51
|
Zhang L, Yang J, Huang Y, You T, Huang Q, Shen X, Xue X, Feng S. Comprehensive landscape of gastric cancer-targeted therapy and identification of CSNK2A1 as a potential target. Heliyon 2024; 10:e36205. [PMID: 39253198 PMCID: PMC11382053 DOI: 10.1016/j.heliyon.2024.e36205] [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: 04/03/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024] Open
Abstract
Objective To conduct a comprehensive analysis of the landscape of gastric cancer (GC)-targeted therapy clinical trials and identify potential therapeutic targets. Methods A systematic search and analysis of the Cochrane Central Register of Controlled Trials (CENTRAL) was performed to retrieve all GC clinical trials published up to June 30, 2022. Approved therapeutic targets for 11 common cancers were compiled and analyzed. The role of CSNK2A1 in GC was investigated using bioinformatics tools such as GEPIA, KMPLOT, SangerBox, STRING, ACLBI, and TIMER. Four gastric cancer cell lines (AGS, HGC, MGC, BGC) and one normal gastric mucosa cell line (GES-1) were utilized to assess the sensitivity to the CSNK2A1 inhibitor CX-4945. Quantitative real-time polymerase chain reaction (qPCR) was employed to quantify the cellular expression of CSNK2A1. Cellular apoptosis was evaluated using flow cytometry and Western blot analysis. Results The failure rate of GC randomized controlled clinical trials (RCTs) was strikingly high, accounting for 74.29 % (26/35) of the trials. Among the 35 approved targets in 11 different cancers, 13 targets were rigorously evaluated and identified as potential therapeutic targets for GC. Bioinformatics analysis revealed that CSNK2A1 is closely associated with multiple biological characteristics in GC, and its increased expression correlated significantly with enhanced sensitivity to CX-4945 treatment. Flow cytometry and Western blot analysis consistently demonstrated concentration-dependent apoptosis induced by CX-4945 in GC cell lines. Conclusions The high failure rate of GC clinical trials highlights the need for a more scientific and precise approach in target identification and clinical trial design. CSNK2A1 emerges as a promising therapeutic target for GC, and its expression level could potentially serve as a biomarker for predicting sensitivity to CX-4945 treatment. Further research is warranted to elucidate the underlying molecular mechanisms and validate the clinical significance of CSNK2A1 in GC therapy.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, China
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, China
| | - Jiaqi Yang
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, China
| | - Yingpeng Huang
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, China
| | - Tao You
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, China
| | - Qunjia Huang
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, China
| | - Xian Shen
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, China
| | - Xiangyang Xue
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, China
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, China
| | - Shiyu Feng
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, China
| |
Collapse
|
52
|
Isakov V. Autoimmune gastritis studies and gastric cancer: True renaissance or bibliometric illusion. World J Gastroenterol 2024; 30:3783-3790. [PMID: 39221066 PMCID: PMC11362875 DOI: 10.3748/wjg.v30.i32.3783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/26/2024] Open
Abstract
A bibliometric analysis of studies dedicated to autoimmune gastritis (AIG) recently published demonstrated a noteworthy surge in publications over the last three years. This can be explained by numerous publications from different regions of the world reporting the results of several studies that stimulated reassessment of our view of AIG as a precancerous condition. Follow-up studies and retrospective analyses showed that the risk of gastric cancer (GC) in AIG patients is much lower than expected if the patients ever being infected with Helicobacter pylori (H. pylori) were excluded. The low prevalence of precancerous lesions, such as the incomplete type of intestinal metaplasia, may explain the low risk of GC in AIG patients because the spasmolytic polypeptide-expressing metaplasia commonly observed in AIG does not involve clonal reprogramming of the gastric gland and can be considered as an adaptive change rather than a true precancerous lesion. However, changes in gastric secretion due to the progression of gastric atrophy during the course of AIG cause changes in the gastric mic-robiome, stimulating the growth of bacterial species such as streptococci, which may promote the development of precancerous lesions and GC. Thus, Streptococcus anginosus exhibited a robust proinflammatory response and induced the gastritis-atrophy-metaplasia-dysplasia sequence in mice, reproducing the well-established process for carcinogenesis associated with H. pylori. Prospective studies in H. pylori-naïve patients evaluating gastric microbiome changes during the long-term course of AIG might provide an explanation for the enigmatic increase in GC incidence in the last decades in younger cohorts, which has been reported in economically developed countries.
Collapse
Affiliation(s)
- Vasily Isakov
- Department of Gastroenterology and Hepatology, Federal Research Center of Nutrition, Biotechnology and Food Safety, Moscow 115446, Russia
| |
Collapse
|
53
|
Cai B, Xu G, Zhang Z, Tao K, Wang W. Early Oral Feeding is Safe and Comfortable in Patients with Gastric Cancer Undergoing Radical Total Gastrectomy. Nutr Cancer 2024; 77:79-85. [PMID: 39188190 DOI: 10.1080/01635581.2024.2396150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 08/28/2024]
Abstract
Data supporting the safety and clinical efficacy of early oral feeding (EOF) after total gastrectomy are limited. The aim of this prospective randomized controlled study was to explore the safety and clinical efficacy of two early enteral nutrition approaches for gastric cancer patients after radical total gastrectomy. The EOF group had faster postoperative recovery of intestinal function than the enteral tube feeding (ETF) group. The times to first flatus and first defecation were shorter in the EOF group (p < 0.05). In addition, the EOF protocol effectively avoided abdominal distension (p < 0.05). The hospitalization cost of the EOF group was lower than that of the ETF group (p < 0.05). Moreover, oral nutrition satisfied the physiological need for oral intake. People were more satisfied with EOF (p < 0.01). Furthermore, it is worth noting that compared with ETF, EOF did not increase the risk of anastomotic complications such as leakage and bleeding. Most obviously, EOF not only avoided the risk of complications during tube insertion, but also avoided the discomfort experience of nasal feeding tube. In summary, compared with ETF, EOF promotes early bowel recovery effectively without increasing the risk of postoperative complications. It is safe and comfortable for gastric cancer patients undergoing radical total gastrectomy.
Collapse
Affiliation(s)
- Bin Cai
- Department of Quality Management, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Clinical Nutriology, Shaoxing People's Hospital, Shaoxing, China
| | - Guangen Xu
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital, Shaoxing, China
| | - Zhenxing Zhang
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital, Shaoxing, China
| | - Kelong Tao
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital, Shaoxing, China
| | - Wei Wang
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital, Shaoxing, China
| |
Collapse
|
54
|
Inoue M. Epidemiology of Gastric Cancer-Changing Trends and Global Disparities. Cancers (Basel) 2024; 16:2948. [PMID: 39272806 PMCID: PMC11394435 DOI: 10.3390/cancers16172948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
Overall, the past century has seen a substantial decline in gastric cancer, attributable to decreases in risk factors such as H. pylori infection, tobacco smoking, and the intake of salt-preserved food. One potential preventive strategy for those at high risk is H. pylori eradication for infected subjects, but confirmation of this effect awaits longer follow-up. Obesity continues to advance and may cause increases in cardia cancer, particularly in Western populations, and careful monitoring of this outcome is warranted in both Western and Asian populations. These changes in gastric cancer epidemiology foreshadow a new era in gastric cancer control and warrant further monitoring of descriptive patterns and risk factors.
Collapse
Affiliation(s)
- Manami Inoue
- National Cancer Center Institute for Cancer Control, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|
55
|
Wang X, Gao X, Yu J, Zhang X, Nie Y. Emerging trends in early-onset gastric cancer. Chin Med J (Engl) 2024:00029330-990000000-01179. [PMID: 39148190 PMCID: PMC11407816 DOI: 10.1097/cm9.0000000000003259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Indexed: 08/17/2024] Open
Abstract
ABSTRACT The incidence of early-onset gastric cancer (EOGC) is consistently increasing, and its etiology is notably complex. This increase may be attributed to distinctive factors that differ from those associated with late-onset gastric cancer (LOGC), including genetic predispositions, dietary factors, gastric microbiota dysbiosis, and screening of high-risk cases. These factors collectively contribute to the onset of cancer. EOGC significantly differs from LOGC in terms of clinicopathological and molecular characteristics. Moreover, multiple differences in prognosis and clinical management also exist. This study aimed to systematically review the latest research advancements in the epidemiological characteristics, etiological factors, clinicopathological and molecular features, prognosis, and treatment modalities of EOGC.
Collapse
Affiliation(s)
- Xinlin Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi 710032, China
- National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xianchun Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi 710032, China
- National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jun Yu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi 710032, China
- National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xiaotian Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi 710032, China
- Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yongzhan Nie
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi 710032, China
- National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
56
|
Tan N, Wu H, Cao M, Yang F, Yan X, He S, Cao M, Zhang S, Teng Y, Li Q, Wang J, Xia C, Chen W. Global, regional, and national burden of early-onset gastric cancer. Cancer Biol Med 2024; 21:j.issn.2095-3941.2024.0159. [PMID: 39109684 PMCID: PMC11359495 DOI: 10.20892/j.issn.2095-3941.2024.0159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/04/2024] [Indexed: 08/30/2024] Open
Abstract
OBJECTIVE The burden of gastric cancer (GC) across different age groups needs updating. We determined the GC global, regional, and national burden profiles and changes in incidence for 3 sequential 5-year intervals from 2003 to 2017. METHODS The latest incidence and mortality estimates of GC from 185 countries and regions were extracted from the GLOBOCAN 2022 database. The 5-year interval age-standardised incidence rates (ASIRs) were evaluated using cancer registry data from volumes X-XII of the Cancer Incidence in Five Continents (CI5). Correlation analysis was used to evaluate the relationship between ASIR or the age-standardised mortality rate (ASMR) and the Human Development Index (HDI). RESULTS There was an estimated global 968,000 new GC cases and 660,000 deaths in 2022, with male predominance. GC ASIRs and ASMRs were 9.2 and 6.1 per 100,000 persons, respectively. East Asia had the highest burden, with 53.8% of cases and 48.2% of deaths among all geographic regions. There was a significant correlation between ASIR and HDI. Over three 5-year intervals from 2003 to 2017, the incidence of GC notably decreased in most countries but peaked at 2008-2012 in New Zealand, Turkey, and South Africa. Several countries in Europe, Oceania, and America suggest an increasingly concerning trend among younger individuals, especially females. CONCLUSIONS GC is a significant health issue, especially among males and in geographic regions with an HDI, such as eastern Asia. While the incidence of GC is decreasing in many countries due to prevention efforts and improved treatments, a rising trend persists among younger individuals. Comprehensive prevention strategies tailored to different age patterns are clearly needed.
Collapse
Affiliation(s)
- Nuopei Tan
- Department 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 100021, China
| | - Hongliang Wu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Maomao Cao
- Department 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 100021, China
| | - Fan Yang
- Department 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 100021, China
| | - Xinxin Yan
- Department 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 100021, China
| | - Siyi He
- Department 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 100021, China
| | - Mengdi Cao
- Department 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 100021, China
| | - Shaoli Zhang
- Department 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 100021, China
| | - Yi Teng
- Department 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 100021, China
| | - Qianru Li
- Department 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 100021, China
| | - Jiachen Wang
- Department 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 100021, China
| | - Changfa Xia
- Department 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 100021, China
| | - Wanqing Chen
- Department 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 100021, China
| |
Collapse
|
57
|
Elimam H, Abdel Mageed SS, Hatawsh A, Moussa R, Radwan AF, Elfar N, Alhamshry NAA, Abd-Elmawla MA, Mohammed OA, Zaki MB, Doghish AS. Unraveling the influence of LncRNA in gastric cancer pathogenesis: a comprehensive review focus on signaling pathways interplay. Med Oncol 2024; 41:218. [PMID: 39103705 DOI: 10.1007/s12032-024-02455-w] [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: 05/12/2024] [Accepted: 07/16/2024] [Indexed: 08/07/2024]
Abstract
Gastric cancers (GCs) are among the most common and fatal malignancies in the world. Despite our increasing understanding of the molecular mechanisms underlying GC, further biomarkers are still needed for more in-depth examination, focused prognosis, and treatment. GC is one among the long non-coding RNAs, or lncRNAs, that have emerged as key regulators of the pathophysiology of cancer. This comprehensive review focuses on the diverse functions of long noncoding RNAs (lncRNAs) in the development of GC and their interactions with important intracellular signaling pathways. LncRNAs affect GC-related carcinogenic signaling cascades including pathways for EGFR, PI3K/AKT/mTOR, p53, Wnt/β-catenin, JAK/STAT, Hedgehog, NF-κB, and hypoxia-inducible factor. Dysregulated long non-coding RNA (lncRNA) expression has been associated with multiple characteristics of cancer, such as extended growth, apoptosis resistance, enhanced invasion and metastasis, angiogenesis, and therapy resistance. For instance, lncRNAs such as HOTAIR, MALAT1, and H19 promote the development of GC via altering these pathways. Beyond their main roles, GC lncRNAs exhibit potential as diagnostic and prognostic biomarkers. The overview discusses CRISPR/Cas9 genome-modifying methods, antisense oligonucleotides, small molecules, and RNA interference as potential therapeutic approaches to regulate the expression of long noncoding RNAs (lncRNAs). An in-depth discussion of the intricate functions that lncRNAs play in the development of the majority of stomach malignancies is provided in this review. It provides the groundwork for future translational research in lncRNA-based whole processes toward GC by highlighting their carcinogenic effects, regulatory roles in significant signaling cascades, and practical scientific uses as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt.
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Abdulrahman Hatawsh
- Biotechnology School, Nile University, 26th of July Corridor, Sheikh Zayed City, 12588, Giza, Egypt
| | - Rewan Moussa
- Faculty of Medicine, Helwan University, Cairo, 11795, Egypt
| | - Abdullah F Radwan
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, 11829, Cairo, Egypt
| | - Nourhan Elfar
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, New Administrative Capital, 11578, Cairo, Egypt
- Egyptian Drug Authority (EDA), Ministry of Health and Population, Cairo, 11567, Egypt
| | - Nora A A Alhamshry
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Mai A Abd-Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt.
- Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt.
| |
Collapse
|
58
|
Mülder DT, Hahn AI, Huang RJ, Zhou MJ, Blake B, Omofuma O, Murphy JD, Gutiérrez-Torres DS, Zauber AG, O'Mahony JF, Camargo MC, Ladabaum U, Yeh JM, Hur C, Lansdorp-Vogelaar I, Meester R, Laszkowska M. Prevalence of Gastric Precursor Lesions in Countries With Differential Gastric Cancer Burden: A Systematic Review and Meta-analysis. Clin Gastroenterol Hepatol 2024; 22:1605-1617.e46. [PMID: 38438000 PMCID: PMC11272442 DOI: 10.1016/j.cgh.2024.02.023] [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: 02/09/2024] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/06/2024]
Abstract
BACKGROUND & AIMS The prevalence of precursor lesions for gastric cancer (GC) and the differential burden between countries of varying GC risk is not well-understood. We conducted a systematic review and meta-analysis to estimate the global prevalence of precursor lesions. METHODS We estimated the prevalence of atrophic gastritis (AG), gastric intestinal metaplasia (IM), and dysplasia in regions with low, medium, and high GC incidence. Because IM is an advanced manifestation of AG, we assessed the prevalence of less advanced precursors, regardless of the presence of more advanced lesions. Prevalence was sub-stratified by Helicobacter pylori infection, symptomatology, and period (<2000, 2000-2010, and >2010). RESULTS Among the 582 articles that underwent full-text review, 166 studies met inclusion criteria. The global prevalence estimates of AG, IM, and dysplasia were 25.4%, 16.2%, and 2.0%, respectively, on the basis of 126 studies that reported the prevalence of less advanced precursors, regardless of the presence of more advanced lesions. The prevalence of all precursor lesions was higher in high and medium compared with low GC incidence countries (P < .01). Prevalence of AG and IM was significantly higher among H pylori-infected individuals (P < .01) but not statistically different between symptomatic and asymptomatic individuals (P > .17). All precursors demonstrated a secular decrease in prevalence over time. CONCLUSIONS Gastric precursor lesions have differences in prevalence in regions with differential GC incidence and are associated with H pylori infection. Because of the substantial prevalence of precursor lesions in both symptomatic and asymptomatic individuals, symptomatic evaluation may not be sufficient to identify individuals at risk. These estimates provide important insights for tailoring GC prevention strategies.
