1
|
You Y, Liang Y, Chen L, Li Z, Gao B, Wang X, Yuan M, Xue Y, Liu Y, Gao J. Radiomics analysis of dual-energy CT-derived iodine maps for differentiating between T1/2 and T3/4a in gastric cancer: A multicenter study. Eur J Radiol 2025; 186:112054. [PMID: 40121898 DOI: 10.1016/j.ejrad.2025.112054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 02/23/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
OBJECTIVE To investigate the value of radiomic analysis of dual-energy CT (DECT)-derived iodine maps (IMs) for the differentiation between T1/2 and T3/4a stage tumors in gastric cancer (GC). METHODS A total of 263 patients who received upfront surgery and were pathologically confirmed with gastric adenocarcinoma were enrolled in this study. Dual-phase enhanced CT scans with gemstone spectral imaging (GSI) mode were performed within two weeks before surgery. 151 patients were retrospectively collected for the training (n = 105) and validation (n = 46) cohorts, and 112 patients were prospectively collected for the external test1 (n = 68) and external test2 (n = 44) cohorts. According to the postoperative pathological T stage, patients were classified into T1/2 and T3/4a stage groups. Clinical characteristics were recorded and quantitative iodine concentration (IC) of tumors was measured. Radiomics features were extracted from the venous phase (VP) IMs by three-dimensional region of interest (3D-ROI) segmentation. Feature selection was performed using the least absolute shrinkage and selection operator. Four machine learning algorithms, including random forest, logistic regression, naive Bayes, and support vector machine, were used to construct radiomics models. Finally, the most valuable clinical characteristics, DECT parameters, and the best radiomics model were combined to build a nomogram. The diagnostic performance of nomogram was evaluated by the area under receiver operating characteristic curve (AUC), calibration curve, and decision curve. RESULTS The nomogram combined tumor clinical T stage (cT), tumor thickness, venous-phase iodine concentration (ICVP), normalized arterial-phase iodine concentration (nICAP), and Radscore (derived from logistic regression model). This integrated model demonstrated favorable performance in the differentiation between T1/2 and T3/4a stage tumors in GC, with AUCs of 0.892 (95 %CI: 0.829-0.956), 0.846 (95 %CI: 0.734-0.958), 0.894 (95 %CI: 0.818-0.970) and 0.821 (95 %CI: 0.689-0.952) observed for the training, validation, external test 1, and external test 2 cohorts, respectively. Hosmer-Lemeshow test showed a good fit (all P > 0.05). Decision curves confirmed that the nomogram provided more net clinical benefit than the default simple strategy over a wide range of threshold probabilities. CONCLUSION We have developed and validated a multidimensional personalized nomogram that integrates a radiomics model based on DECT-derived IMs, DECT quantitative parameters, and traditional clinical features. The proposed model demonstrated favorable performance in preoperative identification of T3/4a stage tumors in GC.
Collapse
Affiliation(s)
- Yaru You
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Imaging Diagnosis and Treatment for Digestive System Tumor, Zhengzhou 450052, China
| | - Yan Liang
- Department of Radiology, Sanmenxia Central Hospital, Sanmenxia 472100, China
| | - Lihong Chen
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Zhanzhan Li
- Department of Radiology, Sanmenxia Central Hospital, Sanmenxia 472100, China
| | - Beijun Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Imaging Diagnosis and Treatment for Digestive System Tumor, Zhengzhou 450052, China
| | - Xiangxiang Wang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Imaging Diagnosis and Treatment for Digestive System Tumor, Zhengzhou 450052, China
| | - Mengchen Yuan
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Imaging Diagnosis and Treatment for Digestive System Tumor, Zhengzhou 450052, China
| | - Yunjing Xue
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, China.
| | - Yiyang Liu
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Imaging Diagnosis and Treatment for Digestive System Tumor, Zhengzhou 450052, China.
| | - Jianbo Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Imaging Diagnosis and Treatment for Digestive System Tumor, Zhengzhou 450052, China.
| |
Collapse
|
2
|
Lv M, Hui X, Yang X, Li S, Mao Z, Zhang X, Yang K. Comparison of the diagnostic accuracy of enhanced-CT and double contrast-enhanced ultrasound for preoperative T-staging of gastric cancer: a meta-analysis. Cancer Imaging 2025; 25:48. [PMID: 40181411 PMCID: PMC11966938 DOI: 10.1186/s40644-025-00861-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 03/11/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Accurate preoperative staging of gastric cancer (GC) depends on effective diagnostic methods. Enhanced computed tomography (enhanced-CT) is a widely used and reliable preoperative assessment tool for GC, Double Contrast-Enhanced Ultrasound (DCEUS) can display the structure and layers of the gastric wall more accurately, and has high sensitivity (SE) and specificity (SP). OBJECTIVE The present study aims to conduct a comprehensive meta-analysis comparing the preoperative T-staging accuracy of DCEUS and enhanced-CT. METHODS A systematic literature search was conducted across PubMed, Embase, Web of Science, and Cochrane Library to identify eligible articles from inception to February 19, 2024. The study included both prospective and retrospective studies involving patients with GC who underwent DCEUS or enhanced-CT. This encompassed studies utilizing comparative diagnostic test accuracy (CDTA) with both DCEUS and enhanced-CT, as well as studies employing single diagnostic test accuracy (SDTA) with either DCEUS or enhanced-CT alone. Risk of bias was assessed using the Quality Assessment Of Diagnostic Accuracy Studies-C (QUADAS-C) and Assessment Of Diagnostic Accuracy Studies-2 (QUADAS-2) tool. The quality of evidence for each outcome was assessed using GRADE (Grading of Recommendations Assessment, Development and Evaluation). RESULTS A total of 39 studies involving 6,374 patients were included in this meta-analysis. Among these, 3 studies (319 patients) directly compared dynamic contrast-enhanced ultrasound (DCEUS) and enhanced computed tomography (CT), while 31 studies (5,180 patients) evaluated enhanced CT alone, and 5 studies (875 patients) assessed DCEUS alone. For the direct comparison studies (CDTA), DCEUS demonstrated higher sensitivity (SE) and specificity (SP) for T1-T4 staging compared to enhanced CT, with moderate to low certainty of evidence. Specifically, DCEUS showed superior performance in detecting early-stage (T1) and advanced-stage (T4) tumors. Enhanced CT, while effective, had lower sensitivity across all stages, particularly for T1 tumors. In the single-modality studies (SDTA), DCEUS consistently showed higher sensitivity for T2-T4 staging compared to enhanced CT, with comparable specificity. However, the certainty of evidence for indirect comparisons was very low, highlighting the need for further high-quality comparative studies. Overall, DCEUS appears to be a promising modality for gastric cancer T staging, particularly for early-stage detection, but the limited number of direct comparative studies underscores the need for more robust evidence. CONCLUSION Current evidence indicates that DCEUS significantly outperforms enhanced-CT in terms of SE and diagnostic accuracy for preoperative T-staging of GC, while maintaining comparable SP. However, these findings require further validation through rigorous studies with larger sample sizes and improved methodological quality.
