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
|
Li Y, Li T, Feng J, Liu B, Wang Z, He J, Chen Z, Tao R, Wang H, Fan K, Sun Y, Wang J, Guo B, Zhang G. Acid-responsive contractile hyaluronic acid-based hydrogel loaded with ginsenoside Rg1 for hemostasis and promotion of gastric wound healing. Biomaterials 2025; 321:123320. [PMID: 40209592 DOI: 10.1016/j.biomaterials.2025.123320] [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: 12/23/2024] [Revised: 04/02/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025]
Abstract
Due to constant stimulation by stomach acid and local bleeding, gastric tissue wounds tend to heal slowly and complications such as anastomotic leakage have a high incidence. Suturing is often used to treat gastric wounds in clinic, but it still faces risks such as bleeding, slow healing, and leakage. Recently, hydrogel have been widely used to treat various types of wounds. Although hydrogels have shown promising efficacy in wound healing, it is still a challenge in dealing with wounds in gastric tissue for the poor adaptability of traditional materials in acidic environments. Hence, a series of pH responsive and good tissue adhesive hydrogels (MA-HA/AA) based on methacryloyl hyaluronic acid (MA-HA) and acryloyl-6-aminocaproic acid (AA) via in situ photo-crosslinking were designed, and anti-inflammatory and pro-healing traditional Chinese medicines ginsenoside Rg1 was incorporated into the hydrogel to treat gastric tissue wound. These acid-responsive hydrogels could form effective acid-resistant barriers and could lead to hemostasis rapidly through its strong adhesion. Besides, the hydrogels contracted under an acidic environment, which could tighten the gastric tissue wounds and sustained release the loaded ginsenoside Rg1. In addition, the hydrogels showed excellent biocompatibility and in vivo degradability. In summary, the acid-responsive contractile hyaluronic acid hydrogel loaded with ginsenoside Rg1 had good properties for hemostasis and acid-resistance to facilitate the promotion of gastric wounds healing.
Collapse
Affiliation(s)
- Yixing Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China; Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Tongyang Li
- Frontier Institute of Science and Technology, and State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jinteng Feng
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China; Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Bohao Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China; Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhiyu Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China; Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jiahui He
- Frontier Institute of Science and Technology, and State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhe Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China; Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Runyi Tao
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China; Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Hongyi Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China; Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Kun Fan
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China; Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ye Sun
- Department of Anesthesia and Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jizhao Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China; Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Baolin Guo
- Frontier Institute of Science and Technology, and State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Guangjian Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China; Key Laboratory of Enhanced Recovery After Surgery of Integrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
3
|
Bokhary RY. Prevalence of HER2 expression and its association with clinicopathological parameters in gastric and gastroesophageal junction adenocarcinoma: A 10?year experience of an academic center. Mol Clin Oncol 2025; 22:49. [PMID: 40242368 PMCID: PMC12001012 DOI: 10.3892/mco.2025.2844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
HER2 overexpression is a marker for targeted therapy in adenocarcinoma of the gastroesophageal junction (GEJ) and stomach. The present study aimed to evaluate the frequency of HER2 overexpression with reference to clinicopathological characteristics in subjects from King Abdulaziz University Hospital, Jeddah, Saudi Arabia over a 10-year period. A retrospective cross-sectional study was conducted on all biopsy and resection specimens diagnosed with either gastric cancer (GC) or GEJ adenocarcinomas from patients between January 2014 and December 2023 that had a final pathology report. Demographic characteristics of 122 patients, including age and sex, were collected, along with pathological details such as tumor grade, histological subtype and HER2 status. χ2 test was used to analyze the association between collected clinicopathological characteristics and HER2 status of the tumor. Most patients were aged 40-60 years. Males constituted 66% of the patients, and the ethnic distribution between Saudi and non-Saudi was almost equal. The most common subtype of cancer was the intestinal type (49%), and the majority of cases were poorly differentiated (64%). HER2 status was assessed in only 61% of cases, with 13.5% showing gene amplification. There was no significant association found between HER2 status and clinicopathological features.
