1
|
Bruhm DC, Vulpescu NA, Foda ZH, Phallen J, Scharpf RB, Velculescu VE. Genomic and fragmentomic landscapes of cell-free DNA for early cancer detection. Nat Rev Cancer 2025; 25:341-358. [PMID: 40038442 DOI: 10.1038/s41568-025-00795-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/24/2025] [Indexed: 03/06/2025]
Abstract
Genomic analyses of cell-free DNA (cfDNA) in plasma are enabling noninvasive blood-based biomarker approaches to cancer detection and disease monitoring. Current approaches for identification of circulating tumour DNA typically use targeted tumour-specific mutations or methylation analyses. An emerging approach is based on the recognition of altered genome-wide cfDNA fragmentation in patients with cancer. Recent studies have revealed a multitude of characteristics that can affect the compendium of cfDNA fragments across the genome, collectively called the 'cfDNA fragmentome'. These changes result from genomic, epigenomic, transcriptomic and chromatin states of an individual and affect the size, position, coverage, mutation, structural and methylation characteristics of cfDNA. Identifying and monitoring these changes has the potential to improve early detection of cancer, especially using highly sensitive multi-feature machine learning approaches that would be amenable to broad use in populations at increased risk. This Review highlights the rapidly evolving field of genome-wide analyses of cfDNA characteristics, their comparison to existing cfDNA methods, and recent related innovations at the intersection of large-scale sequencing and artificial intelligence. As the breadth of clinical applications of cfDNA fragmentome methods have enormous public health implications for cancer screening and personalized approaches for clinical management of patients with cancer, we outline the challenges and opportunities ahead.
Collapse
Affiliation(s)
- Daniel C Bruhm
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas A Vulpescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachariah H Foda
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jillian Phallen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert B Scharpf
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Victor E Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Zhan Z, Chen B, Zeng Y, Huang R, Yu J, Guo Z, Lin X. Long-term trends and projections of stomach cancer burden in China: Insights from the GBD 2021 study. PLoS One 2025; 20:e0320751. [PMID: 40198592 PMCID: PMC11978042 DOI: 10.1371/journal.pone.0320751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 02/25/2025] [Indexed: 04/10/2025] Open
Abstract
Stomach cancer continues to be a major public health concern in China, with its incidence, prevalence, mortality, and overall burden showing notable changes over time. This study set out to analyze the long-term trends of stomach cancer from 1990 to 2021, figure out the effects of aging, epidemiological changes, and population growth, and also make projections for the future. To conduct the study, data from the Global Burden of Disease Study 2021 was used. This data allowed for the analysis of various aspects such as age-standardized incidence, prevalence, mortality rates, disability-adjusted life years (DALYs), years lived with disability (YLDs), and years of life lost (YLLs) related to stomach cancer in China. Joinpoint regression analysis was carried out to spot significant trends and turning points. Decomposition analysis was done to assess how much aging, epidemiological changes, and population growth contributed. Additionally, the Bayesian age-period-cohort (BAPC) model was employed to predict what the trends might be from 2021 to 2030. In 2021, there were 611,799 new cases of stomach cancer in China. The age-standardized incidence rate was 29.05 per 100,000 people, with males having a much higher rate of 44.48 compared to females at 15.23. The age-standardized prevalence and mortality rates were 57.22 and 21.51 per 100,000 respectively, and both were higher in males as well. There were also significant gender differences in DALYs, YLDs, and YLLs, with males shouldering a greater burden. From 1990 to 2021, the incidence and mortality rates went down, especially after 2004. Through decomposition analysis, it was found that aging led to a decrease in incidence but an increase in mortality, especially among males. Epidemiological changes caused both the incidence and mortality rates to drop, and the effect was more pronounced in males. The BAPC model forecasts that the incidence and mortality rates will continue to decline for both genders from 2021 to 2030, with a more rapid decrease in males. Overall, this study emphasizes the changing trends of the stomach cancer burden in China, the significant gender differences, and the impacts of aging, epidemiological changes, and population growth. It's crucial to keep monitoring and implement targeted public health strategies to further reduce the burden of stomach cancer.
Collapse
Affiliation(s)
- Zhouwei Zhan
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Bijuan Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Yi Zeng
- Department of Gastric Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Rui Huang
- Digestive Endoscopy Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China,
| | - Jiami Yu
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Zengqing Guo
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Xiaoyan Lin
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350001, China
| |
Collapse
|
3
|
Murano T, Chung KY, Tang YC, Kano Y, Takeuchi K, Sakamoto N, Kuwata T, Gao Y, Cheong JK, Cheng H, Zhou L, Yano T. Novel and Effective Blood-Based miRNA Diagnostic Panel for Gastric Cancer: A Pilot Study in a Japanese Population. Cancer Med 2025; 14:e70790. [PMID: 40249300 PMCID: PMC12007419 DOI: 10.1002/cam4.70790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 01/31/2025] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) has a high prevalence in Asian countries, and there is an unmet need for non-invasive and efficient GC screening methods. This study evaluated the diagnostic efficacy of GASTROClear, a panel of blood serum miRNAs for the detection of GC, in a Japanese population. METHODS We conducted a pilot cohort study, comprising 103 patients with GC and 122 healthy controls. Serum samples were prospectively collected from study participants at two Japanese hospitals using a predefined blood-processing protocol. The diagnostic performance of GASTROClear was analyzed using a receiver operating characteristic curve and cutoff. By applying a logistic regression algorithm, we evaluated the diagnostic efficacy of novel combinations of GC diagnostic biomarker panels, consisting of GASTROClear and alternative serum markers (anti-Helicobacter pylori [Hp] antibody and pepsinogen). RESULTS Most patients had Stage I (58%) GC and were asymptomatic (59%). The area under the curve (AUC) value for the detection of GC using GASTROClear was 0.80, with 70.9% sensitivity and 75.2% specificity. GASTROClear performed equally well within the subgroups based on age, sex, symptoms, Hp status, and tumor characteristics. We improved the diagnostic performance of GASTROClear by combining it with an anti-Hp antibody and pepsinogen. This yielded an AUC value of 0.88, with the highest specificity (86.9%) at a fixed sensitivity (70.9%). CONCLUSIONS GASTROClear demonstrated competent diagnostic efficacy for GC in the detection of GC in our Japanese cohort, even in the early stages of cancer and asymptomatic cases. Its combination with existing serum markers may contribute to efficient risk stratification to detect GC.
Collapse
Affiliation(s)
- Tatsuro Murano
- Department of Gastroenterology and EndoscopyNational Cancer Center Hospital EastChibaJapan
| | | | | | - Yuki Kano
- Department of Gastroenterology and EndoscopyNational Cancer Center Hospital EastChibaJapan
| | - Ken Takeuchi
- Department of GastroenterologyTsujinaka Hospital KashiwanohaChibaJapan
| | - Naoya Sakamoto
- Department of PathologyNational Cancer Center Hospital EastChibaJapan
| | - Takeshi Kuwata
- Department of PathologyNational Cancer Center Hospital EastChibaJapan
- Department of Genetic Medicine and ServicesNational Cancer Center Hospital EastChibaJapan
| | | | | | | | | | - Tomonori Yano
- Department of Gastroenterology and EndoscopyNational Cancer Center Hospital EastChibaJapan
| |
Collapse
|
4
|
Liu YY, Fu YF, Yang WY, Li Z, Lu Q, Su X, Shi J, Wu SQ, Liang D, He YT. DKK3 and SERPINB5 as novel serum biomarkers for gastric cancer: facilitating the development of risk prediction models for gastric cancer. Front Oncol 2025; 15:1536491. [PMID: 40231256 PMCID: PMC11994446 DOI: 10.3389/fonc.2025.1536491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/11/2025] [Indexed: 04/16/2025] Open
Abstract
The existing gastric cancer (GC) risk prediction models based on biomarkers are limited. This study aims to identify new promising biomarkers for GC to develop a risk prediction model for effective assessment, screening, and early diagnosis. This study was conducted utilizing a large combined cohort for upper gastrointestinal cancer that was established in Hebei Province, China. General macro risk factors, Helicobacter pylori (H.pylori) infection status, and protein biomarkers were collected through questionnaire surveys and laboratory tests. Novel GC biomarkers were explored using data-independent acquisition (DIA) proteomics and enzyme-linked immunosorbent assay (ELISA). Multiple machine learning algorithms were used to identify key predictors for the GC risk prediction model, which was validated with an independent external cohort from multiple hospitals. A total of 530 participants aged 40 to 74 were analyzed, with 104 ultimately diagnosed with GC. Significant biomarkers in GC patients were identified by DIA combined ELISA, including elevated Keratin 7 (KRT7) and Mammary fibrostatin (SERPINB5) (P<0.001) and decreased Dickkopf-associated protein 3 (DKK3) (P<0.001). Factors such as sex, age, smoking status, alcohol consumption, family history of GC, H. pylori infection, DKK3 and SERPINB5 were used to create a multidimensional risk prediction model for GC. This model achieved an area under the curve (AUC) of 0.938 (95% confidence interval: 0.913-0.962). The risk prediction model developed in this study shows high accuracy and practical utility, serving as an effective preliminary screening tool for identifying high-risk individuals for GC.
Collapse
Affiliation(s)
- Yan-Yu Liu
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yan-Fang Fu
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wan-Yu Yang
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zheng Li
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qian Lu
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xin Su
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jin Shi
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Si-Qi Wu
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Di Liang
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yu-Tong He
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
5
|
Lan Y, Sun W, Zhong S, Xu Q, Xue Y, Liu Z, Shi L, Han B, Zhai T, Liu M, Sun Y, Xu H. A risk prediction model for gastric cancer based on endoscopic atrophy classification. BMC Cancer 2025; 25:518. [PMID: 40119304 PMCID: PMC11927292 DOI: 10.1186/s12885-025-13860-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 03/04/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUNDS Gastric cancer (GC) is a prevalent malignancy affecting the digestive system. We aimed to develop a risk prediction model based on endoscopic atrophy classification for GC. METHODS We retrospectively collected the data from January 2020 to October 2021 in our hospital and randomly divided the patients into training and validation sets in an 8:2 ratio. We used multiple machine learning algorithms such as logistic regression (LR), Decision tree, Support Vector Machine, Random forest, and so on to establish the models. We employed the Least absolute shrinkage and selection operator (LASSO) to screen variables for the LR model. However, we chose all the variables to construct the models for other machine learning algorithms. All models were evaluated using the receiver operating characteristic curve (ROC), predictive histograms, and decision curve analysis (DCA). RESULTS A total of 1156 patients were selected for the analysis. Five variables, including age, sex, family history of GC, HP infection status, and Kimura-Takemoto Classification (KTC), were screened using LASSO analysis. The area under the curve (AUC) of all the machine learning models ranged from 0.762 to 0.974 in the training set and from 0.608 to 0.812 in the validation set. Among them, the LR model exhibited the highest AUC value (0.812, 95%CI: 0.737-0.887) in the validation set with good calibration and clinical applicability. Finally, we constructed a nomogram to demonstrate the LR model. CONCLUSIONS We established a nomogram based on endoscopic atrophy classification for GC, which might be valuable in predicting GC risk and assisting clinical decision-making.
Collapse
Affiliation(s)
- Yadi Lan
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Weijia Sun
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Shen Zhong
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Qianqian Xu
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Yining Xue
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Zhaoyu Liu
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Lei Shi
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Bing Han
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Tianyu Zhai
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Mingyue Liu
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Yujing Sun
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Hongwei Xu
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
| |
Collapse
|
6
|
Moratorio I, Canavesi A, Olano C, Mönkemüller K. Prevalence and endoscopic-histological correlation of premalignant gastric lesions at a university hospital in Uruguay. Endosc Int Open 2025; 13:a25420880. [PMID: 40109312 PMCID: PMC11922308 DOI: 10.1055/a-2542-0880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
Background and study aims Although chronic atrophic gastritis (CAG), intestinal metaplasia (IM), and dysplasia constitute gastric pre-neoplastic conditions of gastric cancer (GC), data on endoscopic correlation and the prevalence in many South American countries are scarce. The aims of this study were to establish prevalence and perform endoscopic-histological correlation of gastric pre-neoplastic conditions using high-definition white light endoscopy (WLE) and to determine interobserver agreement for endoscopic findings for CAG and IM. Patients and methods A prospective, observational, descriptive, cross-sectional study was carried out at a Uruguayan hospital during a 6-month period. Risk was stratified according to Operative Link for Gastritis Assessment and Operative Link for Gastric Intestinal Metaplasia stage for CAG and IM, respectively. An independent and blinded second observer was included to determine interobserver endoscopic and histologic agreement. Results A total of 102 patients (mean age 57 years ± 1.6 years, 68.6% woman) were included. Prevalence of histological CAG and IM were 38.2% and IM 31.4%, respectively. Endoscopic-histological correlation for CAG had kappa index 0.063, sensitivity 46%, and specificity 60%. For endoscopic IM, the kappa index was 0.216, sensitivity 22%, and specificity 96%. Interobserver variability was good for gastric fold flattening and very good in the presence of whitish-greyish plaques for CAG and IM, respectively. Conclusions The endoscopic-histological correlation of both CAG and IM was low, raising the need for biopsy for diagnosis in all cases, regardless of HD-WLE findings. Although prevalence of gastric pre-neoplastic conditions in this group of Uruguayan patients was comparable to those described in countries with a high incidence of GC, a low proportion of high-risk stages (III and IV) was identified.
Collapse
Affiliation(s)
- Ignacio Moratorio
- Unidad Académica Gastroenterología, Hospital de Clinicas Doctor Manuel Quintela, Montevideo, Uruguay
| | - Adrian Canavesi
- Unidad Académica Gastroenterología, Hospital de Clinicas Doctor Manuel Quintela, Montevideo, Uruguay
| | - Carolina Olano
- Unidad Académica Gastroenterología, Hospital de Clinicas Doctor Manuel Quintela, Montevideo, Uruguay
| | - Klaus Mönkemüller
- Unidad Académica Gastroenterología, Hospital de Clinicas Doctor Manuel Quintela, Montevideo, Uruguay
| |
Collapse
|
7
|
Huang Y, Chen J, Guo Y, Ding Z, Liang X, Zhang W, Xue H, Zhao Y, Li X, Lu H. Staging of operative link on gastritis assessment and operative link on gastric intestinal metaplasia systems for risk assessment of early gastric cancer: a case-control study. J Clin Pathol 2025; 78:117-122. [PMID: 37989553 DOI: 10.1136/jcp-2023-209209] [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: 09/21/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
AIMS Operative link on gastritis assessment (OLGA) and operative link on gastric intestinal metaplasia assessment (OLGIM) systems are histological staging systems of gastritis for gastric cancer (GC) risk estimation. Intermediate OLGA/OLGIM stages are of concern in a region with high incidence of GC. This study aimed to validate OLGA and OLGIM staging systems for early GC (EGC) in Chinese population. METHODS This single-centre, case-control study included 196 patients with EGC and 196 age-matched and sex-matched health screening control subjects. OLGA and OLGIM systems, and other clinical parameters were evaluated using logistic regression analysis. RESULTS OLGA and OLGIM stages II/III/IV were more prevalent in patients with EGC than in the control subjects. Multivariable analysis revealed family history of GC, previous Helicobacter pylori (H. pylori) infection, OLGA stages II and III-IV, OLGIM stages II and III-IV as independent risk factors for EGC (ORs, 4.04, 1.87, 2.52, 6.79, 4.11 and 10.78, respectively). Area under the receiver operating characteristic curve on EGC risk estimation was improved for OLGIM compared with OLGA (0.78 vs 0.71, p<0.001). Autoantibody seropositivity of gastric mucosa was not associated with EGC risk stratified by H. pylori status. CONCLUSIONS Surveillance of intermediate-risk patients (OLGA/OLGIM II) should be emphasised in our region. The OLGIM may be preferred over the OLGA for EGC risk estimation.
