishimoto Y, Inuyama M, Kawai T, Itoi T. Validation of the BEST-J score, a prediction model for bleeding after endoscopic submucosal dissection for early gastric cancer: a multicenter retrospective observational study. Surg Endosc 2022; 36:7240-7249. [PMID: 35194665 DOI: 10.1007/s00464-022-09096-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 02/07/2022] [Indexed: 10/28/2022]
Abstract
BACKGROUND A new scoring system, the BEST-J score, using ten risk factors to assign cases to different post-endoscopic submucosal dissection (ESD) risk groups for bleeding, has been shown to be accurate for risk stratification. We first aimed to validate the BEST-J score at four hospitals not specialized in performing ESD and then aimed to identify other risk factors for post-ESD bleeding. METHODS We evaluated the incidence of post-ESD bleeding in 791 cases of early gastric cancer (EGC) between October 2013 and December 2020 as a retrospective, multi-center observational study conducted at four hospitals. Multivariate logistic regression models to examine the effect of independent variables on post-ESD bleeding firstly included ten possible factors raised by the BEST-J score and secondly included statistically significant (p < 0.01) in univariate analysis. The prediction accuracy of the model was evaluated by receiver-operating characteristic analysis and the areas under the curve (AUC). RESULTS The incidence of post-ESD bleeding was 4.8% (38/791, 95% confidence interval [CI] 3.4-6.5%). On multivariate analysis, the risk factors were P2Y12 receptor antagonist (odds ratio [OR]: 5.870, 95% CI 1.624-21.219), warfarin (8.382, 1.658-42.322), direct oral anticoagulant (DOAC) (8.980, 1.603-50.322), and tumor location in lower third of stomach (2.151, 1.012-4.571), respectively. When we categorized cases into low-risk by BEST-J score, intermediate-risk, high-risk, and very high-risk groups, the bleeding rates were 2.8%, 7.3%, 12.8%, and 19.0%, respectively. The AUC for our cohort was 0.713 (95% CI 0.625-0.802) for the BEST-J score. In the multivariate analysis in our cohort, the risks were age, body mass index, P2Y12 receptor antagonist, warfarin, DOAC, respectively. DISCUSSION The BEST-J score is equally accurate in risk stratification of patients with EGC for post-ESD bleeding at non-specialized facilities for ESD as in specialized hospitals. BMI and age may be helpful additional risk factors at hospitals not specialized.
Collapse
Affiliation(s)
- Yasuyuki Kagawa
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Masakatsu Fukuzawa
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Mitsushige Sugimoto
- Department of Gastroenterological Endoscopy, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan.
| | - Daiki Nemoto
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Takahiro Muramatsu
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Hirokazu Shinohara
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Taisuke Matsumoto
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Akira Madarame
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Hayato Yamaguchi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Kumiko Uchida
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Takashi Morise
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Yohei Koyama
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Akihiko Sugimoto
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Yoshiya Yamauchi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Shin Kono
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Sakiko Naito
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Kei Yamamoto
- Department of Gastroenterology and Hepatology, Tokyo Medical University Hachioji Medical Center, 1163, Tatemachi, Hachioji-shi, Tokyo, 193-0998, Japan
| | - Yoshiko Kishimoto
- Department of Gastroenterology, Todachuo General Hospital, 1-19-3 Honcho, Toda-shi, Saitama, 335-0023, Japan
| | - Mitsuko Inuyama
- Department of Gastroenterology, Tokyo Rosai Hospital, 4-13-21 Omori-minami, Ota-ku, Tokyo, 143-0013, Japan
| | - Takashi Kawai
- Department of Gastroenterological Endoscopy, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Takao Itoi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| |
Collapse
|
37
|
Liu JY, Yao J, Liu JJ, He T, Wang FJ, Xie TY, Cui JX, Yang XD. LASTR is a novel prognostic biomarker and predicts response to cancer immunotherapy in gastric cancer. Front Oncol 2022; 12:1020255. [PMID: 36249015 PMCID: PMC9557225 DOI: 10.3389/fonc.2022.1020255] [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] [Received: 08/16/2022] [Accepted: 09/07/2022] [Indexed: 11/24/2022] Open
Abstract