Collapse
Affiliation(s)
- MingYue Lv
- The First Clinical Medical College of Gansu University of Traditional Chinese Medicine, Lanzhou, 730000, China
- Evidence-Based Medicine Centre, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, China
| | - Xu Hui
- Evidence-Based Medicine Centre, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, China
- Centre for Evidence-Based Social Science/Center for Health Technology Assessment, School of Public Health, Lanzhou University, Lanzhou, 730000, China
- Gansu Key Laboratory of Evidence-Based Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Xin Yang
- The First Clinical Medical College of Gansu University of Traditional Chinese Medicine, Lanzhou, 730000, China
- Evidence-Based Medicine Centre, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, China
| | - SuSu Li
- The First Clinical Medical College of Gansu University of Traditional Chinese Medicine, Lanzhou, 730000, China
| | - ZhiGuo Mao
- The First Clinical Medical College of Gansu University of Traditional Chinese Medicine, Lanzhou, 730000, China
| | - XinHua Zhang
- Department of Ultrasound Medicine, Gansu University of Chinese Medicine, Gansu Provincial People'S Hospital, No. 204, Donggang West Road, Chengguan District, Lanzhou, 730000, China.
| | - KeHu Yang
- Evidence-Based Medicine Centre, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, China.
- Centre for Evidence-Based Social Science/Center for Health Technology Assessment, School of Public Health, Lanzhou University, Lanzhou, 730000, China.
- Gansu Key Laboratory of Evidence-Based Medicine, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
3
|
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
|
4
|
Ofluoğlu CB, Aydın IC, Mülküt F, Uzun O, Senger AS, Gülmez S, Duman U, Polat E, Duman M. Diagnostic Efficacy of Staging Laparoscopy Compared to CT and PET-CT in Gastric Cancer: A Retrospective Cohort Analysis. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:2079. [PMID: 39768958 PMCID: PMC11677282 DOI: 10.3390/medicina60122079] [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: 11/06/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
Background and Objectives: This study aimed to assess the diagnostic accuracy and prognostic significance of staging laparoscopy (SL) compared to computed tomography (CT) and positron emission tomography-computed tomography (PET-CT) in gastric cancer staging. We evaluated the ability of SL to detect occult peritoneal metastases and influence of SL on survival outcomes across cancer stages and treatment approaches. Materials and Methods: In this retrospective cohort study, 95 patients with gastric cancer underwent preoperative assessment using CT, PET-CT, and SL between 2018 and 2024. Diagnostic performance metrics were calculated for SL, CT, and PET/CT across the local, locally advanced, and metastatic stages. Survival outcomes were analyzed using Kaplan-Meier curves, and comparisons were made using log-rank tests. A multivariable Cox proportional hazards model incorporating interaction terms was used to determine the independent prognostic factors affecting survival, focusing on SL findings and cytology results. Results: The cohort comprised 75 males (78.9%) and 20 females (21.1%), with a mean age of 57.4 ± 10.1 years. The tumor location distribution was predominant in the corpus (31.1%) and cardia. Tumor staging revealed that 48.1% were classified as T3 and 28.8% as T4, respectively. Diagnostic accuracy analysis showed that SL outperformed CT and PET-CT in detecting peritoneal metastasis across all stages. Specifically, SL demonstrated a sensitivity and specificity of 85% and 95% for local disease, 92% and 80% for locally advanced disease, and 95% and 99% for metastatic disease, significantly exceeding those of CT and PET-CT. Patients with SL findings had a median overall survival (OS) of 30 months compared with 20 months for those assessed only with CT and PET-CT (p < 0.05). The stage-specific median OS for SL patients was 40 months in the local, 25 months in the locally advanced (p < 0.05), and 15 months in the metastatic disease (p < 0.01) groups, indicating significant survival benefits. Multivariable Cox regression identified SL findings as an independent factor associated with reduced mortality risk (HR: 0.70, 95% CI: 0.50-0.90, p < 0.01), while positive cytology findings predicted poorer survival (HR: 1.80, 95% CI: 1.25-2.60, p < 0.01). Interaction terms revealed that SL yielded greater survival benefits in patients with metastatic disease (hazard ratio [HR]: 0.60, p < 0.01) and those undergoing systemic therapy (HR: 0.75, p = 0.04). Conclusions: SL provides superior diagnostic accuracy and prognostic information for advanced gastric cancer staging compared with CT and PET-CT. By accurately detecting peritoneal metastasis, SL aids in optimizing treatment plans, particularly in advanced stages, thus potentially improving patient outcomes.
Collapse
Affiliation(s)
- Cem Batuhan Ofluoğlu
- Department of Gastroenterology Surgery, Sancaktepe Sehit Prof. Dr. İlhan Varank Training and Research Hospital, University of Health Sciences, 34147 Istanbul, Turkey
| | - Isa Caner Aydın
- Gastroenterology Surgery Clınıc, Zonguldak Atatürk State Hospital, Health Mınıstry of Turkısh Republıc, 34147 Zonguldak, Turkey;
| | - Fırat Mülküt
- Department of General Surgery, Sancaktepe Sehit Prof. Dr. İlhan Varank Training and Research Hospital, University of Health Sciences, 34147 Istanbul, Turkey;
| | - Orhan Uzun
- Gastroenterology Surgery Clınıc, Koşuyolu Hıgh Specıalızatıon Educatıon and Research Hospıtal, University of Health Sciences, 34147 Istanbul, Turkey; (O.U.); (A.S.S.); (S.G.); (E.P.); (M.D.)
| | - Aziz Serkan Senger
- Gastroenterology Surgery Clınıc, Koşuyolu Hıgh Specıalızatıon Educatıon and Research Hospıtal, University of Health Sciences, 34147 Istanbul, Turkey; (O.U.); (A.S.S.); (S.G.); (E.P.); (M.D.)
| | - Selçuk Gülmez
- Gastroenterology Surgery Clınıc, Koşuyolu Hıgh Specıalızatıon Educatıon and Research Hospıtal, University of Health Sciences, 34147 Istanbul, Turkey; (O.U.); (A.S.S.); (S.G.); (E.P.); (M.D.)
