1
|
Hu Y, Wang B, Shi C, Ren P, Zhang C, Wang Z, Zhao J, Zheng J, Wang T, Wei B, Zhang H, Yu R, Shen Y, Ma J, Guo Y. A machine learning approach to risk-stratification of gastric cancer based on tumour-infiltrating immune cell profiles. Ann Med 2025; 57:2489007. [PMID: 40208029 PMCID: PMC11986862 DOI: 10.1080/07853890.2025.2489007] [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: 03/12/2024] [Revised: 03/14/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a highly heterogeneous disease, and the response of patients to clinical treatment varies substantially. There is no satisfactory strategy for predicting curative effects to date. We aimed to explore a new method for predicting the clinical efficacy of GC treatment based on immune variables detected via flow cytometry. METHODS We collected 394 tumour tissues from GC patients for flow cytometry analysis and gating analysis of tumour-infiltrating immune cells (TIICs). Unsupervised consensus clusters were generated from the cohort to classify patients into different phenogroups, and their clinical characteristics were examined. The derived model was evaluated via principal component analysis and t-distributed stochastic neighbourhood embedding analysis. Kaplan-Meier's curves were used to determine the prognosis during a 920-day-long median follow-up period (interquartile range: 834-1071 days). Adjusted multivariate Cox regression analysis was used to evaluate the association of clusters with disease-free survival (DFS) and recurrence. RESULTS All patients were classified based on their TIIC profiles into the C1 (characterized by low CD45 negative cell, high lymphocyte, high neutrophil and low CD3 + T cell levels), C2 (characterized by high CD8 + CD279+ cell and low CD4+ Th and CD8+ Tc cell numbers) and C3 (characterized by high CD4 + CD25+ and Treg cell levels) phenogroups. Patients from the three clusters had varied pathologies, MMR statuses and TIIC distribution patterns (p < .05). Kaplan-Meier's analysis showed that the prognosis of C3 was inferior compared to C1 and C2 (p = .0025). Adjusted Cox proportional hazard models helped us identify that C1 and C2 exhibited a favourable factor of recurrence after surgery, compared to C3. Kaplan-Meier's analysis showed that C1 and C2 were associated with a better DFS than C3 in some GC patient subgroups. CONCLUSIONS The machine learning model developed was found to be effective model at predicting the prognosis of patients with GC and their TIIC profiles for risk stratification in clinical settings.
Collapse
Affiliation(s)
- Yanping Hu
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - Bo Wang
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - Chao Shi
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - Pengfei Ren
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - Chengjuan Zhang
- Center of Repository, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Zhizhong Wang
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - Jiuzhou Zhao
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - Jiawen Zheng
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - Tingjie Wang
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - Bing Wei
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - He Zhang
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Rentao Yu
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yihang Shen
- Central Laboratory, Suzhou Ninth People’s Hospital, Suzhou, China
| | - Jie Ma
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - Yongjun Guo
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| |
Collapse
|
2
|
Zheng HL, Zhang LK, Zheng HH, Lv CB, Xu BB, Lin GT, Chen QY, Lin JX, Zheng CH, Huang CM, Xie JW. Timing of postoperative chemotherapy and prognosis in neoadjuvant-treated gastric cancer patients: a multicenter real-world cohort study. Ann Med 2025; 57:2500690. [PMID: 40329795 PMCID: PMC12064125 DOI: 10.1080/07853890.2025.2500690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND The optimal time to chemotherapy (TTC) in locally advanced gastric cancer (LAGC) patients treated with neoadjuvant chemotherapy (NLAGC) remains unclear. METHODS Consecutive 524 patients with NLAGC between Jan. 2010 and Dec. 2022 were identified. Patients were categorized into three groups: TTC < 6w, 6w ≤ TTC ≤ 8w, and TTC > 8w. Survival analysis was conducted using the Cox proportional hazards model to assess the impact of TTC on gastric cancer-specific mortality (GCSM) and all-cause mortality (ACM). Cumulative competing risk curves were employed to evaluate the incidence of competing events. RESULTS Overall, 451 patients were included.Cumulative competing risk curves showed that the 3-year ACM and GCSM were significantly lower in the 6w ≤ TTC ≤ 8w group (ACM: 19.7% vs. 37.2% vs. 39.7%, GCSM: 19.7% vs. 35.2% vs. 38.8%) compared to the TTC < 6w and TTC > 8w groups. Compared to patients with 6w ≤ TTC ≤ 8w, those with TTC < 6w or >8w had an increased risk of GCSM (HR: 2.792 and HR: 2.343, respectively) and ACM (HR: 3.102 and HR: 2.719, respectively) after adjusting for confounders. Furthermore, 6w ≤ TTC ≤ 8w had later peak recurrence compared to TTC < 6w and TTC > 8w (Peak months: 9.7 vs. 4.3 vs. 3.1). CONCLUSION A postoperative chemotherapy timing of 6-8 weeks was associated with better survival and delayed recurrence in NLAGC patients. These findings suggest that the 6-8 week time-window should be a key timeframe for personalized postoperative adjuvant chemotherapy decisions.
Collapse
Affiliation(s)
- Hua-Long Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Ling-Kang Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Hong-Hong Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Chen-Bin Lv
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Bin-Bin Xu
- Department of Digestive Endoscopy, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Guang-Tan Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| |
Collapse
|
3
|
Zhao YF, Lv JH, Chen DF, Wang ZH, Teng Y, Ntim M, Xia M, Li S, Wang B. Triglyceride-glucose index in predicting gastric cancer prognosis: A need for caution. World J Gastroenterol 2025; 31:104525. [DOI: 10.3748/wjg.v31.i18.104525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/02/2025] [Accepted: 03/27/2025] [Indexed: 05/13/2025] Open
Abstract
Gastric cancer (GC) remains one of the leading causes of cancer-related mortality worldwide. Accurate prognostic assessment, which is essential for enhancing overall survival (OS), currently depends on pathologic and clinical staging. This underscores the urgent need for reliable and real-time prognostic biomarkers. The triglyceride-glucose (TyG) index, a readily available marker of insulin resistance, has recently emerged as a potential prognostic tool in GC. Numerous studies have consistently shown a significant association between elevated TyG levels and inferior OS as well as progression-free survival. Despite these promising findings, several challenges must be addressed before the TyG index can be widely adopted in clinical practice. Firstly, the TyG index lacks cancer-specificity, reflecting broader metabolic disturbances commonly observed in conditions such as obesity, diabetes, and cardiovascular disease. This lack of specificity complicates its interpretation in oncological settings. Additionally, the cutoff values for TyG index vary across studies, hindering the establishment of a standardized threshold for clinical application. While the TyG index provides valuable insights into a patient's metabolic health, its limited cancer specificity necessitates cautious use when evaluating the prognosis of GC treatment.
Collapse
Affiliation(s)
- Yi-Fan Zhao
- Liaoning Provincial Key Laboratory of Cerebral Diseases, College of Basic Medical Sciences, National-Local Joint Engineering Research Center for Drug Research, Dalian Medical University, Dalian 116000, Liaoning Province, China
| | - Jia-Hui Lv
- Liaoning Provincial Key Laboratory of Cerebral Diseases, College of Basic Medical Sciences, National-Local Joint Engineering Research Center for Drug Research, Dalian Medical University, Dalian 116000, Liaoning Province, China
| | - De-Fang Chen
- Department of Emergency Medicine, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai 201700, China
| | - Zhi-Hui Wang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, College of Basic Medical Sciences, National-Local Joint Engineering Research Center for Drug Research, Dalian Medical University, Dalian 116000, Liaoning Province, China
| | - Yun Teng
- Liaoning Provincial Key Laboratory of Cerebral Diseases, College of Basic Medical Sciences, National-Local Joint Engineering Research Center for Drug Research, Dalian Medical University, Dalian 116000, Liaoning Province, China
| | - Michael Ntim
- Liaoning Provincial Key Laboratory of Cerebral Diseases, College of Basic Medical Sciences, National-Local Joint Engineering Research Center for Drug Research, Dalian Medical University, Dalian 116000, Liaoning Province, China
- Department of Physiology, School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi 00233, Ashanti, Ghana
| | - Min Xia
- Liaoning Provincial Key Laboratory of Cerebral Diseases, College of Basic Medical Sciences, National-Local Joint Engineering Research Center for Drug Research, Dalian Medical University, Dalian 116000, Liaoning Province, China
| | - Shao Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases, College of Basic Medical Sciences, National-Local Joint Engineering Research Center for Drug Research, Dalian Medical University, Dalian 116000, Liaoning Province, China
| | - Bin Wang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, College of Basic Medical Sciences, National-Local Joint Engineering Research Center for Drug Research, Dalian Medical University, Dalian 116000, Liaoning Province, China
| |
Collapse
|
4
|
Zhang ZQ, Zhong Q, Sun YQ, Ma YB, Ding FH, Wu D, Xue MQ, Desiderio J, Yu JH, Wu J, Zheng CH, Li P, Chen QY, Huang CM, Xie JW. The minimum number of lymph node retrieval in gastric cancer patients after neoadjuvant therapy: An international multi-institute cohort study. Surgery 2025; 183:109373. [PMID: 40328160 DOI: 10.1016/j.surg.2025.109373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 02/22/2025] [Accepted: 04/01/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND The minimum number of retrieved lymph nodes for radical gastrectomy after neoadjuvant therapy remains controversial. The objective of this study was to determine the minimum threshold for retrieved lymph nodes in patients with gastric cancer after neoadjuvant therapy to accurately evaluate staging and prognosis. METHODS Multivariate models were employed to investigate the correlation between the number of retrieved lymph nodes and survival outcomes and stage migration. The hazard ratio curves for each retrieved lymph node count compared with 1 retrieved lymph node (as a reference), including overall survival and disease-specific survival, were fitted using a LOWESS smoother, and the structural break points were determined by Chow test. RESULTS This international multicenter study analyzed the clinicopathological data of 2,490 patients with gastric cancer who underwent gastrectomy after neoadjuvant therapy. The median follow-up period for the overall population was 90.0 months, with a median of 22 retrieved lymph nodes. The study demonstrated that a greater number of retrieved lymph nodes was linked to a greater probability of detecting positive lymph nodes (odds ratio, 1.005; P = .030) as well as improved overall survival and disease-specific survival (overall survival: hazard ratio, 0.988; P < .001 and disease-specific survival: hazard ratio, 0.987; P < .001). The cutoff point analysis identified 24 as the minimum number of retrieved lymph nodes. The 5-year overall survival rate was significantly greater in the group with ≥24 retrieved lymph nodes (46.0%) compared with the group with 16-23 retrieved lymph nodes (35.3%) and the group with <16 retrieved lymph nodes group (29.3%) (P < .001). Similar trends were observed with regard to disease-specific survival. The time-dependent area under the curve analysis revealed that the group with ≥24 retrieved lymph nodes exhibited superior predictive performance for overall survival and disease-specific survival compared to the group with <24 retrieved lymph nodes, on the basis of ypN staging (overall survival: time-dependent areas under the receiver operating characteristic curve≥24, 0.71 vs time-dependent areas under the receiver operating characteristic curve<24, 0.67; disease-specific survival: time-dependent areas under the receiver operating characteristic curve≥24, 0.72 vs time-dependent areas under the receiver operating characteristic curve<24, 0.68). CONCLUSION The minimum number of retrieved lymph nodes for evaluation prognosis and reducing stage migration in patients with gastric cancer who undergo radical gastrectomy after neoadjuvant therapy was 24.
Collapse
Affiliation(s)
- Zhi-Quan Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Yu-Qin Sun
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China; Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Yu-Bin Ma
- Gastrointestinal Surgery, Qinghai University Affiliated Hospital, Xining, China
| | - Fang-Hui Ding
- General Surgery Department, The First Hospital of Lanzhou University, Lanzhou, China
| | - Dong Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Meng-Qi Xue
- Public Health School, Fujian Medical University, Fuzhou, Fujian, China
| | - Jacopo Desiderio
- Department of Digestive Surgery, St. Mary's Hospital, University of Perugia, Terni, Italy
| | - Jun-Hua Yu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China; Department of General Surgery, The Quzhou Affiliated Hospital of Wenzhou University, Quzhou, China
| | - Ju Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China; Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
| |
Collapse
|
5
|
Tan Y, Xing Y, Yuan S, Sun F, Lin X, Bao S, Jiang D, Zhang J, Sun SL. Potential Value of AURKA and CDK6 Amplification for the Response of Patients With Gastric Cancer to Neoadjuvant Chemotherapy. Mol Carcinog 2025. [PMID: 40222043 DOI: 10.1002/mc.23921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/19/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025]
Abstract
Many patients respond poorly to neoadjuvant chemotherapy (NACT), negatively affecting the surgical success rate. Identifying effective biomarkers and understanding the potential resistance mechanisms are urgently needed. Data of 18 patients with advanced stomach cancer who were treated with NACT categorized according to tumor regression grade into major histological response (MJHR) and nonhistological response (NHR) groups were retrospectively analyzed. Genomic signatures associated with the response to NACT were identified using whole-exome and RNA sequencing. Extraction of molecular signatures revealed increased deficient mismatch repair signature and tumor mutation levels in the NHR group. Compared to the MJHR group, the NHR group was also characterized by a greater number of copy number alterations (p = 0.08), which was further confirmed by RNA sequencing, and upregulation of aurora kinase A (AURKA) (p = 0.05) and cyclin-dependent kinase 6 (CDK6) (p = 0.049). Western blot analysis and immunohistochemical analyses further confirmed high CDK6 (p < 0.01/p < 0.0001) and AURKA (p < 0.01/p < 0.001) expression levels in the NHR group. Finally, palbociclib, an inhibitor of CDK4/6, effectively inhibited the proliferation (p < 0.05) and induced apoptosis of oxaliplatin-resistant gastric cancer cells (p < 0.01) in vitro. These findings support the potential value of AURKA and CDK6 amplification, as well as their effects on the tumor microenvironment, in predicting poor outcomes of NACT in patients with locally advanced gastric cancer. Thus, CDK4/6 inhibitors could be used to treat NACT-resistant patients with gastric cancer.
Collapse
Affiliation(s)
- Yuen Tan
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Yao Xing
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Shuai Yuan
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Fan Sun
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Xiaohui Lin
- School of Computer Science and Technology, Dalian University of Technology, Dalian, China
| | - Simeng Bao
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Dongyue Jiang
- Key Laboratory of Ocean Energy Utilization and Energy Conservation of Ministry of Education, Dalian University of Technology, Dalian, China
| | - Jianjun Zhang
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| | - Shu-Lan Sun
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, China
| |
Collapse
|
6
|
Ma GF, Huang YY, Chen YC, Luo Z, Li XP. Premature death patterns and trends of stomach cancer in Pudong, Shanghai: a population based study. BMC Cancer 2025; 25:618. [PMID: 40188108 PMCID: PMC11972478 DOI: 10.1186/s12885-025-14024-z] [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: 12/05/2023] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Estimating the disease burden of stomach cancer is essential to developing evidence-based prevention and treatment strategies. AIMS To analyze the number of deaths and the temporal trends in the mortality and years of life lost (YLL) in relation to gender, age, and the impact of aging and comorbidity via non-communicable diseases on stomach cancer burden in one of the most developed regions of a transitioning country. METHODS Mortality data of stomach cancer were collected from the Vital Statistics System of the Pudong New Area, Shanghai, China, from 2005 to 2019. The long-term trends in crude mortality rates (CMR), age-standardized mortality rates worldwide (ASMRW), and rate of YLL(YLLr) were analyzed using the Joinpoint regression program. The aging and non-aging factors affecting the mortality rate were evaluated by the decomposition method. RESULTS A total of 11,609 deaths from stomach cancer occurred from 2005 to 2019. The CMR and ASMRW of stomach cancer were 29.83/105 person-year and 12.20/105 person-year, respectively. The CMR, ASMRW, and YLLr in males were nearly twice as higher as those in females(CMR: 35.47/105 vs. 19.83/105, ASMRW: 15.64/105 vs. 7.74/105, YLLr: 378.63/105 vs. 229.13/105). The main co-morbidities involved the circulatory (24.64%) and respiratory system (20.62%). The main metastatic sites were liver (9.08%), lung (2.79%) and peritoneum (2.33%). The long-term trends in CMR and ASMRW were significantly decreasing in males, females, and the total population from 2005 to 2019. A total of 9,460 (81.48%) elderly people aged ≥ 60 years died of stomach cancer. The top three age groups with the highest CMR were ≥ 80 years, 70-79 years, and 60-69 years. The CMR and YLLr of people aged ≥ 80 years showed the largest significantly decreasing trends. The CMR caused by aging showed significantly upward trends [average annual percent changes (AAPC) 95%CI = 37.63(14.95,64.79)%, P < 0.001], which caused by non-aging factor showed significantly downward trends [AAPC 95%CI = -18.17(-22.83,-13.22)%, P < 0.001]. CONCLUSION Age is an important factor affecting the trend of disease burden of stomach cancer. Paying attention to high-risk people may help to reduce the YLL.