Gastric cancer (GC), a malignant tumor of digestive tract, is characterized by a high death rate. Thus, it is of particular importance to clarify the mechanisms of GC and gain new molecular targets for the sake of preventing and treating GC. It was reported that long non-coding RNAs (IncRNAs) are prognostic factors to cancer. Ferroptosis refers to a process of programmed cell death dependent on iron. This study sets out to investigate the expression and function of ferroptosis-related lncRNA (FRlncRNA) in GC. TCGA datasets offered RNA-seq data for 375 GC patients and clinical data for 443 GC patients. Based on Pearson’s correlation analysis, we studied their expression and identified the FRlncRNAs. Differentially expressed prognosis related to FRlncRNA were determined with the help of the Wilcoxon test and univariate Cox regression analysis. To evaluate the accuracy of the prognostic capacity, researchers used the Kaplan-Meier technique, as well as univariate and multivariate Cox regression and receiver operating characteristic (ROC) curve studies. We also carried out the real-time PCR and CCK8 assays to examine the expression and function of FRlncRNA. In this study, we identified 50 ferroptosis-related DEGs which were involved in tumor progression. In addition, we identified 33 survival-related FRlncRNAs. Among them, lncRNA associated with SART3 regulation of splicing(LASTR) was confirmed to be highly expressed in GC specimens compared to non-tumor specimens in this cohort. Survival assays illuminated that the high LASTR expression predicted a shorter overall survival and progression-free survival of GC patients. Based on multivariate Cox regression analyses, it was confirmed that the GC had a worse chance of surviving the disease overall if their tumors expressed LASTR, which was an independent prognostic indication. Then, Loss-of-function tests showed that knocking down LASTR had a significant effect on reducing the proliferation of GC cells. Finally, we found that the expression of LASTR was negatively associated with CD8 T cells, T cells, Th17 cells, and T helper cells. Overall, our findings identified a novel survival-related FRlncRNA, LASTR which possibly can serve as a novel prognostic biomarker predicting response to cancer immunotherapy and therapeutic target for GC patients.
Collapse
Affiliation(s)
- Jun-Yan Liu
- Department of General Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jing Yao
- Department of General Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jia-Jia Liu
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tao He
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fang-Jie Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical Center (Daping Hospital), Army Medical University, Chongqing, China
| | - Tian-Yu Xie
- Department of General Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jian-Xin Cui
- Department of General Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Jian-Xin Cui, ; Xiao-Dong Yang,
| | - Xiao-Dong Yang
- Department of General Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Jian-Xin Cui, ; Xiao-Dong Yang,
| |
Collapse
|
38
|
Hou S, Liu F, Gao Z, Ye Y. Pathological and oncological outcomes of pylorus-preserving versus conventional distal gastrectomy in early gastric cancer: a systematic review and meta-analysis. World J Surg Oncol 2022; 20:308. [PMID: 36153587 PMCID: PMC9508780 DOI: 10.1186/s12957-022-02766-0] [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] [Received: 07/07/2022] [Accepted: 09/06/2022] [Indexed: 01/30/2023] Open
Abstract
Abstract
Background
Pylorus-preserving gastrectomy (PPG) is a function-preserving surgery for the treatment of early gastric cancer (EGC) in the middle third of the stomach. According to the literature reports, PPG decreases the incidence of dumping syndrome, bile reflux, gallstone formation, and nutritional deficit compared with conventional distal gastrectomy (CDG). However, the debates about PPG have been dominated by the incomplete lymphadenectomy and oncological safety. We carried out a systematic review and meta-analysis to evaluate the pathological and oncological outcomes of PPG.
Methods
The protocol was registered in PROSPERO under number CRD42022304677. Databases including PubMed, Embase, Web of Science, and the Cochrane Register of Controlled Trials were searched before February 21, 2022. The outcomes included the pooled odds ratios (ORs) for dichotomous variables and weighted mean differences (WMDs) for continuous variables. For all outcomes, 95% confidence intervals (CIs) were calculated. Meta-analysis was performed using STATA software (Stata 14, Stata Corporation, Texas) and Review Manager 5.4.