| | - Uğur Duman
- General Surgery Clınıc, Bursa Yüksek İhtisas Training and Research Hospital, University of Health Sciences, Bursa Provincial Health Directorate, 16290 Bursa, Turkey;
| | - Erdal Polat
- Gastroenterology Surgery Clınıc, Koşuyolu Hıgh Specıalızatıon Educatıon and Research Hospıtal, University of Health Sciences, 34147 Istanbul, Turkey; (O.U.); (A.S.S.); (S.G.); (E.P.); (M.D.)
| | - Mustafa Duman
- Gastroenterology Surgery Clınıc, Koşuyolu Hıgh Specıalızatıon Educatıon and Research Hospıtal, University of Health Sciences, 34147 Istanbul, Turkey; (O.U.); (A.S.S.); (S.G.); (E.P.); (M.D.)
| |
Collapse
|
5
|
Bangolo A, Wadhwani N, Nagesh VK, Dey S, Tran HHV, Aguilar IK, Auda A, Sidiqui A, Menon A, Daoud D, Liu J, Pulipaka SP, George B, Furman F, Khan N, Plumptre A, Sekhon I, Lo A, Weissman S. Impact of artificial intelligence in the management of esophageal, gastric and colorectal malignancies. Artif Intell Gastrointest Endosc 2024; 5:90704. [DOI: 10.37126/aige.v5.i2.90704] [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: 12/12/2023] [Revised: 01/28/2024] [Accepted: 03/04/2024] [Indexed: 05/11/2024] Open
Abstract
The incidence of gastrointestinal malignancies has increased over the past decade at an alarming rate. Colorectal and gastric cancers are the third and fifth most commonly diagnosed cancers worldwide but are cited as the second and third leading causes of mortality. Early institution of appropriate therapy from timely diagnosis can optimize patient outcomes. Artificial intelligence (AI)-assisted diagnostic, prognostic, and therapeutic tools can assist in expeditious diagnosis, treatment planning/response prediction, and post-surgical prognostication. AI can intercept neoplastic lesions in their primordial stages, accurately flag suspicious and/or inconspicuous lesions with greater accuracy on radiologic, histopathological, and/or endoscopic analyses, and eliminate over-dependence on clinicians. AI-based models have shown to be on par, and sometimes even outperformed experienced gastroenterologists and radiologists. Convolutional neural networks (state-of-the-art deep learning models) are powerful computational models, invaluable to the field of precision oncology. These models not only reliably classify images, but also accurately predict response to chemotherapy, tumor recurrence, metastasis, and survival rates post-treatment. In this systematic review, we analyze the available evidence about the diagnostic, prognostic, and therapeutic utility of artificial intelligence in gastrointestinal oncology.
Collapse
Affiliation(s)
- Ayrton Bangolo
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Nikita Wadhwani
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Vignesh K Nagesh
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Shraboni Dey
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Hadrian Hoang-Vu Tran
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Izage Kianifar Aguilar
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Auda Auda
- Department of Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Aman Sidiqui
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Aiswarya Menon
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Deborah Daoud
- Department of Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - James Liu
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Sai Priyanka Pulipaka
- Department of Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Blessy George
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Flor Furman
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Nareeman Khan
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Adewale Plumptre
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Imranjot Sekhon
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Abraham Lo
- Department of Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Simcha Weissman
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| |
Collapse
|
6
|
Mithany RH, Shahid MH, Manasseh M, Saeed MT, Aslam S, Mohamed MS, Daniel N. Gastric Cancer: A Comprehensive Literature Review. Cureus 2024; 16:e55902. [PMID: 38595903 PMCID: PMC11003650 DOI: 10.7759/cureus.55902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2024] [Indexed: 04/11/2024] Open
Abstract
Gastric cancer stands as a significant global health concern, particularly prevalent in Eastern Asia, with high mortality rates urging urgent attention and research efforts. This article comprehensively explores the epidemiology, anatomy, risk factors, pathophysiology, clinical presentation, diagnosis, staging, treatment modalities, prevention strategies, and survival rates associated with gastric cancer. Notably, Helicobacter pylori infection, dietary choices, and intricate stomach anatomy play pivotal roles in disease development. Early detection, utilizing staging, grading, and genetic testing for personalized treatment approaches is emphasized. Treatment modalities encompass surgery, chemotherapy, radiation therapy, targeted therapy, and immunotherapy. Prevention strategies involve lifestyle changes, screening, and genetic counseling. Survival rates vary by stage, highlighting the need for individualized care. In conclusion, a collaborative global effort is essential to address the impact of gastric cancer and improve outcomes.
Collapse
Affiliation(s)
- Reda H Mithany
- Laparoscopic Colorectal Surgery, Kingston Hospital NHS Foundation Trust, Kingston Upon Thames, GBR
| | | | - Mina Manasseh
- General Surgery, Torbay and South Devon NHS Foundation Trust, Torquay, GBR
| | | | - Samana Aslam
- General Surgery, Lahore General Hospital, Lahore, PAK
| | | | - Nesma Daniel
- Medical Laboratory Science, Ain Shams University Specialized Hospital, Cairo, EGY
| |
Collapse
|
7
|
Sun N, Tan BB, Li Y. Nomogram model including LATS2 expression was constructed to predict the prognosis of advanced gastric cancer after surgery. World J Gastrointest Surg 2024; 16:518-528. [PMID: 38463354 PMCID: PMC10921222 DOI: 10.4240/wjgs.v16.i2.518] [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: 12/12/2023] [Revised: 01/13/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Gastric cancer is a leading cause of cancer-related deaths worldwide. Prognostic assessments are typically based on the tumor-node-metastasis (TNM) staging system, which does not account for the molecular heterogeneity of this disease. LATS2, a tumor suppressor gene involved in the Hippo signaling pathway, has been identified as a potential prognostic biomarker in gastric cancer. AIM To construct and validate a nomogram model that includes LATS2 expression to predict the survival prognosis of advanced gastric cancer patients following radical surgery, and compare its predictive performance with traditional TNM staging. METHODS A retrospective analysis of 245 advanced gastric cancer patients from the Fourth Hospital of Hebei Medical University was conducted. The patients were divided into a training group (171 patients) and a validation group (74 patients) to develop and test our prognostic model. The performance of the model was determined using C-indices, receiver operating characteristic curves, calibration plots, and decision curves. RESULTS The model demonstrated a high predictive accuracy with C-indices of 0.829 in the training set and 0.862 in the validation set. Area under the curve values for three-year and five-year survival prediction were significantly robust, suggesting an excellent discrimination ability. Calibration plots confirmed the high concordance between the predictions and actual survival outcomes. CONCLUSION We developed a nomogram model incorporating LATS2 expression, which significantly outperformed conventional TNM staging in predicting the prognosis of advanced gastric cancer patients postsurgery. This model may serve as a valuable tool for individualized patient management, allowing for more accurate stratification and improved clinical outcomes. Further validation in larger patient cohorts will be necessary to establish its generalizability and clinical utility.