Collapse
Affiliation(s)
- Yu Huang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jinnan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yixian Guo
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhaohui Ding
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiao Liang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Zhang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hanbing Xue
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yunjia Zhao
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaobo Li
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hong Lu
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Ni Q, Yang H, Rao H, Zhang L, Xiong M, Han X, Deng B, Wang L, Chen J, Shi Y. The role of the C5a-C5aR pathway in iron metabolism and gastric cancer progression. Front Immunol 2025; 15:1522181. [PMID: 39850877 PMCID: PMC11754390 DOI: 10.3389/fimmu.2024.1522181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
Gastric cancer continues to be a leading global health concern, with current therapeutic approaches requiring significant improvement. While the disruption of iron metabolism in the advancement of gastric cancer has been well-documented, the underlying regulatory mechanisms remain largely unexplored. Additionally, the complement C5a-C5aR pathway has been identified as a crucial factor in gastric cancer development. The impact of the complement system on iron metabolism and its role in gastric cancer progression is an area warranting further investigation. Our research demonstrates that the C5a-C5aR pathway promotes gastric cancer progression by enhancing iron acquisition in tumor cells through two mechanisms. First, it drives macrophage polarization toward the M2 phenotype, which has a strong iron-release capability. Second, it increases the expression of LCN2, a high-affinity iron-binding protein critical for iron export from tumor-associated macrophages, by activating endoplasmic reticulum stress in these cells. Both mechanisms facilitate the transfer of iron from macrophages to cancer cells, thereby promoting tumor cell proliferation. This study aims to elucidate the connection between the complement C5a-C5aR pathway and iron metabolism within the tumor microenvironment. Our data suggest a pivotal role of the C5a-C5aR pathway in tumor iron management, indicating that targeting its regulatory mechanisms may pave the way for future iron-targeted therapeutic approaches in cancer treatment.
Collapse
Affiliation(s)
- Qinxue Ni
- The First Affiliated Hospital of Army Military Medical University, Department of General Surgery, Chongqing, China
| | - Hong Yang
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Hang Rao
- The First Affiliated Hospital of Army Military Medical University, Department of General Surgery, Chongqing, China
| | - Liyong Zhang
- The First Affiliated Hospital of Army Military Medical University, Department of General Surgery, Chongqing, China
| | - Mengyuan Xiong
- The First Affiliated Hospital of Army Military Medical University, Department of General Surgery, Chongqing, China
| | - Xiao Han
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Boshao Deng
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lulu Wang
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jian Chen
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yan Shi
- The First Affiliated Hospital of Army Military Medical University, Department of General Surgery, Chongqing, China
| |
Collapse
|
9
|
Kim J, Lee HJ. Function Preserving Gastrectomy and Quality of Life. J Gastric Cancer 2025; 25:247-260. [PMID: 39822178 PMCID: PMC11739640 DOI: 10.5230/jgc.2025.25.e7] [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: 12/23/2024] [Revised: 12/24/2024] [Accepted: 12/24/2024] [Indexed: 01/19/2025] Open
Abstract
Advances in gastric cancer screening have enabled earlier detection, shifting the focus of treatment toward preserving patients' quality of life (QoL). Function-preserving gastrectomy (FPG), including pylorus-preserving gastrectomy, proximal gastrectomy, and sentinel node navigation surgery, represents a paradigm shift in the surgical management of early gastric cancer. These techniques aim to balance oncological safety with the preservation of gastric function, mitigating postgastrectomy syndromes such as dumping syndrome, bile reflux, and nutritional deficiencies. QoL assessment tools, including EORTC QLQ-STO22, KOQUSS-40, and PGSAS-45, have become integral for evaluating patient-reported outcomes, providing insights into physical, emotional, and functional recovery. Although current evidence underscores the benefits of FPG, most studies are limited to East Asia, highlighting the need for multinational trials to validate these findings globally. FPG has demonstrated comparable short- and long-term oncological outcomes to conventional gastrectomy. Enhanced nutritional recovery and reduced gastrointestinal sequelae make FPG increasingly attractive. However, its widespread adoption is challenged by technical complexity, resource intensity, and the need for adequate surgical experience. The integration of advanced technologies, such as robotic surgery and artificial intelligence, coupled with personalized approaches, is expected to further optimize FPG outcomes. This review underscores the critical role of standardized QoL assessments, collaborative research, and technological innovations in advancing FPG as a cornerstone of patient-centered gastric cancer care.
Collapse
Affiliation(s)
- Jeesun Kim
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Hyuk-Joon Lee
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
10
|
Yang B, Xie X, Jin X, Huang X, He Y, Yin K, Ji C, Liu L, Feng Z. Identification and validation of serum MUC17 as a non-invasive early warning biomarker for screening of gastric intraepithelial neoplasia. Transl Oncol 2025; 51:102207. [PMID: 39580962 PMCID: PMC11625214 DOI: 10.1016/j.tranon.2024.102207] [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: 06/21/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND The early diagnosis and treatment of Gastric Intraepithelial Neoplasia (GIN) are pivotal for improving the survival rates of patients with gastric cancer (GC). Regrettably, reliable noninvasive biomarkers for GIN screening are currently lacking. METHODS mRNA data from the GEO database, pan-cancer data from the TCGA database, and a gene list of exocrine proteins were subjected to integrated analysis to identify a noninvasive biomarker for GIN. The scRNA-seq data analysis, IHC and Elisa were employed to validate the expression of the biomarker in the serum and tissues of clinical patients across different pathological stages. RESULTS MUC17 has been identified as a non-invasive diagnostic marker for GIN. It is upregulated in GIN prior to the onset of gastric carcinogenesis and downregulated in other tumors, with high GC specificity. The area under the curve values of serum MUC17 for differentiating chronic gastritis (CG) from low-grade intraepithelial neoplasia (LGIN), high-grade intraepithelial neoplasia (HGIN), and early gastric cancer (EGC) were 0.8788, 0.8544, and 0.9513, respectively. Additionally, low plasma MUC17 levels were found to be significantly lower in gastric ulcer (GU), gastric neuroendocrine tumor (GNET), and gastrointestinal stromal tumor (GIST) compared to GIN. The AUC for differentiating between GIN and GU, GNET, or GIST was 0.7803, 0.9244 and 0.9796, respectively. CONCLUSIONS These findings suggest that plasma MUC17 levels hold substantial promise as a screening biomarker for individuals with GIN and EGC, effectively identifying high-risk groups that necessitate further gastroscopy.
Collapse
Affiliation(s)
- Bingxue Yang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China
| | - Xiaoli Xie
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China
| | - Xiaoxu Jin
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China
| | - Xiuhong Huang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China
| | - Yujian He
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China
| | - Kaige Yin
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China
| | - Chenguang Ji
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China
| | - Li Liu
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China
| | - Zhijie Feng
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China.
| |
Collapse
|
11
|
Huang Z, Chen S, Yin S, Shi Z, Gu L, Li L, Yin H, Huang Z, Li B, Chen X, Yang Y, Wang Z, Li H, Zhang C, He Y. Development and validation of a nomogram for predicting the risk of developing gastric cancer based on a questionnaire: a cross-sectional study. Front Oncol 2024; 14:1351967. [PMID: 39588309 PMCID: PMC11586234 DOI: 10.3389/fonc.2024.1351967] [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: 12/07/2023] [Accepted: 10/21/2024] [Indexed: 11/27/2024] Open
Abstract
Background Detection of gastric cancer (GC) at early stages is an effective strategy for decreasing mortality. This study aimed to construct a prediction nomogram based on a questionnaire to assess the risk of developing GC. Methods Our study comprised a total of 4379 participants (2326 participants from outpatient at Fengqing People's Hospital were considered for model development and internal validation, and 2053 participants from outpatients at the endoscopy center at the Seventh Affiliated Hospital of Sun Yat-Sen University were considered for independent external validation) and gastric mucosa status was determined by endoscopy and biopsies. The eligible participants in development cohort from Fengqing people's Hospital were randomly separated into a training cohort (n=1629, 70.0%) and an internal validation cohort (n=697, 30.0%). The relevant features were selected by a least absolute shrinkage and selection operator (LASSO), and the ensuing features were evaluated through multivariable logistic regression analysis. Subsequently, the variables were selected to construct a prediction nomogram. The discriminative ability and predictive accuracy of the nomogram were evaluated by the C-index and calibration plot, respectively. Decision curve analysis (DCA) curves were used for the assessment of clinical benefit of the model. This model was developed to estimate the risk of developing neoplastic lesions according to the "transparent reporting of a multivariable prediction model for individual prognosis or diagnosis" (TRIPOD) statement. Results Six variables, including age, sex, alcohol consumption, cigarette smoking, education level, and Hp infection status, were independent risk factors for the development of neoplastic lesions. Thus, these variables were incorporated into the final nomogram. The AUC of the nomogram were 0.701, 0.657 and 0.699 in the training, internal validation, and external validation cohorts, respectively. The calibration curve showed that the nomogram was in good agreement with the observed outcomes. Compared to treatment of all patients or none, our nomogram showed a notably higher clinical benefit. Conclusion This nomogram proved to be a convenient, cost-effective tool to effectively predict an individual's risk of developing neoplastic lesions, and it can act as a prescreening tool before gastroscopy.
Collapse
Affiliation(s)
- Zhangsen Huang
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Songyao Chen
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Songcheng Yin
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Zhaowen Shi
- General Surgery, Fengqing People’s Hospital, Lincang, China
| | - Liang Gu
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Liang Li
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Haofan Yin
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zhijian Huang
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Bo Li
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Xin Chen
- General Surgery, Fengqing People’s Hospital, Lincang, China
| | - Yilin Yang
- General Surgery, Fengqing People’s Hospital, Lincang, China
| | - Zhengli Wang
- General Surgery, Fengqing People’s Hospital, Lincang, China
| | - Hai Li
- General Surgery, Fengqing People’s Hospital, Lincang, China
| | - Changhua Zhang
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yulong He
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| |
Collapse
|
12
|
Si YT, Xiong XS, Wang JT, Yuan Q, Li YT, Tang JW, Li YN, Zhang XY, Li ZK, Lai JX, Umar Z, Yang WX, Li F, Wang L, Gu B. Identification of chronic non-atrophic gastritis and intestinal metaplasia stages in the Correa's cascade through machine learning analyses of SERS spectral signature of non-invasively-collected human gastric fluid samples. Biosens Bioelectron 2024; 262:116530. [PMID: 38943854 DOI: 10.1016/j.bios.2024.116530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 07/01/2024]
Abstract
The progression of gastric cancer involves a complex multi-stage process, with gastroscopy and biopsy being the standard procedures for diagnosing gastric diseases. This study introduces an innovative non-invasive approach to differentiate gastric disease stage using gastric fluid samples through machine-learning-assisted surface-enhanced Raman spectroscopy (SERS). This method effectively identifies different stages of gastric lesions. The XGBoost algorithm demonstrates the highest accuracy of 96.88% and 91.67%, respectively, in distinguishing chronic non-atrophic gastritis from intestinal metaplasia and different subtypes of gastritis (mild, moderate, and severe). Through blinded testing validation, the model can achieve more than 80% accuracy. These findings offer new possibilities for rapid, cost-effective, and minimally invasive diagnosis of gastric diseases.
Collapse
Affiliation(s)
- Yu-Ting Si
- Medical Technology School, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Xue-Song Xiong
- Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, Jiangsu Province, China
| | - Jin-Ting Wang
- Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, Jiangsu Province, China
| | - Quan Yuan
- School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yu-Ting Li
- Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, Jiangsu Province, China
| | - Jia-Wei Tang
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China; Division of Microbiology and Immunology, School of Biomedical Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Yong-Nian Li
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xin-Yu Zhang
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zheng-Kang Li
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jin-Xin Lai
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zeeshan Umar
- Marshall Laboratory of Biomedical Engineering, School of Medicine, Shenzhen University, Guangdong Province, China
| | - Wei-Xuan Yang
- Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, Jiangsu Province, China
| | - Fen Li
- Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, Jiangsu Province, China.
| | - Liang Wang
- School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China; Division of Microbiology and Immunology, School of Biomedical Sciences, The University of Western Australia, Crawley, Western Australia, Australia; The Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia.
| | - Bing Gu
- Medical Technology School, Xuzhou Medical University, Xuzhou, Jiangsu Province, China; Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
13
|
Lu F, Li L, Shen K, Qian Y, Zhang P, Yang Y, Zhu Q, Huang Y, Yan C, Wei W. An enzymatic reaction-based SERS saliva analysis microporous array chip for chiral differentiation and high-throughput detection of D-amino acids. Mikrochim Acta 2024; 191:653. [PMID: 39375224 DOI: 10.1007/s00604-024-06733-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/29/2024] [Indexed: 10/09/2024]
Abstract
A Raman-active boronate modified surface-enhanced Raman scattering (SERS) microporous array chip based on the enzymatic reaction was constructed for reliable, sensitive, and quantitative monitoring of D-Proline (D-Pro) and D-Alanine (D-Ala) in saliva. Initially, 3-mercaptophenylboronic acid (3-MPBA) was bonded to Au-coated Si nanocrown arrays (Au/SiNCA) via Au-S bonding. Following this, H2O2 obtained from D-amino acid oxidase (DAAO)-specific catalyzed D-amino acids (D-AAs) further reduced 3-MPBA to 3-hydroxythiophenol (3-HTP) with a new Raman peak at 882 cm-1. Meanwhile, the original characteristic peak at 998 cm-1 remained unchanged. Therefore, the I882/I998 ratio increased with increasing content of D-AAs in the sample to be tested, allowing D-AAs to be quantitatively detected. The Au/SiNCA with large-area periodic crown structure prepared provided numerous, uniform "hot spots," and the microporous array chip with 16 detection units was employed as the platform for SERS analysis, realizing high-throughput, high sensitivity, high specificity and high-reliability quantitative detection of D-AAs (D-Pro and D-Ala). The limits of detection (LOD) were down to 10.1 µM and 13.7 µM throughout the linear range of 20-500 µM. The good results of the saliva detection suggested that this SERS sensor could rapidly differentiate between early-stage gastric cancer patients and healthy individuals.