Results
A total of 4500 patients from 16 studies were included. Compared with the CDG group, the PPG group had fewer lymph nodes harvested (WMD= −3.09; 95% CI −4.75 to −1.43; P < 0.001). Differences in the number of resected lymph nodes were observed at stations No. 5, No. 6, No. 9, and No. 11p. There were no differences in lymph node metastasis at each station. Shorter proximal resection margins (WMD = −0.554; 95% CI −0.999 to −0.108; P = 0.015) and distal resection margins (WMD = −1.569; 95% CI −3.132 to −0.007; P = 0.049) were observed in the PPG group. There were no significant differences in pathological T1a stage (OR = 0.99; 95% CI 0.80 to 1.23; P = 0.88), T1b stage (OR = 1.01; 95% CI 0.81 to 1.26; P = 0.88), N0 stage (OR = 0.97; 95% CI 0.63 to 1.48; P = 0.88), tumor size (WMD = −0.10; 95% CI −0.25 to 0.05; P = 0.187), differentiated carcinoma (OR = 1.04; 95% CI 0.74 to 1.47; P = 0.812) or signet ring cell carcinoma (OR = 1.22; 95% CI 0.90 to 1.64; P = 0.198). No significant differences were observed between the groups in terms of overall survival (HR = 0.63; 95% CI 0.24 to 1.67; P = 0.852) or recurrence-free survival (HR = 0.29; 95% CI 0.03 to 2.67; P = 0.900).
Conclusions
The meta-analysis of existing evidence demonstrated that the survival outcomes of PPG may be comparable to those of CDG. However, fewer lymph nodes at stations in No. 5, No. 6, No. 9, and No. 11p were harvested with PPG. We also found shorter proximal resection margins and distal resection margins for PPG, meaning more remnant stomachs would be preserved in PPG.
Collapse
|
39
|
Liu X, Wu X, Zhu J, Yagi K, Ajioka Y, Terai S, Mizuno K, Li H, Tuo B, Di L. Case report: New subtype of gastric adenocarcinoma arising from an H. pylori infection-negative stomach: Foveolar epithelium and mucous neck cell-mixed type adenocarcinoma. Front Oncol 2022; 12:970231. [PMID: 36106101 PMCID: PMC9464979 DOI: 10.3389/fonc.2022.970231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/01/2022] [Indexed: 11/26/2022] Open
Abstract
The characteristics of Helicobacter pylori (H. pylori) infection-negative gastric cancer have not been well documented because of its rarity, despite several types of H. pylori infection-negative gastric cancers being reported. In this report, we describe a case of early gastric cancer that developed without H. pylori infection with characteristic magnifying narrow-band imaging and novel histological findings. The difficulty in making an accurate diagnosis and differential diagnosis is highlighted, with the goal of providing more clinical experience for the diagnosis of H. pylori infection-negative gastric cancer.
Collapse
Affiliation(s)
- Xuemei Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xinglong Wu
- Department of Pathology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiaxing Zhu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Kazuyoshi Yagi
- Uonuma Institute of Community Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yoichi Ajioka
- Division of Molecular and Diagnostic Pathology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Shuji Terai
- Division of Gastroenterology & Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kenichi Mizuno
- Division of Gastroenterology & Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hongping Li
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- *Correspondence: Lianjun Di, ; Biguang Tuo,
| | - Lianjun Di
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- *Correspondence: Lianjun Di, ; Biguang Tuo,
| |
Collapse
|
40
|
Hatta W, Koike T, Uno K, Asano N, Masamune A. Management of Superficial Esophageal Squamous Cell Carcinoma and Early Gastric Cancer following Non-Curative Endoscopic Resection. Cancers (Basel) 2022; 14:3757. [PMID: 35954421 PMCID: PMC9367302 DOI: 10.3390/cancers14153757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/29/2022] [Accepted: 07/29/2022] [Indexed: 12/14/2022] Open
Abstract
According to the European and Japanese guidelines, additional treatment is recommended for cases of superficial esophageal squamous cell carcinoma (ESCC) and early gastric cancer (EGC) that do not meet the curability criteria for endoscopic resection (ER), i.e., non-curative ER, owing to the risk of lymph node metastasis (LNM). However, the rates of LNM in such cases were relatively low (e.g., 8% for EGC). Several recent advances have been made in this field. First, pathological risk stratification for metastatic recurrence following non-curative ER without additional treatment was developed for both superficial ESCC and EGC. Second, the pattern of metastatic recurrence and prognosis after recurrence following non-curative ER without additional treatment was found to be considerably different between superficial ESCC and EGC. Third, a combination of ER and selective chemoradiotherapy was developed as a minimally invasive treatment method for clinical T1b-SM ESCC. These findings may help clinicians decide the treatment strategy for patients following non-curative ER; however, for optimal therapeutic decision-making in such patients, it is also important to predict the prognosis other than SESCC or EGC and impaired quality of life. Thus, a novel algorithm that considers these factors, as well as metastatic recurrence, should be developed.