Collapse
Affiliation(s)
- Duco T Mülder
- Department of Public Health, Erasmus Medical Center, Rotterdam, Netherlands
| | - Anne I Hahn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert J Huang
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Margaret J Zhou
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Benjamin Blake
- Weill Cornell Medical College of Cornell University, New York, New York
| | - Omonefe Omofuma
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - John D Murphy
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | | | - Ann G Zauber
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James F O'Mahony
- Department of Public Health, Erasmus Medical Center, Rotterdam, Netherlands; School of Economics, University College Dublin, Dublin, Ireland
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Uri Ladabaum
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Jennifer M Yeh
- Department of Pediatrics, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts
| | - Chin Hur
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | | | - Reinier Meester
- Department of Public Health, Erasmus Medical Center, Rotterdam, Netherlands; Health Economics & Outcomes Research, Freenome Holdings Inc, San Francisco, California
| | - Monika Laszkowska
- Gastroenterology, Hepatology, and Nutrition Service, Department of Subspecialty Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
59
|
Romatoski KS, de Geus SWL, Miriyam B, Chung SH, Kenzik K, Papageorge MV, Rasic G, Ng SC, Tseng JF, Sachs TE. Overall Volume of Upper Gastrointestinal Surgery Positively Impacts Gastric Cancer Outcomes at Centers with Low Gastrectomy Volume. Ann Surg Oncol 2024; 31:5293-5303. [PMID: 38777899 DOI: 10.1245/s10434-024-15381-y] [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: 02/22/2024] [Accepted: 04/14/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND The relationship between hospital volume and surgical mortality is well documented. However, complete centralization of surgical care is not always feasible. The present study investigates how overall volume of upper gastrointestinal surgery at hospitals influences patient outcomes following resection for gastric adenocarcinoma. PATIENTS AND METHODS National Cancer Database (2010-2019) patients with pathologic stage 1-3 gastric adenocarcinoma who underwent gastrectomy were identified. Three cohorts were created: low-volume hospitals (LVH) for both gastrectomy and overall upper gastrointestinal operations, mixed-volume hospital (MVH) for low-volume gastrectomy but high-volume overall upper gastrointestinal operations, and high-volume gastrectomy hospitals (HVH). Chi-squared tests were used to analyze sociodemographic factors and surgical outcomes and Kaplan-Meier method for survival analysis. RESULTS In total, 26,398 patients were identified (LVH: 20,099; MVH: 539; HVH: 5,760). The 5-year survival was equivalent between MVH and HVH for all stages of disease (MVH: 56.0%, HVH 55.6%; p = 0.9866) and when stratified into early (MVH: 69.9%, HVH: 65.4%; p = 0.1998) and late stages (MVH: 24.7%, HVH: 32.0%; p = 0.1480), while LVH had worse survival. After matching patients, postoperative outcomes were worse for LVH, but there was no difference between MVH and HVH in terms of adequate lymphadenectomy, margin status, readmission rates, and 90-day mortality rates. CONCLUSIONS Despite lower gastrectomy volume for cancer, postoperative gastrectomy outcomes at centers that perform a high number of upper gastrointestinal cancer surgeries were similar to hospitals with high gastrectomy volume. These hospitals offer a blueprint for providing equivalent outcomes to high volume centers while enhancing availability of quality cancer care.
Collapse
Affiliation(s)
- Kelsey S Romatoski
- Department of Surgery, Section of Surgical Oncology, Boston Medical Center, Boston, MA, USA
- Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Susanna W L de Geus
- Department of Surgery, Section of Surgical Oncology, Boston Medical Center, Boston, MA, USA
- Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Bharath Miriyam
- Department of Surgery, Section of Surgical Oncology, Boston Medical Center, Boston, MA, USA
- Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sophie H Chung
- Department of Surgery, Section of Surgical Oncology, Boston Medical Center, Boston, MA, USA
- Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kelly Kenzik
- Department of Surgery, Section of Surgical Oncology, Boston Medical Center, Boston, MA, USA
- Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Marianna V Papageorge
- Department of Surgery, Section of Surgical Oncology, Boston Medical Center, Boston, MA, USA
- Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Surgery, Yale Medical School, Yale New Haven Hospital, New Haven, CT, USA
| | - Gordana Rasic
- Department of Surgery, Section of Surgical Oncology, Boston Medical Center, Boston, MA, USA
- Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sing Chau Ng
- Department of Surgery, Section of Surgical Oncology, Boston Medical Center, Boston, MA, USA
- Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jennifer F Tseng
- Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Teviah E Sachs
- Department of Surgery, Section of Surgical Oncology, Boston Medical Center, Boston, MA, USA.
- Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
60
|
Cai J, Tan X, Hu Q, Pan H, Zhao M, Guo C, Zeng J, Ma X, Zhao Y. Flavonoids and Gastric Cancer Therapy: From Signaling Pathway to Therapeutic Significance. Drug Des Devel Ther 2024; 18:3233-3253. [PMID: 39081701 PMCID: PMC11287762 DOI: 10.2147/dddt.s466470] [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: 03/01/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Gastric cancer (GC) is a prevalent gastrointestinal tumor characterized by high mortality and recurrence rates. Current treatments often have limitations, prompting researchers to explore novel anti-tumor substances and develop new drugs. Flavonoids, natural compounds with diverse biological activities, are gaining increasing attention in this regard. We searched from PubMed, Web of Science, SpringerLink and other databases to find the relevant literature in the last two decades. Using "gastric cancer", "stomach cancers", "flavonoid", "bioflavonoid", "2-Phenyl-Chromene" as keywords, were searched, then analyzed and summarized the mechanism of flavonoids in the treatment of GC. It was revealed that the anti-tumor mechanism of flavonoids involves inhibiting tumor growth, proliferation, invasion, and metastasis, as well as inducing cell death through various processes such as apoptosis, autophagy, ferroptosis, and pyroptosis. Additionally, combining flavonoids with other chemotherapeutic agents like 5-FU and platinum compounds can potentially reduce chemoresistance. Flavonoids have also demonstrated enhanced biological activity when used in combination with other natural products. Consequently, this review proposes innovative perspectives for the development of flavonoids as new anti-GC agents.
Collapse
Affiliation(s)
- Jiaying Cai
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Xiyue Tan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Qichao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Huafeng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Maoyuan Zhao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Cui Guo
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Jinhao Zeng
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Yanling Zhao
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
61
|
Janiczek-Polewska M, Kolenda T, Poter P, Kozłowska-Masłoń J, Jagiełło I, Regulska K, Malicki J, Marszałek A. Diagnostic Potential of miR-143-5p, miR-143-3p, miR-551b-5p, and miR-574-3p in Chemoresistance of Locally Advanced Gastric Cancer: A Preliminary Study. Int J Mol Sci 2024; 25:8057. [PMID: 39125625 PMCID: PMC11311514 DOI: 10.3390/ijms25158057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
Gastric cancer (GC) is one of the most frequently diagnosed cancers in the world. Although the incidence is decreasing in developed countries, the treatment results are still unsatisfactory. The standard treatment for locally advanced gastric cancer (LAGC) is gastrectomy with perioperative chemotherapy. The association of selected microRNAs (miRNAs) with chemoresistance was assessed using archival material of patients with LAGC. Histological material was obtained from each patient via a biopsy performed during gastroscopy and then after surgery, which was preceded by four cycles of neoadjuvant chemotherapy (NAC) according to the FLOT or FLO regimen. The expression of selected miRNAs in the tissue material was assessed, including miRNA-21-3p, miRNA-21-5p, miRNA-106a-5p, miRNA-122-3p, miRNA-122-5p, miRNA-143-3p, miRNA-143-5p, miRNA-203a-3p, miRNA-203-5p, miRNA-551b-3p, miRNA-551b-5p, and miRNA-574-3p. miRNA expression was assessed using quantitative reverse transcription polymerase chain reaction (qRT-PCR). The response to NAC was assessed using computed tomography of the abdomen and chest and histopathology after gastrectomy. The statistical analyses were performed using GraphPad Prism 9. The significance limit was set at p < 0.05. We showed that the expression of miR-143-3p, miR-143-5p, and miR-574-3p before surgery, and miR-143-5p and miR-574-3p after surgery, decreased in patients with GC. The expression of miR-143-3p, miR-143-5p, miR-203a-3p, and miR-551b-5p decreased in several patients who responded to NAC. The miRNA most commonly expressed in these cases was miRNA-551b-5p. Moreover, it showed expression in a patient whose response to chemotherapy was inconsistent between the histopathological results and computed tomography. The expression of miR-143-3p, miR-143-5p, miR-203a-3p, and miR-551b-5p in formalin-fixed paraffin-embedded tissue (FFPET) samples can help differentiate between the responders and non-responders to NAC in LAGC. miR-143-3p, miR-143-5p, and miR-574-3p expression may be used as a potential diagnostic tool in GC patients. The presence of miR-551b-5p may support the correct assessment of a response to NAC in GC via CT.
Collapse
Affiliation(s)
- Marlena Janiczek-Polewska
- Department of Clinical Oncology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
- Department of Electroradiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Tomasz Kolenda
- Laboratory of Cancer Genetics, Greater Poland Cancer Centre, 61-866 Poznan, Poland
- Research and Implementation Unit, Greater Poland Cancer Centre, 61-866 Poznan, Poland;
| | - Paulina Poter
- Department of Clinical Pathology, Poznan University of Medical Sciences and Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Joanna Kozłowska-Masłoń
- Laboratory of Cancer Genetics, Greater Poland Cancer Centre, 61-866 Poznan, Poland
- Research and Implementation Unit, Greater Poland Cancer Centre, 61-866 Poznan, Poland;
- Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University, 61-614 Poznan, Poland
| | - Inga Jagiełło
- Department of Clinical Pathology, Poznan University of Medical Sciences and Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Katarzyna Regulska
- Research and Implementation Unit, Greater Poland Cancer Centre, 61-866 Poznan, Poland;
- Pharmacy, Greater Poland Cancer Centre, 61-866 Poznan, Poland
- Department of Clinical Pharmacy and Biopharmacy, Poznan University of Medical Sciences, Collegium Pharmaceuticum, 60-806 Poznan, Poland
| | - Julian Malicki
- Department of Electroradiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Andrzej Marszałek
- Department of Clinical Pathology, Poznan University of Medical Sciences and Greater Poland Cancer Centre, 61-866 Poznan, Poland
| |
Collapse
|
62
|
Chauhan A, Pathak VM, Yadav M, Chauhan R, Babu N, Chowdhary M, Ranjan A, Mathkor DM, Haque S, Tuli HS, Ramniwas S, Yadav V. Role of ursolic acid in preventing gastrointestinal cancer: recent trends and future perspectives. Front Pharmacol 2024; 15:1405497. [PMID: 39114347 PMCID: PMC11303223 DOI: 10.3389/fphar.2024.1405497] [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: 03/23/2024] [Accepted: 06/03/2024] [Indexed: 08/10/2024] Open
Abstract
Gastrointestinal malignancies are one of the major worldwide health concerns. In the present review, we have assessed the plausible therapeutic implication of Ursolic Acid (UA) against gastrointestinal cancer. By modulating several signaling pathways critical in cancer development, UA could offer anti-inflammatory, anti-proliferative, and anti-metastatic properties. However, being of low oral bioavailability and poor permeability, its clinical value is restricted. To deliver and protect the drug, liposomes and polymer micelles are two UA nanoformulations that can effectively increase medicine stability. The use of UA for treating cancers is safe and appropriate with low toxicity characteristics and a predictable pharmacokinetic profile. Although the bioavailability of UA is limited, its nanoformulations could emerge as an alternative to enhance its efficacy in treating GI cancers. Further optimization and validation in the clinical trials are necessary. The combination of molecular profiling with nanoparticle-based drug delivery technologies holds the potential for bringing UA to maximum efficacy, looking for good prospects with GI cancer treatment.
Collapse
Affiliation(s)
- Abhishek Chauhan
- Amity Institute of Environmental Toxicology Safety and Management, Amity University, Noida, Uttar Pradesh, India
| | | | - Monika Yadav
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Ritu Chauhan
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Neelesh Babu
- Department of Microbiology, Baba Farid Institute of Technology, Dehradun, Uttarakhand, India
| | - Manish Chowdhary
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Anuj Ranjan
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Darin Mansor Mathkor
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Ambala, India
| | - Seema Ramniwas
- University Centre for Research and Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Mohali, India
| | - Vikas Yadav
- Department of Translational Medicine, Clinical Research Centre, Skåne University Hospital, Lund University, Malmö, Sweden
| |
Collapse
|
63
|
Wei X, Liu Q, Liu L, Wang D, Liu J, Zhu Q, Xu Z, Chen Q, Xu W. Discovery of the Natural Bibenzyl Compound Erianin in Dendrobium Inhibiting the Growth and EMT of Gastric Cancer through Downregulating the LKB1-SIK2/3-PARD3 Pathway. Int J Mol Sci 2024; 25:7973. [PMID: 39063214 PMCID: PMC11277420 DOI: 10.3390/ijms25147973] [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: 05/26/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Erianin, a bibenzyl compound found in dendrobium extract, has demonstrated broad anticancer activity. However, its mechanism of action in gastric cancer (GC) remains poorly understood. LKB1 is a tumor-suppressor gene, and its mutation is an important driver of various cancers. Yet some studies have reported contradictory findings. In this study, we combined bioinformatics and in vitro and in vivo experiments to investigate the effect and potential mechanism of Erianin in the treatment of GC. The results show that LKB1 was highly expressed in patients' tumor tissues and GC cells, and it was associated with poor patient prognosis. Erianin could promote GC cell apoptosis and inhibit the scratch repair, migration, invasion, and epithelial-mesenchymal transition (EMT) characteristics. Erianin dose-dependently inhibited the expression of LKB1, SIK2, SIK3, and PARD3 but had no significant effect on SIK1. Erianin also inhibited tumor growth in CDX mice model. Unexpectedly, 5-FU also exhibited a certain inhibitory effect on LKB1. The combination of Erianin and 5-FU significantly improved the anti-tumor efficacy of 5-FU in the growth of GC cells and xenograft mouse models. In summary, Erianin is a potential anti-GC compound that can inhibit GC growth and EMT properties by targeting the LKB1-SIK2/3-PARD3-signaling axis. The synergistic effect of Erianin and 5-FU suggests a promising therapeutic strategy for GC treatment.
Collapse
Affiliation(s)
- Xin Wei
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; (X.W.); (Q.Z.); (Q.C.)
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (Q.L.); (L.L.); (D.W.); (J.L.); (Z.X.)
| | - Qunshan Liu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (Q.L.); (L.L.); (D.W.); (J.L.); (Z.X.)
| | - Liu Liu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (Q.L.); (L.L.); (D.W.); (J.L.); (Z.X.)
| | - Dan Wang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (Q.L.); (L.L.); (D.W.); (J.L.); (Z.X.)
| | - Jiajia Liu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (Q.L.); (L.L.); (D.W.); (J.L.); (Z.X.)
| | - Qizhi Zhu
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; (X.W.); (Q.Z.); (Q.C.)
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (Q.L.); (L.L.); (D.W.); (J.L.); (Z.X.)
| | - Ziming Xu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (Q.L.); (L.L.); (D.W.); (J.L.); (Z.X.)
| | - Qi Chen
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; (X.W.); (Q.Z.); (Q.C.)
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (Q.L.); (L.L.); (D.W.); (J.L.); (Z.X.)
| | - Weiping Xu
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; (X.W.); (Q.Z.); (Q.C.)
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (Q.L.); (L.L.); (D.W.); (J.L.); (Z.X.)
- Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei 230001, China
| |
Collapse
|
64
|
Yan P, Zhao D. Association between serum total cholesterol and the development of gastric cancer: A two-way two-sample Mendelian randomization study. Medicine (Baltimore) 2024; 103:e38900. [PMID: 38996131 PMCID: PMC11245271 DOI: 10.1097/md.0000000000038900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Previous epidemiologic studies have suggested a potential negative correlation between total cholesterol (TC) and Gastric cancer (GC); however, several observational studies have shown conflicting results and have failed to provide definitive evidence for a causal relationship between TC and GC. Therefore, we conducted a 2-sample bidirectional Mendelian randomization (MR) study to explore the genetic correlation and potential causal relationship between the 2 variables. We screened for single nucleotide polymorphisms (SNPs) associated with TC and GC utilizing a large-scale genome-wide association study (GWAS) public database. The causal relationship was analyzed using 5 MR analysis methods: inverse variance weighting (IVW), weighted median, MR-Egger regression, weighted mode, and simple mode. Additionally, reverse MR analysis was performed to evaluate the possibility of reverse causality. Sensitivity analyses were conducted, including heterogeneity tests, horizontal multiple validity tests, and leave-one-out tests. After meticulous screening, 79 SNPs were identified as instrumental variables (IVs). The IVW method revealed a causal relationship between TC and GC (OR = 0.844; 95% CI: 0.741-0.961; P = .01). Sensitivity analyses did not detect significant horizontal pleiotropy. Though heterogeneity was observed in the forward MR analysis (IVW, Qp = 0.0006), the results remained reliable as we utilized the IVW random-effects model as the primary analytical method. Furthermore, inverse MR analysis found no evidence of reverse causality between TC and GC, effectively ruling out the influence of GC on the reverse causality of TC. Our MR study provided evidence of a causal association between TC and GC, suggesting that TC acts as a protective factor against GC due to its negative association with the disease.