Collapse
Affiliation(s)
- Feng Lu
- Affiliated Huishan Hospital of Medical College, Yangzhou University, Wuxi Huishan District People's Hospital, Wuxi, 214187, People's Republic of China
| | - Limao Li
- Department of Gastrointestinal Surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Kang Shen
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Yayun Qian
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Pengfei Zhang
- Department of Gastrointestinal Surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Yan Yang
- Department of Gastrointestinal Surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Qunshan Zhu
- Department of Gastrointestinal Surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Yong Huang
- Department of Gastrointestinal Surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Chunxiang Yan
- Department of Gastrointestinal Surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, People's Republic of China.
| | - Wei Wei
- Department of Gastrointestinal Surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, People's Republic of China.
| |
Collapse
|
14
|
Zheng H, Liu Z, Chen Y, Ji P, Fang Z, He Y, Guo C, Xiao P, Wang C, Yin W, Li F, Chen X, Liu M, Pan Y, Liu F, Liu Y, He Z, Ke Y. Development and external validation of a quantitative diagnostic model for malignant gastric lesions in clinical opportunistic screening: A multicenter real-world study. Chin Med J (Engl) 2024; 137:2343-2350. [PMID: 38403900 PMCID: PMC11441920 DOI: 10.1097/cm9.0000000000002903] [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: 09/22/2023] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Clinical opportunistic screening is a cost-effective cancer screening modality. This study aimed to establish an easy-to-use diagnostic model serving as a risk stratification tool for identification of individuals with malignant gastric lesions for opportunistic screening. METHODS We developed a questionnaire-based diagnostic model using a joint dataset including two clinical cohorts from northern and southern China. The cohorts consisted of 17,360 outpatients who had undergone upper gastrointestinal endoscopic examination in endoscopic clinics. The final model was derived based on unconditional logistic regression, and predictors were selected according to the Akaike information criterion. External validation was carried out with 32,614 participants from a community-based randomized controlled trial. RESULTS This questionnaire-based diagnostic model for malignant gastric lesions had eight predictors, including advanced age, male gender, family history of gastric cancer, low body mass index, unexplained weight loss, consumption of leftover food, consumption of preserved food, and epigastric pain. This model showed high discriminative power in the development set with an area under the receiver operating characteristic curve (AUC) of 0.791 (95% confidence interval [CI]: 0.750-0.831). External validation of the model in the general population generated an AUC of 0.696 (95% CI: 0.570-0.822). This model showed an ideal ability for enriching prevalent malignant gastric lesions when applied to various scenarios. CONCLUSION This easy-to-use questionnaire-based model for diagnosis of prevalent malignant gastric lesions may serve as an effective prescreening tool in clinical opportunistic screening for gastric cancer.
Collapse
Affiliation(s)
- Hongchen Zheng
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhen Liu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yun Chen
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, Guangdong 516473, China
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 516473, China
| | - Ping Ji
- Clinical Research Institute, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 516473, China
| | - Zhengyu Fang
- Clinical Research Institute, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 516473, China
| | - Yujie He
- Endoscopy Center, Hua County People's Hospital, Anyang, Henan 456483, China
| | - Chuanhai Guo
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ping Xiao
- Clinical Research Institute, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 516473, China
| | - Chengwen Wang
- Endoscope Group, Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 516473, China
| | - Weihua Yin
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 516473, China
| | - Fenglei Li
- Hua County People's Hospital, Anyang, Henan 456483, China
| | - Xiujian Chen
- Department of Pathology, Hua County People's Hospital, Anyang, Henan 456483, China
| | - Mengfei Liu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yaqi Pan
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Fangfang Liu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ying Liu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhonghu He
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yang Ke
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
15
|
Farinati F, Pelizzaro F. Gastric cancer screening in Western countries: A call to action. Dig Liver Dis 2024; 56:1653-1662. [PMID: 38403513 DOI: 10.1016/j.dld.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/11/2024] [Accepted: 02/12/2024] [Indexed: 02/27/2024]
Abstract
Gastric cancer is a major cause of cancer-related death worldwide, despite the reduction in its incidence. The disease is still burdened with a poor prognosis, particularly in Western countries. The main risk factor is the infection by Helicobacter pylori, classified as a class I carcinogen by the IARC, and It is well-known that primary prevention of gastric cancer can be achieved with the eradication of the infection. Moreover, non-invasive measurement of pepsinogens (PGI and PGI/PGII ratio) allows the identification of patients that should undergo upper gastrointestinal (GI) endoscopy. Gastric non-cardia adenocarcinoma is indeed preceded by a well-defined precancerous process that involves consecutive stages, described for the first time by Correa et al. more than 40 years ago, and patients with advance stages of gastric atrophy/intestinal metaplasia and with dysplastic changes should be followed-up periodically with upper GI endoscopies. Despite these effective screening and surveillance methods, national-level screening campaigns have been adopted only in few countries in eastern Asia (Japan and South Korea). In this review, we describe primary and secondary preventive measures for gastric cancer, discussing the need to introduce screening also in Western countries. Moreover, we propose a simple algorithm for screening that could be easily applied in clinical practice.
Collapse
Affiliation(s)
- Fabio Farinati
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, Padova 35128, Italy; Gastroenterology Unit, Azienda Ospedale-Università di Padova, Via Giustiniani 2, Padova 35128, Italy.
| | - Filippo Pelizzaro
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, Padova 35128, Italy; Gastroenterology Unit, Azienda Ospedale-Università di Padova, Via Giustiniani 2, Padova 35128, Italy
| |
Collapse
|
16
|
Alkhateeb MA, Aljarba NH, Yousafi Q, Anwar F, Biswas P. Elucidating gastric cancer mechanisms and therapeutic potential of Adociaquinone A targeting EGFR: A genomic analysis and Computer Aided Drug Design (CADD) approach. J Cell Mol Med 2024; 28:e70133. [PMID: 39434198 PMCID: PMC11493557 DOI: 10.1111/jcmm.70133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/05/2024] [Accepted: 09/09/2024] [Indexed: 10/23/2024] Open
Abstract
Gastric cancer predominantly adenocarcinoma, accounts for over 85% of gastric cancer diagnoses. Current therapeutic options are limited, necessitating the discovery of novel drug targets and effective treatments. The Affymetrix gene expression microarray dataset (GSE64951) was retrieved from NCBI-GEO data normalization and DEGs identification was done by using R-Bioconductor package. Gene Ontology (GO) analysis of DEGs was performed using DAVID. The protein-protein interaction network was constructed by STRING database plugin in Cytoscape. Subclusters/modules of important interacting genes in main network were extracted by using MCODE. The hub genes from in the network were identified by using Cytohubba. The miRNet tool built a hub gene/mRNA-miRNA network and Kaplan-Meier-Plotter conducted survival analysis. AutoDock Vina and GROMACS MD simulations were used for docking and stability analysis of marine compounds against the 5CNN protein. Total 734 DEGs (507 up-regulated and 228 down-regulated) were identified. Differentially expressed genes (DEGs) were enriched in processes like cell-cell adhesion and ATP binding. Eight hub genes (EGFR, HSPA90AA1, MAPK1, HSPA4, PPP2CA, CDKN2A, CDC20, and ATM) were selected for further analysis. A total of 23 miRNAs associated with hub genes were identified, with 12 of them targeting PPP2CA. EGFR displayed the highest expression and hazard rate in survival analyses. The kinase domain of EGFR (PDBID: 5CNN) was chosen as the drug target. Adociaquinone A from Petrosia alfiani, docked with 5CNN, showed the lowest binding energy with stable interactions across a 50 ns MD simulation, highlighting its potential as a lead molecule against EGFR. This study has identified crucial DEGs and hub genes in gastric cancer, proposing novel therapeutic targets. Specifically, Adociaquinone A demonstrates promising potential as a bioactive drug against EGFR in gastric cancer, warranting further investigation. The predicted miRNA against the hub gene/proteins can also be used as potential therapeutic targets.
Collapse
Affiliation(s)
| | - Nada H. Aljarba
- Department of Biology, College of SciencePrincess Nourah bint Abdulrahman UniversityRiyadhSaudi Arabia
| | - Qudsia Yousafi
- Department of BiosciencesCOMSATS University Islamabad, Sahiwal CampusSahiwalPakistan
| | - Fatima Anwar
- Department of BiosciencesCOMSATS University Islamabad, Sahiwal CampusSahiwalPakistan
| | - Partha Biswas
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and BiotechnologyJashore University of Science and TechnologyJashoreBangladesh
| |
Collapse
|
17
|
Yang Y, Du P, Hou X, Yan K, Dai Y, Sun Z, Wu Q, Li S, Yan Y, Wang Z, Qi L, Chen M, Zheng H, Gao W, Gao M, Xue W, Zhang X. Early cancer screening surveillance in one medical center of China. PeerJ 2024; 12:e18179. [PMID: 39351369 PMCID: PMC11441387 DOI: 10.7717/peerj.18179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 09/04/2024] [Indexed: 10/04/2024] Open
Abstract
Objectives Cancer screening aims to detect and treat malignant lesions at an early stage and to prolong patients' lifetime. There is still a lack of effective cancer screening programs in China. We initiated a screening project in 2018 and this study presented the cancer screening status in China. Methods We conducted a cross-sectional study in one cancer-care medical center of China. The screening program included routine blood tests, plasma tumor markers, gastric endoscopy, colonoscopy, ultrasound, and computed tomography (CT) scans. Screening results were presented as sensitivity, specificity and positive predictive values (PPVs). Results Twenty-three (1.46%) out of 1,576 participants were eventually diagnosed with malignant tumors or high-grade intraepithelial neoplasia (HGIN). A family history of malignancy (78.26% in diagnosed cancer and HGIN vs. 46.36% in the others) was the only statistically significant parameter associated with cancer detection (p = 0.002). None of the common tumor markers were associated with the cancers screened. Except for colonoscopy (50.00%) and ultrasound for renal cancer (66.67%), the sensitivities of most screening methods were 100%. The specificities of all the screening means were above 96%. Most PPVs ranged from 30-60%. Conclusion We emphasized risk stratification for early cancer screening, such as a family history of cancer. The survey illustrated that gastric endoscopy, colonoscopy, ultrasound, and lung CT for early cancer screening had high specificity, reasonable sensitivity, and PPV. We anticipated this report would motivate larger-sample studies to estimate the risk-to-benefit ratio of cancer screening and urge the establishment of a native Chinese screening project and even guidelines.
Collapse
Affiliation(s)
- Ying Yang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), The VIP-II Gastrointestinal Cancer Division of Medical Department, Peking University Cancer Hospital & Institute, Beijing, China
| | - Peng Du
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaolu Hou
- Western Beijing Cancer Hospital, Beijing, China
| | - Kun Yan
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Ultrasonography, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ying Dai
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Ultrasonography, Peking University Cancer Hospital & Institute, Beijing, China
| | - ZhiYing Sun
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Ultrasonography, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qi Wu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Endoscopy, Peking University Cancer Hospital & Institute, Beijing, China
| | - Shijie Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Endoscopy, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yan Yan
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Endoscopy, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhilong Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Liping Qi
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Mailin Chen
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hong Zheng
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Gynecologic Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Weijiao Gao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Gynecologic Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Min Gao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Gynecologic Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Weicheng Xue
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaodong Zhang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), The VIP-II Gastrointestinal Cancer Division of Medical Department, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
18
|
Minamitani M, Tatemichi M, Mukai T, Katano A, Ohira S, Nakagawa K. Adherence to national guidelines for colorectal, breast, and cervical cancer screenings in Japanese workplaces: a survey-based classification of enterprises' practices into "overscreening," "underscreening," and "guideline-adherence screening". BMC Public Health 2024; 24:2223. [PMID: 39148101 PMCID: PMC11325713 DOI: 10.1186/s12889-024-19775-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 08/12/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Workplace cancer screening programs are determined as part of an employee's benefits package and health checkups are perceived positively. However, the current status of workplace cancer screening programs in Japan is unavailable. This study aimed to assess the adherence to national guidelines for colorectal, breast, and cervical cancer screenings in the workplace among Japanese enterprises and identify factors associated with excessive or inadequate screenings. METHODS A cross-sectional study design was employed. Data were obtained from a survey conducted by the "Corporate Action to Promote Cancer Control" between November and December 2022 among registered partner enterprises in Japan. The survey included questions on background characteristics, cancer screening practices, and intervention approaches. The analysis included 432 enterprises that provided complete responses regarding colorectal, breast, and cervical cancer screenings. RESULTS The guideline-adherence rates for colorectal, breast, and cervical cancer screenings in the workplace were 12.7%, 3.0%, and 8.8%, respectively. Enterprises had lower adherence to screening guidelines than local governments. Colorectal (70.8%) and breast (67.1%) cancer screenings were predominantly categorized as "overscreening" and cervical (60.6%) cancer screening, as "underscreening." Factors such as enterprise scale, health insurance associations, and the number of interventional approaches were significantly associated with increased "overscreening" (101-1000: β = 0.13, p = 0.01; ≥ 1000: β = 0.17, p < 0.01; health insurance association: β = 0.23, p < 0.01; and approaches: β = 0.42, p < 0.01) and reduced "underscreening" (101-1000: β = -0.13, p = 0.01; ≥ 1000: β = -0.17, p < 0.01; health insurance association: β = -0.18, p < 0.01; and approaches: β = -0.48, p < 0.01). CONCLUSION Adherence to national guidelines for colorectal, breast, and cervical cancer screenings in the workplace was suboptimal among Japanese enterprises. Therefore, appropriate cancer screening measures and interventions to ensure guideline adherence and optimization of screening benefits while minimizing potential harms should be expeditiously implemented.
Collapse
Affiliation(s)
- Masanari Minamitani
- Department of Comprehensive Radiation Oncology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan.
- Department of Radiology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.
| | - Masayuki Tatemichi
- Department of Preventive Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara City, Kanagawa, 259-1193, Japan
| | - Tomoya Mukai
- Department of Psychology, Fukuyama University, 985-1, Sanzo, Higashimura-Machi, Fukuyama-City, Hiroshima, 729-0292, Japan
| | - Atsuto Katano
- Department of Radiology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Shingo Ohira
- Department of Comprehensive Radiation Oncology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
- Department of Radiological Science, Graduate School of Human Health Science, Tokyo Metropolitan University, 7-2-10 Higashi-Ogu, Arakawa-Ku, Tokyo, 116-8551, Japan
| | - Keiichi Nakagawa
- Department of Comprehensive Radiation Oncology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| |
Collapse
|
19
|
Kusano C, Gotoda T, Ishikawa H, Suzuki S, Ikehara H, Matsuyama Y. Gastric cancer detection rates using GI endoscopy with serologic risk stratification: a randomized controlled trial. Gastrointest Endosc 2024; 100:55-63.e1. [PMID: 38272280 DOI: 10.1016/j.gie.2024.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/28/2023] [Accepted: 01/17/2024] [Indexed: 01/27/2024]
Abstract
BACKGROUND AND AIMS Efforts have been made to develop an endoscopic screening system incorporating serologic gastric cancer (GC) risk stratification (ABC classification) alongside annual population-based GC screening using barium. We conducted a randomized controlled trial (RCT) to compare GC detection rates between the Ba-Endo group, which underwent annual barium tests for primary screening followed by detailed endoscopic examinations, and the ABC-Endo group, where endoscopy intervals were determined based on individual gastric cancer risk in the ABC classification. METHODS In total, 1206 individuals from Yurihonjo and Nikaho City, Akita Prefecture, were randomized through the minimization method using sex and age as allocation factors. The intervention study was conducted for both groups over 5 years. The Ba-Endo group received annual barium tests, and the ABC-Endo group underwent EGD at different intervals: group A, EGD only at entry; group B, EGD once every 3 years; group C, EGD once every 2 years; and group D, EGD every year. RESULTS There were 24 detected GC lesions, with a GC detection rate of 1.9%. GC detection rates in the Ba-Endo and ABC-Endo groups were 2.0% and 1.8%, respectively, with no significant differences between groups (P = 1.0). However, the rate of GC cured by endoscopic resection alone was 41.6% in the Ba-Endo group and was significantly higher at 90.9% in the ABC-Endo group (P = .02). CONCLUSIONS There were no differences between the Ba-Endo and ABC-Endo groups in GC detection rates. However, the rate of detected GCs that could be cured by endoscopic resection alone was significantly higher in the ABC-Endo group. (Clinical trial registration number: UMIN000005962.).