Collapse
Affiliation(s)
| | | | | | | | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan; (W.H.); (T.K.); (K.U.); (N.A.)
| |
Collapse
|
41
|
Zhou M, Dong J, Huang J, Ye W, Zheng Z, Huang K, Pan Y, Cen J, Liang Y, Shu G, Ye S, Lu X, Zhang J. Chitosan-Gelatin-EGCG Nanoparticle-Meditated LncRNA TMEM44-AS1 Silencing to Activate the P53 Signaling Pathway for the Synergistic Reversal of 5-FU Resistance in Gastric Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105077. [PMID: 35717675 PMCID: PMC9353463 DOI: 10.1002/advs.202105077] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/31/2022] [Indexed: 05/16/2023]
Abstract
Chemoresistance is one of the leading causes of therapeutic failure in gastric cancer (GC) treatment. Recent studies have shown lncRNAs play pivotal roles in regulating GC chemoresistance. Nanocarriers delivery of small interfering RNAs (siRNAs) to silence cancer-related genes has become a novel approach to cancer treatment research. However, finding target genes and developing nanosystems capable of selectively delivering siRNAs for disease treatment remains a challenge. In this study, a novel lncRNA TMEM44-AS1 that is related to 5-FU resistance is identified. TMEM44-AS1 has the ability to bind to and sponge miR-2355-5p, resulting in the upregulated PPP1R13L expression and P53 pathway inhibition. Next, a new nanocarrier called chitosan-gelatin-EGCG (CGE) is developed, which has a higher gene silencing efficiency than lipo2000, to aid in the delivery of a si-TMEM44-AS1 can efficiently silence TMEM44-AS1 expression to synergistically reverse 5-FU resistance in GC, leading to a markedly enhanced 5-FU therapeutic effect in a xenograft mouse model of GC. These findings indicate that TMEM44-AS1 may estimate 5-FU therapy outcome among GC cases, and that systemic si-TMEM44-AS1 delivery combined with 5-FU therapy is significant in the treatment of patients with recurrent GC.
Collapse
MESH Headings
- Animals
- Antimetabolites, Antineoplastic/pharmacology
- Antimetabolites, Antineoplastic/therapeutic use
- Antineoplastic Agents/metabolism
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Catechin/analogs & derivatives
- Catechin/pharmacology
- Catechin/therapeutic use
- Cell Line, Tumor
- Chitosan/pharmacology
- Chitosan/therapeutic use
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/physiology
- Fluorouracil/pharmacology
- Fluorouracil/therapeutic use
- Gelatin/pharmacology
- Gelatin/therapeutic use
- Gene Expression Regulation, Neoplastic
- Gene Silencing/drug effects
- Gene Silencing/physiology
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- MicroRNAs/genetics
- Nanoparticles/therapeutic use
- RNA/genetics
- RNA/metabolism
- RNA, Antisense/genetics
- RNA, Antisense/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction/genetics
- Stomach Neoplasms/drug therapy
- Stomach Neoplasms/genetics
- Stomach Neoplasms/metabolism
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Mi Zhou
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Jiaqi Dong
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Junqing Huang
- Guangzhou Key Laboratory of Formula‐Pattern of Traditional Chinese MedicineFormula‐Pattern Research CenterSchool of Traditional Chinese MedicineJinan UniversityGuangzhou510632P. R. China
| | - Wen Ye
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Zhousan Zheng
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Kangbo Huang
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Yihui Pan
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Junjie Cen
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Yanping Liang
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Guannan Shu
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Sheng Ye
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Xuanxuan Lu
- Department of Food Science and EngineeringJinan UniversityGuangzhou510632P. R. China
| | - Jiaxing Zhang
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| |
Collapse
|
42
|
Lee YH, Chan WH, Lai YC, Chen AH, Chen CM. Gastric hydrodistension CT versus CT without gastric distension in preoperative TN staging of gastric carcinoma: analysis of single-center cancer registry. Sci Rep 2022; 12:11321. [PMID: 35790760 PMCID: PMC9256680 DOI: 10.1038/s41598-022-15619-3] [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] [Received: 02/23/2022] [Accepted: 06/27/2022] [Indexed: 11/09/2022] Open
Abstract
Accurate staging of gastric cancer is essential for the selection and optimization of therapy. Hydrodistension of the stomach is recommended to improve the accuracy of preoperative staging with contrast-enhanced multidetector computed tomography (MDCT). This study compares the performance of contrast-enhanced gastric water distension versus a nondistension MDCT protocol for T and N staging and serosal invasion in comparison to surgical histopathology. After propensity score matching, 86 patients in each group were included for analysis. The overall accuracy of distension versus nondistension group in T staging was 45% (95% CI 35-56) and 55% (95% CI 44-65), respectively (p = 0.29). There was no difference in the sensitivity and specificity in individual T staging and assessment of serosal invasion (all p > 0.41). Individual stage concordance with pathology was not significantly different (all p > 0.41). The overall accuracy of N staging was the same for distension and nondistension groups (51% [95% CI 40-62]). The majority of N0 staging (78-81%) were correctly staged, whereas N3 staging cases (63-68%) were predominantly understaged. In summary, there was no significant difference in the diagnostic performance of individual TN staging and assessment of serosal invasion using MDCT with or without gastric water distension.