Collapse
Affiliation(s)
- Gui-Fen Ma
- Department of Radioation Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Center for Cancer Prevention and Treatment, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Radioation Oncology, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, 362000, China
| | - Ya-Yu Huang
- Department of Radioation Oncology, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, 362000, China
| | - Yi-Chen Chen
- Center for Disease Control and Prevention, Pudong New Area, Shanghai, 200136, China
- Pudong Institute of Preventive Medicine, Fudan University, Shanghai, 200136, China
| | - Zheng Luo
- Zhoupu Hospital, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - Xiao-Pan Li
- Department of Health Management Center, Zhongshan Hospital, Fudan University, No. 180 Fenling Rd., Xuhui, Shanghai, 200032, China.
| |
Collapse
|
7
|
Sun D, Cui X, Yang W, Wei M, Yan Z, Zhang M, Yu W. Simvastatin inhibits PD-L1 via ILF3 to induce ferroptosis in gastric cancer cells. Cell Death Dis 2025; 16:208. [PMID: 40140647 PMCID: PMC11947124 DOI: 10.1038/s41419-025-07562-8] [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: 10/28/2024] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025]
Abstract
The treatment of gastric cancer remains challenging, with immunotherapy serving as a critical component of the holistic approach to its treatment. The results of this study indicated that statins could decrease the serum levels of interleukin-enhancing binding factor 3 (ILF3) and programmed cell death ligand 1(PD-L1) in GC patients and improve their prognosis. Functional experiments demonstrated that simvastatin induced ferroptosis by inhibiting ILF3 in GC cells and enhanced the killing effect of activated CD8+ T cells on GC cells. The CUT&Tag assay revealed that, mechanistically, simvastatin inhibited ILF3 expression by reducing the acetylation level at residue site H3K14 in ILF3. Next-generation sequencing and Kyoto Encyclopedia of Genes and Genomes analysis revealed that ILF3 regulated PD-L1 expression through the DEPTOR/mTOR signaling pathway. Overall, simvastatin induced ferroptosis in GC cells by inhibiting ILF3 expression while promoting the activation of CD8+ T cells to augment antitumor immune responses, thereby facilitating synergistic immunotherapy.
Collapse
Affiliation(s)
- Danping Sun
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Xiaohan Cui
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Wenshuo Yang
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Meng Wei
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Zhibo Yan
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Mingxiang Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Wenbin Yu
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China.
| |
Collapse
|
8
|
Ji X, Wang G, Pan D, Xu S, Lei X. Efficacy and safety of pembrolizumab in advanced gastric and gastroesophageal junction cancer: a systematic review and meta-analysis. BMC Gastroenterol 2025; 25:173. [PMID: 40087572 PMCID: PMC11908035 DOI: 10.1186/s12876-025-03754-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Pembrolizumab, a PD-1 inhibitor, has shown potential for treating advanced gastric and gastroesophageal junction (GEJ) cancer. This meta-analysis evaluates its efficacy and safety, alone or combined with chemotherapy, in this population. METHODS A systematic review and meta-analysis were conducted in accordance with PRISMA guidelines. Databases including PubMed, Embase, the Cochrane Central Register of Controlled Trials, and Web of Science were searched up to October 31, 2024. Twelve studies comprising 4,069 patients were included. The primary outcomes were overall survival (OS) and progression-free survival (PFS); secondary outcomes included objective response rate (ORR), adverse events (AEs), and grade ≥ 3 AEs. Effect sizes were calculated using mean differences (MDs) and odds ratios (ORs) with 95% confidence intervals (CIs). RESULTS Pembrolizumab combined with chemotherapy significantly improved OS (MD = 1.92 months; 95% CI: 0.94 to 2.91) and ORR (MD = 11.05%; 95% CI: 6.29 to 15.82) compared to chemotherapy alone. Pembrolizumab monotherapy did not show a significant effect on OS (MD = 0.24 months; 95% CI: -1.15 to 1.63) and was associated with a significant reduction in PFS (MD = -2.28 months; 95% CI: -2.85 to -1.71) compared to chemotherapy alone. For safety, pembrolizumab monotherapy significantly reduced the risk of AEs (OR = 0.68; 95% CI: 0.57 to 0.81) and grade ≥ 3 AEs (OR = 0.39; 95% CI: 0.30 to 0.51) compared to chemotherapy. Pembrolizumab combined with chemotherapy did not significantly alter the risk of AEs (OR = 1.01; 95% CI: 0.90 to 1.13) or grade ≥ 3 AEs (OR = 1.12; 95% CI: 0.99 to 1.27) compared to chemotherapy alone. CONCLUSION Pembrolizumab combined with chemotherapy improves survival and response rates with a manageable safety profile in advanced gastric and GEJ cancers. Monotherapy shows limited efficacy, highlighting the need for combination strategies and patient selection.
Collapse
Affiliation(s)
- Xiaoying Ji
- Department of Pharmacy, Yiwu Central Hospital, Yiwu, Zhejiang, 322000, China
| | - Guoping Wang
- Department of Pharmacy, Yiwu Central Hospital, Yiwu, Zhejiang, 322000, China
| | - Dandan Pan
- Department of Pharmacy, Yiwu Central Hospital, Yiwu, Zhejiang, 322000, China
| | - Shanxia Xu
- Quzhou Zhong Da Lang Yuan Nursing Home, Quzhou, Zhejiang, 324000, China
| | - Xinming Lei
- The Quzhou Afliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, 324000, China.
| |
Collapse
|
9
|
He QC, Huang ZN, Lv CB, Wu YH, Qiu WW, Ma YB, Wu J, Zheng CY, Lin GS, Li P, Wang JB, Lin JX, Lin M, Tu RH, Zheng CH, Huang CM, Cao LL, Xie JW. Effect of Helicobacter pylori infection on survival outcomes of patients undergoing radical gastrectomy after neoadjuvant chemotherapy: a multicenter study in China. BMC Cancer 2025; 25:460. [PMID: 40082850 PMCID: PMC11907980 DOI: 10.1186/s12885-025-13840-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy (NAC) has been confirmed to improve the prognosis of patients with advanced gastric cancer (AGC). However, no study has investigated whether Helicobacter pylori (HP) infection affects the postoperative survival of patients who receive NAC. METHODS This retrospective cohort study included 307 patients with AGC who underwent laparoscopic radical gastrectomy after NAC at three hospitals in China between January 1, 2016, and April 31, 2020. Cox regression was used to assess prognostic factors for survival. Kaplan-Meier was used for survival analysis. RESULTS The HP + and the HP- group included 141 and 166 cases. The 3-year overall survival (OS) and disease-free survival (DFS) of the HP + group were significantly better than the HP- group (3-year OS: 75.9% vs. 60.2%, 3-year DFS: 70.2% vs. 52.3%; All P < 0.001). For the HP + group, ypTNM Stage III (HR, 4.00; 95% CI, 1.11-14.39; P = 0.034), NAC ≥ 4 cycles (HR, 0.43; 95% CI, 0.20-0.90; P = 0.026), and adjuvant chemotherapy (AC) ≥ 4 cycles (HR, 0.20; 95% CI, 0.09-0.48; P < 0.001) are independent prognostic factors for OS. In the cohort of HP + patients who received ≥ 4 cycles of NAC, the prognosis of patients who received ≥ 4 cycles of AC after surgery was better than that of patients who received < 4 cycles of AC (3-year OS: 92.5% vs 71.4%; P = 0.042). CONCLUSIONS Following NAC, HP + patients with AGC exhibit better prognosis than that of HP- counterparts. For potentially resectable HP + AGC patients, radical surgery following ≥ 4 cycles of NAC with ≥ 4 cycles of sequential AC might be recommended to improve survival.
Collapse
Affiliation(s)
- Qi-Chen He
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Ze-Ning Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Chen-Bin Lv
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Yong-He Wu
- Department of Pathology, Zhangzhou Affiliated Hospital of Fujian Medical University, ZhangZhou, China
| | - Wen-Wu Qiu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Yu-Bin Ma
- Department of Gastrointestinal Surgery, Qinghai University Affiliated Hospital, Xining, China
| | - Ju Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Chang-Yue Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Putian University, Putian, China
| | - Guo-Sheng Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Ru-Hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China.
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China.
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Rd, Fuzhou, 350001, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
10
|
Carpio AR, Talubo ND, Tsai PW, Chen BY, Tayo LL. Berries as Nature's Therapeutics: Exploring the Potential of Vaccinium Metabolites in Gastric Cancer Treatment Through Computational Insights. Life (Basel) 2025; 15:406. [PMID: 40141751 PMCID: PMC11944152 DOI: 10.3390/life15030406] [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: 01/20/2025] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
Berries from the Vaccinium genus, known for their rich array of bioactive metabolites, are recognized for their antioxidant, anti-inflammatory, and anticancer properties. These compounds, including anthocyanins, flavonoids, and phenolic acids, have attracted significant attention for their potential health benefits, particularly in cancer prevention and treatment. Gastric cancer (GC), a leading cause of cancer-related deaths worldwide, remains challenging to treat, especially in its advanced stages. This study investigates the therapeutic potential of Vaccinium species in GC treatment using computational methods. RNA sequencing revealed upregulated genes associated with GC, while network pharmacology and molecular docking approaches identified strong interactions between cyanidin 3-O-glucoside (C3G), a key bioactive metabolite. Furthermore, molecular dynamics simulations of the HSP90AA1-C3G complex demonstrated stable binding and structural integrity, suggesting that C3G may inhibit HSP90AA1, a protein involved in cancer progression. These findings highlight the therapeutic potential of Vaccinium metabolites, offering a novel approach to GC treatment by targeting key molecular pathways. This research provides valuable insights into the role of berries as natural therapeutics, supporting their integration into future gastric cancer treatment strategies.
Collapse
Affiliation(s)
- Angelica Rachel Carpio
- School of Chemical, Biological, and Materials Engineering and Sciences, School of Graduate Studies, Mapúa University, Manila 1002, Philippines; (A.R.C.); (N.D.T.)
| | - Nicholas Dale Talubo
- School of Chemical, Biological, and Materials Engineering and Sciences, School of Graduate Studies, Mapúa University, Manila 1002, Philippines; (A.R.C.); (N.D.T.)
| | - Po-Wei Tsai
- Department of Food Science, National Taiwan Ocean University, Keelung 202, Taiwan;
| | - Bor-Yann Chen
- Department of Chemical and Materials Engineering, National I-Lan University, I-Lan 260, Taiwan
| | - Lemmuel L. Tayo
- Department of Biology, School of Health Sciences, Mapúa University, Makati 1200, Philippines
| |
Collapse
|
11
|
Vásquez-Jaico ER, Yan-Quiroz EF, Bonilla-Feria N, Gonzalez MNV, Guzmán LS, Vásquez-Tirado GA, Serna-Alarcon V. Upfront surgery versus chemotherapy neoadjuvant in the survival of patients with locally advanced gastric signet-ring-cell adenocarcinoma. A scoping review. Ecancermedicalscience 2025; 19:1843. [PMID: 40248270 PMCID: PMC12003976 DOI: 10.3332/ecancer.2025.1843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Indexed: 04/19/2025] Open
Abstract
Background Recent research suggests that neoadjuvant chemotherapy is not effective for gastric cancer with signet ring cells. Objective The present study performs a scoping review of research that seeks to determine whether neoadjuvant chemotherapy is more effective than upfront surgery in the survival of locally advanced signet ring gastric adenocarcinoma. Design Online databases such as Pubmed, scopus and embase were used to identify articles from the last 20 years that used survival, as an initial or secondary outcome variable, after upfront surgery or neoadjuvant chemotherapy as initial treatment in locally advanced gastric signet ring cells adenocarcinoma. Results After a systematic selection process, five primary studies were selected that evaluated neoadjuvant chemotherapy compared to primary surgery. Conclusion Neoadjuvant chemotherapy does not appear to have greater benefit than initial surgery in gastric adenocarcinoma with locally advanced sign ring cells, it is necessary to define which is the most appropriate qt scheme for adenocarcinoma with sign ring cells, clinical trials type studies are required to improve the evidence. Finally, a national clinical practice guide is required as an interpretative map for the management of gastric cancer which may be appropriate as a first step to know the reality.
Collapse
Affiliation(s)
- Erick R Vásquez-Jaico
- School of Medicine, Faculty of Medicine, Antenor Orrego Private University, Trujillo 13008, Perú
- Virgen De La Puerta High Complexity Hospital Essalud, Trujillo 13013, Perú
- https://orcid.org/0000-0003-4350-7461
| | - Edgar Fermín Yan-Quiroz
- School of Medicine, Faculty of Medicine, Antenor Orrego Private University, Trujillo 13008, Perú
- Virgen De La Puerta High Complexity Hospital Essalud, Trujillo 13013, Perú
- https://orcid.org/0000-0002-9128-4760
| | - Nicol Bonilla-Feria
- School of Medicine, Faculty of Medicine, Antenor Orrego Private University, Trujillo 13008, Perú
- https://orcid.org/0009-0006-3659-7277
| | | | - Luis Salas Guzmán
- National University of San Marcos, Lima 15001, Perú
- https://orcid.org/0009-0008-2893-272X
| | - Gustavo Adolfo Vásquez-Tirado
- School of Medicine, Faculty of Medicine, Antenor Orrego Private University, Trujillo 13008, Perú
- https://orcid.org/0000-0002-2109-6430
| | - Victor Serna-Alarcon
- School of Medicine, Faculty of Medicine, Antenor Orrego Private University, Trujillo 13008, Perú
- Jose Cayetano Heredia EsSalud Regional Hospital, Piura 20002, Perú
- https://orcid.org/0000-0002-9803-6217
| |
Collapse
|
12
|
Matsui R, Yonezu K, Rifu K, Watanabe J, Inaki N, Fukunaga T, Nunobe S. Impact of sarcopenic obesity on postoperative outcomes in patients with oesophago-gastric cancer: A systematic review and meta-analysis. World J Surg 2025; 49:459-471. [PMID: 39725406 DOI: 10.1002/wjs.12451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 11/23/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND We elucidated the influence of sarcopenic obesity on postoperative outcomes in patients with oesophago-gastric cancer. METHODS We conducted a systematic search on MEDLINE, the Cochrane Central Register of Controlled Trials, EMBASE, the World Health Organization International Clinical Trials Platform Search Portal, and ClinicalTrials.gov to identify observational studies published from their inception to September 26, 2024. Studies involving patients who underwent radical resection for oesophago-gastric cancer and were evaluated for visceral fat mass and skeletal muscle mass through body composition were included in our analysis. The primary outcomes assessed were overall survival (OS) and postoperative complications. This protocol was registered in PROSPERO (CRD42023418403). RESULTS Ultimately, 13 studies (involving 4912 patients) were included in our qualitative and quantitative analyses. Among these studies, three were prospective cohort studies, while the remaining 10 were retrospective cohort studies. Twelve studies specifically investigated gastric cancer, while one focused on esophageal cancer. The prevalence of sarcopenic obesity ranged from 5.7% to 28.7%. Compared to the absence of sarcopenic obesity, its presence worsens OS (hazard ratio: 1.52, 95% confidence interval: 1.08-2.15, heterogeneity (I2) = 66%, certainty of the evidence: low) and increases the risk of postoperative complications (relative risk ratio: 1.88, 95% CI: 1.29-2.73, I2 = 77%, certainty of the evidence: moderate). The risk of bias in each study was deemed moderate to high. CONCLUSIONS Sarcopenic obesity worsens OS and increases the risk of postoperative complications in patients with oesophago-gastric cancer undergoing radical resection.
Collapse
Affiliation(s)
- Ryota Matsui
- Department of Gastroenterological Surgery, Cancer Institute Ariake Hospital, Tokyo, Japan
- Department of Upper Gastrointesitinal Surgery, Juntendo University Hospital, Tokyo, Japan
- Department of Gastrointestinal Surgery/Breast Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Keisuke Yonezu
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University, Oita, Japan
| | - Kazuma Rifu
- Department of Surgery, Division of Gastroenterological, General and Transplant Surgery, Jichi Medical University, Shimotsuke City, Tochigi, Japan
| | - Jun Watanabe
- Department of Surgery, Division of Gastroenterological, General and Transplant Surgery, Jichi Medical University, Shimotsuke City, Tochigi, Japan
- Center for Community Medicine, Jichi Medical University, Shimotsuke City, Tochigi, Japan
| | - Noriyuki Inaki
- Department of Gastrointestinal Surgery/Breast Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Tetsu Fukunaga
- Department of Upper Gastrointesitinal Surgery, Juntendo University Hospital, Tokyo, Japan
| | - Souya Nunobe
- Department of Gastroenterological Surgery, Cancer Institute Ariake Hospital, Tokyo, Japan
| |
Collapse
|
13
|
Zhang G, Qu W, Huang X, Yi J, Gao H, He J, Xue W. HECW1-Mediated Ubiquitination of HIPK2 Drives Metastasis in Gastric Cancer Through the AKT Signaling Pathway. J Transl Med 2025; 105:102202. [PMID: 39615883 DOI: 10.1016/j.labinv.2024.102202] [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: 05/23/2024] [Revised: 11/18/2024] [Accepted: 11/24/2024] [Indexed: 12/29/2024] Open
Abstract
E3 ubiquitin ligases, crucial enzymes in the ubiquitination pathway, significantly influence the development of malignant tumors, including gastric cancer (GC), by regulating the stability of oncogenic and tumor-suppressive proteins. This study employed bioinformatics analysis of public databases alongside various experimental techniques-tissue arrays, real-time reverse-transcription polymerase chain reaction, western blot, immunofluorescence, and coimmunoprecipitation-to identify and explore the role of HECW1, a pivotal NEDD4 family E3 ubiquitin ligase, in GC progression. The results demonstrated that HECW1 is markedly overexpressed in GC tissues relative to normal gastric tissues, and its elevated expression correlates with poor prognosis in GC patients. In vitro experiments revealed that HECW1 overexpression significantly enhances the metastatic capabilities of GC cells. Mechanistically, HECW1 interacts with HIPK2 to facilitate its ubiquitination and degradation, thereby activating AKT and promoting the expression of downstream epithelial mesenchymal transition-related genes. In vivo experiments confirmed HECW1's role in promoting GC cell metastasis, highlighting the HECW1-HIPK2-AKT signaling axis as critical in GC metastasis. These findings not only elucidate a novel metastasis mechanism of GC but also suggest potential molecular targets for developing new therapeutic strategies against GC.