Collapse
Affiliation(s)
- Peng Yan
- Department of Medical Oncology, Lixin County People’s Hospital, Bozhou, Anhui, China
| | - Dong Zhao
- Department of Medical Oncology, Lixin County People’s Hospital, Bozhou, Anhui, China
| |
Collapse
|
65
|
Lin JL, Lin JX, Lin GT, Huang CM, Zheng CH, Xie JW, Wang JB, Lu J, Chen QY, Li P. Global incidence and mortality trends of gastric cancer and predicted mortality of gastric cancer by 2035. BMC Public Health 2024; 24:1763. [PMID: 38956557 PMCID: PMC11221210 DOI: 10.1186/s12889-024-19104-6] [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: 07/18/2023] [Accepted: 06/11/2024] [Indexed: 07/04/2024] Open
Abstract
OBJECTIVE To study the historical global incidence and mortality trends of gastric cancer and predicted mortality of gastric cancer by 2035. METHODS Incidence data were retrieved from the Cancer Incidence in Five Continents (CI5) volumes I-XI, and mortality data were obtained from the latest update of the World Health Organization (WHO) mortality database. We used join-point regression analysis to examine historical incidence and mortality trends and used the package NORDPRED in R to predict the number of deaths and mortality rates by 2035 by country and sex. RESULTS More than 1,089,000 new cases of gastric cancer and 769,000 related deaths were reported in 2020. The average annual percent change (AAPC) in the incidence of gastric cancer from 2003 to 2012 among the male population, South Korea, Japan, Malta, Canada, Cyprus, and Switzerland showed an increasing trend (P > 0.05); among the female population, Canada [AAPC, 1.2; (95%Cl, 0.5-2), P < 0.05] showed an increasing trend; and South Korea, Ecuador, Thailand, and Cyprus showed an increasing trend (P > 0.05). AAPC in the mortality of gastric cancer from 2006 to 2015 among the male population, Thailand [3.5 (95%cl, 1.6-5.4), P < 0.05] showed an increasing trend; Malta Island, New Zealand, Turkey, Switzerland, and Cyprus had an increasing trend (P > 0.05); among the male population aged 20-44, Thailand [AAPC, 3.4; (95%cl, 1.3-5.4), P < 0.05] showed an increasing trend; Norway, New Zealand, The Netherlands, Slovakia, France, Colombia, Lithuania, and the USA showed an increasing trend (P > 0.05). It is predicted that the mortality rate in Slovenia and France's female population will show an increasing trend by 2035. It is predicted that the absolute number of deaths in the Israeli male population and in Chile, France, and Canada female population will increase by 2035. CONCLUSION In the past decade, the incidence and mortality of gastric cancer have shown a decreasing trend; however, there are still some countries showing an increasing trend, especially among populations younger than 45 years. Although mortality in most countries is predicted to decline by 2035, the absolute number of deaths due to gastric cancer may further increase due to population growth.
Collapse
Affiliation(s)
- Ju-Li Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Guang-Tan Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China.
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou , Fujian Province, 350001, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China.
| |
Collapse
|
66
|
Sun C, Huang J, Guo X, Zhang C, Wei L, Wong KI, Yang Z, Zhao G, Lu M, Yao W. An all-in-one therapeutic platform for the treatment of resistant Helicobacter pylori infection. Biomaterials 2024; 308:122540. [PMID: 38537343 DOI: 10.1016/j.biomaterials.2024.122540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/26/2024] [Accepted: 03/19/2024] [Indexed: 05/03/2024]
Abstract
Helicobacter pylori (H. pylori) infection is a major cause of gastric diseases. Currently, bismuth-based quadruple therapy is widely adopted for eradicating H. pylori infection. However, this first-line strategy faces several challenges such as drug resistance, intestinal dysbacteriosis, and patients' poor compliance. To overcome these problems, an all-in-one therapeutic platform (CLA-Bi-ZnO2@Lipo) that composed of liposomes loading clarithromycin (CLA), Bi, and ZnO2 hybrid nanoparticles was developed for eradicating multidrug-resistant (MDR) H. pylori. The in vitro and in vivo results showed that CLA-Bi-ZnO2@Lipo could target the infection-induced inflammatory mucosa through liposome mediated nanoparticle-tissue surface charge interaction and quickly respond to the gastric acid environment to release CLA, Bi3+, Zn2+, and H2O2. By oral administration per day, the acid triggered decomposition of CLA-Bi-ZnO2@Lipo could significantly increase intragastric pH to 6 within 30 min; The released CLA, Zn2+, and H2O2 further exerted synergistical anti-bacterial effects in which a ∼2 order higher efficacy in reducing MDR H. pylori burden was achieved in comparison with standard quadruple therapy (p < 0.05); The released Zn2+ and Bi3+ could also alleviate mucosal inflammation. Most importantly, the CLA-Bi-ZnO2@Lipo exhibited superior biosafety and nearly no side effects on intestinal flora. Overall, this study developed a highly integrated and safe anti-MDR H. pylori agent which had great potential to be used as an alternative treatment for MDR H. pylori eradication.
Collapse
Affiliation(s)
- Chao Sun
- Department of General Practice, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Jia Huang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Xiaoqian Guo
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Chenli Zhang
- Department of General Practice, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Li Wei
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Ka Ioi Wong
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Ziyun Yang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Gang Zhao
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China.
| | - Min Lu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China.
| | - Weiyan Yao
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China.
| |
Collapse
|
67
|
Kronsteiner-Gicevic S, Thompson AS, Gaggl M, Bell W, Cassidy A, Kühn T. Adding salt to food at table as an indicator of gastric cancer risk among adults: a prospective study. Gastric Cancer 2024; 27:714-721. [PMID: 38630317 PMCID: PMC11193689 DOI: 10.1007/s10120-024-01502-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/03/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND While dietary salt intake has been linked with gastric cancer risk in Asian studies, findings from Western populations are sparse and limited to case-control studies. Our aim was to evaluate the frequency of adding salt to food at table in relation to gastric cancer risk among UK adults. METHODS We evaluated associations between the frequency of adding salt to food and the risk of gastric cancer in the UK Biobank (N = 471,144) using multivariable Cox regression. Frequency of adding salt to food was obtained from a touchscreen questionnaire completed at baseline (2006-2010). 24-h urinary sodium excretion was estimated using INTERSALT formulae. Cancer incidence was obtained by linkage to national cancer registries. RESULTS During a median follow-up period of 10.9 years, 640 gastric cancer cases were recorded. In multivariable models, the gastric cancer risk among participants reporting adding salt to food at table "always" compared to those who responded "never/rarely" was HR = 1.41 (95% CI: 1.04, 1.90). There was a positive linear association between estimated 24-h urinary sodium levels and the frequency of adding salt to food (p-trend <0 .001). However, no significant association between estimated 24-h urinary sodium with gastric cancer was observed (HR = 1.19 (95% CI: 0.87, 1.61)). CONCLUSIONS "Always adding salt to food" at table was associated with a higher gastric cancer risk in a large sample of UK adults. High frequency of adding salt to food at table can potentially serve as a useful indicator of salt intake for surveillance purposes and a basis for devising easy-to-understand public health messages.
Collapse
Affiliation(s)
- Selma Kronsteiner-Gicevic
- Department of Nutritional Sciences, University of Vienna, Josef-Holaubek-Platz 2 (UZA II), 1090, Vienna, Austria.
- Center for Public Health, Medical University of Vienna, Kinderspitalgasse 15, 1090, Vienna, Austria.
| | - Alysha S Thompson
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast, BT9 5DL, UK
| | - Martina Gaggl
- Center for Public Health, Medical University of Vienna, Kinderspitalgasse 15, 1090, Vienna, Austria
| | - William Bell
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast, BT9 5DL, UK
| | - Aedín Cassidy
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast, BT9 5DL, UK
| | - Tilman Kühn
- Department of Nutritional Sciences, University of Vienna, Josef-Holaubek-Platz 2 (UZA II), 1090, Vienna, Austria
- Center for Public Health, Medical University of Vienna, Kinderspitalgasse 15, 1090, Vienna, Austria
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast, BT9 5DL, UK
| |
Collapse
|
68
|
Skryabin GO, Beliaeva AA, Enikeev AD, Tchevkina EM. Extracellular Vesicle miRNAs in Diagnostics of Gastric Cancer. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1211-1238. [PMID: 39218020 DOI: 10.1134/s0006297924070058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/24/2024] [Accepted: 05/30/2024] [Indexed: 09/04/2024]
Abstract
Gastric cancer (GC) poses a significant global health challenge because of its high mortality rate attributed to the late-stage diagnosis and lack of early symptoms. Early cancer diagnostics is crucial for improving the survival rates in GC patients, which emphasizes the importance of identifying GC markers for liquid biopsy. The review discusses a potential use of extracellular vesicle microRNAs (EV miRNAs) as biomarkers for the diagnostics and prognostics of GC. Methods. Original articles on the identification of EV miRNA as GC markers published in the Web of Science and Scopus indexed issues were selected from the PubMed and Google Scholar databases. We focused on the methodological aspects of EV analysis, including the choice of body fluid, methods for EV isolation and validation, and approaches for EV miRNA analysis. Conclusions. Out of 33 found articles, the majority of authors investigated blood-derived extracellular vesicles (EVs); only a few utilized EVs from other body fluids, including tissue-specific local biofluids (washing the tumor growth areas), which may be a promising source of EVs in the context of cancer diagnostics. GC-associated miRNAs identified in different studies using different methods of EV isolation and analysis varied considerably. However, three miRNAs (miR-10b, miR-21, and miR-92a) have been found in several independent studies and shown to be associated with GC in experimental models. Further studies are needed to determine the optimal miRNA marker panel. Another essential step necessary to improve the reliability and reproducibility of EV-based diagnostics is standardization of methodologies for EV handling and analysis of EV miRNA.
Collapse
Affiliation(s)
- Gleb O Skryabin
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115522, Russia.
| | - Anastasiya A Beliaeva
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115522, Russia
| | - Adel D Enikeev
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115522, Russia
| | - Elena M Tchevkina
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115522, Russia
| |
Collapse
|
69
|
Herrera-Pariente C, Bonjoch L, Muñoz J, Fernàndez G, Soares de Lima Y, Mahmood R, Cuatrecasas M, Ocaña T, Lopez-Prades S, Llargués-Sistac G, Domínguez-Rovira X, Llach J, Luzko I, Díaz-Gay M, Lazaro C, Brunet J, Castillo-Manzano C, García-González MA, Lanas A, Carrillo M, Hernández San Gil R, Quintero E, Sala N, Llort G, Aguilera L, Carot L, Diez-Redondo P, Jover R, Ramon Y Cajal T, Cubiella J, Castells A, Balaguer F, Bujanda L, Castellví-Bel S, Moreira L. CTNND1 is involved in germline predisposition to early-onset gastric cancer by affecting cell-to-cell interactions. Gastric Cancer 2024; 27:747-759. [PMID: 38796558 PMCID: PMC11193828 DOI: 10.1007/s10120-024-01504-7] [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: 12/07/2023] [Accepted: 04/20/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND CDH1 and CTNNA1 remain as the main genes for hereditary gastric cancer. However, they only explain a small fraction of gastric cancer cases with suspected inherited basis. In this study, we aimed to identify new hereditary genes for early-onset gastric cancer patients (EOGC; < 50 years old). METHODS After germline exome sequencing in 20 EOGC patients and replication of relevant findings by gene-panel sequencing in an independent cohort of 152 patients, CTNND1 stood out as an interesting candidate gene, since its protein product (p120ctn) directly interacts with E-cadherin. We proceeded with functional characterization by generating two knockout CTNND1 cellular models by gene editing and introducing the detected genetic variants using a lentiviral delivery system. We assessed β-catenin and E-cadherin levels, cell detachment, as well as E-cadherin localization and cell-to-cell interaction by spheroid modeling. RESULTS Three CTNND1 germline variants [c.28_29delinsCT, p.(Ala10Leu); c.1105C > T, p.(Pro369Ser); c.1537A > G, p.(Asn513Asp)] were identified in our EOGC cohorts. Cells encoding CTNND1 variants displayed altered E-cadherin levels and intercellular interactions. In addition, the p.(Pro369Ser) variant, located in a key region in the E-cadherin/p120ctn binding domain, showed E-cadherin mislocalization. CONCLUSIONS Defects in CTNND1 could be involved in germline predisposition to gastric cancer by altering E-cadherin and, consequently, cell-to-cell interactions. In the present study, CTNND1 germline variants explained 2% (3/172) of the cases, although further studies in larger external cohorts are needed.
Collapse
Affiliation(s)
- Cristina Herrera-Pariente
- Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), CIBEREHD, Universitat de Barcelona, Hospital Clínic, Villarroel 170, 08036, Barcelona, Spain
| | - Laia Bonjoch
- Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), CIBEREHD, Universitat de Barcelona, Hospital Clínic, Villarroel 170, 08036, Barcelona, Spain
| | - Jenifer Muñoz
- Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), CIBEREHD, Universitat de Barcelona, Hospital Clínic, Villarroel 170, 08036, Barcelona, Spain
| | | | - Yasmin Soares de Lima
- Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), CIBEREHD, Universitat de Barcelona, Hospital Clínic, Villarroel 170, 08036, Barcelona, Spain
| | - Romesa Mahmood
- Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), CIBEREHD, Universitat de Barcelona, Hospital Clínic, Villarroel 170, 08036, Barcelona, Spain
| | - Miriam Cuatrecasas
- Pathology, Hospital Clínic, FRCB-IDIBAPS, CIBEREHD, 08036, Barcelona, Spain
| | - Teresa Ocaña
- Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), CIBEREHD, Universitat de Barcelona, Hospital Clínic, Villarroel 170, 08036, Barcelona, Spain
| | | | - Gemma Llargués-Sistac
- Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), CIBEREHD, Universitat de Barcelona, Hospital Clínic, Villarroel 170, 08036, Barcelona, Spain
| | - Xavier Domínguez-Rovira
- Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), CIBEREHD, Universitat de Barcelona, Hospital Clínic, Villarroel 170, 08036, Barcelona, Spain
| | - Joan Llach
- Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), CIBEREHD, Universitat de Barcelona, Hospital Clínic, Villarroel 170, 08036, Barcelona, Spain
| | - Irina Luzko
- Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), CIBEREHD, Universitat de Barcelona, Hospital Clínic, Villarroel 170, 08036, Barcelona, Spain
| | - Marcos Díaz-Gay
- Department of Cellular and Molecular Medicine and Department of Bioengineering and Moores Cancer Center, UC San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Conxi Lazaro
- Hereditary Cancer Program, Catalan Institute of Oncology, IDIBELL, CIBERONC, 08908, Barcelona, Spain
| | - Joan Brunet
- Hereditary Cancer Program, Catalan Institute of Oncology, IDIBELL, CIBERONC, 08908, Barcelona, Spain
- Hereditary Cancer Program, Catalan Institute of Oncology, IDIBGI, 17190, Girona, Spain
| | | | - María Asunción García-González
- Instituto de Investigación Sanitaria Aragón, Instituto Aragonés de Ciencias de La Salud, CIBEREHD, 50009, Zaragoza, Spain
| | - Angel Lanas
- Instituto de Investigación Sanitaria Aragón, Instituto Aragonés de Ciencias de La Salud, CIBEREHD, 50009, Zaragoza, Spain
- Gastroenterology, Hospital Clínico Universitario de Zaragoza, Instituto de Investigación Sanitaria Aragón, Universidad de Zaragoza, CIBEREHD, 50009, Zaragoza, Spain
| | - Marta Carrillo
- Gastroenterology, Centro de Investigación Biomédica de Canarias (CIBICAN), Hospital Universitario de Canarias, Instituto Universitario de Tecnologías Biomédicas (ITB), Universidad de La Laguna, 38320, Santa Cruz de Tenerife, Spain
| | | | - Enrique Quintero
- Gastroenterology, Centro de Investigación Biomédica de Canarias (CIBICAN), Hospital Universitario de Canarias, Instituto Universitario de Tecnologías Biomédicas (ITB), Universidad de La Laguna, 38320, Santa Cruz de Tenerife, Spain
| | - Nuria Sala
- Unit of Nutrition and Cancer, Translational Research Laboratory, Catalan Institute of Oncology (ICO) and Bellvitge Biomedical Research Institute (IDIBELL), 08908, Barcelona, Spain
| | - Gemma Llort
- Medical Oncology, Parc Taulí University Hospital, 08208, Sabadell, Spain
| | - Lara Aguilera
- Gastroenterology, Vall d'Hebron Research Institute, 08035, Barcelona, Spain
| | - Laura Carot
- Gastroenterology, Hospital del Mar, 08003, Barcelona, Spain
| | | | - Rodrigo Jover
- Gastroenterology, Departamento de Medicina Clínica, Hospital General Universitario Dr. Balmis, Instituto de Investigación Sanitaria ISABIAL, Universidad Miguel Hernández, 03010, Alicante, Spain
| | | | - Joaquín Cubiella
- Gastroenterology, Complexo Hospitalario de Ourense, CIBEREHD, 32005, Ourense, Spain
| | - Antoni Castells
- Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), CIBEREHD, Universitat de Barcelona, Hospital Clínic, Villarroel 170, 08036, Barcelona, Spain
| | - Francesc Balaguer
- Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), CIBEREHD, Universitat de Barcelona, Hospital Clínic, Villarroel 170, 08036, Barcelona, Spain
| | - Luis Bujanda
- Department of Hepatology and Gastroenterology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Biodonostia Health Research Institute - Donostia University Hospital, Universidad del País Vasco (UPV/EHU), 20014, San Sebastián, Spain
| | - Sergi Castellví-Bel
- Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), CIBEREHD, Universitat de Barcelona, Hospital Clínic, Villarroel 170, 08036, Barcelona, Spain
| | - Leticia Moreira
- Gastroenterology, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi I Sunyer (FRCB-IDIBAPS), CIBEREHD, Universitat de Barcelona, Hospital Clínic, Villarroel 170, 08036, Barcelona, Spain.