Collapse
Affiliation(s)
- Yu-Hsien Lee
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, and Chang Gung University College of Medicine, 5 Fuxing Street, Guishan District, Taoyuan, Taiwan
| | - Wen-Hui Chan
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, and Chang Gung University College of Medicine, 5 Fuxing Street, Guishan District, Taoyuan, Taiwan
| | - Ying-Chieh Lai
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, and Chang Gung University College of Medicine, 5 Fuxing Street, Guishan District, Taoyuan, Taiwan
| | - An-Hsin Chen
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, and Chang Gung University College of Medicine, 5 Fuxing Street, Guishan District, Taoyuan, Taiwan
| | - Chien-Ming Chen
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, and Chang Gung University College of Medicine, 5 Fuxing Street, Guishan District, Taoyuan, Taiwan.
| |
Collapse
|
43
|
Gu R, Xia Y, Li P, Zou D, Lu K, Ren L, Zhang H, Sun Z. Ferroptosis and its Role in Gastric Cancer. Front Cell Dev Biol 2022; 10:860344. [PMID: 35846356 PMCID: PMC9280052 DOI: 10.3389/fcell.2022.860344] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/28/2022] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer (GC) is the fifth most common cancer and the third leading cause of cancer-related deaths worldwide. Currently, surgery is the treatment of choice for GC. However, the associated expenses and post-surgical pain impose a huge burden on these patients. Furthermore, disease recurrence is also very common in GC patients, thus necessitating the discovery and development of other potential treatment options. A growing body of knowledge about ferroptosis in different cancer types provides a new perspective in cancer therapeutics. Ferroptosis is an iron-dependent form of cell death. It is characterized by intracellular lipid peroxide accumulation and redox imbalance. In this review, we summarized the current findings of ferroptosis regulation in GC. We also tackled on the action of different potential drugs and genes in inducing ferroptosis for treating GC and solving drug resistance. Furthermore, we also explored the relationship between ferroptosis and the tumor microenvironment in GC. Finally, we discussed areas for future studies on the role of ferroptosis in GC to accelerate the clinical utility of ferroptosis induction as a treatment strategy for GC.
Collapse
Affiliation(s)
- Renjun Gu
- Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Provincial Second Chinese Medicine Hospital, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yawen Xia
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China
| | - Pengfei Li
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Defang Zou
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China
| | - Keqin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lang Ren
- Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Provincial Second Chinese Medicine Hospital, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongru Zhang
- School of Basic Medical Sciences, Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Hongru Zhang, ; Zhiguang Sun,
| | - Zhiguang Sun
- Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Provincial Second Chinese Medicine Hospital, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Hongru Zhang, ; Zhiguang Sun,
| |
Collapse
|
44
|
Na JE, Lee YC, Kim TJ, Lee H, Won HH, Min YW, Min BH, Lee JH, Rhee PL, Kim JJ. Utility of a deep learning model and a clinical model for predicting bleeding after endoscopic submucosal dissection in patients with early gastric cancer. World J Gastroenterol 2022; 28:2721-2732. [PMID: 35979158 PMCID: PMC9260866 DOI: 10.3748/wjg.v28.i24.2721] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 03/25/2022] [Accepted: 05/08/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bleeding is one of the major complications after endoscopic submucosal dissection (ESD) in early gastric cancer (EGC) patients. There are limited studies on estimating the bleeding risk after ESD using an artificial intelligence system.