Collapse
Affiliation(s)
- Chika Kusano
- Department of Gastroenterology, Kitasato University School of Medicine, Kanagawa, Japan.
| | - Takuji Gotoda
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan; Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hideki Ishikawa
- Department of Molecular-Targeting Cancer Prevention, Kyoto Prefectural University of Medicine, Osaka, Japan
| | - Sho Suzuki
- Department of Gastroenterology, International University of Health and Welfare Ichikawa Hospital, Chiba, Japan
| | - Hisatomo Ikehara
- Department of Gastroenterology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Yutaka Matsuyama
- Department of Biostatistics, School of Public Health, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
20
|
Fu Y, Jiang J, Wu Y, Cao D, Jia Z, Zhang Y, Li D, Cui Y, Zhang Y, Cao X. Genome-wide 5-hydroxymethylcytosines in circulating cell-free DNA as noninvasive diagnostic markers for gastric cancer. Gastric Cancer 2024; 27:735-746. [PMID: 38584223 DOI: 10.1007/s10120-024-01493-7] [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: 11/13/2023] [Accepted: 03/14/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND 5-Hydroxymethylcytosine-enriched gene profiles and regions show tissue-specific and tumor specific. There is a potential value to explore cell-free DNA 5-hydroxymethylcytosine feature biomarkers for early gastric cancer detection. METHODS A matched case‒control study design with 50 gastric cancer patients and 50 controls was performed to sequence the different 5-hydroxymethylcytosine modification features of cell free DNA. Significantly differential 5-hydroxymethylcytosine modification genes were identified to construct a gastric cancer diagnostic model. Data set from GEO was used as an external testing set to test the robustness of the diagnostic model. RESULTS Accounting for more than 90% of 5-hydroxymethylcytosine peaks were distributed in the gene body in both the gastric cancer and control groups. The diagnostic model was developed based on five different 5-hydroxymethylcytosine modification genes, FBXL7, PDE3A, TPO, SNTG2 and STXBP5. The model could effectively distinguish gastric cancer patients from controls in the training (AUC = 0.95, sensitivity = 88.6%, specificity = 94.3%), validation (AUC = 0.87, sensitivity = 73.3%, specificity = 93.3%) and testing (AUC = 0.90, sensitivity = 81.9%, specificity = 90.2%) sets. The risk scores of the controls from the model were significantly lower than those of gastric cancer patients in both our own data (P < 0.001) and GEO external testing data (P < 0.001), and no significant difference between different TNM stage patients (P = 0.09 and 0.66). Furthermore, there was no significant difference between the healthy control and benign gastric disease patients in the testing set from GEO (P = 0.10). CONCLUSIONS The characteristics of 5-hydroxymethylcytosine in cell free DNA are specific to gastric cancer patients, and the diagnostic model constructed by five genes' 5-hydroxymethylcytosine features could effectively identify gastric cancer patients.
Collapse
Affiliation(s)
- Yingli Fu
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Jing Jiang
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Yanhua Wu
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Donghui Cao
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Zhifang Jia
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Yangyu Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Dongming Li
- Department of Hospital Infection Management, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yingnan Cui
- Department of Hospital Infection Management, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yuzheng Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
- Department of Hospital Infection Management, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xueyuan Cao
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
21
|
Wei H, Li W, Zeng L, Ding N, Li K, Yu H, Jiang F, Yin H, Xia Y, Deng C, Cai N, Chen X, Gu L, Chen H, Zhang F, He Y, Li J, Zhang C. OLFM4 promotes the progression of intestinal metaplasia through activation of the MYH9/GSK3β/β-catenin pathway. Mol Cancer 2024; 23:124. [PMID: 38849840 PMCID: PMC11157765 DOI: 10.1186/s12943-024-02016-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/04/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Intestinal metaplasia (IM) is classified into complete intestinal metaplasia (CIM) and incomplete intestinal metaplasia (IIM). Patients diagnosed with IIM face an elevated susceptibility to the development of gastric cancer, underscoring the critical need for early screening measures. In addition to the complexities associated with diagnosis, the exact mechanisms driving the progression of gastric cancer in IIM patients remain poorly understood. OLFM4 is overexpressed in several types of tumors, including colorectal, gastric, pancreatic, and ovarian cancers, and its expression has been associated with tumor progression. METHODS In this study, we used pathological sections from two clinical centers, biopsies of IM tissues, precancerous lesions of gastric cancer (PLGC) cell models, animal models, and organoids to explore the role of OLFM4 in IIM. RESULTS Our results show that OLFM4 expression is highly increased in IIM, with superior diagnostic accuracy of IIM when compared to CDX2 and MUC2. OLFM4, along with MYH9, was overexpressed in IM organoids and PLGC animal models. Furthermore, OLFM4, in combination with Myosin heavy chain 9 (MYH9), accelerated the ubiquitination of GSK3β and resulted in increased β-catenin levels through the Wnt signaling pathway, promoting the proliferation and invasion abilities of PLGC cells. CONCLUSIONS OLFM4 represents a novel biomarker for IIM and could be utilized as an important auxiliary means to delimit the key population for early gastric cancer screening. Finally, our study identifies cell signaling pathways involved in the progression of IM.
Collapse
Affiliation(s)
- Hongfa Wei
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Jinping, Shantou, Guangdong, 515041, P.R. China
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Wenchao Li
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- The Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Leli Zeng
- Scientific Research Center, The Biobank, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, P.R. China
| | - Ni Ding
- Scientific Research Center, The Biobank, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, P.R. China
- The Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Kuan Li
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Hong Yu
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Fei Jiang
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Haofan Yin
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- Department of Laboratory Medicine, Shenzhen People's Hospital, (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Yu Xia
- Scientific Research Center, The Biobank, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, P.R. China
| | - Cuncan Deng
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Nan Cai
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Xiancong Chen
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Liang Gu
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Huanjie Chen
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Jinping, Shantou, Guangdong, 515041, P.R. China
| | - Feiran Zhang
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Jinping, Shantou, Guangdong, 515041, P.R. China.
| | - Yulong He
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Jia Li
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
- Scientific Research Center, The Biobank, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, P.R. China.
| | - Changhua Zhang
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
22
|
Lin J, Zhu F, Dong X, Li R, Liu J, Xia J. Enhancing gastric cancer early detection: A multi-verse optimized feature selection model with crossover-information feedback. Comput Biol Med 2024; 175:108535. [PMID: 38714049 DOI: 10.1016/j.compbiomed.2024.108535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/05/2024] [Accepted: 04/28/2024] [Indexed: 05/09/2024]
Abstract
Gastric cancer (GC), an acknowledged malignant neoplasm, threatens life and digestive system functionality if not detected and addressed promptly in its nascent stages. The indispensability of early detection for GC to augment treatment efficacy and survival prospects forms the crux of this investigation. Our study introduces an innovative wrapper-based feature selection methodology, referred to as bCIFMVO-FKNN-FS, which integrates a crossover-information feedback multi-verse optimizer (CIFMVO) with the fuzzy k-nearest neighbors (FKNN) classifier. The primary goal of this initiative is to develop an advanced screening model designed to accelerate the identification of patients with early-stage GC. Initially, the capability of CIFMVO is validated through its application to the IEEE CEC benchmark functions, during which its optimization efficiency is measured against eleven cutting-edge algorithms across various dimensionalities-10, 30, 50, and 100. Subsequent application of the bCIFMVO-FKNN-FS model to the clinical data of 1632 individuals from Wenzhou Central Hospital-diagnosed with either early-stage GC or chronic gastritis-demonstrates the model's formidable predictive accuracy (83.395%) and sensitivity (87.538%). Concurrently, this investigation delineates age, gender, serum gastrin-17, serum pepsinogen I, and the serum pepsinogen I to serum pepsinogen II ratio as parameters significantly associated with early-stage GC. These insights not only validate the efficacy of our proposed model in the early screening of GC but also contribute substantively to the corpus of knowledge facilitating early diagnosis.
Collapse
Affiliation(s)
- Jiejun Lin
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University (Wenzhou Central Hospital), Wenzhou, Zhejiang, 325000, China.
| | - Fangchao Zhu
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University (Wenzhou Central Hospital), Wenzhou, Zhejiang, 325000, China.
| | - Xiaoyu Dong
- Department of Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| | - Rizeng Li
- Department of General Surgery, The Dingli Clinical College of Wenzhou Medical University (Wenzhou Central Hospital), Wenzhou, Zhejiang, 325000, China.
| | - Jisheng Liu
- Department of General Surgery, The Dingli Clinical College of Wenzhou Medical University (Wenzhou Central Hospital), Wenzhou, Zhejiang, 325000, China.
| | - Jianfu Xia
- Department of General Surgery, The Dingli Clinical College of Wenzhou Medical University (Wenzhou Central Hospital), Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
23
|
Wu BU, Dong EY, Chen Q, Luong TQ, Lustigova E, Jeon CY, Chen W. Stomach Cancer Prediction Model (SCoPM): An approach to risk stratification in a diverse U.S. population. PLoS One 2024; 19:e0303153. [PMID: 38771811 PMCID: PMC11108155 DOI: 10.1371/journal.pone.0303153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 04/20/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND AND AIMS Population-based screening for gastric cancer (GC) in low prevalence nations is not recommended. The objective of this study was to develop a risk-prediction model to identify high-risk patients who could potentially benefit from targeted screening in a racial/ethnically diverse regional US population. METHODS We performed a retrospective cohort study from Kaiser Permanente Southern California from January 2008-June 2018 among individuals age ≥50 years. Patients with prior GC or follow-up <30 days were excluded. Censoring occurred at GC, death, age 85 years, disenrollment, end of 5-year follow-up, or study conclusion. Cross-validated LASSO regression models were developed to identify the strongest of 20 candidate predictors (clinical, demographic, and laboratory parameters). Records from 12 of the medical service areas were used for training/initial validation while records from a separate medical service area were used for testing. RESULTS 1,844,643 individuals formed the study cohort (1,555,392 training and validation, 289,251 testing). Mean age was 61.9 years with 53.3% female. GC incidence was 2.1 (95% CI 2.0-2.2) cases per 10,000 person-years (pyr). Higher incidence was seen with family history: 4.8/10,000 pyr, history of gastric ulcer: 5.3/10,000 pyr, H. pylori: 3.6/10,000 pyr and anemia: 5.3/10,000 pyr. The final model included age, gender, race/ethnicity, smoking, proton-pump inhibitor, family history of gastric cancer, history of gastric ulcer, H. pylori infection, and baseline hemoglobin. The means and standard deviations (SD) of c-index in validation and testing datasets were 0.75 (SD 0.03) and 0.76 (SD 0.02), respectively. CONCLUSIONS This prediction model may serve as an aid for pre-endoscopic assessment of GC risk for identification of a high-risk population that could benefit from targeted screening.
Collapse
Affiliation(s)
- Bechien U. Wu
- Center for Digestive Health, Department of Gastroenterology, Los Angeles Medical Center, Los Angeles, CA, United States of America
| | - Elizabeth Y. Dong
- Department of Gastroenterology, Southern California Permanente Medical Group, Los Angeles, CA, United States of America
| | - Qiaoling Chen
- Department of Research & Evaluation, Southern California Permanente Medical Group, Pasadena, CA, United States of America
| | - Tiffany Q. Luong
- Department of Research & Evaluation, Southern California Permanente Medical Group, Pasadena, CA, United States of America
| | - Eva Lustigova
- Department of Research & Evaluation, Southern California Permanente Medical Group, Pasadena, CA, United States of America
| | - Christie Y. Jeon
- Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Wansu Chen
- Department of Research & Evaluation, Southern California Permanente Medical Group, Pasadena, CA, United States of America
| |
Collapse
|
24
|
Li M, Gao N, Wang SL, Guo YF, Liu Z. Hotspots and trends of risk factors in gastric cancer: A visualization and bibliometric analysis. World J Gastrointest Oncol 2024; 16:2200-2218. [PMID: 38764808 PMCID: PMC11099465 DOI: 10.4251/wjgo.v16.i5.2200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/08/2024] [Accepted: 03/11/2024] [Indexed: 05/09/2024] Open
Abstract
BACKGROUND The lack of specific symptoms of gastric cancer (GC) causes great challenges in its early diagnosis. Thus it is essential to identify the risk factors for early diagnosis and treatment of GC and to improve the survival rates. AIM To assist physicians in identifying changes in the output of publications and research hotspots related to risk factors for GC, constructing a list of key risk factors, and providing a reference for early identification of patients at high risk for GC. METHODS Research articles on risk factors for GC were searched in the Web of Science core collection, and relevant information was extracted after screening. The literature was analyzed using Microsoft Excel 2019, CiteSpace V, and VOSviewer 1.6.18. RESULTS A total of 2514 papers from 72 countries and 2507 research institutions were retrieved. China (n = 1061), National Cancer Center (n = 138), and Shoichiro Tsugane (n = 36) were the most productive country, institution, or author, respectively. The research hotspots in the study of risk factors for GC are summarized in four areas, namely: Helicobacter pylori (H. pylori) infection, single nucleotide polymorphism, bio-diagnostic markers, and GC risk prediction models. CONCLUSION In this study, we found that H. pylori infection is the most significant risk factor for GC; single-nucleotide polymorphism (SNP) is the most dominant genetic factor for GC; bio-diagnostic markers are the most promising diagnostic modality for GC. GC risk prediction models are the latest current research hotspot. We conclude that the most important risk factors for the development of GC are H. pylori infection, SNP, smoking, diet, and alcohol.
Collapse
Affiliation(s)
- Meng Li
- Department of Gastroenterology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ning Gao
- Department of Acupuncture and Moxibustion, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Shao-Li Wang
- Department of Gastroenterology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yu-Feng Guo
- Department of Acupuncture and Moxibustion, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Zhen Liu
- Department of Gastroenterology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| |
Collapse
|
25
|
Sun YX, Tang T, Zou JY, Yue QQ, Hu LF, Peng T, Meng XR, Feng GH, Huang LL, Zeng Y. Interventions to Improve Endoscopic Screening Adherence of Cancer in High-Risk Populations: A Scoping Review. Patient Prefer Adherence 2024; 18:709-720. [PMID: 38524198 PMCID: PMC10961025 DOI: 10.2147/ppa.s443607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 03/07/2024] [Indexed: 03/26/2024] Open
Abstract
Background Colorectal, and gastric cancers have the second, and fourth mortality rates worldwide, respectively. Endoscopic screening is a crucial diagnostic tool for colorectal, and gastric cancers. Effective interventions can improve adherence to endoscopic screening in high-risk populations, which is important for cancer prevention and mortality reduction. This study aimed to identify interventions that could improve adherence to endoscopic screening for cancer in high-risk populations. Methods Combination keywords including colorectal cancer, gastric cancer, screening adherence, and interventions were used to search for articles in PubMed, Web of Science, Cochrane Library, and MEDLINE Complete. The review methodology was guided by the Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-SCR). Results A total of 12 articles were included in this review: 9 randomized controlled trials(RCT) and 3 quasi-experimental studies(QEDs). Among the extracted studies, 11 were about colorectal cancer, and 1 was about gastric cancer. Most studies used lecture-based or Information Technology-based health education interventions. Narrative interventions have proven to be novel and effective approaches for promoting adherence to endoscopic screening. Health education interventions included cancer epidemiology, cancer risk factors, warning symptoms, and screening methods. Conclusion All interventions involved were effective in increasing individual knowledge of cancer-related endoscopic screening, willingness to undergo screening, and screening behaviors. These findings provide a reference for designing endoscopy-related cancer screening interventions.