Collapse
Affiliation(s)
- Nan Sun
- Department of Blood Transfusion, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hubei Province, China
| | - Bi-Bo Tan
- Third Department of General Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Yong Li
- Third Department of General Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| |
Collapse
|
8
|
Zhu Z, Gong H, Gu J, Dai Y, Yang C, Mao M, Song A, Feng F. Development and validation of a preoperative CT-based risk scoring system for predicting recurrence-free survival in patients undergoing curative surgery for gastric cancer. Eur J Radiol 2024; 171:111303. [PMID: 38215532 DOI: 10.1016/j.ejrad.2024.111303] [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: 11/11/2023] [Revised: 12/30/2023] [Accepted: 01/07/2024] [Indexed: 01/14/2024]
Abstract
PURPOSE The objective of this study was to establish and validate a preoperative risk scoring system that incorporated both clinical and computed tomography(CT) variables to predict recurrence-free survival (RFS) in gastric cancer(GC) patients who underwent curative resection. METHOD We retrospectively included consecutive patients with surgically confirmed GC who underwent preoperative CT scans between October 2017 and January 2022. Multivariate Cox regression analysis was employed in the derivation set to identify clinical and CT variables associated with RFS and to construct a risk score. This risk score was subsequently validated in an independent test set. RESULTS A total of 346 patients were included in the study, with 213 in the derivation set and 133 in the test set. Five variables, namely ctEMVI, ctBorrmann, visceral obesity, sarcopenia, and NLR, were independently associated with RFS. In the test set, the preoperative risk score exhibited a c-index of 0.741, which outperformed the predictive accuracy of pathological tumor staging (c-index of 0.673, p = 0.021) at various time points. The preoperative risk score effectively stratified patients into low and high-risk groups. CONCLUSION The developed preoperative risk scoring system demonstrated the ability to predict RFS following curative resection in GC patients.
Collapse
Affiliation(s)
- Zhengqi Zhu
- Radiology Department, Jiangsu Province Nantong City Cancer Hospital, Nantong 226300, Jiangsu Province, China
| | - Haipeng Gong
- Radiology Department, Jiangsu Province Nantong City Cancer Hospital, Nantong 226300, Jiangsu Province, China
| | - Jianan Gu
- Radiology Department, Jiangsu Province Nantong City Cancer Hospital, Nantong 226300, Jiangsu Province, China
| | - Yongfeng Dai
- Radiology Department, Jiangsu Province Nantong City Cancer Hospital, Nantong 226300, Jiangsu Province, China
| | - Chunyan Yang
- Radiology Department, Jiangsu Province Nantong City Cancer Hospital, Nantong 226300, Jiangsu Province, China
| | - Mimi Mao
- Radiology Department, Jiangsu Province Nantong City Cancer Hospital, Nantong 226300, Jiangsu Province, China
| | - Anyi Song
- Radiology Department, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Feng Feng
- Radiology Department, Jiangsu Province Nantong City Cancer Hospital, Nantong 226300, Jiangsu Province, China.
| |
Collapse
|
9
|
Wu J, Wang H, Yin X, Wang Y, Lu Z, Zhang J, Zhang Y, Xue Y. Normalization weighted combination scores re-evaluate TNM staging of gastric cancer: a retrospective cohort study based on a multicenter database. Int J Surg 2024; 110:11-22. [PMID: 38000074 PMCID: PMC10793834 DOI: 10.1097/js9.0000000000000726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/21/2023] [Indexed: 11/26/2023]
Abstract
BACKGROUND The pathological depth of tumor invasion (pT) and lymph node metastasis (pN) are critical independent prognostic factors for patients with gastric cancer (GC), representing effective methods for evaluating prognosis. In this study, the authors employed a normalization weight combination score to calculate the weight ratio of the pT stage and pN stage. Subsequently, the authors established a novel weighted TN (wTN) staging model based on these T and N weights, evaluating its prognostic capacity. METHODS This study utilized a training cohort from A Medical University Cancer Hospital and a validation cohort from the SEER database. Least absolute shrinkage and selection operator (LASSO) and Cox regression were employed to screen clinical characteristics. Multivariate linear regression and cluster analysis calculated the weight ratio of T stage and N stage in the training and validation cohorts, respectively, followed by re-staging. Prognostic value was evaluated using C-index, likelihood ratio, Wald, and Score tests for wTN stage and tumor-node-metastasis (TNM) stage. A nomogram model was developed, and accuracy was assessed using receiver operating characteristic curve (ROC), decision curve analysis (DCA), and restricted cubic spline (RCS) analyses. RESULTS LASSO was used for initial screening, selecting eight potential features for Cox analysis. Age, tumor size, metastasis lymph nodes (MLNs), and tumor location were confirmed as independent prognostic factors. wTN was calculated in the training and validation cohorts, and nomograms were established with the independent factors. N stage had a higher weight proportion than T stage in both cohorts (0.625/0.375 in training cohort, 0.556/0.444 in validation cohort). wTN outperformed the 8th TNM stage in C-index, likelihood ratio, Wald, and Score tests in the training cohort, with successful validation in the validation cohort. Stratified analysis of distinct pathological types further demonstrates that wTN staging exhibits superior prognostic performance. CONCLUSION The wTN staging model based on T stage and N stage weights has a good prognostic value for GC patients. The same conclusion was obtained in different pathological stratification.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yingwei Xue
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, People’s Republic of China
| |
Collapse
|
10
|
Liu H, Zhao KY. Application of CD34 expression combined with three-phase dynamic contrast-enhanced computed tomography scanning in preoperative staging of gastric cancer. World J Gastrointest Surg 2023; 15:2513-2524. [PMID: 38111775 PMCID: PMC10725531 DOI: 10.4240/wjgs.v15.i11.2513] [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: 09/19/2023] [Revised: 10/26/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Accurate preoperative staging of gastric cancer (GC), a common malignant tumor worldwide, is critical for appropriate treatment plans and prognosis. Dynamic three-phase enhanced computed tomography (CT) scanning for preoperative staging of GC has limitations in evaluating tumor angiogenesis. CD34, a marker on vascular endothelial cell surfaces, is promising in evaluating tumor angiogenesis. We explored the value of their combination for preoperative staging of GC to improve the efficacy and prognosis of patients with GC. AIM To explore the evaluation value of CD34 expression + dynamic three-phase enhanced CT scanning in preoperative staging of GC. METHODS Medical records of 106 patients with GC treated at the First People's Hospital of Lianyungang between February 2021 and January 2023 were retrospectively studied. All patients underwent three-phase dynamic contrast-enhanced CT scanning before surgery, and CD34 was detected in gastroscopic biopsy specimens. Using surgical and pathological results as the gold standard, the diagnostic results of three-phase dynamic contrast-enhanced CT scanning at different T and N stages were analyzed, and the expression of CD34-marked microvessel density (MVD) at different T and N stages was determined. The specificity and sensitivity of three-phase dynamic contrast-enhanced CT and CD34 in T and N staging were calculated; those of the combined diagnosis of the two were evaluated in parallel. Independent factors affecting lymph node metastasis were analyzed using multiple logistic regression. RESULTS The accuracy of three-phase dynamic contrast-enhanced CT scanning in diagnosing stages T1, T2, T3 and T4 were 68.00%, 75.00%, 79.41%, and 73.68%, respectively, and for diagnosing stages N0, N1, N2, and N3 were 75.68%, 74.07%, 85.00%, and 77.27%, respectively. CD34-marked MVD expression increased with increasing T and N stages. Specificity and sensitivity of three-phase dynamic contrast-enhanced CT in T staging were 86.79% and 88.68%; for N staging, 89.06% and 92.86%; for CD34 in T staging, 64.15% and 88.68%; and for CD34 in N staging, 84.38% and 78.57%, respectively. Specificity and sensitivity of joint diagnosis in T staging were 55.68% and 98.72%, and N staging were 75.15% and 98.47%, respectively, with the area under the curve for diagnosis improving accordingly. According to multivariate analysis, a longer tumor diameter, higher pathological T stage, lower differentiation degree, and higher expression of CD34-marked MVD were independent risk factors for lymph node metastasis in patients with GC. CONCLUSION With high accuracy in preoperatively determining the invasion depth and lymph node metastasis of GC, CD34 expression and three-phase dynamic contrast-enhanced CT can provide a reliable basis for surgical resection.