Collapse
Affiliation(s)
- Rana Y. Bokhary
- Department of Pathology, Faculty of Medicine, King Abdulaziz University & King Abdulaziz University Hospital, Jeddah 21589, Saudi Arabia
| |
Collapse
|
4
|
Shamim M, Ali MS, Hossain MA, Hossain MA, Kavey MRH, Hoque MM, Rahman MA, Harrath AH, Rahman MH. Discovery of novel PARP1 inhibitors through computational drug design approaches. Comput Biol Chem 2025; 116:108366. [PMID: 39987745 DOI: 10.1016/j.compbiolchem.2025.108366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is the most frequent malignancy in women. It is a prevalent condition, representing 15-20 % of all breast cancer cases, characterized by its aggressive subtype and unfavorable prognosis. OBJECTIVES The main aim of this study is to find and develop a potential novel therapeutic candidate for TNBC treatment utilizing luteolin derivatives compounds. METHODS In this study, we used the stable TNBC protein structure from the Protein Data Bank (PDB) and selected luteolin, a bioactive compound known for its anti-cancer properties, to design potential anti-cancer drugs using computational methods. Structural activity relationship methodologies were used to evaluate active and inactive outcomes using pass prediction scores. Furthermore, we employed in-silico methods such as ADMET, drug-likeness evaluation, DFT quantum calculations, and Frontier Molecular Orbitals (HOMO and LUMO). Afterwards, we performed molecular docking for binding affinity and molecular dynamics simulations over 200 ns to validate interactions with TNBC protein RESULTS: Our results demonstrated that the ligands DM02, DM06, DM07, and DM09 did not violate Lipinski rules, and their reduced HOMO-LUMO energy gap indicates enhanced chemical reactivity and interaction with biological targets. The drug's maximum softness and minimum hardness values showed rapid metabolism and no hepatotoxicity, carcinogenicity, skin sensitization, or aquatic toxicity. Molecular docking studies revealed that DM02 and DM09, luteolin derivatives, have the highest binding affinity with the TNBC protein (PDB ID 5HA9) and our study confirms their stable interactions with the protein, suggesting potential therapeutic agents for TNBC. CONCLUSIONS Our computational data suggest that Luteolin derivatives have the potential to be utilized as therapeutic agents for TNBC. However, further experimental validation is needed to validate these findings.
Collapse
Affiliation(s)
- Md Shamim
- Department of Pharmacy, Faculty of biological science, Islamic University, Kushtia 7003, Bangladesh
| | - Md Sumon Ali
- Department of Pharmacy, Faculty of biological science, Islamic University, Kushtia 7003, Bangladesh
| | - Md Arju Hossain
- Department of Biochemistry and Biotechnology, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh
| | - Md Alfaz Hossain
- Department of Pharmacy, Faculty of biological science, Islamic University, Kushtia 7003, Bangladesh
| | - Md Reduanul Haque Kavey
- Department of Pharmacy, Faculty of biological science, Islamic University, Kushtia 7003, Bangladesh
| | - Md Mobinul Hoque
- Department of Biomedical Engineering, Faculty of Engineering and Technology, Islamic University, Kushtia 7003, Bangladesh
| | - Md Ataur Rahman
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, School of Medicine, Detroit, Michigan 48201, USA
| | - Abdel Halim Harrath
- King Saud University, Department of Zoology, College of Science, Riyadh 11451, Saudi Arabia
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Faculty of Engineering and Technology, Islamic University, Kushtia 7003, Bangladesh; Center for Advanced Bioinformatics and Artificial Intelligence Research, Islamic University, Kushtia 7003, Bangladesh.