Collapse
Affiliation(s)
- Guangze Zhang
- Department of Gastrointestinal Surgery, Affiliated Hospital and Medical School of Nantong University, Nantong, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China; Nantong Key Laboratory of Gastrointestinal Oncology, Nantong, China
| | - Weilong Qu
- Department of Gastrointestinal Surgery, Affiliated Hospital and Medical School of Nantong University, Nantong, China; Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| | - Xinkun Huang
- Department of General Surgery, Nantong Tumor Hospital and Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Jianfeng Yi
- Department of Gastrointestinal Surgery, Affiliated Hospital and Medical School of Nantong University, Nantong, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China; Nantong Key Laboratory of Gastrointestinal Oncology, Nantong, China
| | - Hanxu Gao
- Department of Gastrointestinal Surgery, Affiliated Hospital and Medical School of Nantong University, Nantong, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China; Nantong Key Laboratory of Gastrointestinal Oncology, Nantong, China
| | - Jiancheng He
- Department of Gastrointestinal Surgery, Affiliated Hospital and Medical School of Nantong University, Nantong, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China; Nantong Key Laboratory of Gastrointestinal Oncology, Nantong, China.
| | - Wanjiang Xue
- Department of Gastrointestinal Surgery, Affiliated Hospital and Medical School of Nantong University, Nantong, China; Nantong Key Laboratory of Gastrointestinal Oncology, Nantong, China.
| |
Collapse
|
14
|
Yu C, Jiang H, Wang L, Jiang Z, Jin C. Baseline (derived) neutrophil-lymphocyte ratio associated with survival in gastroesophageal junction or gastric cancer treated with ICIs. Front Oncol 2025; 15:1404695. [PMID: 39926278 PMCID: PMC11802431 DOI: 10.3389/fonc.2025.1404695] [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: 03/21/2024] [Accepted: 01/06/2025] [Indexed: 02/11/2025] Open
Abstract
Objective We carried out the meta-analysis to determine the predictive value of baseline neutrophil to lymphocyte ratio (NLR) and derived neutrophil to lymphocyte ratio (dNLR) levels in patients with gastroesophageal junction or gastric cancer (GJGC) who underwent immune checkpoint inhibitor (ICI) treatment. Methods Eligible articles were obtained through PubMed, the Cochrane Library, EMBASE, and Google Scholar, until April 15, 2023. The clinical outcomes evaluated in this study encompassed overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and disease control rate (DCR). Results A total of 24 articles with 2221 patients were included in this meta-analysis. The pooled results demonstrated that patients with high NLR levels had significantly poorer OS (HR: 1.860, 95% CI: 1.564-2.213, p < 0.001) and PFS (HR: 1.678, 95% CI: 1.354-2.079, p < 0.001), and lower ORR (OR: 0.754, 95% CI: 0.621-0.915, p = 0.004) and DCR (OR: 0.391, 95% CI: 0.262-0.582, p < 0.001). Besides, we also found that high dNLR levels were significantly associated with shorter OS (HR: 2.117, 95% CI: 1.590-2.820, p < 0.001) and PFS (HR: 1.803, 95% CI: 1.415-2.297, p < 0.001). Conclusion Low baseline (Derived) NLR has the potential to predict the good efficacy of ICIs and survival outcomes in patients with GJGC. (Derived) NLR could be useful in determining the optimal treatment strategies for these patients.
Collapse
Affiliation(s)
| | | | | | | | - Chong Jin
- Department of General Surgery, Taizhou Central Hospital, Taizhou University, Taizhou, Zhejiang, China
| |
Collapse
|
15
|
Chen Y, Zhou Y, Xiong H, Wei Z, Zhang D, Li S. Exploring the Efficacy of Hyperthermic Intraperitoneal Perfusion Chemotherapy in Gastric Cancer: Meta-analysis Based on Randomized Controlled Trials. Ann Surg Oncol 2025; 32:240-248. [PMID: 39373926 DOI: 10.1245/s10434-024-16298-2] [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: 12/15/2023] [Accepted: 09/20/2024] [Indexed: 10/08/2024]
Abstract
PURPOSE Based on randomized clinical trials, this meta-analysis evaluated the efficacy and safety of intent-to-cure or prophylactic hyperthermic intraperitoneal chemotherapy (HIPEC) for the treatment of gastric cancer. METHODS PubMed, Cochrane Library, Medline, and Embase databases were all systemically searched from 2004 to 2023. The quality of the study was assessed by using the Cochrane risk of bias method. The certainty of the evidence was determined by using the GRADE evaluation. RESULTS In this study, 12 articles with a total of 1181 patients were analyzed based on the inclusion criteria. The findings revealed that HIPEC had a higher survival rate (risk ratio [RR] 0.60; 95% confidence interval [CI] [0.43, 0.86], P = 0.005, RR 0.82; 95% CI [0.70, 0.97], P = 0.02, RR 0.83; 95% CI [0.71, 0.96], P < 0.01, and RR 0.63 [0.54, 0.73], P < 0.00001) after 1, 2, 3, and 5 years compared with the control group. The RR was statistically significant for 1, 2, 3, and 5 years. Furthermore, the observed overall recurrence rate for the HIPEC group was lower than control group and statistically significant (RR 0.59; 95% CI [0.50, 0.68], P < 0.0001). Higher disease-free survival rate (RR 1.42; 95% CI [1.07, 1.89], P < 0.01) was observed in the HIPEC group and statistically significant. CONCLUSIONS Gastric cancer patients treated with HIPEC have shown promising outcomes with regard to survival, recurrence, disease-free survival, and adverse reactions. However, multicenter trials with larger sample sizes consisting of different ethnicities is suggested.
Collapse
Affiliation(s)
- Ying Chen
- Oncology Department of Siyang Hospital of Traditional Chinese Medicine, Siyang, China
| | - Yang Zhou
- General Surgery Department, Siyang County First Peoples's Hospital, Siyang, China
| | - Huaping Xiong
- Oncology Department of Siyang Hospital of Traditional Chinese Medicine, Siyang, China
| | - Zhen Wei
- Oncology Department of Siyang Hospital of Traditional Chinese Medicine, Siyang, China
| | - Dong Zhang
- General Surgery Department, Siyang County First Peoples's Hospital, Siyang, China
| | - Shoushan Li
- Oncology Department of Siyang Hospital of Traditional Chinese Medicine, Siyang, China.
| |
Collapse
|
16
|
Dominguez RL, Montalvan-Sanchez EE, Norwood DA, Rodriguez-Murillo A, Dominguez L, Estevez Ordoñez D, Beasley T, Bravo LE, Morgan DR. Population-Based Study of Gastric Cancer Survival and Associations in Rural Western Honduras. Cancer Epidemiol Biomarkers Prev 2024; 33:1578-1585. [PMID: 38949525 PMCID: PMC12103253 DOI: 10.1158/1055-9965.epi-23-1461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/06/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024] Open
Abstract
BACKGROUND Two-thirds of global cancers occur in low/middle income countries (LMIC). Northern Central America is the largest LMIC region in the Western Hemisphere and lack cancer registries to guide cancer control. We conducted a gastric cancer survival study in rural Western Honduras, characterized as having among the highest gastric cancer incidence rates in Latin America. METHODS The cohort of incident gastric cancer diagnosed between 2002 and 2015 was studied with active follow-up with household visits. The regional gastric cancer registry was primary for case identification, with completeness examination with hospital data and national death certificates. Cox regression models were used for survival calculations. RESULTS Survival follow-up was achieved in 741/774 patients (95.7%). Household interviews were conducted in 74.1% (n = 549); 65.7% were male, median age at diagnosis was 64 years, 24.5% were <55 years; 43.9% of tumors had pyloric obstruction; 45.2%, 43.2%, and 7.3% of histology was intestinal, diffuse, and mixed, respectively. A total of 24.7% patients received treatment. The 5-year survival rates were 9.9% for both males and females, 7.7% for age <45, and 7.9% for diffuse gastric cancer. Median survival time was 4.8 months [95% confidence interval (CI), 4.2-5.6]. In the final Cox regression model including age, sex, Lauren subtype, and poverty index, only treatment was significantly associated with survival (hazard ratio = 2.43, 95% CI, 1.8-3.2). CONCLUSIONS Markedly low gastric cancer 5-year survival rates are observed in rural Central America. The majority of patients present with advanced disease and a minority have access to therapy. IMPACT The findings have implications for cancer control in the Central America LMICs and for US Latino populations. See related commentary by Riquelme and Abnet, p. 1550.
Collapse
Affiliation(s)
- Ricardo L Dominguez
- Western Honduras Gastric Cancer Program, Ministry of Health, Santa Rosa de Copan, Honduras
| | - Eleazar E Montalvan-Sanchez
- Western Honduras Gastric Cancer Program, Ministry of Health, Santa Rosa de Copan, Honduras
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Dalton A Norwood
- Western Honduras Gastric Cancer Program, Ministry of Health, Santa Rosa de Copan, Honduras
- Division of Preventive Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Aida Rodriguez-Murillo
- Western Honduras Gastric Cancer Program, Ministry of Health, Santa Rosa de Copan, Honduras
| | - Lucia Dominguez
- Western Honduras Gastric Cancer Program, Ministry of Health, Santa Rosa de Copan, Honduras
| | | | - Timothy Beasley
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama
| | - Luis E Bravo
- Departamento de Patología, Facultad de Salud, Escuela de Medicina, Universidad del Valle, Cali, Colombia
| | - Douglas R Morgan
- Division of Gastroenterology and Hepatology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
17
|
Sun Z, Liu L, Chen J. Targeting non-histone methylation in gastrointestinal cancers: From biology to clinic. Eur J Med Chem 2024; 278:116802. [PMID: 39213938 DOI: 10.1016/j.ejmech.2024.116802] [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: 07/22/2024] [Revised: 08/24/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Gastrointestinal (GI) cancers, encompassing a range of malignancies within the digestive tract, present significant challenges in both diagnosis and treatment, reflecting a dire need for innovative therapeutic strategies. This article delves into the profound influence of non-histone methylation on the pathogenesis and evolution of gastrointestinal (GI) cancers. Non-histone proteins, undergoing methylation by enzymes such as Protein Arginine Methyltransferases (PRMTs) and Lysine Methyltransferases (KMTs), play pivotal roles in cellular signaling, metabolism, chromatin remodeling, and other processes crucial for cancer development. This review illuminates the complex mechanisms by which non-histone methylation affects key aspects of tumor biology, including oncogenesis, growth, proliferation, invasion, migration, metabolic reprogramming, and immune escape in GI malignancies. Highlighting recent discoveries, this work underscores the importance of non-histone methylation in cancer biology and its potential as a target for innovative therapeutic strategies aimed at improving outcomes for patients with GI cancers.
Collapse
Affiliation(s)
- Zhanbo Sun
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China
| | - Lixian Liu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China
| | - Jun Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China.
| |
Collapse
|
18
|
Feng QX, Zhu ZN, Li Q, Liu XS. Dual-energy CT quantitative parameters to evaluate occult peritoneal metastasis in advanced gastric cancer preoperatively. Abdom Radiol (NY) 2024; 49:3309-3318. [PMID: 38634880 DOI: 10.1007/s00261-024-04303-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE To explore whether dual-energy CT (DECT) quantitative parameters could provide analytic value for the diagnosis of patients with occult peritoneal metastasis (OPM) in advanced gastric cancer preoperatively. MATERIALS AND METHODS This retrospective study included 219 patients with advanced gastric cancer and DECT scans. The patient's clinical data and DECT related iodine concentration (IC) parameters and effective atomic number (Zeff) were collated and analyzed among noun-peritoneal metastasis (NPM), OPM and radiologically peritoneal metastasis (RPM) groups. The predictive performance of the DECT parameters was compared with that of the conventional CT features and clinical characteristics through evaluating area under curve of the precision-recall (AUC-PR), F1 score, balanced accuracy, sensitivity, specificity, positive predictive value (PPV) and negative predictive value (NPV). RESULTS Borrmann IV type diagnosed on CT and serum tumor indicator CA125 index were statistically different between the NPM and OPM groups. DECT parameters included IC, normalized IC (NIC), and Zeff of PM group were lower than the NPM group. The DECT predictive nomogram combined three independent DECT parameters produced a better diagnostic performance than the conventional CT feature Borrmann IV type and serum CA125 index in AUC-PR with 0.884 vs 0.368 vs 0.189, but similar to the combined indicator which was based on the DECT parameters, the conventional CT feature, and serum CA125 index in AUC-PR with 0.884 vs 0.918. CONCLUSION The lower quantitative NIC, IC ratio, and Zeff on DECT was associated with peritoneal metastasis in advanced gastric cancer and was promising to identify patients with OPM noninvasively.
Collapse
Affiliation(s)
- Qiu-Xia Feng
- Department of Radiology, The First Affiliated Hospital With Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210009, Jiangsu Province, China
| | - Zhen-Ning Zhu
- Department of Radiology, The First Affiliated Hospital With Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210009, Jiangsu Province, China
| | - Qiong Li
- Department of Radiology, The First Affiliated Hospital With Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210009, Jiangsu Province, China
| | - Xi-Sheng Liu
- Department of Radiology, The First Affiliated Hospital With Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210009, Jiangsu Province, China.
| |
Collapse
|
19
|
Wang Y, Ding G, Chu C, Cheng XD, Qin JJ. Genomic biology and therapeutic strategies of liver metastasis from gastric cancer. Crit Rev Oncol Hematol 2024; 202:104470. [PMID: 39111457 DOI: 10.1016/j.critrevonc.2024.104470] [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: 12/15/2023] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
The liver is a frequent site of metastasis in advanced gastric cancer (GC). Despite significant advancements in diagnostic and therapeutic techniques, the overall survival rate for patients afflicted with gastric cancer liver metastasis (GCLM) remains dismally low. Precision oncology has made significant progress in identifying therapeutic targets and enhancing our understanding of metastasis mechanisms through genome sequencing and molecular characterization. Therefore, it is crucial to have a comprehensive understanding of the various molecular processes involved in GCLM and the fundamental principles of systemic therapy to develop new treatment approaches. This paper aims to review recent findings on the diagnosis, potential biomarkers, and therapies targeting the multiple molecular processes of GCLM, with the goal of improving treatment strategies for patients with GCLM.
Collapse
Affiliation(s)
- Yichao Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 313200, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Guangyu Ding
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Chu Chu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 313200, China
| | - Xiang-Dong Cheng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China.
| | - Jiang-Jiang Qin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Key Laboratory for Molecular Medicine and Chinese Medicine Preparations, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China.
| |
Collapse
|
20
|
Mamun TI, Younus S, Rahman MH. Gastric cancer-Epidemiology, modifiable and non-modifiable risk factors, challenges and opportunities: An updated review. Cancer Treat Res Commun 2024; 41:100845. [PMID: 39357127 DOI: 10.1016/j.ctarc.2024.100845] [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: 05/29/2024] [Revised: 08/27/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024]
Abstract
Gastric cancer represents a significant global health challenge due to its high mortality and incidence rates, particularly in Eastern Asia, Eastern Europe, and South America. This comprehensive review synthesizes the latest epidemiological data and explores both modifiable and non-modifiable risk factors associated with gastric cancer, aiming to delineate the multifactorial etiology of this disease. Modifiable risk factors include Helicobacter pylori infection, obesity, dietary habits, smoking and alcohol consumption, whereas nonmodifiable factors comprise genetic predispositions, age, family history and male gender. The interplay of these factors significantly impacts the risk and progression of gastric cancer, suggesting potential preventive strategies. The challenges in treating gastric cancer are considerable, largely because of the late-stage diagnosis and the heterogeneity of the disease, which complicate effective treatment regimens. Current treatment strategies involve a combination of surgery, chemotherapy, radiotherapy, and targeted therapies. The FLOT regimen (5-FU, Leucovorin, Oxaliplatin and Docetaxel) is now a standard for resectable cases in Europe and the US, showing superior survival and response rates over ECF and ECX regimens. For HER2-positive gastric cancer, trastuzumab combined with chemotherapy improves overall survival, as demonstrated by the ToGA trial. Additionally, immune checkpoint inhibitors like pembrolizumab and nivolumab offer promising results. However, the five-year survival rate remains low, underscoring the urgency for improved therapeutic approaches. Recent advancements in molecular biology and cancer genomics have begun to pave the way for personalized medicine in gastric cancer care, focusing on molecular targeted therapies and immunotherapy. This review also highlights the critical need for better screening methods that could facilitate early detection and treatment, potentially improving the prognosis. By integrating epidemiological insights with new therapeutic strategies, this article aims to thoroughly understand of gastric cancer's dynamics and outline a framework for future research and clinical management, advocating for a multidisciplinary approach to tackle this formidable disease.