Collapse
Affiliation(s)
- Hua Liu
- Department of Pathology, The First People's Hospital of Lianyungang, Lianyungang 222000, Jiangsu Province, China
| | - Kang-Yan Zhao
- Department of Radiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Sciences, Xiangyang 441021, Hubei Province, China
| |
Collapse
|
11
|
Liu F, Wu X, Wang W, Chang J. A novel immunohistochemical score predicts the postoperative prognosis of gastric cancer patients. World J Surg Oncol 2023; 21:220. [PMID: 37491274 PMCID: PMC10369746 DOI: 10.1186/s12957-023-03113-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/12/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND AND AIM Immunohistochemistry indicators are increasingly being used to predict the survival prognosis of cancer patients after surgery. This study aimed to combine some markers to establish an immunohistochemical score (MSI-P53-Ki-67[MPK]) and stratify postoperative patients with gastric cancer according to the score. METHODS We used 245 patients who underwent surgery at one center as the training cohort and 111 patients from another center as the validation cohort. All patients were treated between January 2012 and June 2018. The training cohort was screened for prognostic factors, and MPK scores were established using univariate and multifactorial COX risk proportional models. Patients were prognostically stratified according to the MPK score after gastrectomy for gastric cancer. Overall survival (OS) and recurrence-free survival (RFS) rates were compared among low-, intermediate-, and high-risk groups using the Kaplan-Meier method, and survival curves were plotted. Finally, the MPK score was validated using the validation cohort. RESULTS In the training group, there were statistically significant differences in OS and RFS in the low, medium, and high-risk groups (P < 0.001). Thirty patients were in the high-risk group (12.2%). The median survival times of the three groups were 64.0, 44.0, and 23.0, respectively, and median times to recurrence were 54.0, 35.0, and 16.0 months, respectively. In the validation group, the prognosis in the three risk groups remained significantly different (P < 0.001). CONCLUSIONS The novel MPK score could effectively predict the postoperative OS and RFS of gastric cancer patients, risk-stratify postoperative patients, and identify postoperative high-risk patients for refined management.
Collapse
Affiliation(s)
- Feng Liu
- Department of Gastrointestinal Surgery, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu, 215300, People's Republic of China
| | - Xiaoyang Wu
- Department of Gastrointestinal Surgery, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu, 215300, People's Republic of China
| | - Weiping Wang
- Department of General Surgery, Kunshan Second People's Hospital, Suzhou, 215300, People's Republic of China
| | - Jun Chang
- Department of General Surgery, Kunshan Second People's Hospital, Suzhou, 215300, People's Republic of China.
| |
Collapse
|
12
|
Liang J, Jiang Y, Abboud Y, Gaddam S. Role of Endoscopy in Management of Upper Gastrointestinal Cancers. Diseases 2022; 11:diseases11010003. [PMID: 36648868 PMCID: PMC9844461 DOI: 10.3390/diseases11010003] [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: 09/21/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Upper gastrointestinal (GI) malignancy is a leading cause of cancer-related morbidity and mortality. Upper endoscopy has an established role in diagnosing and staging upper GI cancers, screening for pre-malignant lesions, and providing palliation in cases of advanced malignancy. New advances in endoscopic techniques and technology have improved diagnostic accuracy and increased the therapeutic potential of upper endoscopy. We aim to describe the different types of endoscopic technology used in cancer diagnosis, summarize the current guidelines for endoscopic diagnosis and treatment of malignant and pre-malignant lesions, and explore new potential roles for endoscopy in cancer therapy.
Collapse
|
13
|
Li J, Wang Y, Wang R, Gao JB, Qu JR. Spectral CT for preoperative prediction of lymphovascular invasion in resectable gastric cancer: With external prospective validation. Front Oncol 2022; 12:942425. [PMID: 36267965 PMCID: PMC9577143 DOI: 10.3389/fonc.2022.942425] [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: 06/20/2022] [Accepted: 09/16/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives To develop and externally validate a spectral CT based nomogram for the preoperative prediction of LVI in patients with resectable GC. Methods The two centered study contained a retrospective primary dataset of 224 pathologically confirmed gastric adenocarcinomas (161 males, 63 females; mean age: 60.57 ± 10.81 years, range: 20-86 years) and an external prospective validation dataset from the second hospital (77 males and 35 females; mean age, 61.05 ± 10.51 years, range, 31 to 86 years). Triple-phase enhanced CT scans with gemstone spectral imaging mode were performed within one week before surgery. The clinicopathological characteristics were collected, the iodine concentration (IC) of the primary tumours at arterial phase (AP), venous phase (VP), and delayed phase (DP) were measured and then normalized to aorta (nICs). Univariable analysis was used to compare the differences of clinicopathological and IC values between LVI positive and negative groups. Independent predictors for LVI were screened by multivariable logistic regression analysis in primary dataset and used to develop a nomogram, and its performance was evaluated by using ROC analysis and tested in validation dataset. Its clinical use was evaluated by decision curve analysis (DCA). Results Tumor thickness, Borrmann classification, CT reported lymph node (LN) status and nICDP were independent predictors for LVI, and the nomogram based on these indicators was significantly associated with LVI (P<0.001). It yielded an AUC of 0.825 (95% confidence interval [95% CI], 0.769-0.872) and 0.802 (95% CI, 0.716-0.871) in primary and validation datasets (all P<0.05), with promising clinical utility by DCA. Conclusion This study presented a dual energy CT quantification based nomogram, which enables preferable preoperative individualized prediction of LVI in patients with GC.