| |
Collapse
|
5
|
Baez-Navarro X, Groenendijk FH, Oudijk L, von der Thüsen J, Fusco N, Curigliano G, van Deurzen CHM. HER2-low across solid tumours: different incidences and definitions. Pathology 2025; 57:403-414. [PMID: 40221332 DOI: 10.1016/j.pathol.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/22/2025] [Accepted: 02/10/2025] [Indexed: 04/14/2025]
Abstract
Antibody-drug conjugates, particularly trastuzumab deruxtecan (T-DXd), have emerged as effective therapies for various solid tumours. Clinical trials show that T-DXd improves survival in both HER2-positive and HER2-low breast cancer patients. Additionally, it improves survival in HER2-positive gastro-oesophageal cancer and elicits objective responses in HER2-low tumours. Responses have also been noted in lung and gynaecological cancers with HER2 expression, although subgroup analyses for HER2-low cases are lacking. This review assesses HER2 protein expression levels and gene amplification across solid tumours where T-DXd shows potential benefits. We focus on the accuracy and limitations of HER2 testing methods, particularly for identifying HER2-low cancer. A semi-systematic approach was employed, searching EMBASE, Medline, Cochrane, and PubMed databases. We calculated median incidences of HER2-positive, HER2-low, and HER2-0 by immunohistochemistry (IHC), and HER2 amplification by in situ hybridisation (ISH). A total of 144 studies were included, covering breast (n=57), gastro-oesophageal (n=33), lung (n=17), gynaecological (n=24), and various other carcinomas (n=13). The median incidences of HER2-low were 52%, 16%, 58%, and 17% in breast, gastro-oesophageal, endometrial, and ovarian cancers, respectively, with unknown incidences in lung and cervical cancers. Factors influencing HER2-low detection include tumour heterogeneity, antibody clones, observer variability, and lack of validated scoring criteria. Given the significant proportion of HER2-low cases, many patients could benefit from T-DXd, but limitations in detection accuracy necessitate further research and standardisation in diagnostic methods and criteria to advance the clinical utility of T-DXd for HER2-low tumours.
Collapse
Affiliation(s)
- Ximena Baez-Navarro
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands.
| | | | - Lindsey Oudijk
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jan von der Thüsen
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | | |
Collapse
|
6
|
Zeng R, Lin Z, Feng F, Li Y, Liu W, He W, Huang Y, Lin X, Mei Y, Wu H, Zhang Q. Metabolic alterations and immune heterogeneity in gastric cancer metastasis. iScience 2025; 28:112296. [PMID: 40276776 PMCID: PMC12018583 DOI: 10.1016/j.isci.2025.112296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/11/2024] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Cellular metabolic reprogramming supports tumor proliferation, invasion, and metastasis by enhancing resistance to stress and immune clearance. Understanding these metabolic changes within the tumor microenvironment is vital to developing effective therapies. We conducted single-cell RNA sequencing on 11 gastric cancer (GC) samples and eight metastatic lesions, analyzing 92,842 cells across eight cell types, including cancer cells, stromal cells, and immune cells. Our findings highlight that the mitochondrial ATP synthase subunit ATP5MC2 uniquely alters during early GC metastasis. Experiments and clinical data confirmed that ATP5MC2 upregulation facilitates cancer cell proliferation, invasion, and metastasis. Constructing a single-cell atlas revealed significant immune cell heterogeneity associated with GC metastasis and its molecular subtypes. This study underscores the role of ATP5MC2-driven metabolic changes and diverse immune landscapes in promoting GC metastasis, offering new avenues for anti-metastatic treatment development.