Collapse
Affiliation(s)
- Tajul Islam Mamun
- Department of Epidemiology and Public Health, Sylhet Agricultural University, Sylhet 3100, Bangladesh.
| | - Sabrina Younus
- Department of Pharmacy, University of Chittagong, Chattogram 4331, Bangladesh
| | - Md Hashibur Rahman
- Department of Physiology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| |
Collapse
|
21
|
Akhtar S, Ahmad F, Alam M, Ansari AW, Uddin S, Steinhoff M, Buddenkotte J, Ahmad A, Datsi A. Interleukin-31: The Inflammatory Cytokine Connecting Pruritus and Cancer. FRONT BIOSCI-LANDMRK 2024; 29:312. [PMID: 39344323 DOI: 10.31083/j.fbl2909312] [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: 05/22/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 10/01/2024]
Abstract
Interleukin 31 (IL-31) is a proinflammatory cytokine, mainly secreted by Type II helper T cells. It signals through a heterodimeric receptor complex composed of IL-31 receptor α and oncostatin-M receptor β chain. The hallmark feature of IL-31, in its pathological role, is its ability to induce pruritus in mammals. Pruritus is a common symptom and major reason of morbidity in cancer patients, compromising their quality of life. Although, IL-31 is differentially expressed in different tumor types and could promote or inhibit cancer progression, high expression of IL-31 is a contributing factor to advanced stage tumor and severity of pruritus. The simultaneous existence of pruritus and cancer could either result from the aberrations in common proteins that co-exist in both cancer and pruritus or the therapeutic treatment of cancer could indirectly induce pruritus. Although the biology of IL-31 has predominantly been described in skin diseases such as atopic dermatitis and other inflammatory diseases, the precise role of IL-31 in the tumor biology of different cancer types remains elusive. Herein, we summarize the current understanding on the role of this cytokine in the pathogenesis of different cancers.
Collapse
Affiliation(s)
- Sabah Akhtar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, 3050 Doha, Qatar
| | - Fareed Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, 3050 Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, 3050 Doha, Qatar
| | - Majid Alam
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, 3050 Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, 3050 Doha, Qatar
| | - Abdul Wahid Ansari
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, 3050 Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, 3050 Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, 3050 Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, 3050 Doha, Qatar
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, 3050 Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, 3050 Doha, Qatar
- Department of Dermatology and Venereology, Hamad Medical Corporation, 3050 Doha, Qatar
| | - Joerg Buddenkotte
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, 3050 Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, 3050 Doha, Qatar
- Department of Dermatology and Venereology, Hamad Medical Corporation, 3050 Doha, Qatar
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, 3050 Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, 3050 Doha, Qatar
| | - Angeliki Datsi
- Institute of Transplantation Diagnostics and Cell Therapeutics, University Hospital Dusseldorf, 40225 Dusseldorf, Germany
| |
Collapse
|
22
|
Botsen D, Chabaud S, Perrier H, Ammarguellat H, Jestin-Le-Tallec V, Olesinski J, Toullec C, Aparicio T, Ben Abdelghani M, Borg C, Bouche O, Coutzac C, Devaud H, Di Fiore F, Dubreuil O, Evesque L, Huguenin B, Muller M, Poureau PG, Oularue E, Tougeron D, Zaanan A, Ammari S, De Sousa Carvalho N, Decazes P, De La Fouchardiere C. Trifluridine/tipiracil + oxaliplatin ± nivolumab vs FOLFOX ± nivolumab in HER2 negative advanced oesogastric adenocarcinoma: The PRODIGE73-UCGI40-LOGICAN trial. Dig Liver Dis 2024; 56:1281-1287. [PMID: 38762353 DOI: 10.1016/j.dld.2024.04.032] [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: 04/11/2024] [Accepted: 04/29/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Trifluridine/tipiracil (FTD/TPI) is approved in third-line treatment of patients with advanced/metastatic gastric and gastroesophageal junction adenocarcinomas (aGA/GEJA). The association of oxaliplatin with FTD/TPI is promising and the combination of FTD/TPI + oxaliplatin + nivolumab has shown a predictable and manageable safety profile. AIMS The aim is to evaluate the efficacy and safety of FTD/TPI plus oxaliplatin with or without nivolumab in patients, with HER2 negative aGA/GEJA, unfit for triplet chemotherapy (TFOX/mFLOT regimen), in the first-line metastatic setting in comparison with the standard of care FOLFOX with or without nivolumab. METHODS This study is a prospective randomised, open label, comparative, multicentre, phase II trial designed to include 118 patients. The primary objective is to evaluate the superiority of FTD/TPI plus oxaliplatin with or without nivolumab over FOLFOX regimen with or without nivolumab in terms of PFS in a population of patients non candidate for triplet chemotherapy. Nivolumab will be used for patients whose tumour express PD-L1 with a CPS score ≥5. DISCUSSION PRODIGE73-UCGI40-LOGICAN study will provide efficacy and safety data on the association of FTD/TPI plus oxaliplatin with or without nivolumab versus FOLFOX regimen with or without nivolumab in first-line palliative setting, in patients with aGA/GEJA (NCT05476796).
Collapse
Affiliation(s)
- Damien Botsen
- Medical Oncology Department, Godinot Cancer Institute, 1 avenue General Koenig, Reims 51100, France
| | - Sylvie Chabaud
- Department of Clinical Research and Innovation, Centre Léon Bérard, Lyon, France
| | - Hervé Perrier
- Medical Oncology Department, Saint-Joseph Hospital, Marseille, France
| | | | | | - Jonathan Olesinski
- Gastroenterology Department, Villefranche Sur Saone North West Hospital, Gleize, France
| | - Clémence Toullec
- Medical Oncology Department, Sainte Catherine Institute, Avignon, France
| | - Thomas Aparicio
- Gastroenterology Department, Saint Louis Hospital, Paris, France
| | | | - Christophe Borg
- Medical Oncology Department, Hopital Nord Franche Comté, Montbeliard France
| | - Olivier Bouche
- Hepato-Gastroenterology and Digestive Oncology Department, CHU, Reims, France
| | - Clélia Coutzac
- Medical Oncology Department, Centre Léon Bérard, Lyon France
| | - Hervé Devaud
- Medical Oncology Department, Centre Jean Perrin, Clermont Ferrand, France
| | - Frédéric Di Fiore
- Hepato-Gastroenterology Department, Charles Nicolles Hospital, Rouen, France
| | - Olivier Dubreuil
- Oncology Department, GH Diaconesses - Croix St Simon, Paris, France
| | - Ludovic Evesque
- Medical Oncology Department, Centre Antoine Lacassagne, Nice, France
| | - Bruno Huguenin
- Hepato-Gastroenterology Department, Private Hospital Arras Les Bonnettes, Arras, France
| | - Marie Muller
- Gastroenterology and Hepatology Department, Nancy University Hospital, Vandoeuvre Les Nancy, France
| | | | - Emilie Oularue
- Medical Oncology Department, Institut Mutualiste Montsouris, Paris, France
| | - David Tougeron
- Hepato-Gastroenterology Department, Poitiers University Hospital, Poitiers, France
| | - Aziz Zaanan
- Hepato-Gastroenterology and Digestive Oncology Department, Hopital Européen Georges Pompidou, Paris, France
| | - Samy Ammari
- Biomaps, UMR1281 INSERM, CEA, CNRS, University of Paris-Saclay, 94800 Villejuif, France; Department of Imaging, Gustave Roussy Cancer Campus, University of Paris-Saclay, 94800 Villejuif, France
| | | | - Pierre Decazes
- Department of Nuclear Medicine, Henri Becquerel Cancer Center, 76000 Rouen, France; QuantIF-LITIS (EA[Equipe d'Accueil] 4108), Faculty of Medicine, University of Rouen, 76000 Rouen, France
| | | |
Collapse
|
23
|
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
|
24
|
Han Y, Chang Y, Wang J, Li N, Yu Y, Yang Z, Lv W, Liu W, Yin J, Wu J. A study predicting long-term survival capacity in postoperative advanced gastric cancer patients based on MAOA and subcutaneous muscle fat characteristics. World J Surg Oncol 2024; 22:184. [PMID: 39010072 PMCID: PMC11251287 DOI: 10.1186/s12957-024-03466-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/10/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND The prognosis of advanced gastric cancer (AGC) is relatively poor, and long-term survival depends on timely intervention. Currently, predicting survival rates remains a hot topic. The application of radiomics and immunohistochemistry-related techniques in cancer research is increasingly widespread. However, their integration for predicting long-term survival in AGC patients has not been fully explored. METHODS We Collected 150 patients diagnosed with AGC at the Affiliated Zhongshan Hospital of Dalian University who underwent radical surgery between 2015 and 2019. Following strict inclusion and exclusion criteria, 90 patients were included in the analysis. We Collected postoperative pathological specimens from enrolled patients, analyzed the expression levels of MAOA using immunohistochemical techniques, and quantified these levels as the MAOAHScore. Obtained plain abdominal CT images from patients, delineated the region of interest at the L3 vertebral body level, and extracted radiomics features. Lasso Cox regression was used to select significant features to establish a radionics risk score, convert it into a categorical variable named risk, and use Cox regression to identify independent predictive factors for constructing a clinical prediction model. ROC, DCA, and calibration curves validated the model's performance. RESULTS The enrolled patients had an average age of 65.71 years, including 70 males and 20 females. Multivariate Cox regression analysis revealed that risk (P = 0.001, HR = 3.303), MAOAHScore (P = 0.043, HR = 2.055), and TNM stage (P = 0.047, HR = 2.273) emerged as independent prognostic risk factors for 3-year overall survival (OS) and The Similar results were found in the analysis of 3-year disease-specific survival (DSS). The nomogram developed could predict 3-year OS and DSS rates, with areas under the ROC curve (AUCs) of 0.81 and 0.797, respectively. Joint calibration and decision curve analyses (DCA) confirmed the nomogram's good predictive performance and clinical utility. CONCLUSION Integrating immunohistochemistry and muscle fat features provides a more accurate prediction of long-term survival in gastric cancer patients. This study offers new perspectives and methods for a deeper understanding of survival prediction in AGC.
Collapse
Grants
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
- No. 243, 2021 Dalian Deng Feng Program
Collapse
Affiliation(s)
- Yubo Han
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Yaoyuan Chang
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Jiaqi Wang
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Nanbo Li
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Yang Yu
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Zhengbo Yang
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Weipeng Lv
- Department of Pathology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Wenfei Liu
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Jiajun Yin
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China.
| | - Ju Wu
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China.
| |
Collapse
|
25
|
Morgagni P, Bencivenga M, Carneiro F, Cascinu S, Derks S, Di Bartolomeo M, Donohoe C, Eveno C, Gisbertz S, Grimminger P, Gockel I, Grabsch H, Kassab P, Langer R, Lonardi S, Maltoni M, Markar S, Moehler M, Marrelli D, Mazzei MA, Melisi D, Milandri C, Moenig PS, Mostert B, Mura G, Polkowski W, Reynolds J, Saragoni L, Van Berge Henegouwen MI, Van Hillegersberg R, Vieth M, Verlato G, Torroni L, Wijnhoven B, Tiberio GAM, Yang HK, Roviello F, de Manzoni G. International consensus on the management of metastatic gastric cancer: step by step in the foggy landscape : Bertinoro Workshop, November 2022. Gastric Cancer 2024; 27:649-671. [PMID: 38634954 PMCID: PMC11193703 DOI: 10.1007/s10120-024-01479-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 02/05/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Many gastric cancer patients in Western countries are diagnosed as metastatic with a median overall survival of less than twelve months using standard chemotherapy. Innovative treatments, like targeted therapy or immunotherapy, have recently proved to ameliorate prognosis, but a general agreement on managing oligometastatic disease has yet to be achieved. An international multi-disciplinary workshop was held in Bertinoro, Italy, in November 2022 to verify whether achieving a consensus on at least some topics was possible. METHODS A two-round Delphi process was carried out, where participants were asked to answer 32 multiple-choice questions about CT, laparoscopic staging and biomarkers, systemic treatment for different localization, role and indication of palliative care. Consensus was established with at least a 67% agreement. RESULTS The assembly agreed to define oligometastases as a "dynamic" disease which either regresses or remains stable in response to systemic treatment. In addition, the definition of oligometastases was restricted to the following sites: para-aortic nodal stations, liver, lung, and peritoneum, excluding bones. In detail, the following conditions should be considered as oligometastases: involvement of para-aortic stations, in particular 16a2 or 16b1; up to three technically resectable liver metastases; three unilateral or two bilateral lung metastases; peritoneal carcinomatosis with PCI ≤ 6. No consensus was achieved on how to classify positive cytology, which was considered as oligometastatic by 55% of participants only if converted to negative after chemotherapy. CONCLUSION As assessed at the time of diagnosis, surgical treatment of oligometastases should aim at R0 curativity on the entire disease volume, including both the primary tumor and its metastases. Conversion surgery was defined as surgery on the residual volume of disease, which was initially not resectable for technical and/or oncological reasons but nevertheless responded to first-line treatment.
Collapse
Affiliation(s)
- Paolo Morgagni
- Department of General Surgery, Morgagni-Pierantoni Hospital, Forlì, Italy
| | - Maria Bencivenga
- General and Upper GI Surgery, Department of Surgery, University Hospital Verona, University of Verona, Verona, Italy.
| | - Fatima Carneiro
- Department of Pathology, Centro Hospitalar de São João, Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), Porto, Portugal
| | - Stefano Cascinu
- Department of Medical Oncology, Comprehensive Cancer Center, Università Vita-Salute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Sarah Derks
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Maria Di Bartolomeo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Claire Donohoe
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Clarisse Eveno
- Department of Digestive and Oncologic Surgery, Claude Huriez University Hospital, Centre Hospitalier Universitaire (CHU) Lille, Université de Lille, Lille, France
| | - Suzanne Gisbertz
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Peter Grimminger
- Department of General, Visceral and Transplant Surgery, University Medical Center, University of Mainz, Mainz, Germany
| | - Ines Gockel
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Heike Grabsch
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Paulo Kassab
- Gastric Surgery Division, BP Gastric Surgery Department, Santa Casa Medical School, São Paulo, Brazil
| | - Rupert Langer
- Institute of Pathology and Microbiology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040, Linz, Austria
| | - Sara Lonardi
- Istituto Oncologico Veneto IOV-IRCCS, Padua, Italy
| | - Marco Maltoni
- Unit of Palliative Care, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Forlì-Cesena, Italy
| | - Sheraz Markar
- Surgical Interventional Trials Unit, University of Oxford, Oxford, UK
| | - Markus Moehler
- Department of Medicine, Johannes-Gutenberg University Clinic, Mainz, Germany
| | - Daniele Marrelli
- Unit of General Surgery and Surgical Oncology, Department of Medicine Surgery and Neurosciences, University of Siena, 53100, Siena, Italy
| | - Maria Antonietta Mazzei
- Unit of Diagnostic Imaging, Department of Medical, Surgical and Neuro Sciences and of Radiological Sciences, Azienda Ospedaliero-Universitaria Senese, University of Siena, 53100, Siena, Italy
| | - Davide Melisi
- Medical Oncology at the Department of Medicine, University of Verona, Verona, Italy
| | - Carlo Milandri
- Department of Oncology, San Donato Hospital, 52100, Arezzo, Italy
| | | | - Bianca Mostert
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Gianni Mura
- Department of Surgery, San Donato Hospital, Arezzo, Italy
| | - Wojciech Polkowski
- Department of Surgical Oncology, Medical University of Lublin, Radziwiłłowska 13 St, 20-080, Lublin, Poland
| | | | - Luca Saragoni
- Pathology Unit, Santa Maria delle Croci Ravenna Hospital, Ravenna, Italy
| | - Mark I Van Berge Henegouwen
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Michael Vieth
- Institute of Pathology, Klinikum Bayreuth, Bayreuth, Germany
| | - Giuseppe Verlato
- Department of Diagnostics and Public Health, Section of Epidemiology and Medical Statistics, University of Verona, Verona, Italy
| | - Lorena Torroni
- Department of Diagnostics and Public Health, Section of Epidemiology and Medical Statistics, University of Verona, Verona, Italy
| | - Bas Wijnhoven
- Department of Surgery, Erasmus MC-University Medical Centre Rotterdam, Rotterdam, Netherlands
| | | | - Han-Kwang Yang
- Surgical Department, SNUH National Cancer Center, Seoul, Korea
| | - Franco Roviello
- Unit of General Surgery and Surgical Oncology, Department of Medicine Surgery and Neurosciences, University of Siena, 53100, Siena, Italy
| | - Giovanni de Manzoni
- General and Upper GI Surgery, Department of Surgery, University Hospital Verona, University of Verona, Verona, Italy
| |
Collapse
|
26
|
Kutlu Y, Dae SA, Yilmaz F, Erdem D, Sendur MAN, Akbas S, Senocak Tasci E, Bas O, Dane F, Sakin A, Kaya AO, Aykan MB, Ergun Y, Biter S, Disel U, Korkmaz M, Selcukbiricik F, Kose F, Olmez OF, Bilici A, Demir G, Yalcin S. Real-World Efficacy and Safety of First-Line Nivolumab Plus Chemotherapy in Patients with Advanced Gastric, Gastroesophageal Junction, and Esophageal Adenocarcinoma: A Nationwide Observational Turkish Oncology Group (TOG) Study. Cancers (Basel) 2024; 16:2251. [PMID: 38927957 PMCID: PMC11202017 DOI: 10.3390/cancers16122251] [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: 05/04/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
Based on the CheckMate 649 trial, nivolumab plus chemotherapy is the recommended first-line treatment for HER2-negative unresectable advanced or metastatic gastric, gastroesophageal junction (GEJ), or esophageal adenocarcinoma. This nationwide, multicenter, retrospective study evaluated the real-world effectiveness of this regimen in Turkish patients and identified subgroups that may experience superior outcomes. Conducted across 16 oncology centers in Turkey, this study retrospectively reviewed the clinical charts of adult patients diagnosed with HER2-negative unresectable advanced or metastatic gastric, GEJ, or esophageal adenocarcinoma from 2016 to 2023. This study included 111 patients (54 women, 57 men) with a median age of 58 years. The median progression-free survival (PFS) and overall survival (OS) were 11.7 months and 18.2 months, respectively, whereas the objective response rate (ORR) was 70.3%. Multivariable analyses revealed that previous curative surgery was a favorable independent prognostic factor for both PFS and OS. Conversely, an Eastern Cooperative Oncology Group performance status of 2 emerged as an adverse independent prognostic factor for OS. The safety profile of nivolumab plus chemotherapy was found to be manageable. Our findings support the use of nivolumab plus chemotherapy for the first-line treatment of Turkish patients with HER2-negative unresectable advanced or metastatic gastric, GEJ, or esophageal adenocarcinoma. Patient selection based on clinical characteristics is crucial for optimizing treatment outcomes.