Collapse
Affiliation(s)
- Ying-Xue Sun
- Department of International and Humanistic Nursing, School of Nursing, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Tian Tang
- Department of International and Humanistic Nursing, School of Nursing, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Jin-Yu Zou
- Department of International and Humanistic Nursing, School of Nursing, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Qian-Qian Yue
- Department of International and Humanistic Nursing, School of Nursing, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Li-Feng Hu
- Department of International and Humanistic Nursing, School of Nursing, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Tong Peng
- Department of International and Humanistic Nursing, School of Nursing, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Xin-Ru Meng
- Department of International and Humanistic Nursing, School of Nursing, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Ge-Hui Feng
- Department of International and Humanistic Nursing, School of Nursing, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Li-Li Huang
- Department of International and Humanistic Nursing, School of Nursing, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Ying Zeng
- Department of International and Humanistic Nursing, School of Nursing, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, People's Republic of China
| |
Collapse
|
26
|
Vu NTH, Urabe Y, Quach DT, Oka S, Hiyama T. Population-based X-ray gastric cancer screening in Hiroshima prefecture, Japan. World J Clin Oncol 2024; 15:271-281. [PMID: 38455140 PMCID: PMC10915947 DOI: 10.5306/wjco.v15.i2.271] [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: 10/23/2023] [Revised: 12/11/2023] [Accepted: 01/10/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND X-ray gastric cancer (GC) screening has been shown to decrease mortality. Population-based X-ray GC screening has been performed in Hiroshima Prefecture, Japan, since 1983 but time trends and the efficacy of the method over 39 years have not been assessed. AIM To evaluate time trends and efficacy of population-based X-ray GC screening and identify challenges and suggested solutions for the future. METHODS This was a population-based retrospective study. The data were derived from aggregated data of the Hiroshima Regional Health Medical Promotion Organization, including the number and rate of participants and those requiring esophagogastroduodenoscopies (EGDs), the number and rate of participants diagnosed as having GC, and the positive predictive value of the abnormal findings detected by X-ray and confirmed by EGDs. The number and rate of esophageal cancers were also collected. Further, the cost of detecting one GC was evaluated. RESULTS The number of participants has decreased during the last four decades, from 39925 in 1983 to 12923 in 2021. The rate of those requiring EGDs decreased significantly in recent years (P < 0.001). The number of participants diagnosed as having GC has also declined, from 76 to 10 cases. However, the rate of cases diagnosed as GC among the participants remained around 0.1%. The positive predictive value increased significantly in recent years except during 1983-1991. The number and rate of accidentally detected esophageal cancers have risen recently, from 0% in 2008 to 0.02% in 2021, one-fifth of the diagnosis rate of GC. One GC diagnosis costs approximately 4200000 Japanese Yen (30000 United States Dollars) for the X-ray screenings and EGDs. CONCLUSION X-ray GC screening in Hiroshima has been efficient, but one challenge is the cost. Esophageal cancers may also need to be considered because they have gradually increased in recent years.
Collapse
Affiliation(s)
- Nhu Thi Hanh Vu
- Department of Internal Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh 700000, Viet Nam
- Department of Endoscopy, Hiroshima University Hospital, Hiroshima 734-8551, Japan
| | - Yuji Urabe
- Department of Gastrointestinal Endoscopy and Medicine, Hiroshima University Hospital, Hiroshima 734-8551, Japan
| | - Duc Trong Quach
- Department of Internal Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh 700000, Viet Nam
| | - Shiro Oka
- Department of Gastroenterology, Hiroshima University Hospital, Hiroshima 734-8551, Japan
| | - Toru Hiyama
- Health Service Center, Hiroshima University, Higashihiroshima 739-8514, Japan
| |
Collapse
|
27
|
Wang CY, Fan XJ, Wang FL, Ge YY, Cai Z, Wang W, Zhou XP, Du J, Dai DW. Clinical value of oral contrast-enhanced ultrasonography in diagnosis of gastric tumors. World J Gastrointest Oncol 2024; 16:110-117. [PMID: 38292839 PMCID: PMC10824109 DOI: 10.4251/wjgo.v16.i1.110] [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: 09/14/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND The incidence of gastric cancer remains high, and it is the sixth most common cancer and the fourth leading cause of cancer deaths worldwide. Oral contrast-enhanced ultrasonography is a simple, non-invasive, and painless method for the diagnosis of gastric tumors. AIM To explore the diagnostic value of oral contrast-enhanced ultrasonography for the detection of gastric tumors. METHODS The screening results based on oral contrast-enhanced ultrasonography and electronic gastroscopy were compared with those of the postoperative pathological examination. RESULTS Among 42 patients with gastric tumors enrolled in the study, the diagnostic accordance rate was 95.2% for oral contrast-enhanced ultrasonography (n = 40) and 90.5% for electronic gastroscopy (n = 38) compared with postoperative pathological examination. The Kappa value of consistency test with pathological findings was 0.812 for oral contrast-enhanced ultrasonography and 0.718 for electronic gastroscopy, and there was no significant difference between them (P = 0.397). For the TNM staging of gastric tumors, the accuracy rate of oral contrast-enhanced ultrasonography was 81.9% for the overall T staging and 50%, 77.8%, 100%, and 100% for T1, T2, T3, and T4 staging, respectively. The sensitivity and specificity were both 100% for stages T3 and T4. The diagnostic accuracy rate of oral contrast-enhanced ultrasonography was 93.8%, 80%, 100%, and 100% for stages N0, N1-N3, M0, and M1, respectively. CONCLUSION The accordance rate of qualitative diagnosis by oral contrast-enhanced ultrasonography is comparable to that of gastroscopy, and it could be used as the preferred method for the early screening of gastric tumors.
Collapse
Affiliation(s)
- Chuan-Yu Wang
- Department of Ultrasound Medicine, Beijing Hospital, Beijing 100005, China
| | - Xiao-Jing Fan
- Department of Ultrasound Medicine, Beijing Hospital, Beijing 100005, China
| | - Fei-Liang Wang
- Department of Ultrasound Medicine, Beijing Hospital, Beijing 100005, China
| | - Yue-Yue Ge
- Department of Ultrasound Medicine, Beijing Hospital, Beijing 100005, China
| | - Zhao Cai
- Department of Ultrasound Medicine, Beijing Hospital, Beijing 100005, China
| | - Wei Wang
- Department of Gastroscopy, Beijing Hospital, Beijing 100005, China
| | - Xin-Ping Zhou
- Department of General Surgery, Beijing Hospital, Beijing 100005, China
| | - Jun Du
- Department of Pathology, Beijing Hospital, Beijing 100005, China
| | - De-Wei Dai
- Department of Ultrasound Medicine, Beijing Hospital, Beijing 100005, China
| |
Collapse
|
28
|
Yan SY, Fu XY, Tang SP, Qi RB, Liang JW, Mao XL, Ye LP, Li SW. A feasibility study on utilizing machine learning technology to reduce the costs of gastric cancer screening in Taizhou, China. Digit Health 2024; 10:20552076241277713. [PMID: 39247098 PMCID: PMC11378168 DOI: 10.1177/20552076241277713] [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: 01/28/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024] Open
Abstract
Aim To optimize gastric cancer screening score and reduce screening costs using machine learning models. Methods This study included 228,634 patients from the Taizhou Gastric Cancer Screening Program. We used three machine learning models to optimize Li's gastric cancer screening score: Gradient Boosting Machine (GBM), Distributed Random Forest (DRF), and Deep Learning (DL). The performance of the binary classification models was evaluated using the area under the curve (AUC) and area under the precision-recall curve (AUCPR). Results In the binary classification model used to distinguish low-risk and moderate- to high-risk patients, the AUC in the GBM, DRF, and DL full models were 0.9994, 0.9982, and 0.9974, respectively, and the AUCPR was 0.9982, 0.9949, and 0.9918, respectively. Excluding Helicobacter pylori IgG antibody, pepsinogen I, and pepsinogen II, the AUC in the GBM, DRF, and DL models were 0.9932, 0.9879, and 0.9900, respectively, and the AUCPR was 0.9835, 0.9716, and 0.9752, respectively. Remodel after removing variables IgG, PGI, PGII, and G-17, the AUC in GBM, DRF, and DL was 0.8524, 0.8482, 0.8477, and AUCPR was 0.6068, 0.6008, and 0.5890, respectively. When constructing a tri-classification model, we discovered that none of the three machine learning models could effectively distinguish between patients at intermediate and high risk for gastric cancer (F1 scores in the GBM model for the low, medium and high risk: 0.9750, 0.9193, 0.5334, respectively; F1 scores in the DRF model for low, medium, and high risks: 0.9888, 0.9479, 0.6694, respectively; F1 scores in the DL model for low, medium, and high risks: 0.9812, 0.9216, 0.6394, respectively). Conclusion We concluded that gastric cancer screening indicators could be optimized when distinguishing low-risk and moderate to high-risk populations, and detecting gastrin-17 alone can achieve a good discriminative effect, thus saving huge expenditures.
Collapse
Affiliation(s)
- Si-Yan Yan
- Taizhou Hospital of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xin-Yu Fu
- Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Shen-Ping Tang
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Rong-Bin Qi
- Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Jia-Wei Liang
- Department of Thoracic Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai Zhejiang, China
| | - Xin-Li Mao
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Li-Ping Ye
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Shao-Wei Li
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| |
Collapse
|
29
|
Ferrari C, Tadros M. Enhancing the Quality of Upper Gastrointestinal Endoscopy: Current Indicators and Future Trends. GASTROENTEROLOGY INSIGHTS 2023; 15:1-18. [DOI: 10.3390/gastroent15010001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
The quality of upper gastrointestinal endoscopy (EGD) is crucial and carries significant consequences for patient outcomes, the employment of healthcare resources, and the future course of gastroenterology as a medical specialty. In this review, we navigate through the terrain of the Quality Indicators (QIs) for EGD, shedding light on their indispensable function in ensuring and augmenting the quality of patient care throughout the pre-procedural, intra-procedural, post-procedural, and outcome-oriented facets of the practice. We delve into the comprehensive scope of the QIs and the challenges impeding the delivery of high-quality EGD, from variability in practitioner training and patient compliance to the systemic limitations of current QIs and the barriers hindering the adoption of advanced techniques. Future directions for bolstering the quality of EGD are highlighted, encapsulating the integration of emergent endoscopic technologies, the evolution of patient-centered metrics, the refinement of endoscopist training and credentialing processes, and the promise held by Artificial Intelligence (AI). Particular emphasis is placed on the role of advanced endoscopic techniques and equipment in enhancing EGD quality. This article presents a cogent narrative, promoting the pursuit of excellence in EGD as an ever-evolving endeavor that necessitates the collective dedication of clinicians, researchers, educators, and policymakers.
Collapse
Affiliation(s)
- Caesar Ferrari
- MD Program, Albany Medical College, Albany, NY 12208, USA
| | - Micheal Tadros
- Department of Gastroenterology and Hepatology, Albany Medical College, Albany, NY 12208, USA
| |
Collapse
|
30
|
Huang KK, Ma H, Chong RHH, Uchihara T, Lian BSX, Zhu F, Sheng T, Srivastava S, Tay ST, Sundar R, Tan ALK, Ong X, Lee M, Ho SWT, Lesluyes T, Ashktorab H, Smoot D, Van Loo P, Chua JS, Ramnarayanan K, Lau LHS, Gotoda T, Kim HS, Ang TL, Khor C, Lee JWJ, Tsao SKK, Yang WL, Teh M, Chung H, So JBY, Yeoh KG, Tan P. Spatiotemporal genomic profiling of intestinal metaplasia reveals clonal dynamics of gastric cancer progression. Cancer Cell 2023; 41:2019-2037.e8. [PMID: 37890493 PMCID: PMC10729843 DOI: 10.1016/j.ccell.2023.10.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 08/08/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023]
Abstract
Intestinal metaplasia (IM) is a pre-malignant condition of the gastric mucosa associated with increased gastric cancer (GC) risk. Analyzing 1,256 gastric samples (1,152 IMs) across 692 subjects from a prospective 10-year study, we identify 26 IM driver genes in diverse pathways including chromatin regulation (ARID1A) and intestinal homeostasis (SOX9). Single-cell and spatial profiles highlight changes in tissue ecology and IM lineage heterogeneity, including an intestinal stem-cell dominant cellular compartment linked to early malignancy. Expanded transcriptome profiling reveals expression-based molecular subtypes of IM associated with incomplete histology, antral/intestinal cell types, ARID1A mutations, inflammation, and microbial communities normally associated with the healthy oral tract. We demonstrate that combined clinical-genomic models outperform clinical-only models in predicting IMs likely to transform to GC. By highlighting strategies for accurately identifying IM patients at high GC risk and a role for microbial dysbiosis in IM progression, our results raise opportunities for GC precision prevention and interception.
Collapse
Affiliation(s)
- Kie Kyon Huang
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Haoran Ma
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Roxanne Hui Heng Chong
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Tomoyuki Uchihara
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Benedict Shi Xiang Lian
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Feng Zhu
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Taotao Sheng
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Supriya Srivastava
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Su Ting Tay
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Raghav Sundar
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; Department of Haematology-Oncology, National University Health System, Singapore 119074, Singapore
| | - Angie Lay Keng Tan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Xuewen Ong
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Minghui Lee
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Shamaine Wei Ting Ho
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | | | | | - Duane Smoot
- Department of Internal Medicine, Meharry Medical College, Nashville, TN, USA
| | - Peter Van Loo
- The Francis Crick Institute, London, UK; Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joy Shijia Chua
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Kalpana Ramnarayanan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Louis Ho Shing Lau
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Takuji Gotoda
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Hyun Soo Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Seoul, Korea
| | - Tiing Leong Ang
- Department of Gastroenterology & Hepatology, Changi General Hospital, Singapore 529889, Singapore
| | - Christopher Khor
- Department of Gastroenterology & Hepatology, Singapore General Hospital, Singapore 169854, Singapore
| | - Jonathan Wei Jie Lee
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; iHealthtech, National University of Singapore, Singapore, Singapore; SynCTI, National University of Singapore, Singapore 117599, Singapore; Department of Gastroenterology & Hepatology, National University Hospital, Singapore 119074, Singapore
| | - Stephen Kin Kwok Tsao
- Department of Gastroenterology & Hepatology, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - Wei Lyn Yang
- Department of Gastroenterology & Hepatology, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - Ming Teh
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Hyunsoo Chung
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.
| | - Jimmy Bok Yan So
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Division of Surgical Oncology, National University Cancer Institute of Singapore (NCIS), Singapore, Singapore.
| | - Khay Guan Yeoh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; Department of Gastroenterology & Hepatology, National University Hospital, Singapore 119074, Singapore.
| | - Patrick Tan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore; Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; Cellular and Molecular Research, National Cancer Centre, Singapore, Singapore; Singhealth/Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore 168752, Singapore.
| |
Collapse
|
31
|
Peng D, Xiang YC, Tang KL, Qiu YY. Impact of preoperative type 2 diabetes mellitus on the outcomes of patients with gastric cancer following gastrectomy: Analysis of 834 patients using propensity score matching. Biomed Rep 2023; 19:97. [PMID: 37954633 PMCID: PMC10633813 DOI: 10.3892/br.2023.1679] [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: 06/29/2023] [Accepted: 09/26/2023] [Indexed: 11/14/2023] Open
Abstract
The purpose of the current study was to compare the outcomes of patients with gastric cancer (GC) between the type 2 diabetes mellitus (T2DM) group and the non-T2DM group. The PubMed, Embase and Cochrane Library databases were searched from inception to March 8, 2022, to identify propensity score matching (PSM) studies that analyzed the effect of T2DM on the outcomes of patients with GC. Total complications, overall survival (OS), disease-free survival (DFS) and cancer-specific survival (CSS) were compared between the T2DM group and the non-T2DM group. A total of four PSM studies with 834 patients were included in the current study. There were 311 and 523 patients in the T2DM group and the non-T2DM group, respectively. Baseline characteristics of the two groups were adjusted with PSM in all the four studies, however, no significant difference was found in baseline characteristics (P>0.05). DFS was significantly worse in the T2DM group compared with that in the non-T2DM group [hazard ratio (HR), 1.45; 95% confidence interval (CI), 1.10-1.90; P=0.007)]. However, after pooling up the data, there was no significant difference between the T2DM group and the non-T2DM group in terms of OS (HR, 1.41; 95% CI, 0.92-2.16; P=0.11), CSS (HR, 1.29; 95% CI, 0.92-1.81; P=0.14) and total complications (odds ratio, 1.01; 95% CI, 0.64-1.60; P=0.95). Patients with GC and T2DM are associated with poor DFS. However, there were no significant differences between the T2DM group and the non-T2DM group in terms of OS, CSS and total complications.