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain.
| |
Collapse
|
70
|
Abbas M, Ramspott JP, Chourio Barboza DE, Pascher A, Wardelmann E, Sporn JC. Modified scoring system for the quantitative assessment of histological regression in peritoneal carcinomatosis after pressurized intraperitoneal aerosol chemotherapy: A pilot study. Oncol Lett 2024; 28:308. [PMID: 38784603 PMCID: PMC11112145 DOI: 10.3892/ol.2024.14441] [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: 01/12/2024] [Accepted: 04/11/2024] [Indexed: 05/25/2024] Open
Abstract
Peritoneal carcinomatosis is one of the leading causes of death in patients with gastrointestinal cancer. Newer locoregional treatment concepts include pressurized intraperitoneal aerosol chemotherapy (PIPAC), the regional application of pressurized chemotherapeutic agents to the abdominal cavity, which is usually performed every 4 to 8 weeks. One of the main challenges of PIPAC therapy remains the objective assessment of treatment response. The present study describes a new scoring system to histologically assess the regression of peritoneal cancer following PIPAC therapy, quantitative assessment of histological regression in peritoneal carcinomatosis (QARP). Peritoneal biopsies from 27 patients with peritoneal metastases undergoing PIPAC were obtained and processed in a standardized fashion. Biopsies were scored according to the QARP grading system. The five-tiered system was graded as follows, Grade 0, no residual tumor cells with regressive changes present; grade 1, 1-25% viable tumor cells per tumor focus with regressive changes present; grade 2, 26-50% viable tumor cells per tumor focus with regressive changes present; grade 3, 51-75% viable tumor cells per tumor focus with few regressive changes; grade 4, >75% viable tumor cells per tumor focus with minimal or no regressive changes. Based on the new grading system, the study cohort was divided into QARP responders and QARP non-responders following PIPAC treatment. Higher QARP scores were significantly correlated with higher PCI scores (r=0.32; P=0.007). However, no difference in overall survival was detected between QARP responders and QARP non-responders. Further studies are required to ascertain the reproducibility and prognostic significance of QARP.
Collapse
Affiliation(s)
- Mahmoud Abbas
- Department of Pathology, University Hospital Muenster, D-48149 Muenster, Germany
| | - Jan Philipp Ramspott
- Department of General, Visceral and Transplant Surgery, University Hospital Muenster, D-48149 Muenster, Germany
| | | | - Andreas Pascher
- Department of General, Visceral and Transplant Surgery, University Hospital Muenster, D-48149 Muenster, Germany
| | - Eva Wardelmann
- Department of Pathology, University Hospital Muenster, D-48149 Muenster, Germany
| | - Judith C. Sporn
- Department of General, Visceral and Transplant Surgery, University Hospital Muenster, D-48149 Muenster, Germany
| |
Collapse
|
71
|
Xu Q, Xue S, Zhang Y, Li J, Qian P, Zhang Y, Feng L. Identification and validation of Cystatin A as a novel promising therapeutic target for gastric cancer. J Gastrointest Oncol 2024; 15:873-889. [PMID: 38989439 PMCID: PMC11231850 DOI: 10.21037/jgo-23-941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 06/24/2024] [Indexed: 07/12/2024] Open
Abstract
Background The effect of pharmacological treatment of gastric cancer (GC) is limited, thus, it holds significant scientific importance to thoroughly investigate the molecular mechanisms underlying GC development and identify novel molecules capable of substantially extending patients' survival. This study utilized bioinformatics techniques to identify 11 genes associated with recurrence-free survival (RFS) in GC patients and investigated the potential biological functions of these genes through single-cell transcriptomic analysis. Subsequently, a single gene Cystatin A (CSTA) was selected for further analysis to explore its impact on signaling pathways and treatment. Methods Differentially expressed genes (DEGs) were identified and overlapped in the analysis of RFS to identify potential prognostic genes for GC patients, based on data from the Cancer Genome Atlas-stomach adenocarcinoma (TCGA-STAD) and GSE54129. Subsequently, a prognostic model based on RFS in GC patients was established. Single-cell sequencing data were employed to explore the potential functions of these model genes. CSTA, one of the RFS-related genes, was further investigated using immunohistochemistry (IHC), Cell Counting Kit 8 (CCK-8), transwell, scratch, colony formation assays, flow cytometry, and Western blotting methods. Results Through bioinformatics analysis, we identified 23 RFS-related genes in GC. Using the least absolute shrinkage and selection operator (LASSO)-Cox method, an RFS prognostic model was developed which pinpointed 11 GC prognosis-related (GPR) genes as significant factors influencing RFS in GC patients. The single-cell analysis revealed their potential role in affecting differentiation and maturation of pre-fibroblasts thereby impacting RFS in GC patients. CSTA exhibited low expression levels in GC tissues. Overexpression of CSTA promoted apoptosis in GC cells through the caspase-dependent apoptotic pathway and enhanced their response to cisplatin via this same pathway. Conclusions The 11 GPR genes are primarily enriched within a specific type of stromal cell exhibiting heightened communication, metabolism, and differentiation levels. The gene signature of these stromal cells has implications for patient prognosis. Additionally, CSTA, a gene related to prognosis, has been shown to influence apoptosis levels in GC cells.
Collapse
Affiliation(s)
- Qingyu Xu
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai, China
| | - Shuai Xue
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai, China
| | - Yaqiong Zhang
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai, China
| | - Jian Li
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai, China
| | - Peiyu Qian
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai, China
| | - Yanyan Zhang
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai, China
| | - Li Feng
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
72
|
Han KM, Kwon MJ, Kim JH, Kim JH, Bang WJ, Choi HG, Yoo DM, Lee NE, Kim NY, Kang HS. Association between Gastric Cancer and Osteoporosis: A Longitudinal Follow-Up Study Using a National Health Sample Cohort. Cancers (Basel) 2024; 16:2291. [PMID: 39001355 PMCID: PMC11240602 DOI: 10.3390/cancers16132291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 07/16/2024] Open
Abstract
Gastric cancer (GC) survivors may be more likely to develop osteoporosis. However, few studies on the relationship between GC and osteoporosis have been conducted on large patient populations. We aimed to determine the incidence of osteoporosis and identify related factors by comparing patients with GC and matched controls using the Korean National Health Insurance Service-National Sample Cohort (KNHIS-NSC). This study included 9078 patients with GC and 36,312 controls (1:4 propensity score-matched for sex, age, residence, and income). The hazard ratio (HR) for osteoporosis was significantly greater for GC patients than for controls according to Charlson Comorbidity Index (CCI) score-adjusted models (adjusted HR = 1.13). Kaplan-Meier analysis revealed that the cumulative incidence of osteoporosis during the follow-up period commencing from the index date was significantly greater in GC patients than in the controls (p = 0.0087). A positive correlation of osteoporosis with GC was detected for those aged < 65 years, males, and those with CCI scores = 0. In conclusion, the study findings suggest that men with GC aged < 65 years may be at an increased risk for osteoporosis. Research into additional risk factors and the optimal timing of interventions are needed to prevent fractures and minimize bone loss in GC survivors.
Collapse
Affiliation(s)
- Kyeong Min Han
- Hallym Data Science Laboratory, Hallym University College of Medicine, Anyang 14068, Republic of Korea; (K.M.H.); (D.M.Y.); (N.-E.L.)
| | - Mi Jung Kwon
- Department of Pathology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Republic of Korea;
| | - Joo-Hee Kim
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Republic of Korea;
| | - Ji Hee Kim
- Department of Neurosurgery, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Republic of Korea;
| | - Woo Jin Bang
- Department of Urology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Republic of Korea;
| | - Hyo Geun Choi
- Suseo Seoul E.N.T. Clinic, 10, Bamgogae-ro 1-gil, Gangnam-gu, Seoul 06349, Republic of Korea;
| | - Dae Myoung Yoo
- Hallym Data Science Laboratory, Hallym University College of Medicine, Anyang 14068, Republic of Korea; (K.M.H.); (D.M.Y.); (N.-E.L.)
| | - Na-Eun Lee
- Hallym Data Science Laboratory, Hallym University College of Medicine, Anyang 14068, Republic of Korea; (K.M.H.); (D.M.Y.); (N.-E.L.)
| | - Nan Young Kim
- Hallym Institute of Translational Genomics and Bioinformatics, Hallym University Medical Center, Anyang 14068, Republic of Korea;
| | - Ho Suk Kang
- Division of Gastroenterology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Republic of Korea
| |
Collapse
|
73
|
Romańczyk M, Osmola M, Link A, Druet A, Hémont C, Martin J, Chapelle N, Matysiak-Budnik T. Non-Invasive Markers for the Detection of Gastric Precancerous Conditions. Cancers (Basel) 2024; 16:2254. [PMID: 38927959 PMCID: PMC11202181 DOI: 10.3390/cancers16122254] [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: 05/12/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Gastric cancer (GC) is still one of the most prevalent cancers worldwide, with a high mortality rate, despite improvements in diagnostic and therapeutic strategies. To diminish the GC burden, a modification of the current diagnostic paradigm, and especially endoscopic diagnosis of symptomatic individuals, is necessary. In this review article, we present a broad review and the current knowledge status on serum biomarkers, including pepsinogens, gastrin, Gastropanel®, autoantibodies, and novel biomarkers, allowing us to estimate the risk of gastric precancerous conditions (GPC)-atrophic gastritis and gastric intestinal metaplasia. The aim of the article is to emphasize the role of non-invasive testing in GC prevention. This comprehensive review describes the pathophysiological background of investigated biomarkers, their status and performance based on available data, as well as their clinical applicability. We point out future perspectives of non-invasive testing and possible new biomarkers opportunities.
Collapse
Affiliation(s)
- Marcin Romańczyk
- Department of Gastroenterology, Academy of Silesia, 40-555 Katowice, Poland
- H-T. Medical Center, 43-100 Tychy, Poland
| | | | - Alexander Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Amaury Druet
- IMAD, Hepato-Gastroenterology & Digestive Oncology, University Hospital of Nantes, F-44093 Nantes, France
| | - Caroline Hémont
- CHU de Nantes, Laboratoire d’Immunologie, Center for ImmunoMonitoring Nantes-Atlantique (CIMNA), F-44000 Nantes, France
| | - Jerome Martin
- CHU de Nantes, Laboratoire d’Immunologie, Center for ImmunoMonitoring Nantes-Atlantique (CIMNA), F-44000 Nantes, France
- University of Nantes, INSERM, Centre de Recherche Translationnel en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
| | - Nicolas Chapelle
- IMAD, Hepato-Gastroenterology & Digestive Oncology, University Hospital of Nantes, F-44093 Nantes, France
- University of Nantes, INSERM, Centre de Recherche Translationnel en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
| | - Tamara Matysiak-Budnik
- IMAD, Hepato-Gastroenterology & Digestive Oncology, University Hospital of Nantes, F-44093 Nantes, France
- University of Nantes, INSERM, Centre de Recherche Translationnel en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
| |
Collapse
|
74
|
Kim WS, Kim SH, Joo MK, Park JJ, Lee BJ, Chun HJ. Early gastric cancer of young patients treated by endoscopic submucosal dissection: focusing on the different characteristics and prognosis of elderly patients. Surg Endosc 2024:10.1007/s00464-024-10981-x. [PMID: 38886228 DOI: 10.1007/s00464-024-10981-x] [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: 02/13/2024] [Accepted: 06/02/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND A small portion of patients are diagnosed with early gastric cancer (EGC) and undergo endoscopic submucosal dissection (ESD) at a young age. However, their clinical outcomes are rarely known. AIM We investigated to identify the feasibility and clinical outcomes of ESD for EGC focusing on young patients. METHODS We analyzed the clinical characteristics and outscomes of patients who had undergone ESD for the treatment of EGC at < 50 years of age. We enrolled patients who had been diagnosed with EGC and had undergone ESD between 2006 and 2020. We divided them by age as follows: ≤ 50 and > 50 years into the young age (YA) and other age (OA) groups, respectively. RESULTS Altogether, 1681 patients underwent ESD for EGC (YA group: 124 [7.4%], OA group: 1557 [92.6%]). The YA group had less severe atrophy and more undifferentiated (37.1% vs. 13.9%, P < 0.001) and diffuse type (25% vs. 7.7%, P < 0.001) histology. The curative resection rate was not significantly different between the groups. However, among 1075 patients who had achieved curative resection and had been followed-up for > 12 months, the YA group had a lower incidence of MGN (5.2% vs. 17.5%, P = 0.004) and MGC (2.6% vs. 10.9%, P = 0.019) than those exhibited by the OA group. The YA group was a significant negative predictor of MGN (odds ratio [OR]: 2.983, 95% confidence interval [CI] 1.060-8.393, P = 0.038), and marginally negative predictor in MGC (OR: 3.909, 95% CI: 0.939-16.281, P = 0.061). CONCLUSION ESD is a favorable and effective therapeutic modality for EGC patients aged < 50 years, once curative resection is achieved.
Collapse
Affiliation(s)
- Won Shik Kim
- Division of Gastroenterology, Department of Internal Medicine/Korea, University College of Medicine/Korea, University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, Republic of Korea
| | - Seung Han Kim
- Division of Gastroenterology, Department of Internal Medicine/Korea, University College of Medicine/Korea, University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, Republic of Korea
| | - Moon Kyung Joo
- Division of Gastroenterology, Department of Internal Medicine/Korea, University College of Medicine/Korea, University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, Republic of Korea.
| | - Jong-Jae Park
- Division of Gastroenterology, Department of Internal Medicine/Korea, University College of Medicine/Korea, University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, Republic of Korea
| | - Beom Jae Lee
- Division of Gastroenterology, Department of Internal Medicine/Korea, University College of Medicine/Korea, University Guro Hospital, 148, Gurodong-Ro, Guro-Gu, Seoul, 08308, Republic of Korea
| | - Hoon Jai Chun
- Division of Gastroenterology, Department of Internal Medicine/Korea, University College of Medicine/Korea, University Anam Hospital, 73, Inchon-Ro, Seongbuk-Gu, Seoul, Republic of Korea
| |
Collapse
|
75
|
Zeng Z, Zhu Q. Progress and prospects of biomarker-based targeted therapy and immune checkpoint inhibitors in advanced gastric cancer. Front Oncol 2024; 14:1382183. [PMID: 38947886 PMCID: PMC11211377 DOI: 10.3389/fonc.2024.1382183] [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: 02/05/2024] [Accepted: 05/24/2024] [Indexed: 07/02/2024] Open
Abstract
Gastric cancer and gastroesophageal junction cancer represent the leading cause of tumor-related death worldwide. Although advances in immunotherapy and molecular targeted therapy have expanded treatment options, they have not significantly altered the prognosis for patients with unresectable or metastatic gastric cancer. A minority of patients, particularly those with PD-L1-positive, HER-2-positive, or MSI-high tumors, may benefit more from immune checkpoint inhibitors and/or HER-2-directed therapies in advanced stages. However, for those lacking specific targets and unique molecular features, conventional chemotherapy remains the only recommended effective and durable regimen. In this review, we summarize the roles of various signaling pathways and further investigate the available targets. Then, the current results of phase II/III clinical trials in advanced gastric cancer, along with the superiorities and limitations of the existing biomarkers, are specifically discussed. Finally, we will offer our insights in precision treatment pattern when encountering the substantial challenges.