Collapse
Affiliation(s)
- Yasin Kutlu
- Department of Medical Oncology, Faculty of Medicine, Medipol University, Istanbul 34083, Turkey
| | - Shute Ailia Dae
- Department of Medical Oncology, Faculty of Medicine, Baskent University, Adana 01140, Turkey
| | - Feride Yilmaz
- Department of Medical Oncology, Faculty of Medicine, Hacettepe University, Ankara 06800, Turkey
| | - Dilek Erdem
- Department of Medical Oncology, VM Medical Park Samsun Hospital, Samsun 55200, Turkey
| | | | - Sinem Akbas
- Department of Medical Oncology, Faculty of Medicine, Koc University, Istanbul 34460, Turkey
| | - Elif Senocak Tasci
- Department of Medical Oncology, Acibadem Atakent Hospital, Istanbul 34303, Turkey
| | - Onur Bas
- Department of Medical Oncology, Faculty of Medicine, Hacettepe University, Ankara 06800, Turkey
| | - Faysal Dane
- Department of Medical Oncology, Acibadem Altunizade Hospital, Istanbul 34662, Turkey
| | - Abdullah Sakin
- Department of Medical Oncology, Medipol University Bahcelievler Hospital, Istanbul 34196, Turkey
| | - Ali Osman Kaya
- Department of Medical Oncology, Medicana International Hospital, Istanbul 34520, Turkey
| | - Musa Baris Aykan
- Department of Medical Oncology, Gulhane Training and Research Hospital, Ankara 06010, Turkey
| | - Yakup Ergun
- Department of Medical Oncology, Antalya City Hospital, Antalya 07200, Turkey
| | - Sedat Biter
- Department of Medical Oncology, Faculty of Medicine, Cukurova University, Adana 01330, Turkey
| | - Umut Disel
- Department of Medical Oncology, Acibadem Adana Hospital, Adana 01130, Turkey
| | - Mustafa Korkmaz
- Department of Medical Oncology, Tokat State Hospital, Tokat 60100, Turkey
| | - Fatih Selcukbiricik
- Department of Medical Oncology, Faculty of Medicine, Koc University, Istanbul 34460, Turkey
| | - Fatih Kose
- Department of Medical Oncology, Faculty of Medicine, Baskent University, Adana 01140, Turkey
| | - Omer Fatih Olmez
- Department of Medical Oncology, Faculty of Medicine, Medipol University, Istanbul 34083, Turkey
| | - Ahmet Bilici
- Department of Medical Oncology, Faculty of Medicine, Medipol University, Istanbul 34083, Turkey
| | - Gokhan Demir
- Department of Medical Oncology, Acibadem Maslak Hospital, Istanbul 34398, Turkey
| | - Suayib Yalcin
- Department of Medical Oncology, Faculty of Medicine, Hacettepe University, Ankara 06800, Turkey
| |
Collapse
|
27
|
Zhang LK, Zheng HL, Zheng HH, Ma YB, Lin JX, Xu BB, Xue Z, Zheng ZW, Zheng CH, Huang CM, Xie JW. Effects of tumor marker regression load score on long-term prognosis of gastric cancer patients undergoing radical surgery after neoadjuvant chemotherapy. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108367. [PMID: 38718701 DOI: 10.1016/j.ejso.2024.108367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/14/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND The effects of the dynamics of serum tumor markers (CA72-4, CEA, CA19-9, CA125 and AFP) before and after neoadjuvant chemotherapy (NACT) on the prognosis of gastric cancer(GC) patients remain unclear. METHODS The training set contained 334 GC patients from Fujian Medical University Union Hospital (FJMUUH) and 113 GC patients in Qinghai University Affiliated Hospital (QhUAH) were used as an external validation set. Tumor marker regression load (ΔTMRL) indicator, including ΔCA72-4, ΔCEA, ΔCA19-9, ΔCA125, and ΔAFP, is defined as [(postNACT marker- preNACT marker)/preNACT marker]. Tumor marker regression load score (TMRLS) consists of ΔCA72-4, ΔCEA and ΔCA125. The predictive performance of the nomogram-TMRLS was evaluated using the area under the receiver operating characteristic(ROC) curve(AUC), decision curve analysis(DCA), and C-index. RESULTS Patients from FJMUUH were divided into two groups, TMRLS-low and TMRLS-high, determined by R package maxstat. Survival analysis revealed a higher 3-year overall survival(OS) in the TMRLS-low than in the TMRLS-high group. The TMRLS-high group who received postoperative adjuvant chemotherapy(AC) showed a significantly higher 3-year OS rate than those who did not. Multivariate COX regression analysis indicated that TMRLS was an independent prognostic factor for OS. A nomogram for predicting OS based on TMRLS showed a significantly higher C-index and AUC than the ypTNM stage. The above results were also found in the QhUAH external validation cohort. CONCLUSION TMRLS is a novel independent prognostic factor for GC who underwent NACT and a radical gastrectomy. Furthermore, the TMRLS-high group, who received postoperative AC, may achieve better survival outcomes. Notably, the predictive performance of the nomogram-TMRLS significantly outperformed that of the ypTNM stage.
Collapse
Affiliation(s)
- Ling-Kang Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Hua-Long Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Hong-Hong Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Yu-Bin Ma
- Department of Gastrointestinal Surgery, Qinghai University Affiliated Hospital, Xining, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Bin-Bin Xu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Zhen Xue
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Zhi-Wei Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350001, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350001, China.
| |
Collapse
|
28
|
van der Sluis K, Guchelaar NAD, Triemstra L, Mathijssen RHJ, Ruurda JP, Wijnhoven BPL, van Sandick JW. Staging laparoscopy in gastric cancer patients: From a Dutch nationwide Delphi consensus towards a standardized protocol. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108278. [PMID: 38531232 DOI: 10.1016/j.ejso.2024.108278] [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: 01/23/2024] [Revised: 03/08/2024] [Accepted: 03/16/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND Staging laparoscopy is a common diagnostic tool in gastric cancer, but its performance varies widely. The aim of this study was to gain Dutch nationwide consensus regarding the indications for and execution of staging laparoscopy in patients with gastric cancer. METHODS All surgeons in the Netherlands specialized in gastric cancer surgery (n = 52) were asked to participate in a Delphi consensus study. The study involved an initial questionnaire with a 3-point Likert scale, an online consensus meeting, and a second questionnaire using a 2-point Likert scale (agree/disagree). Consensus was defined as 70% or more agreement among participants. RESULTS In total, 45 experts completed both questionnaires (87% response rate). Consensus was reached on the indication to perform staging laparoscopy in cT3-4 or cN + or diffuse-type gastric cancer, including Siewert type III oesophagogastric junctional cancer. The experts agreed that if preoperative scans suggest infiltration of surrounding organs (cT4), the tumour's resectability should explicitly be investigated. Consensus was also reached for a systematic peritoneal cavity inspection according to Sugarbaker's Peritoneal Cancer Index (PCI) score. All regions should be inspected routinely, although the omental bursa may be inspected on indication. Aspiration of ascites or peritoneal washing should be performed for cytology. The experts agreed that restaging laparoscopy should be performed before resection in case of progressive disease on preoperative imaging. Without progression, global inspection was considered sufficient. CONCLUSIONS The results of this Dutch nationwide Delphi consensus study exposed the variability of performing staging laparoscopy in patients with gastric cancer and provided the concept for a standardized protocol.
Collapse
Affiliation(s)
| | - Niels A D Guchelaar
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| | - Lianne Triemstra
- Department of Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Jelle P Ruurda
- Department of Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Bas P L Wijnhoven
- Department of Surgery, Erasmus Medical Center, Rotterdam, the Netherlands
| | | |
Collapse
|
29
|
Yuan N, Zhang Y, Lv K, Liu Y, Yang A, Hu P, Yu H, Han X, Guo X, Li J, Wang T, Lei B, Ma G. HCA-DAN: hierarchical class-aware domain adaptive network for gastric tumor segmentation in 3D CT images. Cancer Imaging 2024; 24:63. [PMID: 38773670 PMCID: PMC11107051 DOI: 10.1186/s40644-024-00711-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/11/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND Accurate segmentation of gastric tumors from CT scans provides useful image information for guiding the diagnosis and treatment of gastric cancer. However, automated gastric tumor segmentation from 3D CT images faces several challenges. The large variation of anisotropic spatial resolution limits the ability of 3D convolutional neural networks (CNNs) to learn features from different views. The background texture of gastric tumor is complex, and its size, shape and intensity distribution are highly variable, which makes it more difficult for deep learning methods to capture the boundary. In particular, while multi-center datasets increase sample size and representation ability, they suffer from inter-center heterogeneity. METHODS In this study, we propose a new cross-center 3D tumor segmentation method named Hierarchical Class-Aware Domain Adaptive Network (HCA-DAN), which includes a new 3D neural network that efficiently bridges an Anisotropic neural network and a Transformer (AsTr) for extracting multi-scale context features from the CT images with anisotropic resolution, and a hierarchical class-aware domain alignment (HCADA) module for adaptively aligning multi-scale context features across two domains by integrating a class attention map with class-specific information. We evaluate the proposed method on an in-house CT image dataset collected from four medical centers and validate its segmentation performance in both in-center and cross-center test scenarios. RESULTS Our baseline segmentation network (i.e., AsTr) achieves best results compared to other 3D segmentation models, with a mean dice similarity coefficient (DSC) of 59.26%, 55.97%, 48.83% and 67.28% in four in-center test tasks, and with a DSC of 56.42%, 55.94%, 46.54% and 60.62% in four cross-center test tasks. In addition, the proposed cross-center segmentation network (i.e., HCA-DAN) obtains excellent results compared to other unsupervised domain adaptation methods, with a DSC of 58.36%, 56.72%, 49.25%, and 62.20% in four cross-center test tasks. CONCLUSIONS Comprehensive experimental results demonstrate that the proposed method outperforms compared methods on this multi-center database and is promising for routine clinical workflows.
Collapse
Affiliation(s)
- Ning Yuan
- Department of Medical Imaging, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Yongtao Zhang
- School of Biomedical Engineering, Health Science Centers, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Marshall Laboratory of Biomedical Engineering, Shenzhen University, Shenzhen, China
| | - Kuan Lv
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yiyao Liu
- School of Biomedical Engineering, Health Science Centers, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Marshall Laboratory of Biomedical Engineering, Shenzhen University, Shenzhen, China
| | - Aocai Yang
- Department of Radiology, China-Japan Friendship Hospital, No. 2 East Yinghua Road, Chaoyang District, Beijing, 100029, China
| | - Pianpian Hu
- Department of Radiology, China-Japan Friendship Hospital, No. 2 East Yinghua Road, Chaoyang District, Beijing, 100029, China
| | - Hongwei Yu
- Department of Radiology, China-Japan Friendship Hospital, No. 2 East Yinghua Road, Chaoyang District, Beijing, 100029, China
| | - Xiaowei Han
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xing Guo
- Department of Medical Imaging, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Junfeng Li
- Department of Medical Imaging, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Tianfu Wang
- School of Biomedical Engineering, Health Science Centers, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Marshall Laboratory of Biomedical Engineering, Shenzhen University, Shenzhen, China
| | - Baiying Lei
- School of Biomedical Engineering, Health Science Centers, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Marshall Laboratory of Biomedical Engineering, Shenzhen University, Shenzhen, China
- AI Research Center for Medical Image Analysis and Diagnosis, Shenzhen University, Guangdong, China
| | - Guolin Ma
- Department of Radiology, China-Japan Friendship Hospital, No. 2 East Yinghua Road, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
30
|
Chen W, Liu X, Wang H, Dai J, Li C, Hao Y, Jiang D. Exploring the immune escape mechanisms in gastric cancer patients based on the deep AI algorithms and single-cell sequencing analysis. J Cell Mol Med 2024; 28:e18379. [PMID: 38752750 PMCID: PMC11097712 DOI: 10.1111/jcmm.18379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 03/30/2024] [Accepted: 04/08/2024] [Indexed: 05/19/2024] Open
Abstract
Gastric cancer is a prevalent and deadly malignancy, and the response to immunotherapy varies among patients. This study aimed to develop a prognostic model for gastric cancer patients and investigate immune escape mechanisms using deep machine learning and single-cell sequencing analysis. Data from public databases were analysed, and a prediction model was constructed using 101 algorithms. The high-AIDPS group, characterized by increased AIDPS expression, exhibited worse survival, genomic variations and immune cell infiltration. These patients also showed immunotherapy tolerance. Treatment strategies targeting the high-AIDPS group identified three potential drugs. Additionally, distinct cluster groups and upregulated AIDPS-associated genes were observed in gastric adenocarcinoma cell lines. Inhibition of GHRL expression suppressed cancer cell activity, inhibited M2 polarization in macrophages and reduced invasiveness. Overall, AIDPS plays a critical role in gastric cancer prognosis, genomic variations, immune cell infiltration and immunotherapy response, and targeting GHRL expression holds promise for personalized treatment. These findings contribute to improved clinical management in gastric cancer.
Collapse
Affiliation(s)
- Wenli Chen
- Department of General SurgeryThe Affiliated Bozhou Hospital of Anhui Medical UniversityBozhouAnhuiChina
| | - Xiaohui Liu
- Department of Nursing, Xiangya HospitalCentral South UniversityChangshaChina
| | - Houhong Wang
- Department of General SurgeryThe Affiliated Bozhou Hospital of Anhui Medical UniversityBozhouAnhuiChina
| | - Jingyou Dai
- Department of Pediatric SurgeryThe Affiliated Bozhou Hospital of Anhui Medical UniversityBozhouAnhuiChina
| | - Changquan Li
- Department of General SurgeryThe Affiliated Bozhou Hospital of Anhui Medical UniversityBozhouAnhuiChina
| | - Yanyan Hao
- Department of Articular SurgeryThe Affiliated Bozhou Hospital of Anhui Medical UniversityBozhouAnhuiChina
| | - Dandan Jiang
- The Second Affiliated Hospital, Department of Emergency, Hengyang Medical SchoolUniversity of South ChinaHengyangChina
| |
Collapse
|
31
|
Tao C, Hong W, Yin P, Wu S, Fan L, Lei Z, Yu Y. Nomogram Based on Body Composition and Prognostic Nutritional Index Predicts Survival After Curative Resection of Gastric Cancer. Acad Radiol 2024; 31:1940-1949. [PMID: 37981487 DOI: 10.1016/j.acra.2023.10.057] [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: 09/15/2023] [Revised: 10/17/2023] [Accepted: 10/29/2023] [Indexed: 11/21/2023]
Abstract
RATIONALE AND OBJECTIVES This study aimed to identify independent prognostic factors for gastric cancer (GC) patients after curative resection using quantitative computed tomography (QCT) combined with prognostic nutritional index (PNI), and to develop a nomogram prediction model for individualized prognosis. MATERIALS AND METHODS This study retrospectively analyzed 119 patients with GC who underwent curative resection from January 2016 to March 2018. The patients' preoperative clinical pathological data were recorded, and all patients underwent QCT scans before and after curative resection to obtain QCT parameters: bone mineral density (BMD), skeletal muscle area (SMA), visceral fat area (VFA), subcutaneous fat area (SFA) and CT fat fraction (CTFF), then relative rate of change in each parameter (ΔBMD, ΔSMA, ΔVFA, ΔSFA, ΔCTFF) was calculated after time normalization. Multivariate Cox proportional hazards was used to establish a nomogram model that based on independent prognostic factors. The concordance index (C-index), area under the time-dependent receiver operating characteristic (ROC) curve and clinical decision curve were used to evaluate the predictive performance and clinical benefit of the nomogram model. RESULTS: This study found that ΔCTFF, ΔVFA, ΔBMD and PNI are independent prognostic factors for overall survival (OS) (hazard ratio: 1.034, 0.895, 0.976, 2.951, respectively, all p < 0.05). The established nomogram model could predict the area under the ROC curve of OS at 1, 3 and 5 years as 0.816, 0.815 and 0.881, respectively. The C-index was 0.743 (95% CI, 0.684-0.801), and the decision curve analysis showed that this model has good clinical net benefit. CONCLUSION The nomogram model based on body composition and PNI is reliable in predicting the individualized survival of underwent curative resection for GC patients.