Collapse
Affiliation(s)
- Dong Peng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Ying-Chun Xiang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Kai-Lin Tang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yan-Yu Qiu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
32
|
Xia C, Basu P, Kramer BS, Li H, Qu C, Yu XQ, Canfell K, Qiao Y, Armstrong BK, Chen W. Cancer screening in China: a steep road from evidence to implementation. Lancet Public Health 2023; 8:e996-e1005. [PMID: 38000379 PMCID: PMC10665203 DOI: 10.1016/s2468-2667(23)00186-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/19/2023] [Accepted: 08/08/2023] [Indexed: 11/26/2023]
Abstract
Cancer screening has the potential to decrease mortality from several common cancer types. The first cancer screening programme in China was initiated in 1958 and the Cancer High Incidence Fields established in the 1970s have provided an extensive source of information for national cancer screening programmes. From 2012 onwards, four ongoing national cancer screening programmes have targeted eight cancer types: cervical, breast, colorectal, lung, oesophageal, stomach, liver, and nasopharyngeal cancers. By synthesising evidence from pilot screening programmes and population-based studies for various screening tests, China has developed a series of cancer screening guidelines. Nevertheless, challenges remain for the implementation of a fully successful population-based programme. The aim of this Review is to highlight the key milestones and the current status of cancer screening in China, describe what has been achieved to date, and identify the barriers in transitioning from evidence to implementation. We also make a set of implementation recommendations on the basis of the Chinese experience, which might be useful in the establishment of cancer screening programmes in other countries.
Collapse
Affiliation(s)
- Changfa Xia
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Partha Basu
- Early Detection, Prevention & Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - Barnett S Kramer
- The Lisa Schwartz Foundation for Truth in Medicine, Hanover, NH, USA
| | - He Li
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunfeng Qu
- State Key Lab of Molecular Oncology and Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xue Qin Yu
- The Daffodil Centre, The University of Sydney, A Joint Venture with Cancer Council NSW, Sydney, NSW, Australia
| | - Karen Canfell
- The Daffodil Centre, The University of Sydney, A Joint Venture with Cancer Council NSW, Sydney, NSW, Australia
| | - Youlin Qiao
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bruce K Armstrong
- School of Public Health, University of Sydney, Sydney, NSW, Australia; School of Global and Population Health, University of Western Australia, Perth, WA, Australia
| | - Wanqing Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
33
|
Wang S, Qian M, Wu M, Feng S, Zhang K. The prediction model of operative link on gastric intestinal metaplasia stage III-IV: A multicenter study. Heliyon 2023; 9:e21905. [PMID: 38027917 PMCID: PMC10665748 DOI: 10.1016/j.heliyon.2023.e21905] [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: 07/29/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Intestinal metaplasia plays a crucial role in the risk stratification of gastric cancer development. The objective of the study was to develop a prediction model for Operative Link on Gastric Intestinal Metaplasia (OLGIM) Stage III-IV. Methods We analyzed 7945 high-risk gastric cancer individuals from 115 hospitals who underwent questionnaires and gastroscope. The participants were assigned to either the development or validation cohort randomly. Demographics and clinical characteristics were obtained. The outcome measurement was OLGIM III-IV. Univariate logistic regression was used for feature selection and multivariate logistic analysis was performed to develop the nomogram. Area under the curves, calibration plots, decision curve and clinical impact analysis were used to assess the performance of the nomogram. Results 4600 individuals and 3345 individuals were included in the development and validation cohort, of which 124 and 86 individuals were diagnosed with OLGIM III-IV, respectively. Parameters in the training validation cohort matched well and there was no significant difference between two cohorts. A nomogram model for predicting OLGIM Stage III-IV consisted of 4 significantly associated variables, including age, gender, PG I and G-17 (AUC 0.723 and 0.700 for the 2 cohorts). The nomogram demonstrated excellent performance in the calibration curve. Decision curve and clinical impact analysis suggested clinical benefit of the prediction model. Conclusions This reliable individualized nomogram might contribute to more accurate management for patients with OLGIM III-IV. Therefore, we suggest that this study be used as an incentive to promote the application.
Collapse
Affiliation(s)
- Song Wang
- Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Meng Qian
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Min Wu
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuo Feng
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kaiguang Zhang
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
34
|
Luu XQ, Lee K, Jun JK, Suh M, Choi KS. Socioeconomic inequality in organized and opportunistic screening for gastric cancer: results from the Korean National Cancer Screening Survey 2009-2022. Front Public Health 2023; 11:1256525. [PMID: 37876718 PMCID: PMC10591186 DOI: 10.3389/fpubh.2023.1256525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/15/2023] [Indexed: 10/26/2023] Open
Abstract
Objectives This study aimed to evaluate the socioeconomic inequality in gastric cancer (GC) screening in Korea. Socioeconomic inequality was assessed using both organized and opportunistic screening according to income and educational level. Methods GC screening data were obtained from the 2009-2022 Korean National Cancer Screening Survey. The final analysis included 47,163 cancer-free men and women. The weighted cancer screening rate was estimated using joinpoint regression. The inequality indices were measured in terms of both the absolute slope index of inequality (SII) and the relative index of inequality (RII) using the Poisson regression model. Results The organized screening rate for GC increased from 38.2% in 2009 to 70.8% in 2022, whereas the opportunistic screening rate decreased from 18.8 to 4.5%. Regarding educational inequality, a negative SII value was observed [-3.5, 95% confidence interval (CI), -7.63-0.83%] in organized screening, while a positive SII (9.30%; 95% CI, 6.69-11.91%) and RII (1.98%; 95% CI, 1.59-2.46) were observed in opportunistic screening. Furthermore, income inequality was not found in organized GC screening; however, overall SII and RII for opportunistic screening were 7.72% (95% CI, 5.39-10.5) and 1.61 (95% CI, 1.42-1.81), respectively. Conclusion Organized screening rates have grown gradually over time and account for the majority of GC screenings in South Korea. While no socioeconomic inequalities were found in organized screening, significant socioeconomic inequalities were found in opportunistic screening.
Collapse
Affiliation(s)
- Xuan Quy Luu
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Kyeongmin Lee
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Jae Kwan Jun
- National Cancer Control Institute, National Cancer Center, Goyang, Republic of Korea
| | - Mina Suh
- National Cancer Control Institute, National Cancer Center, Goyang, Republic of Korea
| | - Kui Son Choi
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| |
Collapse
|
35
|
Komori E, Kato-Kogoe N, Imai Y, Sakaguchi S, Taniguchi K, Omori M, Ohmichi M, Nakamura S, Nakano T, Lee SW, Ueno T. Changes in salivary microbiota due to gastric cancer resection and its relation to gastric fluid microbiota. Sci Rep 2023; 13:15863. [PMID: 37740058 PMCID: PMC10516953 DOI: 10.1038/s41598-023-43108-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/20/2023] [Indexed: 09/24/2023] Open
Abstract
Gastric cancer is one of the leading causes of death worldwide, and resections are performed to cure the disease. We have previously reported the changes in the gastric microbiota after gastric cancer resection, which may be associated with the oral microbiota; however, the changes in the oral microbiota remain uncharacterized. This study aimed to characterize the changes in the salivary microbiota caused by gastric cancer resection and to evaluate their association with the gastric fluid microbiota. Saliva and gastric fluid samples were collected from 63 patients who underwent gastrectomy before and after surgery, and a 16S rRNA metagenomic analysis was performed to compare the microbiota composition. The number of bacterial species in the salivary microbiota decreased, and the bacterial composition changed after the resection of gastric cancer. In addition, we identified several bacterial genera that varied significantly in the salivary microbiota, some of which also showed similar changes in the gastric fluid microbiota. These findings indicate that changes in the gastric environment affect the oral microbiota, emphasizing the close association between the oral and gastric fluid microbiota. Our study signifies the importance of focusing on the oral microbiota in the perioperative period of gastrectomy in patients with gastric cancer.
Collapse
Affiliation(s)
- Eri Komori
- Department of Dentistry and Oral Surgery, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki City, Osaka, 569-8686, Japan
| | - Nahoko Kato-Kogoe
- Department of Dentistry and Oral Surgery, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki City, Osaka, 569-8686, Japan.
| | - Yoshiro Imai
- Department of General and Gastroenterological Surgery, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki City, Osaka, 569-8686, Japan
| | - Shoichi Sakaguchi
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki City, Osaka, 569-8686, Japan
| | - Kohei Taniguchi
- Translational Research Program, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki City, Osaka, 569-8686, Japan
| | - Michi Omori
- Department of Dentistry and Oral Surgery, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki City, Osaka, 569-8686, Japan
| | - Mayu Ohmichi
- Department of Dentistry and Oral Surgery, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki City, Osaka, 569-8686, Japan
| | - Shota Nakamura
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Takashi Nakano
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki City, Osaka, 569-8686, Japan
| | - Sang-Woong Lee
- Department of General and Gastroenterological Surgery, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki City, Osaka, 569-8686, Japan
| | - Takaaki Ueno
- Department of Dentistry and Oral Surgery, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki City, Osaka, 569-8686, Japan
| |
Collapse
|
36
|
Shinozuka T, Kanda M, Shimizu D, Umeda S, Takami H, Inokawa Y, Hattori N, Hayashi M, Tanaka C, Nakayama G, Kodera Y. Identification of stromal cell-derived factor 4 as a liquid biopsy-based diagnostic marker in solid cancers. Sci Rep 2023; 13:15540. [PMID: 37730904 PMCID: PMC10511445 DOI: 10.1038/s41598-023-42201-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/06/2023] [Indexed: 09/22/2023] Open
Abstract
There is a need for serum diagnostic biomarkers to improve the prognosis of solid malignant tumors. Here, we conducted a single-institutional study to evaluate the diagnostic performance of serum stromal cell-derived factor 4 (SDF4) levels in cancer patients. Serum samples were collected from a total of 582 patients with solid cancers including gastric cancer (GC) and 80 healthy volunteers. SDF4 protein levels in sera, and conditioned media or lysates of human GC cell lines were measured by enzyme-linked immunosorbent assay, and those in GC tissue by immunohistochemistry. Serum SDF4 levels were higher in patients with cancer than the healthy control in all cancer type. Regarding GC, serum SDF4 levels distinguished healthy controls from GC patients with the area under the curve value of 0.973, sensitivity of 89%, and specificity of 99%. Serum SDF4 levels were significantly elevated in patient with early stage GC. In immunohistochemistry, the frequency of SDF4-positive GC tumors did not vary significantly between GC stages. The ability of human GC cell lines to both produce and secrete SDF4 was confirmed in vitro. In conclusion, serum SDF4 levels could be a promising candidate for a novel diagnostic biomarker for GC and other malignancies.
Collapse
Affiliation(s)
- Takahiro Shinozuka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan.
| | - Dai Shimizu
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Shinichi Umeda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Hideki Takami
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Yoshikuni Inokawa
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Norifumi Hattori
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Masamichi Hayashi
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Chie Tanaka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Goro Nakayama
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| |
Collapse
|
37
|
Fu XY, Mao XL, Wu HW, Lin JY, Ma ZQ, Liu ZC, Cai Y, Yan LL, Sun Y, Ye LP, Li SW. Development and validation of LightGBM algorithm for optimizing of Helicobacter pylori antibody during the minimum living guarantee crowd based gastric cancer screening program in Taizhou, China. Prev Med 2023; 174:107605. [PMID: 37419420 DOI: 10.1016/j.ypmed.2023.107605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 06/22/2023] [Accepted: 07/02/2023] [Indexed: 07/09/2023]
Abstract
Gastric cancer continues to be a significant health concern in China, with a high incidence rate. To mitigate its impact, early detection and treatment is key. However, conducting large-scale endoscopic gastric cancer screening is not feasible in China. Instead, a more appropriate approach would be to initially screen high-risk groups and follow up with endoscopic testing as needed. We conducted a study on 25,622 asymptomatic participants aged 45-70 years from a free gastric cancer screening program in the Taizhou city government's Minimum Living Guarantee Crowd (MLGC) initiative. Participants completed questionnaires, blood tests, and underwent gastrin-17 (G-17), pepsinogen I and II (PGI and PGII), and H. pylori IgG antibody (IgG) assessments. Using the light gradient boosting machine (lightGBM) algorithm, we developed a predictive model for gastric cancer risk. In the full model, F1 score was 2.66%, precision was 1.36%, and recall was 58.14%. In the high-risk model, F1 score was 2.51%, precision was 1.27%, and recall was 94.55%. Excluding IgG, the F1 score was 2.73%, precision was 1.40%, and recall was 68.62%. We conclude that H. pylori IgG appears to be able to be excluded from the prediction model without significantly affecting its performance, which is important from a health economic point of view. It suggests that screening indicators can be optimized, and expenditures reduced. These findings can have important implications for policymakers, as we can focus resources on other important aspects of gastric cancer prevention and control.