AIM To derivate and verify the performance of the deep learning model and the clinical model for predicting bleeding risk after ESD in EGC patients.
METHODS Patients with EGC who underwent ESD between January 2010 and June 2020 at the Samsung Medical Center were enrolled, and post-ESD bleeding (PEB) was investigated retrospectively. We split the entire cohort into a development set (80%) and a validation set (20%). The deep learning and clinical model were built on the development set and tested in the validation set. The performance of the deep learning model and the clinical model were compared using the area under the curve and the stratification of bleeding risk after ESD.
RESULTS A total of 5629 patients were included, and PEB occurred in 325 patients. The area under the curve for predicting PEB was 0.71 (95% confidence interval: 0.63-0.78) in the deep learning model and 0.70 (95% confidence interval: 0.62-0.77) in the clinical model, without significant difference (P = 0.730). The patients expected to the low- (< 5%), intermediate- (≥ 5%, < 9%), and high-risk (≥ 9%) categories were observed with actual bleeding rate of 2.2%, 3.9%, and 11.6%, respectively, in the deep learning model; 4.0%, 8.8%, and 18.2%, respectively, in the clinical model.
CONCLUSION A deep learning model can predict and stratify the bleeding risk after ESD in patients with EGC.
Collapse
Affiliation(s)
- Ji Eun Na
- Department of Internal Medicine, Inje University Haeundae Paik Hospital, Busan 48108, South Korea
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Yeong Chan Lee
- Department of Digital Health, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University, Seoul 06351, South Korea
| | - Tae Jun Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Hyuk Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Hong-Hee Won
- Department of Digital Health, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University, Seoul 06351, South Korea
| | - Yang Won Min
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Byung-Hoon Min
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Jun Haeng Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Poong-Lyul Rhee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Jae J Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| |
Collapse
|
45
|
Isomoto H, Suzuki H. Experimental and translational research in gastrointestinal endoscopy, the Japan Gastroenterological Endoscopy Society and perspective. Dig Endosc 2022; 34 Suppl 2:129-131. [PMID: 34558127 DOI: 10.1111/den.14129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 02/08/2023]
Affiliation(s)
- Hajime Isomoto
- Division of Gastroenterology and Nephrology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Hidekazu Suzuki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tokai University School of Medicine, Kanagawa, Japan
| |
Collapse
|
46
|
LRG1 mediated by ATF3 promotes growth and angiogenesis of gastric cancer by regulating the SRC/STAT3/VEGFA pathway. Gastric Cancer 2022; 25:527-541. [PMID: 35094168 DOI: 10.1007/s10120-022-01279-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/15/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Increasing evidence indicates that leucine-rich-alpha-2-glycoprotein 1 (LRG1) is associated with multiple malignancies, but whether it participates in gastric cancer (GC) angiogenesis remains unclear. METHODS The expression levels of LRG1 were assessed in GC samples. Endothelial tube formation analysis, HUVEC migration assay, chorioallantoic membrane assay (CAM), and xenograft tumor model were used to investigate the effect of LRG1 on angiogenesis in gastric cancer. The involvement of activating transcription factor 3 (ATF3) was analyzed by chromatin immunoprecipitation (ChIP) and dual-luciferase reporter assay. Western blot and enzyme-linked immunosorbent assay were performed to measure the SRC/STAT3/VEGFA pathway. RESULTS LRG1 was overexpressed in GC tissues and associated with cancer angiogenesis. In addition, LRG1 markedly promoted GC cell proliferation in vitro and in vivo. Moreover, overexpression of LRG1 could stimulate GC angiogenesis in vitro and in vivo. Then, we identified ATF3 promotes the transcription of LRG1 and is a positive regulator of angiogenesis. Additionally, LRG1 could activate VEGFA expression via the SRC/STAT3/ VEGFA pathway in GC cells, thus contributing to the angiogenesis of GC. CONCLUSIONS The present study suggests LRG1 plays a crucial role in the regulation of angiogenesis in GC and could be a potential therapeutic target for GC.