Collapse
Affiliation(s)
- Jing Li
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, China
| | - Yi Wang
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, China
| | - Rui Wang
- Department of Radiology, The first Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jian-bo Gao
- Department of Radiology, The first Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jin-rong Qu
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, China
- *Correspondence: Jin-rong Qu,
| |
Collapse
|
14
|
Qian Y, Zhai E, Chen S, Liu Y, Ma Y, Chen J, Liu J, Qin C, Cao Q, Chen J, Cai S. Single-cell RNA-seq dissecting heterogeneity of tumor cells and comprehensive dynamics in tumor microenvironment during lymph nodes metastasis in gastric cancer. Int J Cancer 2022; 151:1367-1381. [PMID: 35716132 DOI: 10.1002/ijc.34172] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 05/23/2022] [Accepted: 06/03/2022] [Indexed: 12/24/2022]
Abstract
Lymph node metastasis is the common metastasis route of gastric cancer. However, until now, heterogeneities of tumor cells and tumor microenvironment in primary tumors (PT) and metastatic lymph nodes (MLN) of gastric cancer (GC) remains uncharacterized. In this study, scRNA-seq was performed on tissues from PT and MLN of gastric cancer. Trajectory analysis and function enrichment analyses were conducted to decode the underlying mechanisms contributing to LN metastasis of gastric cancer. Heterogeneous composition of immune cells and distant intercellular interactions in PT and MLN were analyzed. Based on the generated single cell transcriptome profiles, dynamics of gene expressions in cancer cells between PT and MLN were characterized. Moreover, we reconstructed the developmental trajectory of GC cells' metastasis to LN and identified two sub-types of GC cells with distinct potentials of having malignant biological behaviors. We characterized the repression of neutrophil polarization associated genes, like LCN2, which would contribute to LN metastasis, and histochemistry experiments validated our findings. Additionally, heterogeneity in neutrophils, rather than macrophages, was characterized. Immune checkpoint associated interaction of SPP1 was found active in MLN. In conclusion, we decode the dynamics of tumor cells during LN metastasis in GC and to identify a sub-type of GC cells with potentials of LN metastasis. Our data indicated that the disordering the neutrophils polarization and maturation and the activation of immune checkpoint SPP1 might contribute to LN metastasis in GC, providing a novel insight on the mechanism and potential therapeutic targets of LN metastasis in GC. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yan Qian
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ertao Zhai
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Sile Chen
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yinan Liu
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Ma
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junting Chen
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianqiu Liu
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | - Qin Cao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Jianhui Chen
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shirong Cai
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
15
|
Lee SS, Park J, Oh S, Kwack K. Downregulation of LOC441461 Promotes Cell Growth and Motility in Human Gastric Cancer. Cancers (Basel) 2022; 14:cancers14051149. [PMID: 35267457 PMCID: PMC8909665 DOI: 10.3390/cancers14051149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 11/16/2022] Open
Abstract
Gastric cancer is a common tumor, with a high mortality rate. The severity of gastric cancer is assessed by TNM staging. Long noncoding RNAs (lncRNAs) play a role in cancer treatment; investigating the clinical significance of novel biomarkers associated with TNM staging, such as lncRNAs, is important. In this study, we investigated the association between the expression of the lncRNA LOC441461 and gastric cancer stage. LOC441461 expression was lower in stage IV than in stages I, II, and III. The depletion of LOC441461 promoted cell proliferation, cell cycle progression, apoptosis, cell motility, and invasiveness. LOC441461 downregulation increased the epithelial-to-mesenchymal transition, as indicated by increased TRAIL signaling and decreased RUNX1 interactions. The interaction of the transcription factors RELA, IRF1, ESR1, AR, POU5F1, TRIM28, and GATA1 with LOC441461 affected the degree of the malignancy of gastric cancer by modulating gene transcription. The present study identified LOC441461 and seven transcription factors as potential biomarkers and therapeutic targets for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Sang-soo Lee
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea; (S.-s.L.); (J.P.)
| | - JeongMan Park
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea; (S.-s.L.); (J.P.)
| | - Sooyeon Oh
- Chaum Life Center, CHA University School of Medicine, Seoul 06062, Korea;
| | - KyuBum Kwack
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea; (S.-s.L.); (J.P.)
- Correspondence: ; Tel.: +82-31-881-7141
| |
Collapse
|
16
|
The Value of Gastric Cancer Staging by Endoscopic Ultrasonography Features in the Diagnosis of Gastroenterology. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:6192190. [PMID: 35222686 PMCID: PMC8881163 DOI: 10.1155/2022/6192190] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/25/2022] [Indexed: 12/22/2022]
Abstract
This research was aimed at exploring the application value of endoscopic ultrasonography (EUS) in the diagnosis of gastric cancer staging and the correlation between staging and clinical features of gastric cancer. A total of 72 patients with gastric cancer were selected and randomly divided into two groups. The patients in the pathological group underwent postoperative pathological examination, while those in the EUS group received preoperative EUS examination. The results showed that the staging accuracy of EUS was 73.33% for T1, 78.57% for T2, 27% for T3, and 100% for T4, compared with the pathological staging. The accuracy of N- and N+ was 42.5% and 82.3% in EUS, respectively, and the total accuracy was 55.7%. There was no considerable difference in the accuracy of T staging between early gastric cancer and advanced gastric cancer (P > 0.05), but there was a considerable difference in N staging (P < 0.05). Lymph node metastasis affected the accuracy of N staging (P < 0.05). The number and location of metastatic lymph nodes did not affect the judgment of metastatic lymph nodes (P > 0.05). In addition, the proportion of understaging and overstaging was greatly different among different lesion sizes and histological types of gastric cancer (P < 0.05). To sum up, the accuracy of EUS for T and N staging of gastric cancer needed to be improved. The location of gastric cancer lesions affected the accuracy of T staging, while the depth of invasion and lymph node metastasis affected the accuracy of N staging.