Collapse
Affiliation(s)
- Chao Tao
- Department of Radiology, the First Affiliated Hospital of Wannan Medical College, Wuhu, China (T.C., W.H., P.Y., S.W., Z.L., Y.Y.)
| | - Wei Hong
- Department of Radiology, the First Affiliated Hospital of Wannan Medical College, Wuhu, China (T.C., W.H., P.Y., S.W., Z.L., Y.Y.)
| | - Pengzhan Yin
- Department of Radiology, the First Affiliated Hospital of Wannan Medical College, Wuhu, China (T.C., W.H., P.Y., S.W., Z.L., Y.Y.)
| | - Shujian Wu
- Department of Radiology, the First Affiliated Hospital of Wannan Medical College, Wuhu, China (T.C., W.H., P.Y., S.W., Z.L., Y.Y.)
| | - Lifang Fan
- School of Medical imaging, Wannan Medical College, Wuhu, China (L.F.)
| | - Zihao Lei
- Department of Radiology, the First Affiliated Hospital of Wannan Medical College, Wuhu, China (T.C., W.H., P.Y., S.W., Z.L., Y.Y.)
| | - Yongmei Yu
- Department of Radiology, the First Affiliated Hospital of Wannan Medical College, Wuhu, China (T.C., W.H., P.Y., S.W., Z.L., Y.Y.).
| |
Collapse
|
32
|
Zhang SY, Luo Q, Xiao LR, Yang F, Zhu J, Chen XQ, Yang S. Role and mechanism of NCAPD3 in promoting malignant behaviors in gastric cancer. Front Pharmacol 2024; 15:1341039. [PMID: 38711992 PMCID: PMC11070777 DOI: 10.3389/fphar.2024.1341039] [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: 11/19/2023] [Accepted: 03/30/2024] [Indexed: 05/08/2024] Open
Abstract
Background Gastric cancer (GC) is one of the major malignancies threatening human lives and health. Non-SMC condensin II complex subunit D3 (NCAPD3) plays a crucial role in the occurrence of many diseases. However, its role in GC remains unexplored. Materials and Methods The Cancer Genome Atlas (TCGA) database, clinical samples, and cell lines were used to analyze NCAPD3 expression in GC. NCAPD3 was overexpressed and inhibited by lentiviral vectors and the CRISPR/Cas9 system, respectively. The biological functions of NCAPD3 were investigated in vitro and in vivo. Gene microarray, Gene set enrichment analysis (GSEA) and ingenuity pathway analysis (IPA) were performed to establish the potential mechanisms. Results NCAPD3 was highly expressed in GC and was associated with a poor prognosis. NCAPD3 upregulation significantly promoted the malignant biological behaviors of gastric cancer cell, while NCAPD3 inhibition exerted a opposite effect. NCAPD3 loss can directly inhibit CCND1 and ESR1 expression to downregulate the expression of downstream targets CDK6 and IRS1 and inhibit the proliferation of gastric cancer cells. Moreover, NCAPD3 loss activates IRF7 and DDIT3 to regulate apoptosis in gastric cancer cells. Conclusion Our study revealed that NCAPD3 silencing attenuates malignant phenotypes of GC and that it is a potential target for GC treatment.
Collapse
Affiliation(s)
- Su-Yun Zhang
- Departments of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Qiong Luo
- Departments of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Li-Rong Xiao
- Departments of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Fan Yang
- Departments of Respiratory and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jian Zhu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiang-Qi Chen
- Departments of Respiratory and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Fuzhou, Fujian, China
| | - Sheng Yang
- Departments of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Fuzhou, Fujian, China
| |
Collapse
|
33
|
Degu A, Karimi PN, Opanga SA, Nyamu DG. Drug-related problems among esophageal, gastric and colorectal cancer patients at the National and referral hospital in Kenya. J Oncol Pharm Pract 2024; 30:493-506. [PMID: 37272031 DOI: 10.1177/10781552231178297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
INTRODUCTION Cancer therapy has remarkable potential for drug-related problems due to the high cytotoxicity and narrow therapeutic index of most anti-neoplastic regimens. However, there is a lack of comprehensive studies on drug-related problems in patients with gastrointestinal cancer in Kenya. Therefore, the present study aimed to investigate the prevalence, types and predictors of drug-related problems among gastrointestinal cancer patients at Kenyatta National Hospital. METHODS A cross-sectional study was used to assess the prevalence of drug-related problems among a random sample of 160 esophageal, 103 gastric, and 96 colorectal cancer patients. Data were collected using a researcher-administered questionnaire and data abstraction tool after training the data collectors. Patient-specific details such as socio-demographic features, histological cancer types, cancer stage, comorbidity types, and treatment regimen were recorded after the review of medical records and patient interviews. The potential of drug-related problems was determined as per the standard guidelines. The data were entered and analysed using version 26.0 SPSS statistical software. RESULTS Most esophageal (51.9%), gastric (59.2%), and colorectal (62.5%) cancer patients had a high prevalence of drug-related problems. The need for additional drug therapy and adverse drug reactions were the predominant categories of drug-related problems. Most adverse drug reactions identified had possible categories of causality score, mild severity levels, and definitely preventable types of adverse drug reactions among all gastrointestinal cancer patients. Comorbidity and advanced-stage disease were significant predictors of drug-related problems. CONCLUSIONS Drug-related problems were prevalent among gastrointestinal cancer patients in our setting. Comorbidity and advanced stages of disease were significant predictors of drug-related problems.
Collapse
Affiliation(s)
- Amsalu Degu
- Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, Nairobi, Kenya
- Department of Pharmacy, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
| | - Peter N Karimi
- Department of Pharmacy, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
| | - Sylvia A Opanga
- Department of Pharmacy, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
| | - David G Nyamu
- Department of Pharmacy, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
34
|
Kim S, Lee HH, Song KY, Seo HS. Peritoneal Washing Cytology Positivity in Gastric Cancer: Role of Lymph Node Metastasis as a Risk Factor. J Gastric Cancer 2024; 24:185-198. [PMID: 38575511 PMCID: PMC10995825 DOI: 10.5230/jgc.2024.24.e14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/28/2023] [Accepted: 11/12/2023] [Indexed: 04/06/2024] Open
Abstract
PURPOSE Peritoneal washing cytology (PWC) is a widely used diagnostic tool for detecting peritoneal metastasis of advanced gastric cancer. However, the prognosis of patients with positive PWC remains poor even after gastrectomy, and treatments vary among institutions and eras. In this study, we identified the clinical factors that can help predict cytology-positive (CY(+)) gastric cancer. MATERIALS AND METHODS We retrospectively reviewed the national data of patients with gastric cancer from 2019, as provided by the Information Committee of the Korean Gastric Cancer Association. Of the 13,447 patients with gastric cancer, 3,672 underwent PWC. Based on cytology results, we analyzed the clinicopathological characteristics and assessed the possibility of CY(+) outcomes in relation to T and N stages. RESULTS Of the 3,270 patients who underwent PWC without preoperative chemotherapy, 325 were CY(+), whereas 2,945 were negative. CY(+) was more commonly observed in patients with Borrmann type IV gastric cancer, an undifferentiated histological type, and advanced pathological stages. Multivariate analysis revealed Borrmann type IV (odds ratio [OR], 1.821), tumor invasion to T3-4 (OR, 2.041), and lymph node metastasis (OR, 3.155) as independent predictors of CY(+). Furthermore, for circular tumor location, the N stage emerged as a significant risk factor for CY(+), particularly when the tumor was located on the posterior wall (PW) side. CONCLUSIONS Lymph node metastasis significantly affects CY(+) outcomes, particularly when the tumor is located on the PW side. Therefore, PWC should be considered not only in suspected serosal exposure cases but also in cases of lymph node metastasis.
Collapse
Affiliation(s)
- Sojung Kim
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Han Hong Lee
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kyo Young Song
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ho Seok Seo
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
35
|
Ji J, Zhang X, Yuan S, Liu H, Yang L. Survival impact of gastrectomy and chemotherapy on gastric signet ring-cell carcinoma with different metastatic lesions: A population-based study. Asian J Surg 2024; 47:1769-1775. [PMID: 38302357 DOI: 10.1016/j.asjsur.2024.01.129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/26/2022] [Accepted: 01/19/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND A comprehensive understanding of gastric signet ring cell carcinoma (SRCC) is limited. The aim of our study was to analyze metastatic patterns of gastric SRCC and evaluate impacts of gastrectomy and chemotherapy for metastatic gastric SRCC. METHODS We obtained data of gastric cancer patients between 2010 and 2017 in the Surveillance, Epidemiology, and End Results database. Chi-square tests were used to compare data significance. Kaplan-Meier, Cox proportional hazards regression and Fine-Gray competing risk analysis were used to analyze the difference in the overall survival (OS) and cancer-specific survival (CSS). Propensity-score matching was used to adjust numerical difference. RESULTS Among 36,459 eligible gastric cancer patients, 6264 (17.2 %) were SRCC patients. Bone metastasis was more common in SRCC patients than in non-SRCC patients. The multivariate analysis showed that chemotherapy (HR = 0.30, 95 %CI = 0.27-0.33, p < 0.01) and gastrectomy (HR = 0.51, 95 %CI = 0.45-0.59, p < 0.01) were protective prognostic factors in certain stage Ⅳ SRCC patients. For the effect of gastrectomy, survival benefits could be found in patients with liver metastasis. The gastrectomy was not associated with improved OS in patients with lung or multiple metastases. In subgroup analysis, SRCC patients with metastasis who received gastrectomy and chemotherapy (HR = 0.17, p < 0.01; HR = 0.03, p < 0.01) had a better OS and CSS than those who had chemotherapy only (HR = 0.30, p < 0.01; HR = 0.18, p < 0.01). CONCLUSION Our study analyzed the unique metastatic patterns of gastric SRCC and recommended chemotherapy as the first choice in metastatic SRCC. For patients with liver metastasis, gastrectomy plus chemotherapy can be considered.
Collapse
Affiliation(s)
- Jiali Ji
- Department of Medical Oncology, Affiliate Tumor Hospital of Nantong University, Nantong, China.
| | - Xunlei Zhang
- Department of Medical Oncology, Affiliate Tumor Hospital of Nantong University, Nantong, China.
| | - Shushu Yuan
- Department of Medical Oncology, Affiliate Tumor Hospital of Nantong University, Nantong, China.
| | - Hong Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, China.
| | - Lei Yang
- Department of Medical Oncology, Affiliate Tumor Hospital of Nantong University, Nantong, China.
| |
Collapse
|
36
|
van Hootegem SJM, Chmelo J, van der Sluis PC, Lagarde SM, Phillips AW, Wijnhoven BPL. The yield of diagnostic laparoscopy with peritoneal lavage in gastric adenocarcinoma: A retrospective cohort study. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108233. [PMID: 38428107 DOI: 10.1016/j.ejso.2024.108233] [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: 10/20/2023] [Revised: 01/24/2024] [Accepted: 02/25/2024] [Indexed: 03/03/2024]
Abstract
INTRODUCTION Diagnostic laparoscopy (DL) with peritoneal lavage has been adopted as a standard staging procedure for patients with gastric cancer (GC). Evaluation of the value of DL is important given ongoing improvements in diagnostic imaging and treatment. As contemporary data from European centres are sparse, this retrospective cohort study aimed to assess the yield of DL in patients with potentially curable gastric cancer, and to identify predictive factors for peritoneal metastases. METHODS Patients with adenocarcinoma of the stomach, treated between January 2016 and December 2018, were identified from institutional databases of two high volume European Upper-GI centres. Patients who underwent a DL with peritoneal lavage for potentially curable disease after clinical staging with imaging (cT1-4N0-3M0) were included. The primary outcome was the proportion of patients with a positive DL, defined as macroscopic metastatic disease, positive peritoneal cytology washings (PC+) or locally irresectable disease. RESULTS Some 80 of 327 included patients (24.5%) had a positive DL, excluding these patients from neoadjuvant treatment (66 of 327; 20.2%) and/or surgical resection (76 of 327; 23.2%). In 34 of 327 patients (10.3%), macroscopic metastatic disease was seen, with peritoneal deposits in 30 of these patients. Only 16 of 30 patients with peritoneal disease had positive cytology. Some 41 of 327 patients (12.5%) that underwent DL had PC+ in the absence of macroscopic metastases and five patients (1.5%) had an irresectable primary tumour. Diffuse type carcinoma had the highest risk of peritoneal dissemination, irrespective of cT and cN categories. CONCLUSION The diagnostic yield of staging laparoscopy is high, changing the management in approximately one quarter of patients. DL should be considered in patients with diffuse type carcinoma irrespective of cT and cN categories.
Collapse
Affiliation(s)
- S J M van Hootegem
- Department of Surgery, Erasmus MC University, Rotterdam, the Netherlands.
| | - J Chmelo
- Northern Oesophagogastric Unit, Royal Victoria Infirmary, Newcastle-Upon-Tyne, UK
| | - P C van der Sluis
- Department of Surgery, Erasmus MC University, Rotterdam, the Netherlands
| | - S M Lagarde
- Department of Surgery, Erasmus MC University, Rotterdam, the Netherlands
| | - A W Phillips
- Northern Oesophagogastric Unit, Royal Victoria Infirmary, Newcastle-Upon-Tyne, UK; School of Medical Education, Newcastle University, UK
| | - B P L Wijnhoven
- Department of Surgery, Erasmus MC University, Rotterdam, the Netherlands
| |
Collapse
|
37
|
Wu R, Qin K, Fang Y, Xu Y, Zhang H, Li W, Luo X, Han Z, Liu S, Li Q. Application of the convolution neural network in determining the depth of invasion of gastrointestinal cancer: a systematic review and meta-analysis. J Gastrointest Surg 2024; 28:538-547. [PMID: 38583908 DOI: 10.1016/j.gassur.2023.12.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/16/2023] [Accepted: 12/30/2023] [Indexed: 04/09/2024]
Abstract
BACKGROUND With the development of endoscopic technology, endoscopic submucosal dissection (ESD) has been widely used in the treatment of gastrointestinal tumors. It is necessary to evaluate the depth of tumor invasion before the application of ESD. The convolution neural network (CNN) is a type of artificial intelligence that has the potential to assist in the classification of the depth of invasion in endoscopic images. This meta-analysis aimed to evaluate the performance of CNN in determining the depth of invasion of gastrointestinal tumors. METHODS A search on PubMed, Web of Science, and SinoMed was performed to collect the original publications about the use of CNN in determining the depth of invasion of gastrointestinal neoplasms. Pooled sensitivity and specificity were calculated using an exact binominal rendition of the bivariate mixed-effects regression model. I2 was used for the evaluation of heterogeneity. RESULTS A total of 17 articles were included; the pooled sensitivity was 84% (95% CI, 0.81-0.88), specificity was 91% (95% CI, 0.85-0.94), and the area under the curve (AUC) was 0.93 (95% CI, 0.90-0.95). The performance of CNN was significantly better than that of endoscopists (AUC: 0.93 vs 0.83, respectively; P = .0005). CONCLUSION Our review revealed that CNN is one of the most effective methods of endoscopy to evaluate the depth of invasion of early gastrointestinal tumors, which has the potential to work as a remarkable tool for clinical endoscopists to make decisions on whether the lesion is feasible for endoscopic treatment.
Collapse
Affiliation(s)
- Ruo Wu
- Nanfang Hospital (The First School of Clinical Medicine), Southern Medical University, Guangzhou, Guangdong, China
| | - Kaiwen Qin
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuxin Fang
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuyuan Xu
- Department of Hepatology Unit and Infectious Diseases, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Haonan Zhang
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenhua Li
- Nanfang Hospital (The First School of Clinical Medicine), Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaobei Luo
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zelong Han
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Side Liu
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Pazhou Lab, Guangzhou, Guangdong, China
| | - Qingyuan Li
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
38
|
Liu D, Quan H, Ma M, Zhou H, Yang X, Wu Z, Luo J, Xiao H, Xiao Y. Nomogram to predict overall survival of patients receiving radical gastrectomy and incomplete peri-operative adjuvant chemotherapy for stage II/III gastric cancer: a retrospective bi-center cohort study. BMC Cancer 2024; 24:344. [PMID: 38500085 PMCID: PMC10946121 DOI: 10.1186/s12885-024-12103-1] [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: 03/29/2023] [Accepted: 03/11/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND To establish a nomogram to predict the probability of survival of patients with stage II/III gastric cancer (GC) who received incomplete peri-operative adjuvant chemotherapy (PAC). METHODS The medical records of stage II/III GC patients who received curative resection and 1 to 5 cycles of PAC from two tertiary hospitals were retrospectively reviewed. Patients were randomly classified into either a training group or validation group at a ratio of 7:3. The nomogram was constructed based on various prognostic factors using Cox regression analysis in the training cohort, and was validated by the validation group. Concordance index and calibration curves were used to evaluate the discrimination and calibration of the nomogram. Additionally, decision curve analysis (DCA) was used to compare the net clinical benefits of the nomogram and eighth version of TNM staging system. RESULTS A total of 1,070 consecutive patients were included and 749 patients were enrolled into the training group. Lower body mass index (< 18.5 kg/m2), total gastrectomy, stage III disease and fewer cycles of PAC were identified to be independent predictors for poorer survival. The area under the curve (AUC) values of receiver operating characteristics (ROC) curve predicting 5-year survival probabilities and C-index were 0.768 and 0.742, 0.700 (95%CI: 0.674-0.726) and 0.689 (95%CI: 0.646-0.732) in the training and validation groups, respectively. The calibration curves in the validation cohort showed good agreement between the prediction and observation of 1-, 3- and 5-year survival probabilities. Furthermore, DCA showed that our model has a better net benefit than that of TNM staging system. CONCLUSIONS The findings emphasize the value of completing PAC. The nomogram which was established to predict survival probability in patients with stage II/III GC receiving radical gastrectomy and incomplete PAC had good accuracy and was verified through both internal and external validation.