Collapse
|
47
|
Abe S, Sekiguchi M. It is time to tailor endoscopic resection for early gastric cancer: Evaluate not only lesion but also patient. Dig Endosc 2022; 34:826-827. [PMID: 35352401 DOI: 10.1111/den.14271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Seiichiro Abe
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Japan
| | - Masau Sekiguchi
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
48
|
Cai F, Dong Y, Wang P, Zhang L, Yang Y, Liu Y, Wang X, Zhang R, Liang H, Sun Y, Deng J. Risk assessment of lymph node metastasis in early gastric cancer: Establishment and validation of a Seven-point scoring model. Surgery 2022; 171:1273-1280. [PMID: 34865863 DOI: 10.1016/j.surg.2021.10.049] [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: 04/20/2021] [Revised: 10/10/2021] [Accepted: 10/25/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Treatment options for early gastric cancer have evolved toward achieving accurate evaluation of lymph node metastasis. This study aimed to investigate risk factors of lymph node metastasis in patients with early gastric cancer and establish a risk score model to guide the selection of optimal treatment. METHODS The clinicopathological characteristics of 351 patients with early gastric cancer from January 2016 to December 2018 were reviewed retrospectively. On the basis of the independent risk factors determined by multivariate binary logistic regression analysis, we established a risk score model for predicting lymph node metastasis and then verified it. The receiver operating characteristic curves were plotted using the test and validation sets. The area under the receiver operating characteristic curve was used to assess the discriminant ability of the model. RESULTS Lymph node metastasis was observed in 10.5% (37/351) of early gastric cancer cases. Patients with early gastric cancer were grouped based on the independent risk factors for lymph node metastasis (tumor size, depth, histological type, and lymphovascular involvement) determined by multivariate analysis. A 7-point risk score model was established to predict the risk of lymph node metastasis. The area under the receiver operating characteristic curve in the development and validation sets were 0.839 (95% confidence interval, 0.769%-0.910%) and 0.820 (95% confidence interval, 0.711%-0.930%), respectively. CONCLUSION A feasible risk score model for lymph node metastasis was established to guide the optimal treatment of patients with early gastric cancer early gastric cancer.
Collapse
Affiliation(s)
- Fenglin Cai
- Department of Gastroenterology, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P.R. China
| | - Yinping Dong
- Department of Gastroenterology, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P.R. China
| | - Pengliang Wang
- Department of Gastroenterology, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P.R. China
| | - Li Zhang
- Department of Gastroenterology, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P.R. China
| | - Yang Yang
- Department of Anesthesiology, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P.R. China
| | - Yong Liu
- Department of Gastroenterology, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P.R. China
| | - Xuejun Wang
- Department of Gastroenterology, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P.R. China
| | - Rupeng Zhang
- Department of Gastroenterology, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P.R. China
| | - Han Liang
- Department of Gastroenterology, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P.R. China
| | - Yan Sun
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P.R. China
| | - Jingyu Deng
- Department of Gastroenterology, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, P.R. China.
| |
Collapse
|
49
|
Ogata Y, Hatta W, Ohara Y, Koike T, Abe H, Saito M, Jin X, Kanno T, Uno K, Asano N, Imatani A, Yamamura A, Tanaka N, Kamei T, Unno M, Nakamura T, Nakaya N, Masamune A. Predictors of early and late mortality after the treatment for early gastric cancers. Dig Endosc 2022; 34:816-825. [PMID: 34662456 DOI: 10.1111/den.14172] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/15/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Although many patients with early gastric cancers (EGCs) die of non-gastric cancer-related causes, the association of the risk categories of lymph node metastasis (LNM) with all-cause mortality remains unclear. We aimed to clarify the predictors of early and late mortality, separately. METHODS Patients with endoscopic resection or gastrectomy for EGCs between 2003 and 2017 were retrospectively enrolled. We analyzed predictors for early and late mortality, including risk categories of LNM, treatment method, and nine non-cancer-related indices, separately, with a cut-off value of 3 years. RESULTS We enrolled 1439 patients with a median follow-up period of 79 months. The 5-year overall survival rate was 86.8%. In the multivariate Cox analysis, the most important predictors for early and late mortality were age ≥85 years (hazard ratio [HR] 2.88 and 4.54, respectively) and Eastern Cooperative Oncology Group Performance Status ≥2 (HR 3.00 and 4.19, respectively). Charlson comorbidity index ≥2 (HR 2.76 and 1.99, respectively), American Society of Anesthesiologists Physical Status ≥3 (HR 2.35 and 1.79, respectively), and C-reactive protein/albumin ratio ≥0.028 (HR 2.30 and 1.58, respectively) were also predictors for both early and late mortality. Male (HR 2.26), intermediate- (HR 2.12)/high-risk (HR 1.85) of LNM in eCura system, and sarcopenia evaluated by the psoas muscle mass index (HR 1.70) were predictors for early mortality. CONCLUSION The combined assessment of multiple predictors might help to predict early and/or late mortality in patients with EGCs. The eCura system was associated with early mortality.