Collapse
|
17
|
Ambrosio MR, Perotti B, Battini A, Fattorini C, Cavazzana A, Pasqua R, Palumbo P, Gia L, Arganini M. Surgeon-Pathologist Team Approach Dramatically Affects Lymph Nodes Detection and Improves Patients' Short-Term Outcome. Cancers (Basel) 2022; 14:cancers14041034. [PMID: 35205783 PMCID: PMC8870551 DOI: 10.3390/cancers14041034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/07/2022] [Accepted: 02/11/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Appropriate lymph node harvesting for patients with gastric cancer is fundamental for a correct staging and is strongly related to survival. In this study, we present a new protocol for on-site macroscopic evaluation and sampling of lymph nodes for gastric cancer patients. With the joint collaboration of surgeons and pathologists, our method aims to provide the largest possible number of analyzed lymph nodes per patient, allowing for a better staging. We are convinced that this approach is routinely feasible, and our preliminary results seem to confirm better patient stratification compared to other lymph node dissection methods. Abstract The downstaging of gastric cancer has recently gained particular attention in the field of gastric cancer surgery. The phenomenon is mainly due to an inappropriate sampling of lymph nodes during standard lymphadenectomy. Hence, collection of the maximum number of lymph nodes is a critical factor affecting the outcome of patients. None of the techniques proposed so far have demonstrated a real efficiency in increasing the number of identified lymph nodes. To harvest the maximum number of lymph nodes, we designed a protocol for on-site macroscopic evaluation and sampling of lymph nodes according to the Japanese Gastric Cancer Association protocol. The procedure was carried out by a surgeon/pathologist team in the operating room. We enrolled one hundred patients, 50 of whom belonged to the study group and 50 to a control group. The study group included patients who underwent lymph node dissection following the proposed protocol; the control group encompassed patients undergoing standard procedures for sampling. We compared the number and maximum diameter of lymph nodes collected in both groups, as well as some postoperative variables, the 30-day mortality and the overall survival. In the study group, the mean number of lymph nodes harvested was higher than the control one (p = 0.001). Moreover, by applying the proposed technique, we sampled lymph nodes with a very small diameter, some of which were metastatic. Noticeably, no difference in terms of postoperative course was identified between the two groups, again supporting the feasibility of an extended lymphadenectomy. By comparing the prognosis of patients, a better overall survival (p = 0.03) was detected in the study group; however, to date, no long-term follow-up is available. Interestingly, patients with metastasis in node stations number 8, 9, 11 or with skip metastasis, experienced a worse outcome and died. Based on our preliminary results, the pathologist/surgeon team approach seems to be a reliable option, despite of a slight increase in sfaff workload and technical cost. It allows for the harvesting of a larger number of lymph nodes and improves the outcome of the patients thanks to more precise staging and therapy. Nevertheless, since a higher number of patients are necessary to confirm our findings and assess the impact of this technique on oncological outcome, our study could serve as a proof-of-concept for a larger, multicentric collaboration.
Collapse
Affiliation(s)
- Maria Raffaella Ambrosio
- Pathology Unit, Azienda Sanitaria Toscana Nord-Ovest, Via Cocchi 1, 56121 Pisa, Italy; (C.F.); (A.C.)
- Correspondence:
| | - Bruno Perotti
- Surgery Unit, Ospedale Unico Versilia and Nuovo Ospedale Apuane, Azienda Sanitaria Toscana Nord Ovest, 56121 Pisa, Italy; (B.P.); (A.B.); (L.G.); (M.A.)
| | - Alda Battini
- Surgery Unit, Ospedale Unico Versilia and Nuovo Ospedale Apuane, Azienda Sanitaria Toscana Nord Ovest, 56121 Pisa, Italy; (B.P.); (A.B.); (L.G.); (M.A.)
| | - Caterina Fattorini
- Pathology Unit, Azienda Sanitaria Toscana Nord-Ovest, Via Cocchi 1, 56121 Pisa, Italy; (C.F.); (A.C.)
| | - Andrea Cavazzana
- Pathology Unit, Azienda Sanitaria Toscana Nord-Ovest, Via Cocchi 1, 56121 Pisa, Italy; (C.F.); (A.C.)
| | - Rocco Pasqua
- Department of Surgical Sciencies, University “La Sapienza”, 00100 Roma, Italy; (R.P.); (P.P.)
| | - Piergaspare Palumbo
- Department of Surgical Sciencies, University “La Sapienza”, 00100 Roma, Italy; (R.P.); (P.P.)
| | - Liano Gia
- Surgery Unit, Ospedale Unico Versilia and Nuovo Ospedale Apuane, Azienda Sanitaria Toscana Nord Ovest, 56121 Pisa, Italy; (B.P.); (A.B.); (L.G.); (M.A.)
| | - Marco Arganini
- Surgery Unit, Ospedale Unico Versilia and Nuovo Ospedale Apuane, Azienda Sanitaria Toscana Nord Ovest, 56121 Pisa, Italy; (B.P.); (A.B.); (L.G.); (M.A.)
| |
Collapse
|
18
|
Seely KD, Morgan AD, Hagenstein LD, Florey GM, Small JM. Bacterial Involvement in Progression and Metastasis of Colorectal Neoplasia. Cancers (Basel) 2022; 14:1019. [PMID: 35205767 PMCID: PMC8870662 DOI: 10.3390/cancers14041019] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 02/06/2023] Open
Abstract
While the gut microbiome is composed of numerous bacteria, specific bacteria within the gut may play a significant role in carcinogenesis, progression, and metastasis of colorectal carcinoma (CRC). Certain microbial species are known to be associated with specific cancers; however, the interrelationship between bacteria and metastasis is still enigmatic. Mounting evidence suggests that bacteria participate in cancer organotropism during solid tumor metastasis. A critical review of the literature was conducted to better characterize what is known about bacteria populating a distant site and whether a tumor depends upon the same microenvironment during or after metastasis. The processes of carcinogenesis, tumor growth and metastatic spread in the setting of bacterial infection were examined in detail. The literature was scrutinized to discover the role of the lymphatic and venous systems in tumor metastasis and how microbes affect these processes. Some bacteria have a potent ability to enhance epithelial-mesenchymal transition, a critical step in the metastatic cascade. Bacteria also can modify the microenvironment and the local immune profile at a metastatic site. Early targeted antibiotic therapy should be further investigated as a measure to prevent metastatic spread in the setting of bacterial infection.