Collapse
Affiliation(s)
| | - Qing Zhu
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
76
|
Gao Y, Liu K, Fang S. Analysis and projections of disease burden for different risk factors and sexes of ischemic stroke in young adults in China. Sci Rep 2024; 14:13339. [PMID: 38858463 PMCID: PMC11164860 DOI: 10.1038/s41598-024-63920-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024] Open
Abstract
To estimate the rate of death, and disability-adjusted life years (DALYs) and project the disease burden of ischemic stroke due to relevant risk factors in young adults age 20-49 years by sex in China. Data from the Global Burden of Diseases, Injuries, and Risk Factors Study (GBD) 2019 were used. The age-standardized mortality (ASMR), age-standardized DALYs rate (ASDR), and estimated annual percentage changes (EAPC) were calculated to evaluate the temporal trends from 1990 to 2019. We also used the NORDPRED model to predict ASMR for ischemic stroke due to related risk factors in Chinese young adults over the next 10 years. From 1990 to 2019, the general age-standardized mortality [from 2.39 (1.97 to 2.99) in 1990 to 1.8 (1.41 to 2.18) in 2019, EAPC = - 1.23] and DALYs rates (from 171.7 (140.34 to 212.36) in 1990 to 144.4 (114.29 to 177.37) in 2019, EAPC = - 0.86) decreased for ischemic stroke in young adults in China. ASMR and ASDR decreased for all level 1 risk factors (including behavioral, environmental/occupational, and metabolic) from 1990 to 2019, with the slightest decrease for metabolic risks [ASMR from 1.86 (1.39 to 2.41) in 1990 to 1.53 (1.15 to 1.92) in 2019, ASDR from 133.68 (99.96 to 173.89) in 1990 to 123.54 (92.96 to 156.98) in 2019] and the largest decrease for environmental/occupational risks [ASMR from 1.57 (1.26 to 1.98) in 1990 to 1.03 (0.78 to 1.29) in 2019, ASDR from 110.91 (88.44 to 138.34) in 1990 to 80.03 (61.87 to 100.33) in 2019]. In general, high body-mass index, high red meat intake, and ambient particulate matter pollution contributed to the large increase in ASMR and ASDR between 1990 and 2019. Significant reductions in ASMR and ASDR were observed in low vegetables intake, household air pollution from solid fuels, lead exposure, and low fiber intake. In addition, there were sex differences in the ranking of ASMR attributable to risks in ischemic stroke. The disease burden of ischemic stroke attributable to relevant risk factors in young adults in China is greater and has a faster growth trend or a slower decline trend in males than in females (except for secondhand smoke). The apparent increasing trend of ASMR attributable to high fasting plasma glucose, high systolic blood pressure, high body-mass index, and high red meat intake was observed in males but not in females. The projected analysis showed an increasing trend in ASMR between 1990 and 2030 for all specific metabolic risks for males, but a decreasing trend for females. ASMR attributable to ambient particulate matter pollution showed an increasing trend from 1990 to 2030 for both males and females. The burden of ischemic stroke in young adults in China showed a downward trend from 1990 to 2019. Specific risk factors associated with the burden of ischemic stroke varied between the sexes. Corresponding measures need to be developed in China to reduce the disease burden of ischemic stroke among young adults.
Collapse
Affiliation(s)
- Ying Gao
- Department of Neurology, Neuroscience Centre, The First Hospital of Jilin University, No. 1 Xinmin Street, Chaoyang District, Changchun, 130021, Jilin Province, People's Republic of China
| | - Kangding Liu
- Department of Neurology, Neuroscience Centre, The First Hospital of Jilin University, No. 1 Xinmin Street, Chaoyang District, Changchun, 130021, Jilin Province, People's Republic of China
| | - Shaokuan Fang
- Department of Neurology, Neuroscience Centre, The First Hospital of Jilin University, No. 1 Xinmin Street, Chaoyang District, Changchun, 130021, Jilin Province, People's Republic of China.
| |
Collapse
|
77
|
Bittar V, Boneli MF, Reis PCA, Felix N, Braga MAP, Rocha KM, Fogaroli LO, Costa GB, Comini AC, Amaral G, Marini DC, Camandaroba MPG. Laparoscopic Versus Open Gastrectomy for Advanced Gastric Cancer: A Meta-Analysis of Randomized Controlled Trials. J Gastrointest Cancer 2024; 55:652-661. [PMID: 38564116 DOI: 10.1007/s12029-024-01048-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Laparoscopy-assisted gastrectomy (LAG) is a well-established surgical technique in treating patients with early gastric cancer. However, the efficacy and safety of LAG versus open gastrectomy (OG) in patients with advanced gastric cancer (AGC) remains unclear. METHODS We systematically searched PubMed, Embase, and Cochrane Library in June 2023 for RCTs comparing LAG versus OG in patients with AGC. We pooled risk ratios (RR) and mean differences (MD) with 95% confidence intervals (CI) for binary and continuous endpoints, respectively. We performed all statistical analyses using R software version 4.3.1 and a random-effects model. RESULTS Nine RCTs comprising 3827 patients were included. There were no differences in terms of intraoperative complications (RR 1.14; 95% CI 0.72 to 1.82), number of retrieved lymph nodes (MD -0.54 lymph nodes; 95% CI -1.18 to 0.09), or mortality (RR 0.91; 95% CI 0.30 to 2.83). LAG was associated with a longer operative time (MD 49.28 minutes; 95% CI 30.88 to 67.69), lower intraoperative blood loss (MD -51.24 milliliters; 95% CI -81.41 to -21.06), shorter length of stay (MD -0.83 days; 95% CI -1.60 to -0.06), and higher incidence of pancreatic fistula (RR 2.44; 95% CI 1.08 to 5.50). Postoperatively, LAG was also superior to OG in reducing bleeding rates (RR 0.44; 95% CI 0.22 to 0.86) and time to first flatus (MD -0.27 days; 95% CI -0.47 to -0.07), with comparable results in anastomotic leakage, wound healing issues, major complications, time to ambulation, or time to first liquid intake. In the long-term analyses at 3 and 5 years, there were no significant differences between LAG and OG in terms of overall survival (RR 0.99; 95% CI 0.96 to 1.03) or relapse-free survival (RR 0.99; 95% CI 0.94 to 1.04). CONCLUSION This meta-analysis of RCTs suggests that LAG may be an effective and safe alternative to OG for treating AGC; albeit, it may be associated with an increased risk for pancreatic fistula.
Collapse
Affiliation(s)
- Vinicius Bittar
- Centro Universitário das Faculdades Associadas de Ensino, São João da Boa Vista, São Paulo, Brazil.
| | - Mauricio Ferreira Boneli
- Centro Universitário das Faculdades Associadas de Ensino, São João da Boa Vista, São Paulo, Brazil
| | | | - Nicole Felix
- Universidade Federal de Campina Grande, Campina Grande, Brazil
| | | | - Kian M Rocha
- Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo O Fogaroli
- Centro Universitário das Faculdades Associadas de Ensino, São João da Boa Vista, São Paulo, Brazil
| | - Gamaliel B Costa
- Centro Universitário das Faculdades Associadas de Ensino, São João da Boa Vista, São Paulo, Brazil
| | | | - Gustavo Amaral
- Centro Universitário das Faculdades Associadas de Ensino, São João da Boa Vista, São Paulo, Brazil
| | - Danyelle Cristine Marini
- Centro Universitário das Faculdades Associadas de Ensino, São João da Boa Vista, São Paulo, Brazil
| | | |
Collapse
|
78
|
Yang M. Interaction between intestinal flora and gastric cancer in tumor microenvironment. Front Oncol 2024; 14:1402483. [PMID: 38835386 PMCID: PMC11148328 DOI: 10.3389/fonc.2024.1402483] [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: 03/17/2024] [Accepted: 05/01/2024] [Indexed: 06/06/2024] Open
Abstract
Gastric Cancer (GC) is a prevalent malignancy globally and is the third leading cause of cancer-related deaths. Recent researches focused on the correlation between intestinal flora and GC. Studies indicate that bacteria can influence the development of gastrointestinal tumors by releasing bacterial extracellular vesicles (BEVs). The Tumor microenvironment (TME) plays an important role in tumor survival, with the interaction between intestinal flora, BEVs, and TME directly impacting tumor progression. Moreover, recent studies have demonstrated that intestinal microflora and BEVs can modify TME to enhance the effectiveness of antitumor drugs. This review article provides an overview and comparison of the biological targets through which the intestinal microbiome regulates TME, laying the groundwork for potential applications in tumor diagnosis, treatment, and prognosis.
Collapse
Affiliation(s)
- Mingjin Yang
- Department of Gastrointestinal Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
79
|
Mao C, Zhang Z, Fang R, Yuan W, Wu Y, Cong H. A novel tRNA-derived fragment tRF-17-18VBY9M works as a potential diagnostic biomarker for gastric cancer. J Cancer Res Clin Oncol 2024; 150:263. [PMID: 38767702 PMCID: PMC11106195 DOI: 10.1007/s00432-024-05792-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/08/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Gastric cancer (GC) is one of the most prevalent malignant tumors worldwide. The low effectiveness of common biomarkers for the detection of early GC makes it essential to seek new biomarkers to improve diagnostic efficacy. tsRNAs (transfer RNA-derived small RNAs) are related to the growth of malignant tumors. In this article, we focused on whether tsRNAs may be employed as biomarkers for GC. METHODS tRF-17-18VBY9M was screened in the tsRFun database as a research object. The methodological efficacy of tRF-17-18VBY9M was evaluated using Sanger sequencing, agarose gel electrophoresis assays, and gradient dilution. The χ2 test was applied to assess the interaction between tRF-17-18VBY9M expression and clinicopathologic characteristics. The receiver operating characteristic (ROC) curve was utilized to investigate the clinical efficiency of tRF-17-18VBY9M in GC. RESULTS The Chi-square test demonstrated that high-expressed tRF-17-18VBY9M was closely associated with the T stage, tumor node metastasis stage (TNM), lymph node metastasis, and neurological/vascular invasion. ROC curve analysis revealed that the diagnostic value of tRF-17-18VBY9M in GC was superior to carcinoembryonic antigen (CEA), carbohydrate antigen 199 (CA199), and carbohydrate antigen 724 (CA724). CONCLUSION tRF-17-18VBY9M is up-regulated in both GC sera and tissues. Differential tRF-17-18VBY9M expression distinguishes GC patients from healthy donors and gastritis patients, which suggests tRF-17-18VBY9M could act as a diagnostic biomarker in GC.
Collapse
Affiliation(s)
- Chunyan Mao
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical School of Nantong University, Nantong University, Nantong, 226001, China
| | - Zhihan Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical School of Nantong University, Nantong University, Nantong, 226001, China
| | - Ronghua Fang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical School of Nantong University, Nantong University, Nantong, 226001, China
| | - Wentao Yuan
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical School of Nantong University, Nantong University, Nantong, 226001, China
| | - Yi Wu
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical School of Nantong University, Nantong University, Nantong, 226001, China
| | - Hui Cong
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China.
- Department of Blood Transfusion, Affiliated Hospital of Nantong University, Nantong, 226001, China.
| |
Collapse
|
80
|
Zhou CM, Zhao SH. Evaluation of the value of combined detection of tumor markers CA724, carcinoembryonic antigen, CA242, and CA19-9 in gastric cancer. World J Gastrointest Oncol 2024; 16:1737-1744. [PMID: 38764828 PMCID: PMC11099441 DOI: 10.4251/wjgo.v16.i5.1737] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/08/2024] [Accepted: 03/20/2024] [Indexed: 05/09/2024] Open
Abstract
BACKGROUND Gastric cancer is a global health concern that poses a significant threat to human well-being. AIM To detecting serum changes in carcinoembryonic antigen (CEA), carbohydrate antigens (CA) 724, CA242, and CA19-9 expression among patients with gastric cancer. METHODS Eighty patients diagnosed with gastric cancer between January 2020 and January 2023 were included in the observation group, while 80 patients with benign gastric diseases were included in the control group. Both groups were tested for tumor markers (CA724, CEA, CA242, and CA19-9]. Tumor marker indicators (CA724, CEA, CA242, and CA19-9) were compared between the two groups, assessing positive rates of tumor markers across various stages in the observation group. Additionally, single and combined detection of various tumor markers were examined. RESULTS The sensitivity, specificity, accuracy, positive predictive value, and negative predictive value observed for the combined detection of CA724, CEA, CA242, and CA19-9 were higher than those of CA724, CEA, CA242, and CA19-9 individually. Therefore, the combined detection of CA724, CEA, CA242, and CA19-9 has a high diagnostic accuracy and could reduce the occurrence of missed or misdiagnosed cases, facilitating the early diagnosis and treatment of patients. CONCLUSION CA724, CEA, CA242, and CA19-9 serum levels in gastric cancer patients significantly surpassed those in non-gastric cancer patients (P < 0.05). Their combined detection can improve the diagnostic accuracy for gastric cancer, warranting clinical promotion.
Collapse
Affiliation(s)
- Chong-Mei Zhou
- Department of Clinical Laboratory, Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, China
| | - Shao-Hua Zhao
- Department of Clinical Laboratory, Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, China
| |
Collapse
|
81
|
Sassano M, Seyyedsalehi MS, Collatuzzo G, Pelucchi C, Bonzi R, Ferraroni M, Palli D, Yu GP, Zhang ZF, López-Carrillo L, Lunet N, Morais S, Zaridze D, Maximovich D, Martín V, Castano-Vinyals G, Vioque J, González-Palacios S, Ward MH, Malekzadeh R, Pakseresht M, Hernández-Ramirez RU, López-Cervantes M, Negri E, Turati F, Rabkin CS, Tsugane S, Hidaka A, Lagiou A, Lagiou P, Camargo MC, Curado MP, Boccia S, La Vecchia C, Boffetta P. Dietary intake of vitamin C and gastric cancer: a pooled analysis within the Stomach cancer Pooling (StoP) Project. Gastric Cancer 2024; 27:461-472. [PMID: 38436761 PMCID: PMC11016516 DOI: 10.1007/s10120-024-01476-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 01/29/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Previous studies suggest that dietary vitamin C is inversely associated with gastric cancer (GC), but most of them did not consider intake of fruit and vegetables. Thus, we aimed to evaluate this association within the Stomach cancer Pooling (StoP) Project, a consortium of epidemiological studies on GC. METHODS Fourteen case-control studies were included in the analysis (5362 cases, 11,497 controls). We estimated odds ratios (ORs) and corresponding 95% confidence intervals (CIs) for the association between dietary intake of vitamin C and GC, adjusted for relevant confounders and for intake of fruit and vegetables. The dose-response relationship was evaluated using mixed-effects logistic models with second-order fractional polynomials. RESULTS Individuals in the highest quartile of dietary vitamin C intake had reduced odds of GC compared with those in the lowest quartile (OR: 0.64; 95% CI: 0.58, 0.72). Additional adjustment for fruit and vegetables intake led to an OR of 0.85 (95% CI: 0.73, 0.98). A significant inverse association was observed for noncardia GC, as well as for both intestinal and diffuse types of the disease. The results of the dose-response analysis showed decreasing ORs of GC up to 150-200 mg/day of vitamin C (OR: 0.54; 95% CI: 0.41, 0.71), whereas ORs for higher intakes were close to 1.0. CONCLUSIONS The findings of our pooled study suggest that vitamin C is inversely associated with GC, with a potentially beneficial effect also for intakes above the currently recommended daily intake (90 mg for men and 75 mg for women).
Collapse
Affiliation(s)
- Michele Sassano
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Monireh Sadat Seyyedsalehi
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Giulia Collatuzzo
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Claudio Pelucchi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Rossella Bonzi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Monica Ferraroni
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Domenico Palli
- Cancer Risk Factors and Life-Style Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network, ISPRO, Florence, Italy
| | - Guo-Pei Yu
- Medical Informatics Center, Peking University, Beijing, China
| | - Zuo-Feng Zhang
- Department of Epidemiology, UCLA Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | | | - Nuno Lunet
- EPIUnit, Instituto de Saúde Pública da Universidade Do Porto, Porto, Portugal
- Laboratório Para a Investigação Integrativa E Translacional Em Saúde Populacional (ITR), Porto, Portugal
- Departamento de Ciências da Saúde Pública E Forenses E Educação Médica, Faculdade de Medicina, Universidade Do Porto, Porto, Portugal
| | - Samantha Morais
- EPIUnit, Instituto de Saúde Pública da Universidade Do Porto, Porto, Portugal
- Laboratório Para a Investigação Integrativa E Translacional Em Saúde Populacional (ITR), Porto, Portugal
- Departamento de Ciências da Saúde Pública E Forenses E Educação Médica, Faculdade de Medicina, Universidade Do Porto, Porto, Portugal
| | - David Zaridze
- Department of Clinical Epidemiology, N.N. Blokhin National Medical Research Center for Oncology, Moscow, Russia
| | - Dmitry Maximovich
- Department of Clinical Epidemiology, N.N. Blokhin National Medical Research Center for Oncology, Moscow, Russia
| | - Vicente Martín
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Instituto de Biomedicina, Universidad de León, León, Spain
| | - Gemma Castano-Vinyals
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Barcelona Institute for Global Health-ISGlobal, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jesús Vioque
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante, Universidad Miguel Hernandez (ISABIAL-UMH), Alicante, Spain
| | - Sandra González-Palacios
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante, Universidad Miguel Hernandez (ISABIAL-UMH), Alicante, Spain
| | - Mary H Ward
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Reza Malekzadeh
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Pakseresht
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, Canada
- Nutritional Epidemiology Group, Centre for Epidemiology and Biostatistics, University of Leeds, Leeds, UK
| | | | | | - Eva Negri
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Federica Turati
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Charles S Rabkin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Shoichiro Tsugane
- Division of Cohort Research, National Cancer Center Institute for Cancer Control, Tokyo, Japan
- International University of Health and Welfare Graduate School of Public Health, Tokyo, Japan
| | - Akihisa Hidaka
- Division of Epidemiology, National Cancer Center Institute for Cancer Control, Tokyo, Japan
- Department of Diabetes and Endocrinology, JCHO Tokyo Yamate Medical Centre, Tokyo, Japan
| | - Areti Lagiou
- Department of Public and Community Health, School of Public Health, University of West Attica, Athens, Greece
| | - Pagona Lagiou
- Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Maria Paula Curado
- Centro Internacional de Pesquisa, A. C. Camargo Cancer Center, São Paulo, Brasil
| | - Stefania Boccia
- Section of Hygiene, University Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italia
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Paolo Boffetta
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy.