Collapse
Affiliation(s)
- Xin-Yu Fu
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Xin-Li Mao
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China; Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China; Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Hao-Wen Wu
- Department of Mathematics and Statistics, York University, Toronto, ON, Canada
| | - Jia-Ying Lin
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Zong-Qing Ma
- Information center, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Zhi-Cheng Liu
- Information center, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Yue Cai
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Ling-Ling Yan
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Yi Sun
- Department of Neurology, Faculty of Medical, University of Toyama, Toyama, Toyama Ken, Japan.
| | - Li-Ping Ye
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China; Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China; Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China.
| | - Shao-Wei Li
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China; Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China; Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China.
| |
Collapse
|
38
|
Quek SXZ, Lee JWJ, Feng Z, Soh MM, Tokano M, Guan YK, So JBY, Tada T, Koh CJ. Comparing artificial intelligence to humans for endoscopic diagnosis of gastric neoplasia: An external validation study. J Gastroenterol Hepatol 2023; 38:1587-1591. [PMID: 37408330 DOI: 10.1111/jgh.16274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 06/03/2023] [Accepted: 06/14/2023] [Indexed: 07/07/2023]
Abstract
OBJECTIVES Artificial intelligence (AI) uses deep learning functionalities that may enhance the detection of early gastric cancer during endoscopy. An AI-based endoscopic system for upper endoscopy was recently developed in Japan. We aim to validate this AI-based system in a Singaporean cohort. METHODS There were 300 de-identified still images prepared from endoscopy video files obtained from subjects that underwent gastroscopy in National University Hospital (NUH). Five specialists and 6 non-specialists (trainees) from NUH were assigned to read and categorize the images into "neoplastic" or "non-neoplastic." Results were then compared with the readings performed by the endoscopic AI system. RESULTS The mean accuracy, sensitivity, and specificity for the 11 endoscopists were 0.847, 0.525, and 0.872, respectively. These values for the AI-based system were 0.777, 0.591, and 0.791, respectively. While AI in general did not perform better than endoscopists on the whole, in the subgroup of high-grade dysplastic lesions, only 29.1% were picked up by the endoscopist rating, but 80% were classified as neoplastic by AI (P = 0.0011). The average diagnostic time was also faster in AI compared with endoscopists (677.1 s vs 42.02 s (P < 0.001). CONCLUSION We demonstrated that an AI system developed in another health system was comparable in diagnostic accuracy in the evaluation of static images. AI systems are faster and not fatigable and may have a role in augmenting human diagnosis during endoscopy. With more advances in AI and larger studies to support its efficacy it would likely play a larger role in screening endoscopy in future.
Collapse
Affiliation(s)
- Sabrina Xin Zi Quek
- Division of Gastroenterology and Hepatology, National University Hospital, Singapore
| | - Jonathan W J Lee
- Division of Gastroenterology and Hepatology, National University Hospital, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- iHealthtech, National University of Singapore, Singapore
| | - Zhu Feng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Min Min Soh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Yeoh Khay Guan
- Division of Gastroenterology and Hepatology, National University Hospital, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jimmy B Y So
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Surgery, National University Hospital, Singapore
| | - Tomohiro Tada
- AI Medical Service Inc, Japan
- Tada Tomohiro Institute of Gastroenterology and Proctology, Japan
| | - Calvin J Koh
- Division of Gastroenterology and Hepatology, National University Hospital, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- The Gastroenterology Group, Gleneagles Hospital, Singapore, Singapore
| |
Collapse
|
39
|
Fang L, Huang H, Lv J, Chen Z, Lu C, Jiang T, Xu P, Li Y, Wang S, Li B, Li Z, Wang W, Xu Z. m5C-methylated lncRNA NR_033928 promotes gastric cancer proliferation by stabilizing GLS mRNA to promote glutamine metabolism reprogramming. Cell Death Dis 2023; 14:520. [PMID: 37582794 PMCID: PMC10427642 DOI: 10.1038/s41419-023-06049-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/30/2023] [Accepted: 08/07/2023] [Indexed: 08/17/2023]
Abstract
Abnormal 5-methylcytosine (m5C) methylation has been proved to be closely related to gastric carcinogenesis, progression, and prognosis. Dysregulated long noncoding RNAs (lncRNAs) participate in a variety of biological processes in cancer. However, to date, m5C-methylated lncRNAs are rarely researched in gastric cancer (GC). Here, we found that RNA cytosine-C(5)-methyltransferase (NSUN2) was upregulated in GC and high NSUN2 expression was associated with poor prognosis. NR_033928 was identified as an NSUN2-methylated and upregulated lncRNA in GC. Functionally, NR_033928 upregulated the expression of glutaminase (GLS) by interacting with IGF2BP3/HUR complex to promote GLS mRNA stability. Increased glutamine metabolite, α-KG, upregulated NR_033928 expression by enhancing its promoter 5-hydroxymethylcytosine (hm5C) demethylation. In conclusion, our results revealed that NSUN2-methylated NR_033928 promoted GC progression and might be a potential prognostic and therapeutic target for GC.
Collapse
Affiliation(s)
- Lang Fang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Hongxin Huang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jialun Lv
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zetian Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chen Lu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Tianlu Jiang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Penghui Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ying Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Sen Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Bowen Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zheng Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Weizhi Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 210029, Nanjing, Jiangsu Province, China.
| |
Collapse
|
40
|
Qin Y, Geng JX, Huang B. Clinical value of serum pepsinogen in the diagnosis and treatment of gastric diseases. World J Gastrointest Oncol 2023; 15:1174-1181. [PMID: 37546552 PMCID: PMC10401465 DOI: 10.4251/wjgo.v15.i7.1174] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/28/2023] [Accepted: 06/14/2023] [Indexed: 07/12/2023] Open
Abstract
Pepsinogen, secreted from the gastric mucosa, is the precursor of pepsin. It is categorized as pepsinogen 1 and pepsinogen 2 based on its immunogenicity. The pepsinogen content that can enter the blood circulation through the capillaries of the gastric mucosa is approximately 1% and remains stable all the time. The pepsinogen content in serum will change with the pathological changes of gastric mucosa. Therefore, the level of pepsinogen in serum can play a role in serologic biopsy to reflect the function and morphology of different regions of gastric mucosa and serve as an indicator of gastric disease. This study conducts relevant research on serum pepsinogen 1, pepsinogen 2, and the ratio of pepsinogen 1 to pepsinogen 2, and reviews their important value in clinical diagnosis of Helicobacter pylori infection, gastric ulcer, and even gastric carcinoma, providing ideas for other researchers.
Collapse
Affiliation(s)
- Yuan Qin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Jia-Xin Geng
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| |
Collapse
|
41
|
Shinozuka T, Kanda M, Kodera Y. Site-specific protein biomarkers in gastric cancer: a comprehensive review of novel biomarkers and clinical applications. Expert Rev Mol Diagn 2023; 23:701-712. [PMID: 37395000 DOI: 10.1080/14737159.2023.2232298] [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: 04/27/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
INTRODUCTION Gastric cancer (GC) is the fifth most common cancer and the fourth leading cause of cancer-related death worldwide, thus representing a significant global health burden. Early detection and monitoring of GC are essential to improve patient outcomes. While traditional cancer biomarkers such as carcinoembryonic antigen, carbohydrate antigen (CA) 19-9, and CA 72-4 are widely used, their limited sensitivity and specificity necessitate the exploration of alternative biomarkers. AREAS COVERED This review comprehensively analyzes the landscape of GC protein biomarkers identified from 2019 to 2022, with a focus on tissue, blood, urine, saliva, gastric juice, ascites, and exhaled breath as sample sources. We address the potential clinical applications of these biomarkers in early diagnosis, monitoring recurrence, and predicting survival and therapeutic response of GC patients. EXPERT OPINION The discovery of novel protein biomarkers holds great promise for improving the clinical management of GC. However, further validation in large, diverse cohorts is needed to establish the clinical utility of these biomarkers. Integrating these biomarkers with existing diagnostic and monitoring approaches will likely lead to improved personalized treatment plans and patient outcomes.
Collapse
Affiliation(s)
- Takahiro Shinozuka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
42
|
Chen Z, Xu P, Wang X, Li Y, Yang J, Xia Y, Wang S, Liu H, Xu Z, Li Z. MSC-NPRA loop drives fatty acid oxidation to promote stemness and chemoresistance of gastric cancer. Cancer Lett 2023; 565:216235. [PMID: 37209945 DOI: 10.1016/j.canlet.2023.216235] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 05/22/2023]
Abstract
Cisplatin (CDDP)-based chemotherapy is the preferred treatment strategy for advanced stage gastric cancer (GC) patients. Despite the efficacy of chemotherapy, the development of chemoresistance negatively affects the prognosis of GC and the underlying mechanism remains poorly understood. Accumulated evidence suggests that mesenchymal stem cells (MSCs) play important roles in drug resistance. The chemoresistance and stemness of GC cells were observed by colony formation, CCK-8, sphere formation and flow cytometry assays. Cell lines and animal models were utilized to investigate related functions. Western blot, quantitative real-time PCR (qRT-PCR) and co-immunoprecipitation were used to explore related pathways. The results showed that MSCs improved the stemness and chemoresistance of GC cells and accounted for the poor prognosis of GC. Natriuretic peptide receptor A (NPRA) was upregulated in GC cells cocultured with MSCs and knockdown of NPRA reversed the MSC-induced stemness and chemoresistance. At the same time, MSCs could be recruited to GC by NPRA, which formed a loop. In addition, NPRA facilitated stemness and chemoresistance through fatty acid oxidation (FAO). Mechanistically, NPRA protected Mfn2 against protein degradation and promoted its mitochondrial localization, which consequently improved FAO. Furthermore, inhibition of FAO with etomoxir (ETX) attenuated MSC-induced CDDP resistance in vivo. In conclusion, MSC-induced NPRA promoted stemness and chemoresistance by upregulating Mfn2 and improving FAO. These findings help us understand the role of NPRA in the prognosis and chemotherapy of GC. NPRA may be a promising target to overcome chemoresistance.
Collapse
Affiliation(s)
- Zetian Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Penghui Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Xinghong Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Ying Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Jing Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Yiwen Xia
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Sen Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Hongda Liu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China; The Institute of Gastric Cancer, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Zheng Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
43
|
Zang H, Wang J, Wang H, Guo J, Li Y, Zhao Y, Song J, Liu F, Liu X, Zhao Y. Metabolic alterations in patients with Helicobacter pylori-related gastritis: The H. pylori-gut microbiota-metabolism axis in progression of the chronic inflammation in the gastric mucosa. Helicobacter 2023:e12984. [PMID: 37186092 DOI: 10.1111/hel.12984] [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: 10/04/2022] [Revised: 02/15/2023] [Accepted: 03/13/2023] [Indexed: 05/17/2023]
Abstract
PURPOSE To characterize the serum metabolism in patients with Helicobacter pylori-positive and H. pylori-negative gastritis. METHODS Clinical data and serum gastric function parameters, PGI (pepsinogen I), PGII, PGR (PGI/II), and G-17 (gastrin-17) of 117 patients with chronic gastritis were collected, including 57 H. pylori positive and 60 H. pylori negative subjects. Twenty cases in each group were randomly selected to collect intestinal mucosa specimens and serum samples. The gut microbiota profiles were generated by 16S rRNA gene sequencing, and the serum metabolites were analyzed by a targeted metabolomics approach based on liquid chromatography-mass spectrometry (LC-MS) technology. RESULTS Altered expression of 20 metabolites, including isovaleric acid, was detected in patients with HPAG. Some taxa of Bacteroides, Fusobacterium, and Prevotella in the gut microbiota showed significant correlations with differentially expressed metabolites between H. pylori positive and H. pylori negative individuals. As a result, an H. pylori-gut microbiota-metabolism (HGM) axis was proposed. CONCLUSION Helicobacter pylori infection may influence the progression of mucosal diseases and the emergence of other complications in the host by altering the gut microbiota, and thus affecting the host serum metabolism.
Collapse
Affiliation(s)
- Hongmin Zang
- Hebei University of Chinese Medicine, Shijiazhuang, China
- The Traditional Chinese Medicine Hospital of Shijiazhuang, Shijiazhuang, China
| | - Jin Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Huijie Wang
- The Traditional Chinese Medicine Hospital of Shijiazhuang, Shijiazhuang, China
| | - Jiaxuan Guo
- The Traditional Chinese Medicine Hospital of Shijiazhuang, Shijiazhuang, China
| | - Yuchan Li
- The Traditional Chinese Medicine Hospital of Shijiazhuang, Shijiazhuang, China
| | - Yinuo Zhao
- School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Jinzhong Song
- Hebei University of Chinese Medicine, Shijiazhuang, China
- The Traditional Chinese Medicine Hospital of Shijiazhuang, Shijiazhuang, China
| | - Fengshuang Liu
- Hebei University of Chinese Medicine, Shijiazhuang, China
- Hebei Academy of Traditional Chinese Medicine, Shijiazhuang, China
| | - Xuzhao Liu
- North China University of Science and Technology, Tangshan, China
| | - Yubin Zhao
- Hebei University of Chinese Medicine, Shijiazhuang, China
- North China University of Science and Technology, Tangshan, China
- Shijiazhuang People's Hospital, Shijiazhuang, China
| |
Collapse
|
44
|
Luo X, Yao K, Lin X, Lin B, Zhu C, Huang S, Chen Z, Li A, Wang J, Huang Y, Li Z, Liu S, Han Z. Intensive Systematic "Train-the-Trainer" Course as an Effective Strategy to Improve Detection of Early Gastric Cancer: A Multicenter Retrospective Study. J Gastrointest Surg 2023:10.1007/s11605-023-05640-w. [PMID: 36941524 DOI: 10.1007/s11605-023-05640-w] [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: 07/25/2022] [Accepted: 02/18/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND To improve the diagnosis of early gastric cancer (EGC), a train-the-trainer (TTT) course was developed. This trial aimed to investigate whether TTT courses from trained trainers could improve trainees' EGC detection. METHODS In this multi-center, retrospective study, the training was carried out 8 times in one year. Clinical records one year before ("2016"), during ("2017"), and after ("2018") the course were collected. The primary endpoint was the improvement of EGC detection rate after TTT courses. RESULTS Twenty-four trainees from 17 hospitals were included in this study. A total of 123,416 esophagogastroduodenoscopy and 65,570 colonoscopy procedures were analyzed. The early gastric cancer detection rate (EDR) was 0.101% in 2016, which significantly increased to 0.338% in 2018 (p = 0.015). The early gastric cancer ratio (ECR, ratio of newly detected EGCs to all newly detected gastric cancers) in 2016 was 8.440%, which consistently increased to 11.853% and 19.778% in 2017 and 2018 (p = 0.006), respectively. In contrast, the advanced gastric cancer detection rate (ADR) was similar before, during, or after the course (p = 0.987). The 3-year EDR, ECR, and ADR in esophageal and colorectal cancer were not significantly different. CONCLUSIONS The systematic training course can improve EGC detection rate and may be an effective educational strategy to reduce gastrointestinal cancers mortality.
Collapse
Affiliation(s)
- Xiaobei Luo
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
- Department of Gastroenterology, Zhuhai People's Hospital (Zhuhai Hospital Affifiliated With Jinan University), Zhuhai, Guangdong, China
| | - Kenshi Yao
- Department of Endoscopy, Chikushi Hospital, Fukuoka University, Fukuoka, Japan
| | - Xin Lin
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Bitao Lin
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Chaojun Zhu
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Silin Huang
- Department of Gastroenterology, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Zhenyu Chen
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Aimin Li
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Jiahao Wang
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), Singapore, 138669, Singapore
| | - Yin Huang
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Zhihao Li
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Side Liu
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China.