Collapse
Affiliation(s)
- Rui Zeng
- School of Medicine, South China University of Technology, Guangzhou 510006, China
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Zhihao Lin
- School of Medicine, South China University of Technology, Guangzhou 510006, China
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Feiyan Feng
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yanyan Li
- School of Medicine, South China University of Technology, Guangzhou 510006, China
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Weiwei Liu
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Wenting He
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yongjun Huang
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Xingtao Lin
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yan Mei
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Hongmei Wu
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Qingling Zhang
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China; School of Medicine, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
7
|
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
|
8
|
Xiao Q, Liu Y, Li T, Wang C, He S, Zhai L, Yang Z, Zhang X, Wu Y, Liu Y. Viral oncogenesis in cancer: from mechanisms to therapeutics. Signal Transduct Target Ther 2025; 10:151. [PMID: 40350456 DOI: 10.1038/s41392-025-02197-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/22/2025] [Accepted: 03/03/2025] [Indexed: 05/14/2025] Open
Abstract
The year 2024 marks the 60th anniversary of the discovery of the Epstein-Barr virus (EBV), the first virus confirmed to cause human cancer. Viral infections significantly contribute to the global cancer burden, with seven known Group 1 oncogenic viruses, including hepatitis B virus (HBV), human papillomavirus (HPV), EBV, Kaposi sarcoma-associated herpesvirus (KSHV), hepatitis C virus (HCV), human T-cell leukemia virus type 1 (HTLV-1), and human immunodeficiency virus (HIV). These oncogenic viruses induce cellular transformation and cancer development by altering various biological processes within host cells, particularly under immunosuppression or co-carcinogenic exposures. These viruses are primarily associated with hepatocellular carcinoma, gastric cancer, cervical cancer, nasopharyngeal carcinoma, Kaposi sarcoma, lymphoma, and adult T-cell leukemia/lymphoma. Understanding the mechanisms of viral oncogenesis is crucial for identifying and characterizing the early biological processes of virus-related cancers, providing new targets and strategies for treatment or prevention. This review first outlines the global epidemiology of virus-related tumors, milestone events in research, and the process by which oncogenic viruses infect target cells. It then focuses on the molecular mechanisms by which these viruses induce tumors directly or indirectly, including the regulation of oncogenes or tumor suppressor genes, induction of genomic instability, disruption of regular life cycle of cells, immune suppression, chronic inflammation, and inducing angiogenesis. Finally, current therapeutic strategies for virus-related tumors and recent advances in preclinical and clinical research are discussed.
Collapse
Affiliation(s)
- Qing Xiao
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yi Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Tingting Li
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Chaoyu Wang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Sanxiu He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Liuyue Zhai
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Zailin Yang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaomei Zhang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| | - Yongzhong Wu
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| | - Yao Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
9
|
He C, Zhou H, He G, Zhang W, Chen H. Application of accommodated jejunal interposition double tract reconstruction after total gastrectomy for gastric cancer: a retrospective study. Discov Oncol 2025; 16:729. [PMID: 40354014 DOI: 10.1007/s12672-025-02536-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 04/29/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND The aim of this study was to compare the effects of accommodated jejunal interposition double tract reconstruction (aji-DTR) and Roux-en-Y reconstruction after laparoscopic-assisted total gastrectomy on intraoperative and postoperative indicators in advanced gastric cancer (AGC) patients. METHODS A retrospective analysis was performed on 80 AGC patients, including 43 with aji-ATR and 37 with Roux-en-Y reconstruction. Propensity score matching was performed between the two groups. The primary outcome measures included operative time, intraoperative blood loss, postoperative complications, postoperative hospital stay, total hospitalization costs, and survival rate. The secondary outcome measures were postoperative nutritional status, recovery of digestive function, and postoperative gastrointestinal·symptoms. RESULTS There were 24 pairs of patients after matching. There were no significant differences in the operative time, intraoperative blood loss, time of first flatus, time of first defecation, time of liquid diet, time of semi-liquid diet, postoperative complications, postoperative hospital stays, and total hospitalization costs (all P > 0.05). Interestingly, Roux stasis syndrome was significantly more frequent in Roux-en-Y group than aji-DTR group [6 (25.0%) vs 1 (4.2%), P = 0.045]. While no significant difference was observed in survival rates, reflux esophagitis, dumping syndrome and nutritional parameter including hemoglobin, albumin, and prognostic nutritional index (all P > 0.05). CONCLUSIONS Compared with Roux-en-Y reconstruction, aji-DTR had similar surgical parameters, postoperative digestive function recovery, nutritional parameters, and survival rate, but showed an advantage in reducing Roux stasis syndrome. Therefore, aji-DTR after laparoscopic assisted total gastrectomy may be a safe and feasible alternative for AGC patients.