- Stony Brook Cancer Center, Stony Brooke University, Stony Brook, NY, USA.
- Department of Family, Population and Preventive Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
82
|
Wang W, Baral S, Liu B, Sun Q, Wang L, Ren J, Tang D, Wang D. FANCA facilitates G1/S cell cycle advancement, proliferation, migration and invasion in gastric cancer. Acta Biochim Biophys Sin (Shanghai) 2024; 56:973-985. [PMID: 38682160 PMCID: PMC11322876 DOI: 10.3724/abbs.2024045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 01/31/2024] [Indexed: 05/01/2024] Open
Abstract
The present study explores the function of FANCA gene, a pivotal member of the Fanconi anaemia (FA) pathway crucial for preserving genomic stability and preventing cancer, particularly in the context of gastric cancer (GC). Using immunohistochemistry, quantitative real-time PCR, and western blot analysis, we evaluate FANCA mRNA and protein expressions in GC cell lines. The relationship between FANCA expression and clinicopathological characteristics is also explored. Various assays, including CCK8, colony formation, wound healing, and Transwell assays, are used to assess functional changes in cells associated with FANCA. Flow cytometry is utilized to evaluate alterations in the cell cycle resulted from FANCA knockdown and overexpression. Our findings show elevated FANCA expression in GC cell lines, with levels correlated with pathologic stage and lymphatic metastasis. FANCA knockdown impedes cell proliferation, migration, and invasion and induces G1/S phase cell cycle arrest. Conversely, FANCA overexpression stimulates cell proliferation, migration, and invasion. In vivo xenograft experiments confirm the promotional role of FANCA in GC tumor progression. Moreover, FANCA overexpression is associated with the activation of cell cycle. Collectively, our results suggest that FANCA drives malignant cell behaviors in GC through the cell cycle pathway, highlighting its potential as a therapeutic target for the treatment of GC.
Collapse
Affiliation(s)
- Wei Wang
- The Yangzhou School of Clinical Medicine of Dalian Medical UniversityYangzhou225001China
- Northern Jiangsu People’s HospitalYangzhou225001China
- General Surgery Institute of YangzhouYangzhou UniversityYangzhou225001China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic DiseasesYangzhou225001China
| | - Shantanu Baral
- Northern Jiangsu People’s Hospital Affiliated to Yangzhou UniversityYangzhou225001China
- Northern Jiangsu People’s HospitalYangzhou225001China
- General Surgery Institute of YangzhouYangzhou UniversityYangzhou225001China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic DiseasesYangzhou225001China
| | - Bin Liu
- Northern Jiangsu People’s HospitalYangzhou225001China
- General Surgery Institute of YangzhouYangzhou UniversityYangzhou225001China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic DiseasesYangzhou225001China
| | - Qiannan Sun
- Northern Jiangsu People’s HospitalYangzhou225001China
- General Surgery Institute of YangzhouYangzhou UniversityYangzhou225001China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic DiseasesYangzhou225001China
| | - Liuhua Wang
- Northern Jiangsu People’s HospitalYangzhou225001China
- General Surgery Institute of YangzhouYangzhou UniversityYangzhou225001China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic DiseasesYangzhou225001China
| | - Jun Ren
- Northern Jiangsu People’s Hospital Affiliated to Yangzhou UniversityYangzhou225001China
- Northern Jiangsu People’s HospitalYangzhou225001China
- General Surgery Institute of YangzhouYangzhou UniversityYangzhou225001China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic DiseasesYangzhou225001China
| | - Dong Tang
- Northern Jiangsu People’s HospitalYangzhou225001China
- General Surgery Institute of YangzhouYangzhou UniversityYangzhou225001China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic DiseasesYangzhou225001China
| | - Daorong Wang
- The Yangzhou School of Clinical Medicine of Dalian Medical UniversityYangzhou225001China
- Northern Jiangsu People’s Hospital Affiliated to Yangzhou UniversityYangzhou225001China
- Northern Jiangsu People’s HospitalYangzhou225001China
- General Surgery Institute of YangzhouYangzhou UniversityYangzhou225001China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic DiseasesYangzhou225001China
| |
Collapse
|
83
|
Tropeano G, Chiarello MM, Fico V, Brisinda G. How to identify early complications in patients undergoing distal gastrectomy? World J Gastrointest Surg 2024; 16:974-981. [PMID: 38690038 PMCID: PMC11056664 DOI: 10.4240/wjgs.v16.i4.974] [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: 11/12/2023] [Revised: 02/05/2024] [Accepted: 03/25/2024] [Indexed: 04/22/2024] Open
Abstract
In this editorial we comment on the article by Zhang et al published in a recent issue of the World Journal of Gastrointestinal Surgery. Gastrectomy with appropriate lymph node dissection is still standard curative treatment in locally advanced gastric cancer. Several studies point out that gastric cancer surgery is a complex procedure that leads to a high risk of morbidity and mortality. Many factors can contribute to the onset of complications with consequent effects on prognosis and increased mortality. The complications can be divided in complications related to anastomosis, to motility and to surgical site infection. The study presented by Zhang B et al represent an interesting analysis on the possibility to prevent postoperative morbidity. The study was performed on 131 patients with distal gastric cancer who underwent gastrectomy with D2 lymph node dissection. Of these patients, 16% developed early postoperative complications. The univariate analysis showed that prealbumin level, hypertension, diabetes, history of abdominal surgery, R0 resection, and blood transfusion were factors influencing early postoperative complications after distal gastrectomy. Moreover, the inclusion of the above significant variables in the logistic regression analysis revealed that hypertension, diabetes, a history of abdominal surgery, and blood transfusion were independent predictors of postoperative complications. In conclusion, preoperative and intraoperative factors can be used to establish an early postoperative nomogram model. The results of the study presented by Zhang et al suggest that the prediction model can be used to guide the detection of postoperative complications and has clinical reference value.
Collapse
Affiliation(s)
- Giuseppe Tropeano
- Emergency Surgery and Trauma Center, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
| | | | - Valeria Fico
- Emergency Surgery and Trauma Center, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
| | - Giuseppe Brisinda
- Emergency Surgery and Trauma Center, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Department of Medical and Surgical Sciences, Università Cattolica del Sacro Cuore, Rome 00168, Italy
| |
Collapse
|
84
|
Tsukanov V, Tonkikh Y, Vasyutin A. Gastric cancer: incidence, risk factors, screening. RUSSIAN JOURNAL OF PREVENTIVE MEDICINE 2024; 27:135. [DOI: 10.17116/profmed202427121135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Modern literature data on the incidence, risk factors, and screening methods for gastric cancer (GC) were analyzed. Global GC incidence is 9.2 per 100.000 people. The GC incidence in Russia, at 13.7 per 100.000 people, exceeds the global value and amounts to 19.8 per 100.000 males and 9.8 per 100.000 females. GC ranks second in the cancer mortality structure in Russia after lung cancer. Main GC risk factors include Helicobacter pylori (H. pylori) infection, tobacco smoking, a diet high in salt and processed meat, being male, being over 50 years of age, having a family history of the condition, alcohol consumption, and obesity. The main population screening method for GC is an endoscopic examination of people aged 50 and over every 2—3 years. Besides endoscopy, opportunistic screening can include pepsinogen and H. pylori tests. Activating screening and preventive measures is reasonable for reducing GC incidence in Russia.
Collapse
Affiliation(s)
- V.V. Tsukanov
- Federal Research Center «Krasnoyarsk Science Center» of the Siberian Branch of the Russian Academy of Sciences
| | - Yu.L. Tonkikh
- Federal Research Center «Krasnoyarsk Science Center» of the Siberian Branch of the Russian Academy of Sciences
| | - A.V. Vasyutin
- Federal Research Center «Krasnoyarsk Science Center» of the Siberian Branch of the Russian Academy of Sciences
| |
Collapse
|
85
|
Ke Y, Tan C, Zhen J, Dong W. Global status and trends of gastric cancer and gastric microbiota research: a bibliometric analysis. Front Microbiol 2024; 15:1341012. [PMID: 38655079 PMCID: PMC11037409 DOI: 10.3389/fmicb.2024.1341012] [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: 11/19/2023] [Accepted: 03/12/2024] [Indexed: 04/26/2024] Open
Abstract
Background Numerous studies have cast light on the relationship between the gastric microbiota and gastric carcinogenesis. In this study, we conducted a bibliometric analysis of the relevant literature in the field of gastric cancer and the gastric microbiota and clarified its research status, hotspots, and development trends. Materials and methods Publications were retrieved from the Web of Science Core Collection on 18 July 2023. CiteSpace 6.2.R4, VOSviewer 1.6.19.0, and Biblioshiny were used for the co-occurrence and cooperation analyses of countries, institutions, authors, references, and keywords. A keyword cluster analysis and an emergence analysis were performed, and relevant knowledge maps were drawn. Results The number of published papers in this field totaled 215 and showed an increasing trend. The analysis of funding suggested that the input in this field is increasing steadily. China had the highest number of publications, while the United States had the highest betweenness centrality. Baylor College of Medicine published the most articles cumulatively. Both Ferreira RM and Cooker OO had the highest citation frequency. The journal Helicobacter showed the most interest in this field, while Gut provided a substantial research foundation. A total of 280 keywords were obtained using CiteSpace, which were primarily focused on the eradication and pathogenic mechanisms of Helicobacter pylori, as well as the application of the gastric microbiota in the evaluation and treatment of gastric cancer. The burst analysis suggested that in the future, research may focus on the application of gastric microorganisms, particularly Fusobacterium nucleatum, in the diagnosis and treatment of gastric cancer, along with their pathogenic mechanisms. Conclusion Current studies have been tracking the eradication of Helicobacter pylori and its pathogenic mechanisms, as well as changes in the gastric microbiota during gastric carcinogenesis. Future research may focus on the clinical application and pathogenesis of stomach microorganisms through bacteria such as Fusobacterium nucleatum.
Collapse
Affiliation(s)
- Yujia Ke
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Cheng Tan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Junhai Zhen
- Department of General Practice, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
86
|
Zhang K, Wang H, Cheng Y, Liu H, Gong Q, Zeng Q, Zhang T, Wei G, Wei Z, Chen D. Early gastric cancer detection and lesion segmentation based on deep learning and gastroscopic images. Sci Rep 2024; 14:7847. [PMID: 38570595 PMCID: PMC10991264 DOI: 10.1038/s41598-024-58361-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/28/2024] [Indexed: 04/05/2024] Open
Abstract
Gastric cancer is a highly prevalent disease that poses a serious threat to public health. In clinical practice, gastroscopy is frequently used by medical practitioners to screen for gastric cancer. However, the symptoms of gastric cancer at different stages of advancement vary significantly, particularly in the case of early gastric cancer (EGC). The manifestations of EGC are often indistinct, leading to a detection rate of less than 10%. In recent years, researchers have focused on leveraging deep learning algorithms to assist medical professionals in detecting EGC and thereby improve detection rates. To enhance the ability of deep learning to detect EGC and segment lesions in gastroscopic images, an Improved Mask R-CNN (IMR-CNN) model was proposed. This model incorporates a "Bi-directional feature extraction and fusion module" and a "Purification module for feature channel and space" based on the Mask R-CNN (MR-CNN). Our study includes a dataset of 1120 images of EGC for training and validation of the models. The experimental results indicate that the IMR-CNN model outperforms the original MR-CNN model, with Precision, Recall, Accuracy, Specificity and F1-Score values of 92.9%, 95.3%, 93.9%, 92.5% and 94.1%, respectively. Therefore, our proposed IMR-CNN model has superior detection and lesion segmentation capabilities and can effectively aid doctors in diagnosing EGC from gastroscopic images.
Collapse
Affiliation(s)
- Kezhi Zhang
- Guangxi Key Laboratory of Information Functional Materials and Intelligent Information Processing, School of Physics and Electronics, Nanning Normal University, 175 Mingxiu East Road, Nanning, 530001, Guangxi, China
| | - Haibao Wang
- Guangxi Key Laboratory of Information Functional Materials and Intelligent Information Processing, School of Physics and Electronics, Nanning Normal University, 175 Mingxiu East Road, Nanning, 530001, Guangxi, China
| | - Yaru Cheng
- Department of Gastroenterology, Shandong Second Provincial General Hospital, 4 Duan Xing West Road, Jinan, 250022, Shandong, China
| | - Hongyan Liu
- Department of Gastroenterology, Shandong Second Provincial General Hospital, 4 Duan Xing West Road, Jinan, 250022, Shandong, China
| | - Qi Gong
- Department of Gastroenterology, Shandong Second Provincial General Hospital, 4 Duan Xing West Road, Jinan, 250022, Shandong, China
| | - Qian Zeng
- Guangxi Key Laboratory of Information Functional Materials and Intelligent Information Processing, School of Physics and Electronics, Nanning Normal University, 175 Mingxiu East Road, Nanning, 530001, Guangxi, China
| | - Tao Zhang
- Guangxi Key Laboratory of Information Functional Materials and Intelligent Information Processing, School of Physics and Electronics, Nanning Normal University, 175 Mingxiu East Road, Nanning, 530001, Guangxi, China
| | - Guoqiang Wei
- Guangxi Key Laboratory of Information Functional Materials and Intelligent Information Processing, School of Physics and Electronics, Nanning Normal University, 175 Mingxiu East Road, Nanning, 530001, Guangxi, China.
- School of Electronic Engineering, Hunan College of Information, Changsha, 410200, Hunan, China.
| | - Zhi Wei
- Department of Gastroenterology, Shandong Second Provincial General Hospital, 4 Duan Xing West Road, Jinan, 250022, Shandong, China.
| | - Dong Chen
- Guangxi Key Laboratory of Information Functional Materials and Intelligent Information Processing, School of Physics and Electronics, Nanning Normal University, 175 Mingxiu East Road, Nanning, 530001, Guangxi, China.
| |
Collapse
|
87
|
Yoo HJ, Kim JJ. Comparing Pre- and Post-Operative Findings in Patients Who Underwent Laparoscopic Proximal Gastrectomy With a Double-Flap Technique: A Study on High-Resolution Manometry, Impedance pH Monitoring, and Esophagogastroduodenoscopy Findings. J Gastric Cancer 2024; 24:137-144. [PMID: 38575507 PMCID: PMC10995827 DOI: 10.5230/jgc.2024.24.e1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/11/2023] [Accepted: 10/19/2023] [Indexed: 04/06/2024] Open
Abstract
PURPOSE Laparoscopic proximal gastrectomy (LPG) is a viable choice for treating proximal gastric lesions. However, the occurrence of severe reflux has limited its widespread adoption. To address this issue, the double flap technique (DFT), which incorporates artificial lower esophageal sphincteroplasty, has been developed to prevent reflux problems after proximal gastrectomy. In this study, we aimed to investigate the usefulness of this technique using high-resolution manometry (HRM), impedance pH monitoring, and esophagogastroduodenoscopy (EGD). MATERIALS AND METHODS The findings of pre- and postoperative 6-month HRM, pH monitoring, and EGD were compared for 9 patients who underwent LPG with DFT for various proximal gastric lesions at Incheon St. Mary's Hospital from January 2021 to December. RESULTS A total of 9 patients underwent proximal gastrectomy. Approximately half of the patients had Hill's grade under II preoperatively, whereas all patients had Hill's grades I and II in EGD findings. In the HRM test, there was no significant difference between distal contractile integral (1,412.46±1,168.51 vs. 852.66±495.62 mmHg·cm·s, P=0.087) and integrated relaxation pressure (12.54±8.97 vs. 8.33±11.30 mmHg, P=0.27). The average lower esophageal sphincter (LES) pressure was 29.19±14.51 mmHg preoperatively, which did not differ from 19.97±18.03 mmHg after the surgery (P=0.17). DeMeester score (7.02±6.36 vs. 21.92±36.17, P=0.21) and total acid exposure time (1.49±1.48 vs. 5.61±10.17, P=0.24) were slightly higher, but the differences were not statistically significant. CONCLUSIONS There is no significant functional difference in HRM and impedance pH monitoring tests after DFT. DFT appears to be useful in preserving LES function following proximal gastrectomy.