Collapse
Affiliation(s)
- Yohei Ogata
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Waku Hatta
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Yuki Ohara
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Tomoyuki Koike
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Hiroko Abe
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Masahiro Saito
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Xiaoyi Jin
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Takeshi Kanno
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Kaname Uno
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Naoki Asano
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Akira Imatani
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Akihiro Yamamura
- Department of Surgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Naoki Tanaka
- Department of Surgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Takashi Kamei
- Department of Surgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Tomohiro Nakamura
- Department of, Health Record Informatics, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan
| | - Naoki Nakaya
- Department of, Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| |
Collapse
|
50
|
Jiang J, Ye G, Wang J, Xu X, Zhang K, Wang S. The Comparison of Short- and Long-Term Outcomes for Laparoscopic Versus Open Gastrectomy for Patients With Advanced Gastric Cancer: A Meta-Analysis of Randomized Controlled Trials. Front Oncol 2022; 12:844803. [PMID: 35449576 PMCID: PMC9016843 DOI: 10.3389/fonc.2022.844803] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/14/2022] [Indexed: 11/25/2022] Open
Abstract
Objectives The effect of laparoscopic gastrectomy (LG) for the treatment of advanced gastric cancer (AGC) is still controversial. The aim of this meta-analysis was to contrast the short- and long-term outcomes of laparoscopic versus conventional open gastrectomy (OG) for patients with AGC. Methods Databases including PubMed, Embase, Scopus, and Cochrane Library were systematically searched until December 2021 for randomized controlled trial-enrolled patients undergoing LG or OG for the treatment of AGC. Short-term outcomes were overall postoperative complications, anastomotic leakage, number of retrieved lymph node, surgical time, blood loss, length of hospital stay, and short-term mortality. Long-term outcomes were survival rates at 1, 3, and 5 years. Results A total of 12 trials involving 4,101 patients (2,059 in LG group, 2,042 in OG group) were included. No effect on overall postoperative complications (OR 0.84, 95% CI 0.67 to 1.05, p = 0.12, I2 = 34%) and anastomotic leakage (OR 1.26, 95% CI 0.82 to 1.95, p = 0.30, I2 = 0%) was found. Compared with the open approach, patients receiving LG had fewer blood loss (MD -54.38, 95% CI -78.09 to -30.67, p < 0.00001, I2 = 90%) and shorter length of hospital stay (MD -1.25, 95% CI -2.08 to -0.42, p = 0.003, I2 = 86%). However, the LG was associated with a lower number of retrieved lymph nodes (MD -1.02, 95% CI -1.77 to -0.27, p = 0.008, I2 = 0%) and longer surgical time (MD 40.87, 95% CI 20.37 to 54.44, p < 0.00001, I2 = 94%). Furthermore, there were no differences between LG and OG groups in short-term mortality and survival rate at 1, 3, and 5 years. Conclusions LG offers improved short-term outcomes including shorter hospital stays and fewer blood loss, with comparable postoperative complications, short-term mortality, and survival rate at 1, 3, and 5 years when compared to the open approach. Our results support the implementation of LG in patients with AGC. Systematic Review Registration PROSPERO (CRD 42021297141).
Collapse
Affiliation(s)
- Jinyan Jiang
- Department of Nursing, Lishui People's Hospital, Lishui, China
| | - Guanxiong Ye
- Department of General Surgery, Lishui People's Hospital, Lishui, China
| | - Jun Wang
- Department of General Surgery, Lishui People's Hospital, Lishui, China
| | - Xiaoya Xu
- Department of General Surgery, Lishui People's Hospital, Lishui, China
| | - Kai Zhang
- Department of Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shi Wang
- Department of General Surgery, Lishui People's Hospital, Lishui, China
| |
Collapse
|