- Department of Gastroenterology, Zhuhai People's Hospital (Zhuhai Hospital Affifiliated With Jinan University), Zhuhai, Guangdong, China.
| | - Zelong Han
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China.
| |
Collapse
|
45
|
Sheng C, Sun L, Lyu Z, Li L, Zhang Y, Zhang Y, Zhang Y, Dai H, Huang Y, Song F, Yuan Y, Chen K. Development of a modified ABC method among Helicobacter pylori infected but serum pepsinogen test-negative individuals. Helicobacter 2023; 28:e12966. [PMID: 36941759 DOI: 10.1111/hel.12966] [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: 10/07/2022] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND Although the ABC method for gastric cancer (GC) screening has been widely adopted in Japan, it may not be suitable for other countries due to population heterogeneity and different tumor histology. We aim to develop a modified ABC method to improve GC screening performance, especially among Helicobacter pylori (Hp) infected but serum pepsinogen (sPG) test-negative individuals. METHODS A total of 4745 participants were recruited from Tianjin, China, and were classified into four groups by combined assay for Hp infection and sPG concentrations: Group A (Hp [-], PG [-]), Group B (Hp [+], PG [-]), Group C (Hp [+], PG [+]), and Group D (Hp [-], PG [+]). We used receiver-operating characteristic (ROC) curves analysis and minimum p value method to determine the optimal cutoff point for PG II in Group B. We performed logistic regressions to examine the risk of GC across different subgroups. In addition to the derivation set, the performance of the modified ABC method was also evaluated in an external set involving 16,292 participants from Liaoning, China. RESULTS In the modified ABC method, we further classified Group B as low-risk (Group B1) and high-risk subgroups (Group B2) using optimal sPG II cutoff point (20.0 ng/mL) by ROC curves analysis and minimum p value method. Compared with Group B1, Group B2 had a significantly higher risk of GC (adjusted OR = 2.54, 95% CI = 1.94-3.33). The modified ABC method showed good discrimination for GC (AUC = 0.61, 95% CI = 0.59-0.63) and improved risk reclassification (NRI = 0.11, p < .01). Similar results were observed in the validation dataset. CONCLUSIONS The modified ABC method can effectively identify high-risk population for GC among Hp-infected but sPG test-negative participants in China.
Collapse
Affiliation(s)
- Chao Sheng
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Liping Sun
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Zhangyan Lyu
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Limin Li
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Yuhao Zhang
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Yu Zhang
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Yacong Zhang
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Hongji Dai
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Yubei Huang
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Fengju Song
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| |
Collapse
|
46
|
Ji X, He G, Wang K, Zhang Y, Yin J, Wang K. Estimation of gastric cancer burden attributable to Helicobacter pylori infection in Asia. J Public Health (Oxf) 2023; 45:40-46. [PMID: 35137200 DOI: 10.1093/pubmed/fdab410] [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: 05/17/2021] [Revised: 12/22/2021] [Accepted: 01/02/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Helicobacter pylori causes large burden of gastric cancer (GC) in Asia. We aimed to comprehensively quantify the burden of GC attributable to H. pylori infection in Asia. METHODS We searched related articles from January 1998 to December 2020 to obtain the prevalence and relative risks (or odds ratio) of GC associated with H. pylori in Asia. The burden of GC attributable to H. pylori infection was quantified by Population Attributable Fraction (PAF) and Disability-adjusted life-years (DALYs). RESULTS We quantified the burden of GC attributable to H. pylori infection with 415.6 thousand DALYs and 38.03% PAF through the five included Asian countries in 2019. The study found that the burden had obvious regional differences. The DALYs ranged from 298.9 thousand in China to 1.9 thousand in Malaysia, and the PAFs were between 58.00% in Japan and 30.89% in China. The average prevalence of H. pylori in the included general population was estimated to be 56.29%. CONCLUSIONS Helicobacter pylori poses a huge disease burden of GC to the population, and its eradication should receive attention, especially in the countries with high incidence of and mortality due to GC.
Collapse
Affiliation(s)
- Xuanke Ji
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Gui He
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Kunyan Wang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Yuehua Zhang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Jingjing Yin
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Kaijuan Wang
- Medical School, Huanghe Science & Technology College, Zijingshan Road No. 666, Zhengzhou City, Henan 450000, China
| |
Collapse
|
47
|
Zhang Q, Zhang Q, Yan H, You Y, Cao J, Wang W, Zhao D, Zhang X, Wu J, Chen Z, Yang W. Survival Advantages in Gastric Cancer Patients Receiving Preoperative SOX Regimen Chemotherapy. Cancer Biother Radiopharm 2023; 38:81-88. [PMID: 32833547 DOI: 10.1089/cbr.2020.3970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Objective: This study addressed whether preoperative chemotherapy (PECT) plus surgery prolongs overall survival (OS) compared with surgery plus postoperative chemotherapy (POCT) among gastric cancer (GC) patients in Northwest China. Materials and Methods: The authors included 157 GC patients confirmed histologically or by gastroscopic pathological examination treated at the General Hospital of Ningxia Medical University of China between 2012 and 2018. All patients were followed up by telephone in January 2019 within a 2-week period. The endpoint was death due to GC or its complications. Results: Thirty-eight patients received PECT, 41 patients received POCT, 40 patients received surgery alone, and 38 patients received chemotherapy alone. Surgery was performed with R0 resection and subsequent extended lymph node dissection. Chemotherapy was performed with the S-1, oxaliplatin capecitabine regimen. Patients who received PECT had longer OS than those with POCT treatment (hazard ratio = 2.409, p = 0.037). The 5-year OS rate was 32.7% higher in the PECT group than in the POCT group. Conclusions: PECT was associated with better OS in GC patients and should be considered by clinicians in GC treatment, although prospective studies are needed for confirmation.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Cancer Research, The General Hospital and Basic Medical School, Ningxia Medical University, Yinchuan, China
| | - Qinghua Zhang
- Department of Neurosurgery, Shenzhen Nanshan Hospital, Huazhong University of Science and Technology, Shenzhen, China
| | - Hui Yan
- Department of Medical Oncology, The General Hospital, Ningxia Medical University, Yinchuan, China
| | - Yanjie You
- Department of Gastroenterology, The People's Hospital, Yinchuan, China
| | - Juan Cao
- Department of Cancer Research, The General Hospital and Basic Medical School, Ningxia Medical University, Yinchuan, China
| | - Wenfan Wang
- Department of Cancer Research, The General Hospital and Basic Medical School, Ningxia Medical University, Yinchuan, China
| | - Dan Zhao
- Department of Cancer Research, The General Hospital and Basic Medical School, Ningxia Medical University, Yinchuan, China
| | - Xiaoxu Zhang
- Department of Cancer Research, The General Hospital and Basic Medical School, Ningxia Medical University, Yinchuan, China
| | - Jing Wu
- Department of Cancer Research, The General Hospital and Basic Medical School, Ningxia Medical University, Yinchuan, China
| | - Zhiqiang Chen
- Department of Radiology, The General Hospital, Ningxia Medical University, Yinchuan, China.,Department of Radiology, Shenzhen Nanshan Hospital, Huazhong University of Science and Technology, Shenzhen, China
| | - Wenjun Yang
- Department of Clinical Research, Shenzhen Nanshan Hospital, Huazhong University of Science and Technology, Shenzhen, China
| |
Collapse
|
48
|
Min Z, Hui L, Hui R, Jie Z, Hong Y, Yuhan L. Screening Behaviors and Related Factors among the First-Degree Relatives of Chinese Patients with Gastric Cancer. Asia Pac J Oncol Nurs 2023; 10:100220. [PMID: 37181816 PMCID: PMC10173167 DOI: 10.1016/j.apjon.2023.100220] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/20/2023] [Indexed: 03/31/2023] Open
Abstract
Objective This study was aimed at investigating the screening behaviors of the first-degree relatives (FDRs) of Chinese patients with gastric cancer as well as the factors influencing screening behaviors. Methods In a cross-sectional design, 197 FDRs of patients with gastric cancer were enrolled at the Peking University Cancer Hospital. Four questionnaires were used: a demographic questionnaire, a knowledge questionnaire of risk factors and warning symptoms for gastric cancer, the Gastric Cancer Health Belief Scale, and a questionnaire screening for behavioral motivators and barriers. Logistic regression analysis was performed to determine the factors influencing screening behaviors. Results Among the 197 FDRs of patients with gastric cancer, 30.96% (61/197) of patients had undergone gastric cancer screening. Among those who participated in gastric cancer screening, the most common screening methods were gastroscopy and the Helicobacter pylori testing, both of which were applied in 63.93% (39/61) of participants, followed by serum tumor marker testing (55.74%, 34/61) and barium meal examination of the upper digestive tract (29.51%, 18/61). The gastric cancer risk factor knowledge score was 9.02 ± 3.95, and the gastric cancer warning symptom knowledge score was 4.39 ± 1.85. The participants' knowledge score was moderate, at 13.41 ± 5.16. The total health beliefs score was low, at 88.91 ± 12.66. Factors independently associated with the screening behaviors of FDRs included educational background, knowledge of gastric cancer risk factors, and health motivation (P < 0.05). Conclusions The participation rate in gastric cancer screening of FDRs of patients with gastric cancer was relatively low and was affected by multiple factors. Our results highlighted the urgent need for educational campaigns and precision interventions to raise gastric cancer awareness.
Collapse
|
49
|
Jin T, Chen ZH, Liang PP, Li ZD, He FJ, Chen ZW, Hu JK, Yang K. A Gastrectomy for early-stage gastric cancer patients with or without preserving celiac branches of vagus nerves: A meta-analysis. Surgery 2023; 173:375-382. [PMID: 36379744 DOI: 10.1016/j.surg.2022.10.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 08/24/2022] [Accepted: 10/11/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Vagus nerve-preserving gastrectomy is increasingly popular in treating gastric cancer in the early stage, however the long and short-term outcomes after gastrectomy while preserving the celiac branch of the vagus nerve are not well defined. We aimed to summarize and compare perioperative and longer-term outcomes after celiac branch vagus nerve-preserving gastrectomy (CBP, preserving both the celiac and hepatic branches of the vagus nerve), compared to those without CBP (non-CBP, only the hepatic branch of the vagus nerve is preserved). METHODS We searched the Embase, PubMed, Cochrane Library and Web of Science databases for papers published before October 2021. The primary results were evaluated by short-term and long-term postoperative complications, whereas the secondary outcomes included surgery-related parameters, recovery-related parameters and overall survival. Random-effects or fixed-effects model were used to estimate odds ratio, and weighted mean difference for the outcomes. The underlying publication bias was identified via funnel charts, Begg's test and Egger's test. Sensitivity analysis was conducted by removing the research one by one. RESULTS A total of 9 studies consisting of 8 retrospective studies and one randomized control trial were included. The analysis included 1,109 patients, with 568 (51.2%) of patients receiving CBP and 541 (48.8%) patients who received non-CBP. The CBP group had a shorter time in terms of first flatus (weighted mean difference = -0.436, 95% confidence interval: -0.603 to -0.269; P < 0.001) and hospital stay (weighted mean difference = -0.456, 95% confidence interval: -0.874 to -0.037, P = 0.033) than the non-CBP group, but the time to the start of oral intake was comparable between the groups. Regarding short-term complications and surgery-related parameters, between CBP and non-CBP, no evident differences were observed in pancreatic complications, anastomotic leakage, postoperative bleeding, operation time, blood loss or lymph nodes examined. In terms of long-term complications, the incidence of gallstones in CBP was lower than that in non-CBP (odds ratio = 0.582, 95% confidence interval: 0.356-0.953, P = 0.031), and the incidence of bile reflux in CBP was lower than that in non-CBP (odds ratio = 0.473, 95% confidence interval: 0.280-0.800, P = 0.005). However, the prevalence rates of diarrhea, early dumping syndrome, esophageal reflux, and delayed gastric emptying were comparable between CBP and non-CBP. CONCLUSION The present research showed that gastric cancer patients in the early stage under CBP were superior to those without CBP in terms of incidence of gallstones, bile reflux, time of first flatus and hospital stay. Furthermore, it is imperative to conduct randomized control studies with larger sample sizes to determine the oncological survival outcomes when preserving the celiac branch.
Collapse
Affiliation(s)
- Tao Jin
- Gastric Cancer Center, West China Hospital, Sichuan University, China; Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, China
| | - Ze-Hua Chen
- Gastric Cancer Center, West China Hospital, Sichuan University, China; Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, China
| | - Pan-Ping Liang
- Gastric Cancer Center, West China Hospital, Sichuan University, China; Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, China
| | - Ze-Dong Li
- Gastric Cancer Center, West China Hospital, Sichuan University, China; Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, China
| | - Feng-Jun He
- Gastric Cancer Center, West China Hospital, Sichuan University, China; Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, China
| | - Zheng-Wen Chen
- Gastric Cancer Center, West China Hospital, Sichuan University, China; Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, China
| | - Jian-Kun Hu
- Gastric Cancer Center, West China Hospital, Sichuan University, China; Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, China
| | - Kun Yang
- Gastric Cancer Center, West China Hospital, Sichuan University, China; Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, China; Department of Gastrointestinal Surgery, JinTang Hospital, West China Hospital, Sichuan University, China.
| |
Collapse
|
50
|
Chatterjee A, Shah D, Roy B, Ghosh J, Ray S, Patra A, Gehani A, Gupta B, Ghosh P, Mukhopadhyay S, Chandra A, Lingegowda D, Sen S. Imaging Recommendations for Diagnosis, Staging, and Management of Gastric Cancer. Indian J Med Paediatr Oncol 2023. [DOI: 10.1055/s-0042-1759715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
AbstractGastric cancer is the second most common cause of cancer-related death in Indian men and women aged between 15 and 44 years. Most patients present at an advanced stage of disease. Surgically resectable disease usually requires a standard gastric resection and D2 lymphadenectomy. Imaging, especially with computed tomography scan of abdomen as well as thorax, is necessary for localization, nodal mapping, and metastatic workup of gastric cancer. In this review, we discuss current imaging recommendations for gastric carcinoma.
Collapse
Affiliation(s)
- Argha Chatterjee
- Department of Radiology and Imaging, Tata Medical Center, Kolkata, West Bengal, India
| | - Diva Shah
- Consultant Radiologist, HCG Cancer Center, Ahmedabad, Gujarat, India
| | - Bipradas Roy
- Department of GI and HPB Surgery, Department of Radiology and Imaging, Tata Medical Center, Kolkata, West Bengal, India
| | - Joydeep Ghosh
- Department of Medical Oncology, Tata Medical Center, Kolkata, West Bengal, India
| | - Soumendranath Ray
- Department of Nuclear Medicine, Tata Medical Center, Kolkata, West Bengal, India
| | - Anurima Patra
- Department of Radiology and Imaging, Tata Medical Center, Kolkata, West Bengal, India
| | - Anisha Gehani
- Department of Radiology and Imaging, Tata Medical Center, Kolkata, West Bengal, India
| | - Bharat Gupta
- Department of Radiology and Imaging, Tata Medical Center, Kolkata, West Bengal, India
| | - Priya Ghosh
- Department of Radiology and Imaging, Tata Medical Center, Kolkata, West Bengal, India
| | - Sumit Mukhopadhyay
- Department of Radiology and Imaging, Tata Medical Center, Kolkata, West Bengal, India
| | - Aditi Chandra
- Department of Radiology and Imaging, Tata Medical Center, Kolkata, West Bengal, India
| | - Dayananda Lingegowda
- Department of Radiology and Imaging, Tata Medical Center, Kolkata, West Bengal, India
| | - Saugata Sen
- Department of Radiology and Imaging, Tata Medical Center, Kolkata, West Bengal, India
| |
Collapse
|