Collapse
Affiliation(s)
- Hyun Joo Yoo
- Division of Gastrointestinal Surgery, Department of Surgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Korea
| | - Jin-Jo Kim
- Division of Gastrointestinal Surgery, Department of Surgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Korea.
| |
Collapse
|
88
|
Zhu N, Zhang Y, Mi M, Ding Y, Weng S, Zheng J, Tian Y, Yuan Y. The death burden of colorectal cancer attributable to modifiable risk factors, trend analysis from 1990 to 2019 and future predictions. Cancer Med 2024; 13:e7136. [PMID: 38545767 PMCID: PMC10973881 DOI: 10.1002/cam4.7136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 11/14/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2025] Open
Abstract
BACKGROUND The death burden attributable to modifiable risk factors is key to colorectal cancer (CRC) prevention. This study aimed to assess the prevalence and regional distribution of attributable CRC death burden worldwide from 1990 to 2019. METHODS We extracted data from the Global Burden of Disease Study in 2019 and assessed the mortality, age-standardized death rate (ASDR), population attributable fractions, and time trend in CRC attributable to risk factors by geography, socio-demographic index (SDI) quintile, age, and sex. RESULTS Over the past 30 years, from high to low SDI region, the number of deaths increased by 46.56%, 103.55%, 249.64%, 231.89%, 163.11%, and the average annual percentage change (AAPC) for ASDR were -1.06%, -0.01%, 1.32%, 1.19%, and 0.65%, respectively. ASDR in males was 1.88 times than in females in 2019; ASDR in males showed an increasing trend (AAPC 0.07%), whereas ASDR in females showed a decreasing trend (AAPC -0.69%) compared to figures in 1990. In 2019, from high to low SDI region, the 15-49 age group accounted for 3%, 6%, 10%, 11%, and 15% of the total population; dietary and metabolic factors contributed 43.4% and 20.8% to CRC-attributable death worldwide. From high to low SDI region, ASDRs caused by dietary and metabolic factors increased by -23.4%, -5.5%, 25.8%, 29.1%, 13.5%, and 1.4%, 33.3%, 100.8%, 128.4%, 77.7% respectively, compared to 1990. CONCLUSIONS The attributable CRC death burden gradually shifted from higher SDI to lower SDI regions. The limitation in males was more significant, and the gap is expected to be further expanded. In lower SDI regions, the death burden tended to affect younger people. The leading cause of CRC-attributable deaths was the inadequate control of dietary and metabolic risk factors.
Collapse
Affiliation(s)
- Ning Zhu
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Department of Medical Oncology, Cancer InstituteThe Second Affiliated Hospital of Zhejiang University, School of MedicineHangzhouZhejiangChina
| | - Yan Zhang
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Department of Medical Oncology, Cancer InstituteThe Second Affiliated Hospital of Zhejiang University, School of MedicineHangzhouZhejiangChina
| | - Mi Mi
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Department of Medical Oncology, Cancer InstituteThe Second Affiliated Hospital of Zhejiang University, School of MedicineHangzhouZhejiangChina
| | - Yuwei Ding
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Department of Medical Oncology, Cancer InstituteThe Second Affiliated Hospital of Zhejiang University, School of MedicineHangzhouZhejiangChina
| | - Shanshan Weng
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Department of Medical Oncology, Cancer InstituteThe Second Affiliated Hospital of Zhejiang University, School of MedicineHangzhouZhejiangChina
| | - Jia Zheng
- Department of Medical GeriatricsThe Second Affiliated Hospital of Zhejiang University, School of MedicineHangzhouZhejiangChina
| | - Yang Tian
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated Hospital of Zhejiang University, School of MedicineHangzhouZhejiangChina
| | - Ying Yuan
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Department of Medical Oncology, Cancer InstituteThe Second Affiliated Hospital of Zhejiang University, School of MedicineHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for CancerHangzhouZhejiangChina
- Cancer Center, Zhejiang UniversityHangzhouZhejiangChina
| |
Collapse
|
89
|
Wang P, Han Y, Pan W, Du J, Zuo D, Ba Y, Zhang H. Tyrosine phosphatase SHP2 aggravates tumor progression and glycolysis by dephosphorylating PKM2 in gastric cancer. MedComm (Beijing) 2024; 5:e527. [PMID: 38576457 PMCID: PMC10993348 DOI: 10.1002/mco2.527] [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: 02/07/2023] [Revised: 11/26/2023] [Accepted: 12/22/2023] [Indexed: 04/06/2024] Open
Abstract
Gastric cancer (GC) is among the most lethal human malignancies, yet it remains hampered by challenges in fronter of molecular-guided targeted therapy to direct clinical treatment strategies. The protein tyrosine phosphatase Src homology 2 domain-containing phosphatase 2 (SHP2) is involved in the malignant progression of GC. However, the detailed mechanisms of the posttranslational modifications of SHP2 remain poorly understood. Herein, we demonstrated that an allosteric SHP2 inhibitor, SHP099, was able to block tumor proliferation and migration of GC by dephosphorylating the pyruvate kinase M2 type (PKM2) protein. Mechanistically, we found that PKM2 is a bona fide target of SHP2. The dephosphorylation and activation of PKM2 by SHP2 are necessary to exacerbate tumor progression and GC glycolysis. Moreover, we demonstrated a strong correlation between the phosphorylation level of PKM2 and adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) in GC cells. Notably, the low phosphorylation expression of AMPK was negatively correlated with activated SHP2. Besides, we proved that cisplatin could activate SHP2 and SHP099 increased sensitivity to cisplatin in GC. Taken together, our results provide evidence that the SHP2/PKM2/AMPK axis exerts a key role in GC progression and glycolysis and could be a viable therapeutic approach for the therapy of GC.
Collapse
Affiliation(s)
- Peiyun Wang
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin Medical UniversityTianjinChina
| | - Yueting Han
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin Medical UniversityTianjinChina
| | - Wen Pan
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin Medical UniversityTianjinChina
| | - Jian Du
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin Medical UniversityTianjinChina
| | - Duo Zuo
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin Medical UniversityTianjinChina
| | - Yi Ba
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin Medical UniversityTianjinChina
| | - Haiyang Zhang
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin Medical UniversityTianjinChina
- The Institute of Translational MedicineTianjin Union Medical Center of Nankai UniversityTianjinChina
| |
Collapse
|
90
|
Teng A, Stanley J, Jackson C, Koea J, Lao C, Lawrenson R, Meredith I, Sika-Paotonu D, Gurney J. The growing cancer burden: Age-period-cohort projections in Aotearoa New Zealand 2020-2044. Cancer Epidemiol 2024; 89:102535. [PMID: 38280359 DOI: 10.1016/j.canep.2024.102535] [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: 09/19/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 01/29/2024]
Abstract
BACKGROUND Cancer is a major cause of premature death and inequity, and global case numbers are rapidly expanding. This study projects future cancer numbers and incidence rates in Aotearoa New Zealand. METHODS Age-period-cohort modelling was applied to 25-years of national data to project cancer cases and incidence trends from 2020 to 2044. Nationally mandated cancer registry data and official historical and projected population estimates were used, with sub-groups by age, sex, and ethnicity. RESULTS Cancer diagnoses were projected to increase from 25,700 per year in 2015-2019 to 45,100 a year by 2040-44, a 76% increase (2.3% per annum). Across the same period, age-standardised cancer incidence increased by 9% (0.3% per annum) from 348 to 378 cancers per 100,000 person years, with greater increases for males (11%) than females (6%). Projected incidence trends varied substantially by cancer type, with several projected to change faster or in the opposite direction compared to projections from other countries. CONCLUSIONS Increasing cancer numbers reinforces the critical need for both cancer prevention and treatment service planning activities. Investment in developing new ways of working and increasing the workforce are required for the health system to be able to afford and manage the future burden of cancer.
Collapse
Affiliation(s)
- Andrea Teng
- Department of Public Health, University of Otago, PO Box 7343, Wellington, New Zealand.
| | - James Stanley
- Department of Public Health, University of Otago, PO Box 7343, Wellington, New Zealand
| | - Christopher Jackson
- Department of Medicine (Dunedin), University of Otago, PO Box 56, Dunedin, New Zealand
| | - Jonathan Koea
- General Surgery, Waitakere Hospital, Private Bag 92019, Auckland, New Zealand; Medical Surgery, The University of Auckland, Auckland, New Zealand
| | - Chunhuan Lao
- Medical Research Centre, The University of Waikato, Private Bag 3105, Hamilton, New Zealand
| | - Ross Lawrenson
- Medical Research Centre, The University of Waikato, Private Bag 3105, Hamilton, New Zealand; Commissioning, Te Whatu Ora, Hamilton, Waikato, New Zealand
| | - Ineke Meredith
- General Surgery, Wakefield Hospital, 30 Florence Street, Wellington, New Zealand
| | - Dianne Sika-Paotonu
- Dean's Department UOW & Division of Health Sciences, University of Otago, PO Box 7343, Wellington, New Zealand
| | - Jason Gurney
- Department of Public Health, University of Otago, PO Box 7343, Wellington, New Zealand
| |
Collapse
|
91
|
Qiao X, Sun J, Ren P, Guo H, Xu H, Bao C, Jiang C. Integrated single-cell sequencing, spatial transcriptome sequencing and bulk RNA sequencing highlights the molecular characteristics of parthanatos in gastric cancer. Aging (Albany NY) 2024; 16:5471-5500. [PMID: 38499384 PMCID: PMC11006479 DOI: 10.18632/aging.205658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 02/08/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Parthanatos is a novel programmatic form of cell death based on DNA damage and PARP-1 dependency. Nevertheless, its specific role in the context of gastric cancer (GC) remains uncertain. METHODS In this study, we integrated multi-omics algorithms to investigate the molecular characteristics of parthanatos in GC. A series of bioinformatics algorithms were utilized to explore clinical heterogeneity of GC and further predict the clinical outcomes. RESULTS Firstly, we conducted a comprehensive analysis of the omics features of parthanatos in various human tumors, including genomic mutations, transcriptome expression, and prognostic relevance. We successfully identified 7 cell types within the GC microenvironment: myeloid cell, epithelial cell, T cell, stromal cell, proliferative cell, B cell, and NK cell. When compared to adjacent non-tumor tissues, single-cell sequencing results from GC tissues revealed elevated scores for the parthanatos pathway across multiple cell types. Spatial transcriptomics, for the first time, unveiled the spatial distribution characteristics of parthanatos signaling. GC patients with different parthanatos signals often exhibited distinct immune microenvironment and metabolic reprogramming features, leading to different clinical outcomes. The integration of parthanatos signaling and clinical indicators enabled the creation of novel survival curves that accurately assess patients' survival times and statuses. CONCLUSIONS In this study, the molecular characteristics of parthanatos' unicellular and spatial transcriptomics in GC were revealed for the first time. Our model based on parthanatos signals can be used to distinguish individual heterogeneity and predict clinical outcomes in patients with GC.
Collapse
Affiliation(s)
- Xiuli Qiao
- Department of Gastroenterology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiaao Sun
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Pingping Ren
- Department of Gastroenterology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hui Guo
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hua Xu
- Department of Gastroenterology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Chongchan Bao
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Key Laboratory of Molecular Pathology in Tumors of Guangxi, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Chunmeng Jiang
- Department of Gastroenterology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
92
|
Shao Y, Yu X, Shan K, Yan J, Ye G. Defining the biological functions and clinical significance of AKR1C3 in gastric carcinogenesis through multiomics functional analysis and immune infiltration analysis. J Cancer 2024; 15:2646-2658. [PMID: 38577596 PMCID: PMC10988316 DOI: 10.7150/jca.94228] [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: 01/12/2024] [Accepted: 03/09/2024] [Indexed: 04/06/2024] Open
Abstract
Background: Human aldo-keto reductase family 1 member C3 (AKR1C3) is an important molecule that participates in multiple physiological metabolic processes. However, its expression, biological functions and clinical significance in gastric carcinogenesis are unclear. Methods: We collected data from several public data portals and clinical samples and systematically analyzed the clinical significance of tissue and plasma AKR1C3 expression. Then, we filtered prognostic risk factors and established novel prognosis-related nomogram models for predicting overall survival time and postoperative recurrence risk. The application value of the nomogram models was further assessed using clinical samples. Moreover, we explored the potential biological functions of AKR1C3 in gastric carcinogenesis and metastasis through multiomics functional analysis and immune infiltration analysis. Results: AKR1C3 levels were reduced in cancer tissue but increased significantly in the plasma of GC patients; AKR1C3 expression in either sample type was closely associated with multiple clinicopathological characteristics. By combining clinicopathological factors and AKR1C3 levels, two novel nomogram models were developed to predict overall survival time and postoperative recurrence risk. Multiomics functional analysis revealed that when its expression is dysregulated, AKR1C3 can widely participate in gene expression regulation through multiple regulatory modes at the gene, RNA and protein levels and exert various crucial biological effects in carcinogenesis and metastasis. Moreover, AKR1C3 expression was correlated with the infiltration of several immune cell types, and AKR1C3 was predicted to interact with several clinical drugs. Conclusion: Dysregulated AKR1C3 expression is related to gastric carcinogenesis and immunotherapy response and is a promising biomarker and effective biotherapy target in GC.
Collapse
Affiliation(s)
- Yongfu Shao
- Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315020, China
| | - Xuan Yu
- Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Keshu Shan
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315020, China
| | - Jianing Yan
- Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315020, China
| | - Guoliang Ye
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315020, China
| |
Collapse
|
93
|
Christodoulidis G, Koumarelas KE, Kouliou MN, Thodou E, Samara M. Gastric Cancer in the Era of Epigenetics. Int J Mol Sci 2024; 25:3381. [PMID: 38542354 PMCID: PMC10970362 DOI: 10.3390/ijms25063381] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 11/11/2024] Open
Abstract
Gastric cancer (GC) remains a significant contributor to cancer-related mortality. Novel high-throughput techniques have enlightened the epigenetic mechanisms governing gene-expression regulation. Epigenetic characteristics contribute to molecular taxonomy and give rise to cancer-specific epigenetic patterns. Helicobacter pylori (Hp) infection has an impact on aberrant DNA methylation either through its pathogenic CagA protein or by inducing chronic inflammation. The hypomethylation of specific repetitive elements generates an epigenetic field effect early in tumorigenesis. Epstein-Barr virus (EBV) infection triggers DNA methylation by dysregulating DNA methyltransferases (DNMT) enzyme activity, while persistent Hp-EBV co-infection leads to aggressive tumor behavior. Distinct histone modifications are also responsible for oncogene upregulation and tumor-suppressor gene silencing in gastric carcinomas. While histone methylation and acetylation processes have been extensively studied, other less prevalent alterations contribute to the development and migration of gastric cancer via a complex network of interactions. Enzymes, such as Nicotinamide N-methyltransferase (NNMT), which is involved in tumor's metabolic reprogramming, interact with methyltransferases and modify gene expression. Non-coding RNA molecules, including long non-coding RNAs, circular RNAs, and miRNAs serve as epigenetic regulators contributing to GC development, metastasis, poor outcomes and therapy resistance. Serum RNA molecules hold the potential to serve as non-invasive biomarkers for diagnostic, prognostic or therapeutic applications. Gastric fluids represent a valuable source to identify potential biomarkers with diagnostic use in terms of liquid biopsy. Ongoing clinical trials are currently evaluating the efficacy of next-generation epigenetic drugs, displaying promising outcomes. Various approaches including multiple miRNA inhibitors or targeted nanoparticles carrying epigenetic drugs are being designed to enhance existing treatment efficacy and overcome treatment resistance.
Collapse
Affiliation(s)
- Grigorios Christodoulidis
- Department of General Surgery, University Hospital of Larissa, University of Thessaly, Biopolis Campus, 41110 Larissa, Greece; (G.C.); (K.-E.K.); (M.-N.K.)
| | - Konstantinos-Eleftherios Koumarelas
- Department of General Surgery, University Hospital of Larissa, University of Thessaly, Biopolis Campus, 41110 Larissa, Greece; (G.C.); (K.-E.K.); (M.-N.K.)
| | - Marina-Nektaria Kouliou
- Department of General Surgery, University Hospital of Larissa, University of Thessaly, Biopolis Campus, 41110 Larissa, Greece; (G.C.); (K.-E.K.); (M.-N.K.)
| | - Eleni Thodou
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis Campus, 41110 Larissa, Greece;
| | - Maria Samara
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis Campus, 41110 Larissa, Greece;
| |
Collapse
|
94
|
Zullo A, Annibale B, Dinis-Ribeiro M, Fanchellucci G, Esposito G, Hassan C. Gastric juice analysis in clinical practice: why, how, and when. The experience with EndoFaster. Eur J Gastroenterol Hepatol 2024; 36:264-270. [PMID: 38179876 DOI: 10.1097/meg.0000000000002704] [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] [Indexed: 01/06/2024]
Abstract
Gastric juice analysis may be useful for clinical purposes, including the detection of H. pylori infection and diffuse atrophic gastritis on gastric mucosa. EndoFaster is a novel device which performs real-time analysis of gastric juice revealing the infection and hypochlorhydria by measuring ammonium concentrations and pH levels. This review aimed to evaluate the clinical applications of such a tool. By considering data from overall 11 studies, the values of sensitivity, specificity, positive predictive value, negative predictive value, accuracy, positive likelihood ratio, and negative likelihood ratio were 90%, 86%, 67%, 96%, 87%, 8.5, and 0.13, respectively, for H. pylori diagnosis, and 83%, 92%, 58%, 97%, 91%, 9.9 and 0.2, respectively, for suspecting diffuse atrophic gastritis. The very high value of negative predictive values for both H. pylori and mucosal atrophy would allow avoiding to perform useless negative gastric biopsies when the results of the test are negative. Some promising data suggest that gastric juice analysis may be useful also to diagnose H. pylori infection in patients with chronic active gastritis without evidence of bacteria at histology, as well as in predicting persistent acid reflux in patients on proton pump inhibitor therapy for reflux disease.