Collapse
Affiliation(s)
- Cankun He
- Department of General Surgery, Hui'an County Hospital, 582 Huixing Street, Hui'an, 362100, China
| | - Huangming Zhou
- Department of General Surgery, Hui'an County Hospital, 582 Huixing Street, Hui'an, 362100, China
| | - Guobao He
- Department of General Surgery, Hui'an County Hospital, 582 Huixing Street, Hui'an, 362100, China
| | - Weixin Zhang
- Department of General Surgery, Hui'an County Hospital, 582 Huixing Street, Hui'an, 362100, China
| | - Huizhong Chen
- Department of General Surgery, Hui'an County Hospital, 582 Huixing Street, Hui'an, 362100, China.
| |
Collapse
|
10
|
Hua G, Zhao L, Zeng X, Luo L. Synergistic inhibition of gastric cancer cell proliferation by concanavalin A and silibinin via attenuation of the JAK/STAT3 signaling pathway and molecular docking analysis. Hereditas 2025; 162:73. [PMID: 40350446 DOI: 10.1186/s41065-025-00438-z] [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: 03/11/2025] [Accepted: 04/23/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND In the current period of pharmaceutical discovery, herbal remedies have shown to be an unmatched supply of anticancer medications. Plants and their derivatives, through analogues, play a vital role in cancer treatment. OBJECTIVES The current investigation assessed the effectiveness of inhibiting the growth of gastric cancer cells in AGS cells by blocking the JAK/ STAT3 signalling pathways using the natural medicines Concanavalin A (Con-A) and silibinin (SB). MATERIALS AND METHODS After being exposed to various doses of concanavalin A, and silibinin (Con-A + SB) for 24 h (0- 60 µM), the cells were evaluated for multiple studies. The MTT assay was used to examine the combination of Con-A + SB-induced cytotoxicity. To evaluate ROS, DCFH-DA staining was utilized. Dual (AO/EtBr) staining was performed to examine apoptotic modifications, and MMP levels in AGS cells were examined using the appropriate fluorescence staining assays. By using flow cytometry and western blotting, cell cycle, and apoptosis were assessed. RESULTS The relative cytotoxicity of Con-A and SB was found to be approximately 19.6 μM and 16.78 μM, (p < 0.05) correspondingly, according to the findings. After a 24-h incubation period, the combination of Con-A and SB generates significant cytotoxicity in AGS cells, with an IC50 of 10.37 μM (p < 0.01). Furthermore, AGS cells treated with Con-A and SB concurrently showed increased apoptotic signals, exhibited by Bax overexpression, Bcl-2 downregulation, and Caspase-3 activation, as well as considerable ROS generation. CONCLUSION Therefore, the combination usage of Con-A + SB has the potential to serve as a chemotherapeutic agent since it prevents the synthesis of JAK/STAT3 intermediated control of proliferation and cell cycle-regulating proteins.
Collapse
Affiliation(s)
- Gaoyan Hua
- Department of Oncology, The People's Hospital of Chizhou, Chizhou, Anhui, 247001, China
| | - Lisha Zhao
- Department of Gastroenterology, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, 430000, China
| | - Xianjing Zeng
- General Practice Medicine, Affiliated Hospital of Jinggangshan University, Ji 'an, Jiangxi, 343000, China
| | - Liang Luo
- Department of Oncology, Affiliated Hospital of Jinggangshan University, Ji 'an, Jiangxi, 343000, China.