Collapse
Affiliation(s)
- Kevin D. Seely
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA; (A.D.M.); (L.D.H.)
| | - Amanda D. Morgan
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA; (A.D.M.); (L.D.H.)
| | - Lauren D. Hagenstein
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA; (A.D.M.); (L.D.H.)
| | - Garrett M. Florey
- College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA;
| | - James M. Small
- Department of Biomedical Sciences, Rocky Vista University, Parker, CO 80134, USA;
| |
Collapse
|
19
|
Wu F, Sun D, Liao Y, Shang K, Lu C. RPL35A is a key promotor involved in the development and progression of gastric cancer. Cancer Cell Int 2021; 21:497. [PMID: 34535139 PMCID: PMC8447681 DOI: 10.1186/s12935-021-02199-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 09/06/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND RPL35A has been reported to work as a biomarker in tumor angiogenesis. However, little work has been performed on the expression level and functional importance of RPL35A in gastric cancer (GC). METHODS The protein expression level of RPL35A was detected by immunohistochemical staining and western blot analysis. The Celigo cell counting assay was used to assess cell proliferation. Both the wound healing assay and the transwell assay were conducted to evaluate cell migration. Flow cytometric analysis was utilized to detect cell apoptosis and cell cycle. A mouse xenograft model was constructed for in vivo experiments. RESULTS The results demonstrated that RPL35A expression was abundantly up-regulated in GC and positively related to tumor infiltrate. In addition, RPL35A knockdown could significantly suppress cell proliferation, migration, enhance apoptosis and arrest cell cycle. The in vivo study also verified the inhibitory effects of RPL35A knockdown on GC tumorigenesis. CONCLUSIONS The above mentioned results indicated that the knockdown of RPL35A might be a considerable therapeutic strategy for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Fang Wu
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Dachuan Sun
- Senior Department of General Surgery, The First Medical Center of Chinese, PLA General Hospital, Fuxin Road, No. 28, Haidian District, Beijing, 100853, China
| | - Yuqian Liao
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Kai Shang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Canrong Lu
- Senior Department of General Surgery, The First Medical Center of Chinese, PLA General Hospital, Fuxin Road, No. 28, Haidian District, Beijing, 100853, China.
| |
Collapse
|
20
|
Koemans WJ, Lurvink RJ, Grootscholten C, Verhoeven RHA, de Hingh IH, van Sandick JW. Synchronous peritoneal metastases of gastric cancer origin: incidence, treatment and survival of a nationwide Dutch cohort. Gastric Cancer 2021; 24:800-809. [PMID: 33495964 DOI: 10.1007/s10120-021-01160-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The peritoneum is a predilection site for gastric cancer metastases. Current standard treatment for gastric cancer patients with synchronous peritoneal metastases is palliative systemic therapy. However, its efficacy is largely unknown. The aim of this study was to investigate the incidence, treatment and survival patterns of gastric cancer patients with synchronous peritoneal metastases in the Netherlands. METHODS All newly diagnosed gastric adenocarcinoma patients with synchronous peritoneal metastases between 1999 and 2017 were selected from the Netherlands Cancer Registry (NCR). Incidence, treatment and survival patterns were analyzed. RESULTS In total, 3,773 patients were identified from the NCR. The incidence of synchronous peritoneal metastases in gastric cancer patients increased from 18% in 2008 to 27% in 2017. The use of systemic therapy increased from 15% in 1999-2002 to 43% in 2013-2017 (p < 0.001). The median survival of the entire cohort did not significantly increase over time. Median survival of patients treated with systemic therapy increased from 7.4 months in 1999-2002 to 9.4 months in 2013-2017 (p = 0.005). In contrast, median survival of patients not treated with systemic therapy decreased from 3.3 months in 1999-2002 to 2.1 months in 2013-2017 (p < 0.001). Some clinical and pathological data such as the extent of the peritoneal metastases were not available. CONCLUSION Synchronous peritoneal metastases are increasingly diagnosed in gastric cancer patients. In recent years, more patients were treated with systemic treatment and survival of these patients increased. However, as survival of the entire group did not improve over time, the effect of systemic therapy remains unknown.
Collapse
Affiliation(s)
- Willem J Koemans
- Department of Surgical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Robin J Lurvink
- Department of Surgery, Catharina Hospital, Michelangelolaan 2, 5623 EJ, Eindhoven, The Netherlands
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
| | - Cecile Grootscholten
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Rob H A Verhoeven
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ignace H de Hingh
- Department of Surgery, Catharina Hospital, Michelangelolaan 2, 5623 EJ, Eindhoven, The Netherlands
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Johanna W van Sandick
- Department of Surgical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
21
|
Wu P, Wang J, Mao X, Xu H, Zhu Z. PDCD4 regulates apoptosis in human peritoneal mesothelial cells and promotes gastric cancer peritoneal metastasis. Histol Histopathol 2021; 36:447-457. [PMID: 33442866 DOI: 10.14670/hh-18-305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Programmed cell death 4 (PDCD4) is a tumor suppressor gene, however, the function and regulatory mechanism remain to be discovered. The connection between tumorigenesis and apoptosis is one of the most important foci of cancer research. Our study aimed to explore the connections between PDCD4-mediated apoptosis of human peritoneal mesothelial cells (HPMC) and peritoneal metastasis in gastric cancer. METHODS The PDCD4 expression in 31 pairs of HPMC and tumor tissues was assessed by immunohistochemistry and RT-PCR. In cell experiments, we monitored gastric cancer cell migration with a Transwell chamber assay when PDCD4 was silenced in HPMC. Subsequently, apoptosis of HPMC was detected by a flow cytometric assay and western blotting. After analyzing cytokines in culture supernatants from gastric cancer with enzyme-linked immunosorbent assays (ELISAs), transforming growth factor-beta 1 (TGF-β1) was abundant in the culture supernatants of gastric cancer. Then, PDCD4 expression in HMrSV5 cells was analyzed by western blotting after retreatment with different concentrations of TGF-β1. Moreover, apoptosis of peritoneal mesothelial cells treated with TGF-β1 was detected according to the above methods. RESULTS In human metastatic peritoneal tissues, the expression of PDCD4 was significantly lower than that in normal tissues. At the same time, decreased expression of PDCD4 in HPMC was associated with increased migration capacity of gastric cancer cells. Moreover, suppressing the expression of PDCD4 promoted apoptosis in mesothelial cells which may be regulated by TGF-β secreted from gastric cancer cells. CONCLUSIONS These data suggested that decreased expression of PDCD4 significantly promoted apoptosis in human peritoneal mesothelial cells, thus inducing peritoneal metastasis, and that TGF-β1 secreted from gastric cancer cells may have played a crucial role.
Collapse
Affiliation(s)
- Pei Wu
- Department of Surgical Oncology, Department of General Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Jinou Wang
- Department of Pathology, Shengjing Hospital of China Medical University, Senyang, China
| | - Xiaoyun Mao
- Department of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Huimian Xu
- Department of Surgical Oncology, Department of General Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Zhi Zhu
- Department of Surgical Oncology, Department of General Surgery, First Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|