Collapse
Affiliation(s)
- Angelo Zullo
- Gastroenterology Unit, 'Nuovo Regina Margherita' Hospital
| | - Bruno Annibale
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, University Sapienza, Rome, Italy
| | - Mario Dinis-Ribeiro
- Gastroenterology Department, Portuguese Oncology Institute of Porto, and Porto Comprehensive Cancer Center (Porto.CCC) & RISE@CI-IPOP (Health Research Network), Porto, Portugal
| | - Gianluca Fanchellucci
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele
- Endoscopy Unit, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
| | - Gianluca Esposito
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, University Sapienza, Rome, Italy
| | - Cesare Hassan
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele
- Endoscopy Unit, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
| |
Collapse
|
95
|
Guo CH, Zhong JD, Jin XL, Zhang JE. Optimal time to initiate early oral feeding in postoperative patients with upper gastrointestinal malignancy: A network meta-analysis. Int J Nurs Stud 2024; 151:104680. [PMID: 38228066 DOI: 10.1016/j.ijnurstu.2023.104680] [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: 04/03/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND With the development of enhanced recovery after surgery, early oral feeding is likely to become the preferred mode of nutrition after surgery for upper gastrointestinal tract malignancies. However, the optimal time to initiate early oral feeding remains unknown. OBJECTIVE We aimed to compare the effects of different introduction times of early oral feeding in patients with upper gastrointestinal malignancies in terms of safety, tolerance, and effectiveness and to identify the optimal time for early oral feeding after surgery. METHODS A random-effects meta-analysis was performed to identify evidence from relevant randomized controlled trials. Ten electronic databases were searched for randomized controlled trials from their earliest records to May 2023. Data were analyzed using the Stata 16.0 software. RESULTS A total of 22 randomized controlled trials including 2510 patients and seven time points for oral feeding after surgery were considered. Regarding safety, oral feeding initiated on postoperative day 3 may be the safest (high-quality evidence) compared with other times. Regarding tolerance, oral feeding initiated on postoperative day 5 may be the most well-tolerated (moderate-quality evidence) compared with other times. Regarding effectiveness, oral feeding initiated on postoperative day 3 may be the most effective (moderate-quality evidence) compared with other times. CONCLUSIONS Early oral feeding is safe, tolerable, and effective in postoperative patients with upper gastrointestinal malignancies. The optimal time to initiate early oral feeding after surgery was most likely postoperative day 3. The results of this meta-analysis provide evidence-based guidelines for clinical decision-making.
Collapse
Affiliation(s)
- Cong-Hui Guo
- Postoperative Recovery Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China; School of Nursing, Sun Yat-sen University, Guangzhou, China
| | - Jiu-di Zhong
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiao-Lei Jin
- Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China; School of Nursing, Sun Yat-sen University, Guangzhou, China
| | - Jun-E Zhang
- School of Nursing, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
96
|
He F, Wang S, Zheng R, Gu J, Zeng H, Sun K, Chen R, Li L, Han B, Li X, Wei W, He J. Trends of gastric cancer burdens attributable to risk factors in China from 2000 to 2050. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2024; 44:101003. [PMID: 38269331 PMCID: PMC10806286 DOI: 10.1016/j.lanwpc.2023.101003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/07/2023] [Accepted: 12/24/2023] [Indexed: 01/26/2024]
Abstract
Background The incidence of gastric cancer (GC) decreased in past decades, which was thought largely attributable to risk factors control, yet China still accounts for 44% of global GC burdens. We aimed to estimate changing trajectories of proportions of GC burdens attributable to modifiable risk factors from 2000 to 2050 in China, to inform future targeted preventive strategies. Methods The incidence and new cases of GC were predicted to 2050 using Bayesian age-period-cohort model based on incidence data by anatomical subsites drawn from 682 cancer registries from National Central Cancer Registry. Population attributable fractions (PAFs) were calculated based on prevalence of risk factors and relative risks with GC. Temporal trends of PAFs were described by sex and categories of risk factors using joinpoint analysis. Findings We observed declining trends of PAFs of Helicobacter pylori (H. pylori) infection, smoking, pickled vegetable and alcohol consumption, but increasing trends of PAFs of unhealthy body mass index and diabetes for GC in China. The combined PAFs of these risk factors were estimated to decrease by 10.57% from 2000 to 2050 for GC. We estimated there will be 279,707 GC (122,796 cardia gastric cancer [CGC] and 156,911 non-cardia gastric cancer [NCGC]) cases in 2050. Out of these cases, 70.18% of GC cases could be attributable to modifiable risk factors, while H. pylori infection was predicted to be responsible for 40.7% of CGC and 62.1% of NCGC cases in 2050. Interpretation More than half of GC remained attributable to modifiable risk factors in China. Continued effective strategies on risk factors control are needed to reduce the burden of this highly life-threatening cancer in future. Funding Beijing Nova Program (No. Z201100006820069), CAMS Innovation Fund for Medical Sciences (CIFMS, grant No. 2021-I2M-1-023), CAMS Innovation Fund for Medical Sciences (CIFMS, grant No. 2021-I2M-1-010), Talent Incentive Program of Cancer Hospital Chinese Academy of Medical Sciences (Hope Star).
Collapse
Affiliation(s)
- Feifan He
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shaoming Wang
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rongshou Zheng
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianhua Gu
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Hongmei Zeng
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kexin Sun
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ru Chen
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Li
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bingfeng Han
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinqing Li
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenqiang Wei
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
97
|
Wang S, Zheng R, Li J, Zeng H, Li L, Chen R, Sun K, Han B, Bray F, Wei W, He J. Global, regional, and national lifetime risks of developing and dying from gastrointestinal cancers in 185 countries: a population-based systematic analysis of GLOBOCAN. Lancet Gastroenterol Hepatol 2024; 9:229-237. [PMID: 38185129 PMCID: PMC10849975 DOI: 10.1016/s2468-1253(23)00366-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Gastrointestinal cancers account for a quarter of the global cancer incidence and a third of cancer-related deaths. We sought to estimate the lifetime risks of developing and dying from gastrointestinal cancers at the country, world region, and global levels in 2020. METHODS For this population-based systematic analysis, we obtained estimates of gastrointestinal cancer incidence and mortality rates from GLOBOCAN for 185 countries, alongside all-cause mortality and population data from the UN. Countries were categorised into quartiles of the Human Development Index (HDI). The lifetime risk of gastrointestinal cancers was estimated with a standard method that adjusts for multiple primaries, taking into account competing risks of death from causes other than cancer and life expectancy. FINDINGS The global lifetime risks of developing and dying from gastrointestinal cancers from birth to death was 8·20% (95% CI 8·18-8·21) and 6·17% (6·16-6·18) in 2020. For men, the risk of developing gastrointestinal cancers was 9·53% (95% CI 9·51-9·55) and of dying from them 7·23% (7·22-7·25); for women, the risk of developing gastrointestinal cancers was 6·84% (6·82-6·85) and of dying from them 5·09% (5·08-5·10). Colorectal cancer presented the highest risk, accounting for 38·5% of the total lifetime risk of developing, and 28·2% of dying from, gastrointestinal cancers, followed by cancers of the stomach, liver, oesophagus, pancreas, and gallbladder. Eastern Asia has the highest lifetime risks for cancers of the stomach, liver, oesophagus, and gallbladder, Australia and New Zealand for colorectal cancer, and Western Europe for pancreatic cancer. The lifetime risk of gastrointestinal cancers increased consistently with increasing level of HDI; however, high HDI countries (the third HDI quartile) had the highest death risk. INTERPRETATION The global lifetime risk of gastrointestinal cancers translates to one in 12 people developing, and one in 16 people dying from, gastrointestinal cancers. The identified high risk and observed disparities across countries warrants context-specific targeted gastrointestinal cancer control and health systems planning. FUNDING Beijing Nova Program, CAMS Innovation Fund for Medical Sciences, and Talent Incentive Program of Cancer Hospital, CAMS (Hope Star).
Collapse
Affiliation(s)
- Shaoming Wang
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rongshou Zheng
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiayue Li
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongmei Zeng
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Li
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ru Chen
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kexin Sun
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bingfeng Han
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Freddie Bray
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Wenqiang Wei
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
98
|
Díaz del Arco C, Fernández Aceñero MJ, Ortega Medina L. Molecular Classifications in Gastric Cancer: A Call for Interdisciplinary Collaboration. Int J Mol Sci 2024; 25:2649. [PMID: 38473896 PMCID: PMC10931799 DOI: 10.3390/ijms25052649] [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: 01/11/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Gastric cancer (GC) is a heterogeneous disease, often diagnosed at advanced stages, with a 5-year survival rate of approximately 20%. Despite notable technological advancements in cancer research over the past decades, their impact on GC management and outcomes has been limited. Numerous molecular alterations have been identified in GC, leading to various molecular classifications, such as those developed by The Cancer Genome Atlas (TCGA) and the Asian Cancer Research Group (ACRG). Other authors have proposed alternative perspectives, including immune, proteomic, or epigenetic-based classifications. However, molecular stratification has not yet transitioned into clinical practice for GC, and little attention has been paid to alternative molecular classifications. In this review, we explore diverse molecular classifications in GC from a practical point of view, emphasizing their relationships with clinicopathological factors, prognosis, and therapeutic approaches. We have focused on classifications beyond those of TCGA and the ACRG, which have been less extensively reviewed previously. Additionally, we discuss the challenges that must be overcome to ensure their impact on patient treatment and prognosis. This review aims to serve as a practical framework to understand the molecular landscape of GC, facilitate the development of consensus molecular categories, and guide the design of innovative molecular studies in the field.
Collapse
Affiliation(s)
- Cristina Díaz del Arco
- Department of Legal Medicine, Psychiatry and Pathology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (M.J.F.A.); (L.O.M.)
- Department of Pathology, Hospital Clínico San Carlos, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - María Jesús Fernández Aceñero
- Department of Legal Medicine, Psychiatry and Pathology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (M.J.F.A.); (L.O.M.)
- Department of Pathology, Hospital Clínico San Carlos, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Luis Ortega Medina
- Department of Legal Medicine, Psychiatry and Pathology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (M.J.F.A.); (L.O.M.)
- Department of Pathology, Hospital Clínico San Carlos, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| |
Collapse
|
99
|
Rugge M, Genta RM, Malfertheiner P, Dinis-Ribeiro M, El-Serag H, Graham DY, Kuipers EJ, Leung WK, Park JY, Rokkas T, Schulz C, El-Omar EM. RE.GA.IN.: the Real-world Gastritis Initiative-updating the updates. Gut 2024; 73:407-441. [PMID: 38383142 DOI: 10.1136/gutjnl-2023-331164] [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: 09/18/2023] [Accepted: 12/18/2023] [Indexed: 02/23/2024]
Abstract
At the end of the last century, a far-sighted 'working party' held in Sydney, Australia addressed the clinicopathological issues related to gastric inflammatory diseases. A few years later, an international conference held in Houston, Texas, USA critically updated the seminal Sydney classification. In line with these initiatives, Kyoto Global Consensus Report, flanked by the Maastricht-Florence conferences, added new clinical evidence to the gastritis clinicopathological puzzle.The most relevant topics related to the gastric inflammatory diseases have been addressed by the Real-world Gastritis Initiative (RE.GA.IN.), from disease definitions to the clinical diagnosis and prognosis. This paper reports the conclusions of the RE.GA.IN. consensus process, which culminated in Venice in November 2022 after more than 8 months of intense global scientific deliberations. A forum of gastritis scholars from five continents participated in the multidisciplinary RE.GA.IN. consensus. After lively debates on the most controversial aspects of the gastritis spectrum, the RE.GA.IN. Faculty amalgamated complementary knowledge to distil patient-centred, evidence-based statements to assist health professionals in their real-world clinical practice. The sections of this report focus on: the epidemiology of gastritis; Helicobacter pylori as dominant aetiology of environmental gastritis and as the most important determinant of the gastric oncogenetic field; the evolving knowledge on gastric autoimmunity; the clinicopathological relevance of gastric microbiota; the new diagnostic horizons of endoscopy; and the clinical priority of histologically reporting gastritis in terms of staging. The ultimate goal of RE.GA.IN. was and remains the promotion of further improvement in the clinical management of patients with gastritis.
Collapse
Affiliation(s)
- Massimo Rugge
- Department of Medicine-DIMED, University of Padova, Padua, Italy
- Azienda Zero, Veneto Tumour Registry, Padua, Italy
| | - Robert M Genta
- Gastrointestinal Pathology, Inform Diagnostics Research Institute, Dallas, Texas, USA
- Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Peter Malfertheiner
- Medizinische Klinik und Poliklinik II, Ludwig Maximilian Universität Klinikum München, Munich, Germany
- Klinik für Gastroenterologie, Hepatologie und Infektiologie, Otto-von-Guericke Universität Magdeburg, Magdeburg, Germany
| | - Mario Dinis-Ribeiro
- Porto Comprehensive Cancer Center & RISE@CI-IPO, University of Porto, Porto, Portugal
- Gastroenterology Department, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Hashem El-Serag
- Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
- Houston VA Health Services Research & Development Center of Excellence, Michael E DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | - David Y Graham
- Department of Medicine, Michael E DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | - Ernst J Kuipers
- Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Jin Young Park
- International Agency for Research on Cancer, Lyon, France
| | - Theodore Rokkas
- Gastroenterology, Henry Dunant Hospital Center, Athens, Greece
| | | | - Emad M El-Omar
- Microbiome Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
100
|
Morgos DT, Stefani C, Miricescu D, Greabu M, Stanciu S, Nica S, Stanescu-Spinu II, Balan DG, Balcangiu-Stroescu AE, Coculescu EC, Georgescu DE, Nica RI. Targeting PI3K/AKT/mTOR and MAPK Signaling Pathways in Gastric Cancer. Int J Mol Sci 2024; 25:1848. [PMID: 38339127 PMCID: PMC10856016 DOI: 10.3390/ijms25031848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Gastric cancer (GC) is the fourth leading cause of death worldwide, with more than 1 million cases diagnosed every year. Helicobacter pylori represents the main risk factor, being responsible for 78% of the cases. Increased amounts of salt, pickled food, red meat, alcohol, smoked food, and refined sugars negatively affect the stomach wall, contributing to GC development. Several gene mutations, including PIK3CA, TP53, ARID1A, CDH1, Ras, Raf, and ERBB3 are encountered in GC pathogenesis, leading to phosphatidylinositol 3-kinase (PI3K) protein kinase B (AKT)/mammalian target of rapamycin (mTOR)-PI3K/AKT/mTOR-and mitogen-activated protein kinase (MAPK) signaling pathway activation and promoting tumoral activity. Helicobacter pylori, growth factors, cytokines, hormones, and oxidative stress also activate both pathways, enhancing GC development. In clinical trials, promising results have come from monoclonal antibodies such as trastuzumab and ramucirumab. Dual inhibitors targeting the PI3K/AKT/mTOR and MAPK signaling pathways were used in vitro studies, also with promising results. The main aim of this review is to present GC incidence and risk factors and the dysregulations of the two protein kinase complexes together with their specific inhibitors.
Collapse
Affiliation(s)
- Diana-Theodora Morgos
- Discipline of Anatomy, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Constantin Stefani
- Department I of Family Medicine and Clinical Base, “Dr. Carol Davila” Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Daniela Miricescu
- Discipline of Biochemistry, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Maria Greabu
- Discipline of Biochemistry, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Silviu Stanciu
- Department of Internal Medicine and Gastroenterology, Carol Davila University of Medicine and Pharmacy, Central Military Emergency University Hospital, 010825 Bucharest, Romania;
| | - Silvia Nica
- Emergency Discipline, University Hospital of Bucharest, 050098 Bucharest, Romania;
| | - Iulia-Ioana Stanescu-Spinu
- Discipline of Physiology, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (I.-I.S.-S.); (D.G.B.); (A.-E.B.-S.)
| | - Daniela Gabriela Balan
- Discipline of Physiology, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (I.-I.S.-S.); (D.G.B.); (A.-E.B.-S.)
| | - Andra-Elena Balcangiu-Stroescu
- Discipline of Physiology, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (I.-I.S.-S.); (D.G.B.); (A.-E.B.-S.)
| | - Elena-Claudia Coculescu
- Discipline of Oral Pathology, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Dragos-Eugen Georgescu
- Department of General Surgery, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 50474 Bucharest, Romania;
- Department of General Surgery, “Dr. Ion Cantacuzino” Clinical Hospital, 020475 Bucharest, Romania
| | - Remus Iulian Nica
- Central Military Emergency University Hospital “Dr. Carol Davila”, 010825 Bucharest, Romania;
- Discipline of General Surgery, Faculty of Midwifery and Nursing, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|