| |
Collapse
|
11
|
Jiang XH, Chen YH, Ye WF, Lin YJ, Chen XJ, Sun Q, Peng JS. Development and evaluation of an intelligent individualized nutrition management (iNutrition) applet for postdischarge gastric cancer patients following gastrectomy. Support Care Cancer 2025; 33:460. [PMID: 40342161 DOI: 10.1007/s00520-025-09507-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/29/2025] [Indexed: 05/11/2025]
Abstract
PURPOSE This study aimed to develop an intelligent individualized nutrition management (iNutrition) applet for postdischarge gastric cancer patients following gastrectomy based on the Health Action Process Approach (HAPA) theory and using the design thinking method. METHODS We developed the iNutrition applet using the design thinking method, including the following phases: (1) "empathize and define," which focused on understanding patients' needs and establishing the goals of the iNutrition applet through qualitative interviews; (2) "ideate and prototype," during which literature reviews and multidisciplinary interactive workshops to conceptualize innovative solutions were conducted and a fully functional iNutrition applet was developed; and (3) "test," during which the iNutrition applet was evaluated through usability testing. RESULTS Based on qualitative interviews with 15 patients, we determined that the iNutrition applet would need to satisfy patients' needs in the following areas: adjustment to postoperative anatomical changes, evidence-based knowledge, communication with peers, consultation with medical staff, tailored nutritional plans, coping plans, and needs for technology. Consequently, the applet was designed to have eight functions: gastrointestinal symptom management, nutrition knowledge, medical consultation, peer communication, nutrition goals, nutrition diary with calculator, weekly meal plan, and nutritional status score. During short-term usability testing, the patients' task completion rate ranged from 88 to 100%. Long-term usability testing resulted in a System Usability Scale (SUS) score of 75.20 ± 10.63 and a net promoter score (NPS) of 32%. The participants were generally satisfied with the applet's design and functionality, providing feedback that informed further refinements to the final version of the applet. CONCLUSION The iNutrition applet, which has received high ratings for usability and user satisfaction, is an acceptable and feasible method of postdischarge nutritional management for gastric cancer patients following gastrectomy.
Collapse
Affiliation(s)
- Xiao-Han Jiang
- School of Nursing, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yong-He Chen
- Department of Gastric Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
| | - Wen-Feng Ye
- Department of Nutrition, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yi-Jia Lin
- Department of Gastric Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
| | - Xi-Jie Chen
- Department of Gastric Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
| | - Qian Sun
- School of Nursing, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jun-Sheng Peng
- School of Nursing, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Department of Gastric Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China.
| |
Collapse
|
12
|
Hua Y, Gao Y, Luo S, Song G, Tian X, Wang C, Lv S, Zhang X, Shao G. Programmed cell death 1 inhibitor combined with chemotherapy compared to chemotherapy alone as first-line treatment in advanced gastric cancer patients: A real-world study. Int Immunopharmacol 2025; 154:114487. [PMID: 40179586 DOI: 10.1016/j.intimp.2025.114487] [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: 08/22/2024] [Revised: 10/13/2024] [Accepted: 03/11/2025] [Indexed: 04/05/2025]
Abstract
OBJECTIVE Previous trials have revealed better treatment efficacy of programmed cell death 1 (PD-1) inhibitors combined with chemotherapy as first-line treatments than chemotherapy alone in advanced gastric cancer (GC) patients, but real-world evidence is still lacking. Hence, this real-world study aimed to investigate the efficacy and safety of PD-1 inhibitors plus chemotherapy as first-line treatments compared with chemotherapy alone in advanced GC patients. METHODS In total, 102 advanced GC patients were allocated into a combination group (receiving chemotherapy combined with a PD-1 inhibitor as a first-line treatment) (n = 48) or a chemotherapy group (receiving chemotherapy only as a first-line treatment) (n = 54) according to their actual treatment regimens. RESULTS The objective response rate (ORR) was greater in the combination group than in the chemotherapy group (25.0 % versus 9.3 %, P = 0.033), whereas the disease control rate (DCR) was not different between the groups (83.3 % versus 66.7 %, P = 0.054). Progression-free survival (PFS) was prolonged in the combination group than in the chemotherapy group (P = 0.018). The median (95 % confidence interval) PFS was 19.7 (12.2-27.2) months in the combination group and 16.5 (7.3-25.7) months in the chemotherapy group. Multivariate logistic regression analyses revealed that PD-1 inhibitors combined with chemotherapy were independently associated with an increased ORR (odds ratio: 4.180, P = 0.024), increased DCR (odds ratio: 2.928, P = 0.049), and prolonged PFS (hazard ratio: 0.388, P = 0.030). No difference was found in total or each specific grade III-IV adverse reaction between the groups (all P > 0.05). CONCLUSION Treatment with a PD-1 inhibitor plus chemotherapy as a first-line treatment shows better treatment efficacy with similar safety to that of chemotherapy alone in advanced GC patients.