Collapse
Affiliation(s)
- Dian Liu
- Department of Lamphoma and Abdominal Radiotherapy, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410013, Changsha, China
| | - Hu Quan
- Department of Hepatobiliary and Intestinal Surgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410013, Changsha, China
| | - Min Ma
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, 410013, Changsha, China
| | - Huijun Zhou
- Department of Gastroenterology and Urology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410013, Changsha, China
| | - Xiaolin Yang
- Department of Gastroenterology and Urology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410013, Changsha, China
| | - Zhengchun Wu
- Department of Hepatobiliary and Intestinal Surgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410013, Changsha, China
| | - Jia Luo
- Department of Hepatobiliary and Intestinal Surgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410013, Changsha, China
| | - Hua Xiao
- Department of Hepatobiliary and Intestinal Surgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410013, Changsha, China.
- Department of Gastroduodenal and Pancreatic Surgery, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 410013, Changsha, China.
| | - Yanping Xiao
- Department of Scientific Research, Changsha Health Vocational College, 410605, Changsha, China.
| |
Collapse
|
39
|
Wu L, Gao G, Mi H, Luo Z, Wang Z, Liu Y, Wu L, Long H, Shen Y. Validation of CDC45 as a novel biomarker for diagnosis and prognosis of gastric cancer. PeerJ 2024; 12:e17130. [PMID: 38515458 PMCID: PMC10956518 DOI: 10.7717/peerj.17130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/27/2024] [Indexed: 03/23/2024] Open
Abstract
Background Cell division cycle protein 45 (CDC45) has been demonstrated to play vital roles in the progression of various malignancies. However, the clinical significance of CDC45 in gastric cancer (GC) remains unreported. Method In this study, we employed the TCGA database and the TCGA & GTEx dataset to compare the mRNA expression levels of CDC45 between gastric cancer tissues and adjacent or normal tissues (p < 0.05 was considered statistically significant), which was further validated in multiple datasets including GSE13911, GSE29272, GSE118916, GSE66229, as well as RT-qPCR. Furthermore, we harnessed the Human Protein Atlas (HPA) to evaluate the protein expression of CDC45, which was subsequently verified through immunohistochemistry (IHC). To ascertain the diagnostic utility of CDC45, receiver operating characteristic (ROC) curves and the area under the ROC curve (AUC) were calculated in TCGA database, and further validated it in TCGA & GTEx and GSE66229 datasets. The Kaplan-Meier method was used to reveal the prognostic importance of CDC45 in The Cancer Genome Atlas (TCGA) database and authenticated through the GSE66229, GSE84433, and GSE84437 datasets. Through cBioPortal, we identified co-expressed genes of CDC45, and pursued enrichment analysis. Additionally, we availed gene set enrichment analysis (GSEA) to annotate the biological functions of CDC45. Results Differential expression analysis revealed that CDC45 was significantly upregulated at both the mRNA and protein levels in GC (all p < 0.05). Remarkably, CDC45 emerged as a promising prognostic indicator and a novel diagnostic biomarker for GC. In a comprehensive the drug susceptibility analysis, we found that patients with high expression of CDC45 had high sensitivity to various chemotherapeutic agents, among which 5-fluorouracil, docetaxel, cisplatin, and elesclomol were most evident. Furthermore, our findings suggested a plausible association between CDC45 and immune cell infiltration. Enrichment analysis revealed that CDC45 and its associated genes may play crucial roles in muscle biofunction, whereas GSEA demonstrated significant enrichment of gene sets pertaining to G protein-coupled receptor ligand binding and G alpha (i) signaling events. Conclusion Our study elucidates that upregulation of CDC45 is intricately associated with immune cell infiltration and holds promising potential as a favorable prognostic marker and a novel diagnostic biomarker for GC.
Collapse
Affiliation(s)
- Lihua Wu
- Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, China
| | - Gan Gao
- Clinical Laboratory, Liuzhou Hospital of Guangzhou Women and Children’s Medical Center, Liuzhou, Guangxi, China
- Guangxi Clinical Research Center for Obstetrics and Gynecology, liuzhou, Guangxi, China
| | - Hui Mi
- Changzhi People’s Hospital, Changzhi, china
| | - Zhou Luo
- Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, China
| | - Zheng Wang
- Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, China
| | - Yongdong Liu
- Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, China
| | - Liangyan Wu
- Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, China
| | - Haihua Long
- Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, China
| | - Yongqi Shen
- Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, China
| |
Collapse
|
40
|
Liu R, Chu W, Liu X, Hong J, Wang H. Establishment of Golgi apparatus-related genes signature to predict the prognosis and immunotherapy response in gastric cancer patients. Medicine (Baltimore) 2024; 103:e37439. [PMID: 38489711 PMCID: PMC10939665 DOI: 10.1097/md.0000000000037439] [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: 07/20/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 03/17/2024] Open
Abstract
The Golgi apparatus plays a crucial role in intracellular protein transportation, processing, and sorting. Dysfunctions of the Golgi apparatus have been implicated in tumorigenesis and drug resistance. This study aimed to investigate the prognostic and treatment response assessment value of Golgi apparatus-related gene (GARGs) features in gastric cancer patients. Transcriptome data and clinical information of gastric cancer patients were obtained from The Cancer Genome Atlas and Gene Expression Omnibus databases. Cox regression analysis was employed to assess the prognostic significance of GARGs and construct risk features. The immune landscape, drug sensitivity, immune therapy response, gene expression patterns, and somatic mutation characteristics were analyzed between different risk groups. A nomogram model for predicting gastric cancer prognosis was developed and evaluated. Among 1643 GARGs examined, 365 showed significant associations with gastric cancer prognosis. Five independent prognostic GARGs (NGF, ABCG1, CHAC1, GBA2, PCSK7) were selected to construct risk features for gastric cancer patients. These risk features effectively stratified patients into high-risk and low-risk groups, with the former exhibiting worse prognosis than the latter. Patients in the high-risk group displayed higher levels of immune cell infiltration, while the expression levels of NGF, CHAC1, GBA2, PCSK7 were significantly correlated with immune cell infiltration. Notably, the low-risk group exhibited higher sensitivity to epothilone.B, metformin, and tipifarnib compared to the high-risk group. Moreover, patients in the low-risk group demonstrated greater responsiveness to immune therapy than those in the high-risk group. In terms of biological processes and KEGG pathways related to immunity regulation, significant suppression was observed in the high-risk group compared to the low-risk group; meanwhile cell cycle pathways exhibited significant activation in the high-risk group. Furthermore, the low-risk group exhibited a higher tumor mutation burden compared to the high-risk group. The risk features derived from GARGs, in conjunction with age, were identified as independent risk factors for gastric cancer. The nomogram incorporating these factors demonstrated improved performance in predicting gastric cancer prognosis. Our study established risk features derived from GARGs that hold potential clinical utility in prognostic assessment and immune therapy response evaluation of gastric cancer patients.
Collapse
Affiliation(s)
- Rui Liu
- Department of Gastrointestinal Surgery, Hangzhou Third People’s Hospital, Hangzhou, Zhejiang, China
| | - Weiwei Chu
- Department of Gastrointestinal Surgery, Hangzhou Third People’s Hospital, Hangzhou, Zhejiang, China
| | - Xiaojin Liu
- Department of Gastrointestinal Surgery, Hangzhou Third People’s Hospital, Hangzhou, Zhejiang, China
| | - Jie Hong
- Department of Gastrointestinal Surgery, Hangzhou Third People’s Hospital, Hangzhou, Zhejiang, China
| | - Haiming Wang
- Department of Gastrointestinal Surgery, Hangzhou Third People’s Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
41
|
Mirghani H, Alamrani BA, Aljabri MO, Alamrani FO, Saleh Alatawi M, Albalawi MM, Alasmari MAS, Alsharif AFB, Albalawi WMB, Alzamhari OS. A Comparison Between the Transoral Endoscopic Thyroidectomy Vestibular Approach and the Transareolar Approach Regarding Perioperative Complications: A Systematic Review and Meta-Analysis. Cureus 2024; 16:e56438. [PMID: 38646299 PMCID: PMC11026944 DOI: 10.7759/cureus.56438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 04/23/2024] Open
Abstract
The global adoption of remote thyroidectomy is increasing, with the transoral endoscopic thyroidectomy vestibular approach (TOETVA) and transareolar approach (TAA) emerging as predominant methods. However, existing meta-analyses comparing these approaches to operative surgeries and short-term postoperative complications have significant limitations. To address this gap, our meta-analysis provides a comprehensive comparison between the TOETVA and TAA, focusing on operation time, intraoperative blood loss, postoperative drainage, and hospital stay duration. It aims to offer robust insights into their relative efficacy and safety profiles. We searched SCOPUS, PubMed, Web of Science, MEDLINE, and Cochrane Library from June 2015 to January 2024 for studies comparing transoral endoscopic thyroidectomy with the vestibular approach and areolar thyroidectomy using keywords, including "transoral thyroidectomy" and "scarless thyroidectomy." Studies were included if they were randomized controlled trials, case-control studies, or prospective/retrospective cohort studies comparing the TOETVA and TAA. Exclusion criteria removed case series, cross-sectional studies, editorials, non-English language, animal studies, and irrelevant articles. Data on operative time, postoperative drainage, intraoperative blood loss, and hospital stay were extracted. The Newcastle-Ottawa Scale was used to assess study quality (all studies scored 7-8). The findings revealed that the operative time was longer among the TOETVA group, with less intraoperative blood loss (odds ratio (OR) = 13.31, 95% confidence interval (CI) = 4.44-22.19); OR = -1.61, 95% CI = -2.82 to -0.39, respectively). Regarding hospitalization duration and postoperative drainage, no discernible difference was observed between the endoscopic TAA (ETAA) and TOETVA (OR = -0.04, 95% CI = -0.24 to 0.16; OR = -6.74, 95% CI = -20.08 to 6.60, respectively). The TOETVA has advantages over the TAA in terms of intraoperative blood loss and shorter operation times. However, both approaches exhibited comparable outcomes in terms of hospital stay duration and postoperative drainage. Furthermore, extensive randomized trials are warranted.
Collapse
|
42
|
You Y, Wang Y, Yu X, Gao F, Li M, Li Y, Wang X, Jia L, Shi G, Yang L. Prediction of lymph node metastasis in advanced gastric adenocarcinoma based on dual-energy CT radiomics: focus on the features of lymph nodes with a short axis diameter ≥6 mm. Front Oncol 2024; 14:1369051. [PMID: 38496754 PMCID: PMC10940341 DOI: 10.3389/fonc.2024.1369051] [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: 01/11/2024] [Accepted: 02/15/2024] [Indexed: 03/19/2024] Open
Abstract
Objective To explore the value of the features of lymph nodes (LNs) with a short-axis diameter ≥6 mm in predicting lymph node metastasis (LNM) in advanced gastric adenocarcinoma (GAC) based on dual-energy CT (DECT) radiomics. Materials and methods Data of patients with GAC who underwent radical gastrectomy and LN dissection were retrospectively analyzed. To ensure the correspondence between imaging and pathology, metastatic LNs were only selected from patients with pN3, nonmetastatic LNs were selected from patients with pN0, and the short-axis diameters of the enrolled LNs were all ≥6 mm. The traditional features of LNs were recorded, including short-axis diameter, long-axis diameter, long-to-short-axis ratio, position, shape, density, edge, and the degree of enhancement; univariate and multivariate logistic regression analyses were used to establish a clinical model. Radiomics features at the maximum level of LNs were extracted in venous phase equivalent 120 kV linear fusion images and iodine maps. Intraclass correlation coefficients and the Boruta algorithm were used to screen significant features, and random forest was used to build a radiomics model. To construct a combined model, we included the traditional features with statistical significance in univariate analysis and radiomics scores (Rad-score) in multivariate logistic regression analysis. Receiver operating curve (ROC) curves and the DeLong test were used to evaluate and compare the diagnostic performance of the models. Decision curve analysis (DCA) was used to evaluate the clinical benefits of the models. Results This study included 114 metastatic LNs from 36 pN3 cases and 65 nonmetastatic LNs from 28 pN0 cases. The samples were divided into a training set (n=125) and a validation set (n=54) at a ratio of 7:3. Long-axis diameter and LN shape were independent predictors of LNM and were used to establish the clinical model; 27 screened radiomics features were used to build the radiomics model. LN shape and Rad-score were independent predictors of LNM and were used to construct the combined model. Both the radiomics model (area under the curve [AUC] of 0.986 and 0.984) and the combined model (AUC of 0.970 and 0.977) outperformed the clinical model (AUC of 0.772 and 0.820) in predicting LNM in both the training and validation sets. DCA showed superior clinical benefits from radiomics and combined models. Conclusion The models based on DECT LN radiomics features or combined traditional features have high diagnostic performance in determining the nature of each LN with a short-axis diameter of ≥6 mm in advanced GAC.
Collapse
Affiliation(s)
- Yang You
- Department of Computed Tomography and Magnetic Resonance, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yan Wang
- Department of Computed Tomography and Magnetic Resonance, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xianbo Yu
- CT Collaboration, Siemens Healthineers Ltd., Beijing, China
| | - Fengxiao Gao
- Department of Computed Tomography and Magnetic Resonance, Xing Tai People’s Hospital, Xingtai, China
| | - Min Li
- Department of Computed Tomography and Magnetic Resonance, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yang Li
- Department of Computed Tomography and Magnetic Resonance, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiangming Wang
- Department of Computed Tomography and Magnetic Resonance, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Litao Jia
- Department of Computed Tomography and Magnetic Resonance, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Gaofeng Shi
- Department of Computed Tomography and Magnetic Resonance, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Li Yang
- Department of Computed Tomography and Magnetic Resonance, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
43
|
Liu B, Xu YJ, Chu FR, Sun G, Zhao GD, Wang SZ. Development of a clinical nomogram for prediction of response to neoadjuvant chemotherapy in patients with advanced gastric cancer. World J Gastrointest Surg 2024; 16:396-408. [PMID: 38463346 PMCID: PMC10921200 DOI: 10.4240/wjgs.v16.i2.396] [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: 08/25/2023] [Revised: 11/05/2023] [Accepted: 01/19/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND The efficacy of neoadjuvant chemotherapy (NAC) in advanced gastric cancer (GC) is still a controversial issue. AIM To find factors associated with chemosensitivity to NAC treatment and to provide the optimal therapeutic strategies for GC patients receiving NAC. METHODS The clinical information was collected from 230 GC patients who received NAC treatment at the Central South University Xiangya School of Medicine Affiliated Haikou Hospital from January 2016 to December 2020. Least absolute shrinkage and selection operator logistic regression analysis was used to find the possible predictors. A nomogram model was employed to predict the response to NAC. RESULTS In total 230 patients were finally included in this study, including 154 males (67.0%) and 76 females (33.0%). The mean age was (59.37 ± 10.60) years, ranging from 24 years to 80 years. According to the tumor regression grade standard, there were 95 cases in the obvious response group (grade 0 or grade 1) and 135 cases in the poor response group (grade 2 or grade 3). The obvious response rate was 41.3%. Least absolute shrinkage and selection operator analysis showed that four risk factors significantly related to the efficacy of NAC were tumor location (P < 0.001), histological differentiation (P = 0.001), clinical T stage (P = 0.008), and carbohydrate antigen 724 (P = 0.008). The C-index for the prediction nomogram was 0.806. The calibration curve revealed that the predicted value exhibited good agreement with the actual value. Decision curve analysis showed that the nomogram had a good value in clinical application. CONCLUSION A nomogram combining tumor location, histological differentiation, clinical T stage, and carbohydrate antigen 724 showed satisfactory predictive power to the response of NAC and can be used by gastrointestinal surgeons to determine the optimal treatment strategies for advanced GC patients.
Collapse
Affiliation(s)
- Bing Liu
- Department of Gastrointestinal Surgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou 570208, Hainan Province, China
| | - Yu-Jie Xu
- Department of Gastrointestinal Surgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou 570208, Hainan Province, China
| | - Feng-Ran Chu
- Clinical College, Hainan Medical University, Haikou 571199, Hainan Province, China
| | - Guang Sun
- Department of Gastrointestinal Surgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou 570208, Hainan Province, China
| | - Guo-Dong Zhao
- Department of Gastrointestinal Surgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou 570208, Hainan Province, China
| | - Sheng-Zhong Wang
- Department of Gastrointestinal Surgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou 570208, Hainan Province, China
| |
Collapse
|
44
|
Zheng HL, Wang FH, Zhang LK, Li P, Zheng CH, Chen QY, Huang CM, Xie JW. Trajectories of neutrophil-to-lymphocyte ratios during neoadjuvant chemotherapy correlate with short- and long-term outcomes in gastric cancer: a group-based trajectory analysis. BMC Cancer 2024; 24:226. [PMID: 38365617 PMCID: PMC10873962 DOI: 10.1186/s12885-024-11950-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 02/04/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Systemic inflammatory factors can predict the survival prognosis of gastric cancer (GC) patients after neoadjuvant chemotherapy (NACT). However, whether longitudinal changes in systemic inflammatory factors are associated with short - and long-term outcomes has not been reported. METHODS This study is a retrospective analysis of 216 patients with advanced gastric cancer who received NACT between January 2011 and June 2019, comparing receiver operating characteristic (ROC) curves for screening suitable inflammatory markers. Group-based trajectory modeling (GBTM) was used to analyze longitudinal changes in inflammatory markers during NACT to identify different potential subgroups and to compare postoperative complications, recurrence-free survival (RFS), and overall survival (OS) among subgroups. RESULTS Ultimately, neutrophil-lymphocyte ratio (NLR) had the highest area under the curve (AUC) value in predicting prognosis was included in the GBTM analysis. Three trajectories of NLR were obtained: Stable group (SG) (n = 89), Ascent-descend group (ADG) (n = 80) and Continuous descend group (CDG) (n = 47). Compared with SG, ADG and CDG are associated with an increased risk of postoperative recurrence and death. The median time of RFS and OS of SG was longer than that of ADG and CDG (median RFS 81 vs. 44 and 22 months; median OS 69 vs. 41 and 30 months). In addition, CDG had significantly higher postoperative serious complications than SG and ADG (17 (36.2%) vs. 17 (19.1%) and 12 (15.0%); p = 0.005). CONCLUSION There were different trajectories of NLR during NACT, and these potential trajectories were significantly associated with severe postoperative complications, recurrence, and mortality in patients with GC.
Collapse
Affiliation(s)
- Hua-Long Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian Province, 350001, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Fu-Hai Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian Province, 350001, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ling-Kang Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian Province, 350001, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian Province, 350001, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian Province, 350001, China
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian Province, 350001, China.
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian Province, 350001, China.
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian Province, 350001, China.
- Fujian Provincial Minimally Invasive Medical Center, Fuzhou, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| |
Collapse
|
45
|
Ge HT, Chen JW, Wang LL, Zou TX, Zheng B, Liu YF, Xue YJ, Lin WW. Preoperative prediction of lymphovascular and perineural invasion in gastric cancer using spectral computed tomography imaging and machine learning. World J Gastroenterol 2024; 30:542-555. [PMID: 38463023 PMCID: PMC10921149 DOI: 10.3748/wjg.v30.i6.542] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/18/2023] [Accepted: 01/15/2024] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Lymphovascular invasion (LVI) and perineural invasion (PNI) are important prognostic factors for gastric cancer (GC) that indicate an increased risk of metastasis and poor outcomes. Accurate preoperative prediction of LVI/PNI status could help clinicians identify high-risk patients and guide treatment decisions. However, prior models using conventional computed tomography (CT) images to predict LVI or PNI separately have had limited accuracy. Spectral CT provides quantitative enhancement parameters that may better capture tumor invasion. We hypothesized that a predictive model combining clinical and spectral CT parameters would accurately preoperatively predict LVI/PNI status in GC patients. AIM To develop and test a machine learning model that fuses spectral CT parameters and clinical indicators to predict LVI/PNI status accurately. METHODS This study used a retrospective dataset involving 257 GC patients (training cohort, n = 172; validation cohort, n = 85). First, several clinical indicators, including serum tumor markers, CT-TN stages and CT-detected extramural vein invasion (CT-EMVI), were extracted, as were quantitative spectral CT parameters from the delineated tumor regions. Next, a two-step feature selection approach using correlation-based methods and information gain ranking inside a 10-fold cross-validation loop was utilized to select informative clinical and spectral CT parameters. A logistic regression (LR)-based nomogram model was subsequently constructed to predict LVI/PNI status, and its performance was evaluated using the area under the receiver operating characteristic curve (AUC). RESULTS In both the training and validation cohorts, CT T3-4 stage, CT-N positive status, and CT-EMVI positive status are more prevalent in the LVI/PNI-positive group and these differences are statistically significant (P < 0.05). LR analysis of the training group showed preoperative CT-T stage, CT-EMVI, single-energy CT values of 70 keV of venous phase (VP-70 keV), and the ratio of standardized iodine concentration of equilibrium phase (EP-NIC) were independent influencing factors. The AUCs of VP-70 keV and EP-NIC were 0.888 and 0.824, respectively, which were slightly greater than those of CT-T and CT-EMVI (AUC = 0.793, 0.762). The nomogram combining CT-T stage, CT-EMVI, VP-70 keV and EP-NIC yielded AUCs of 0.918 (0.866-0.954) and 0.874 (0.784-0.936) in the training and validation cohorts, which are significantly higher than using each of single independent factors (P < 0.05). CONCLUSION The study found that using portal venous and EP spectral CT parameters allows effective preoperative detection of LVI/PNI in GC, with accuracy boosted by integrating clinical markers.
Collapse
Affiliation(s)
- Hui-Ting Ge
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
- Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors, Fujian Medical University, Fuzhou 350001, Fujian Province, China
- Digestive, Hematological and Breast Malignancies, Clinical Research Center for Radiology and Radiotherapy of Fujian Province, Fuzhou 350001, Fujian Province, China
| | - Jian-Wu Chen
- Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors, Fujian Medical University, Fuzhou 350001, Fujian Province, China
- Digestive, Hematological and Breast Malignancies, Clinical Research Center for Radiology and Radiotherapy of Fujian Province, Fuzhou 350001, Fujian Province, China
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Li-Li Wang
- Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors, Fujian Medical University, Fuzhou 350001, Fujian Province, China
- Digestive, Hematological and Breast Malignancies, Clinical Research Center for Radiology and Radiotherapy of Fujian Province, Fuzhou 350001, Fujian Province, China
- Department of Diagnostic Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Tian-Xiu Zou
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Bin Zheng
- School of Electrical and Computer Engineering, University of Oklahoma, Oklahoma, OK 73019, United States
| | - Yuan-Fen Liu
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Yun-Jing Xue
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Wei-Wen Lin
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| |
Collapse
|
46
|
Ni Y, Zhuang Z. DDX24 promotes tumor progression by mediating hexokinase-1 induced glycolysis in gastric cancer. Cell Signal 2024; 114:110995. [PMID: 38043669 DOI: 10.1016/j.cellsig.2023.110995] [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: 09/05/2023] [Revised: 11/10/2023] [Accepted: 11/26/2023] [Indexed: 12/05/2023]
Abstract
Metabolic reprogramming allows tumor cells to meet high demand of biogenesis and increased energy for rapid proliferation. Gastric cancer (GC) ranks among the most prevalent malignancies globally. Exploring the underlying mechanisms of glycolytic reprogramming in GC could provide new therapeutic target for GC treatment. Here, we showed that DEAD-box helicase 24 (DDX24) played a critical role in hexokinase-1 (HK1) induced glycolysis. DDX24 expression was significantly elevated in GC tissues and was closely associated with worse survival in GC patients. In addition, DDX24 promoted glucose uptake and lactate production in GC cells. Mechanistically, DDX24 could bind the HK1 mRNA and positively regulated HK1 level at the transcriptional level. Moreover, DDX24 promoted the proliferation, migration, and invasion ability of GC cells by upregulating HK1. Collectively, these results suggested that DDX24 was a critical player in the regulation of glycolytic reprogramming and also implicated DDX24 as a valuable therapeutic target for GC.
Collapse
Affiliation(s)
- Yuanyuan Ni
- Department of Oncology, the Second Affiliated Hospital of Soochow University, Suzhou 215008, Jiangsu Province, PR China; Department of Radiation Oncology, the Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, Jiangsu Province, PR China
| | - Zhixiang Zhuang
- Department of Oncology, the Second Affiliated Hospital of Soochow University, Suzhou 215008, Jiangsu Province, PR China.
| |
Collapse
|
47
|
Leibold J, Tsanov KM, Amor C, Ho YJ, Sánchez-Rivera FJ, Feucht J, Baslan T, Chen HA, Tian S, Simon J, Wuest A, Wilkinson JE, Lowe SW. Somatic mouse models of gastric cancer reveal genotype-specific features of metastatic disease. NATURE CANCER 2024; 5:315-329. [PMID: 38177458 PMCID: PMC10899107 DOI: 10.1038/s43018-023-00686-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 11/10/2023] [Indexed: 01/06/2024]
Abstract
Metastatic gastric carcinoma is a highly lethal cancer that responds poorly to conventional and molecularly targeted therapies. Despite its clinical relevance, the mechanisms underlying the behavior and therapeutic response of this disease are poorly understood owing, in part, to a paucity of tractable models. Here we developed methods to somatically introduce different oncogenic lesions directly into the murine gastric epithelium. Genotypic configurations observed in patients produced metastatic gastric cancers that recapitulated the histological, molecular and clinical features of all nonviral molecular subtypes of the human disease. Applying this platform to both wild-type and immunodeficient mice revealed previously unappreciated links between the genotype, organotropism and immune surveillance of metastatic cells, which produced distinct patterns of metastasis that were mirrored in patients. Our results establish a highly portable platform for generating autochthonous cancer models with flexible genotypes and host backgrounds, which can unravel mechanisms of gastric tumorigenesis or test new therapeutic concepts.
Collapse
Affiliation(s)
- Josef Leibold
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medical Oncology and Pneumology, University Hospital Tuebingen, Tuebingen, Germany.
- iFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tuebingen, Tuebingen, Germany.
| | - Kaloyan M Tsanov
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Corina Amor
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Yu-Jui Ho
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Francisco J Sánchez-Rivera
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Judith Feucht
- iFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tuebingen, Tuebingen, Germany
- Department I-General Paediatrics, Haematology/Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany
| | - Timour Baslan
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Biomedical Sciences, School of Veterinary Medicine, The University of Pennsylvania, Philadelphia, PA, USA
| | - Hsuan-An Chen
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sha Tian
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Janelle Simon
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexandra Wuest
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John E Wilkinson
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Scott W Lowe
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
48
|
Yao G, Yuan J, Duan Q, Tan Y, Zhang Q, Chen D, Chen J. Immunoneoadjuvant therapy with immune checkpoint inhibitors of gastric cancer: an emerging exemplification : Immunoneoadjuvant therapy of gastric cancer. Invest New Drugs 2024; 42:1-13. [PMID: 37971628 DOI: 10.1007/s10637-023-01406-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023]
Abstract
Advances in immune checkpoint inhibitors (ICIs) have enabled more effective treatment for individuals with various types of solid tumors. Given the improved survival benefit and acceptable safety profile of ICIs in advanced gastric cancer, there is plenty of interest in the use of ICIs in the neoadjuvant setting with curative intent. Theoretically, immunoneoadjuvant with ICIs could boost the levels of endogenous tumor antigen present in the tumor to enhance T-cell priming and further enhance systemic immunity. This systemic immune response may improve the detection and elimination of the disseminated micrometastatic tumors beyond the resected tumor, which are sources of postsurgical relapse. Numerous clinical studies have begun to explore the application of ICIs in neoadjuvant treatment of gastric cancer. This article reviews the progress in the use of ICI monotherapy and in combination with alternative therapies for the treatment of gastric cancer to aid in the development of gastric cancer immunoneoadjuvant therapy and improve the overall therapeutic benefit.
Collapse
Affiliation(s)
- Guoliang Yao
- Department of hepatobiliary surgery, The first affiliated hospital of Henan university of science and technology, Henan Province, Luoyang, China
| | - Jianyong Yuan
- The 5th Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Qianqian Duan
- The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co, Xuanwu District, Ltd; Building 5, No. 699-18 Xuanwu Avenue, Nanjing, Jiangsu Province, China
| | - Yuan Tan
- The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co, Xuanwu District, Ltd; Building 5, No. 699-18 Xuanwu Avenue, Nanjing, Jiangsu Province, China
| | - Qin Zhang
- The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co, Xuanwu District, Ltd; Building 5, No. 699-18 Xuanwu Avenue, Nanjing, Jiangsu Province, China
| | - Dongsheng Chen
- The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co, Xuanwu District, Ltd; Building 5, No. 699-18 Xuanwu Avenue, Nanjing, Jiangsu Province, China
| | - Jingbo Chen
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong Province, China.
| |
Collapse
|
49
|
Xiong JX, Li YT, Tan XY, Chen T, Liu BH, Fu L. Targeting PRSS23 with tipranavir induces gastric cancer stem cell apoptosis and inhibits growth of gastric cancer via the MKK3/p38 MAPK-IL24 pathway. Acta Pharmacol Sin 2024; 45:405-421. [PMID: 37814123 PMCID: PMC10789761 DOI: 10.1038/s41401-023-01165-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/04/2023] [Indexed: 10/11/2023]
Abstract
Gastric cancer stem cells (GCSCs) contribute to the refractory features of gastric cancer (GC) and are responsible for metastasis, relapse, and drug resistance. The key factors drive GCSC function and affect the clinical outcome of GC patients remain poorly understood. PRSS23 is a novel serine protease that is significantly up-regulated in several types of cancers and cancer stem cells, and related to tumor progression and drug resistance. In this study, we investigated the role of PRSS23 in GCSCs as well as the mechanism by which PRSS23 regulated the GCSC functions. We demonstrated that PRSS23 was critical for sustaining GCSC survival. By screening a collection of human immunodeficiency virus (HIV) protease inhibitors (PIs), we identified tipranavir as a PRSS23-targeting drug, which effectively killed both GCSC and GC cell lines (its IC50 values were 4.7 and 6.4 μM in GCSC1 cells and GCSC2 cells, respectively). Administration of tipranavir (25 mg·kg-1·d-1, i.p., for 8 days) in GCSC-derived xenograft mice markedly inhibited the growth of subcutaneous GCSC tumors without apparent toxicity. In contrast, combined treatment with 5-FU plus cisplatin did not affect the tumor growth but causing significant weight loss. Furthermore, we revealed that tipranavir induced GCSC cell apoptosis by suppressing PRSS23 expression, releasing MKK3 from the PRSS23/MKK3 complex to activate p38 MAPK, and thereby activating the IL24-mediated Bax/Bak mitochondrial apoptotic pathway. In addition, tipranavir was found to kill other types of cancer cell lines and drug-resistant cell lines. Collectively, this study demonstrates that by targeting both GCSCs and GC cells, tipranavir is a promising anti-cancer drug, and the clinical development of tipranavir or other drugs specifically targeting the PRSS23/MKK3/p38MAPK-IL24 mitochondrial apoptotic pathway may offer an effective approach to combat gastric and other cancers.
Collapse
Affiliation(s)
- Ji-Xian Xiong
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China.
| | - Yu-Ting Li
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Xiang-Yu Tan
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Tie Chen
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Bao-Hua Liu
- Department of Biochemistry, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Li Fu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China.
| |
Collapse
|
50
|
Wang X, Zhang J, Wu Y, Zhang Y, Zhang S, Li A, Wang J, Wang Z. RORα inhibits gastric cancer proliferation through attenuating G6PD and PFKFB3 induced glycolytic activity. Cancer Cell Int 2024; 24:12. [PMID: 38184549 PMCID: PMC10770990 DOI: 10.1186/s12935-023-03201-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 12/27/2023] [Indexed: 01/08/2024] Open
Abstract
BACKGROUND Glycolysis is critical for harvesting abundant energy to maintain the tumor microenvironment in malignant tumors. Retinoic acid-related orphan receptor α (RORα) has been identified as a circadian gene. However, the association of glycolysis with RORα in regulating gastric cancer (GC) proliferation remains poorly understood. METHODS Bioinformatic analysis and retrospective study were utilized to explore the role of RORα in cell cycle and glycolysis in GC. The mechanisms were performed in vitro and in vivo including colony formation, Cell Counting Kit-8 (CCK-8), Epithelial- mesenchymal transition (EMT) and subcutaneous tumors of mice model assays. The key drives between RORα and glycolysis were verified through western blot and chip assays. Moreover, we constructed models of high proliferation and high glucose environments to verify a negative feedback and chemoresistance through a series of functional experiments in vitro and in vivo. RESULTS RORα was found to be involved in the cell cycle and glycolysis through a gene set enrichment analysis (GSEA) algorithm. GC patients with low RORα expression were not only associated with high circulating tumor cells (CTC) and high vascular endothelial growth factor (VEGF) levels. However, it also presented a positive correlation with the standard uptake value (SUV) level. Moreover, the SUVmax levels showed a positive linear relation with CTC and VEGF levels. In addition, RORα expression levels were associated with glucose 6 phosphate dehydrogenase (G6PD) and phosphofructokinase-2/fructose-2,6-bisphosphatase (PFKFB3) expression levels, and GC patients with low RORα and high G6PD or low RORα and high PFKFB3 expression patterns had poorest disease-free survival (DFS). Functionally, RORα deletion promoted GC proliferation and drove glycolysis in vitro and in vivo. These phenomena were reversed by the RORα activator SR1078. Moreover, RORα deletion promoted GC proliferation through attenuating G6PD and PFKFB3 induced glycolytic activity in vitro and in vivo. Mechanistically, RORα was recruited to the G6PD and PFKFB3 promoters to modulate their transcription. Next, high proliferation and high glucose inhibited RORα expression, which indicated that negative feedback exists in GC. Moreover, RORα deletion improved fluorouracil chemoresistance through inhibition of glucose uptake. CONCLUSION RORα might be a novel biomarker and therapeutic target for GC through attenuating glycolysis.
Collapse
Affiliation(s)
- Xiaoshan Wang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Junyi Zhang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Yuwei Wu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Yuqing Zhang
- Department of Occupational Health and Environmental Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Siyuan Zhang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Angqing Li
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Jian Wang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Zhengguang Wang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China.
| |
Collapse
|