Collapse
Affiliation(s)
- Yunqi Hua
- Department of Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia 014040, China
| | - Yuqian Gao
- Department of Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia 014040, China
| | - Shuang Luo
- Department of Graduate School, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014030, China
| | - Ge Song
- Department of Graduate School, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014030, China
| | - Xiaoling Tian
- Department of Graduate School, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014030, China
| | - Chenlin Wang
- Department of Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia 014040, China
| | - Shuang Lv
- Department of Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia 014040, China
| | - Xinyi Zhang
- Department of Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia 014040, China
| | - Guo Shao
- Center for Translational Medicine, the Third People's Hospital of Longgang District & Longgang Institute of Medical Imaging, Shantou University Medical College, Shenzhen 518100, China.
| |
Collapse
|
13
|
Fan S, Jiang H, Xu Q, Shen J, Lin H, Yang L, Yu D, Zheng N, Chen L. Risk factors for pneumonia after radical gastrectomy for gastric cancer: a systematic review and meta-analysis. BMC Cancer 2025; 25:840. [PMID: 40336054 PMCID: PMC12060482 DOI: 10.1186/s12885-025-14149-1] [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: 04/23/2024] [Accepted: 04/14/2025] [Indexed: 05/09/2025] Open
Abstract
OBJECTIVE The objective is to systematically gather relevant research to determine and quantify the risk factors and pooled prevalence for pneumonia after a radical gastrectomy for gastric cancer. METHODS The reporting procedures of this meta-analysis conformed to the PRISMA 2020. Chinese Wan Fang data, Chinese National Knowledge Infrastructure (CNKI), Chinese Periodical Full-text Database (VIP), Embase, Scopus, CINAHL, Ovid MEDLINE, PubMed, Web of Science, and Cochrane Library from inception to January 20, 2024, were systematically searched for cohort or case-control studies that reported particular risk factors for pneumonia after radical gastrectomy for gastric cancer. The pooled prevalence of pneumonia was estimated alongside risk factor analysis. The quality was assessed using the Newcastle-Ottawa Scale after the chosen studies had been screened and the data retrieved. RevMan 5.4 and R 4.4.2 were the program used to perform the meta-analysis. RESULTS Our study included data from 20,840 individuals across 27 trials. The pooled prevalence of postoperative pneumonia was 11.0% (95% CI = 8.0% ~ 15.0%). Fifteen risk factors were statistically significant, according to pooled analyses. Several factors were identified to be strong risk factors, including smoking history (OR 2.71, 95% CI = 2.09 ~ 3.50, I2 = 26%), prolonged postoperative nasogastric tube retention (OR 2.25, 95% CI = 1.36-3.72, I2 = 63%), intraoperative bleeding ≥ 200 ml (OR 2.21, 95% CI = 1.15-4.24, I2 = 79%), diabetes mellitus (OR 4.58, 95% CI = 1.84-11.38, I2 = 96%), male gender (OR 3.56, 95% CI = 1.50-8.42, I2 = 0%), total gastrectomy (OR 2.59, 95% CI = 1.83-3.66, I2 = 0%), COPD (OR 4.72, 95% CI = 3.80-5.86, I2 = 0%), impaired respiratory function (OR 2.72, 95% CI = 1.58-4.69, I2 = 92%), D2 lymphadenectomy (OR 4.14, 95% CI = 2.29-7.49, I2 = 0%), perioperative blood transfusion (OR 4.21, 95% CI = 2.51-7.06, I2 = 90%), and hypertension (OR 2.21, 95% CI = 1.29-3.79, I2 = 0%). Moderate risk factors included excessive surgery duration (OR 1.51, 95% CI = 1.25-1.83, I2 = 90%), advanced age (OR 1.91, 95% CI = 1.42-2.58, I2 = 94%), nutritional status (OR 2.62, 95% CI = 1.55-4.44, I2 = 71%), and history of pulmonary disease (OR 1.61, 95% CI = 1.17-2.21, I2 = 79%). CONCLUSIONS This study identified 15 independent risk factors significantly associated with pneumonia after radical gastrectomy for gastric cancer, with a pooled prevalence of 11.0%. These findings emphasize the importance of targeted preventive strategies, including preoperative smoking cessation, nutritional interventions, blood glucose and blood pressure control, perioperative respiratory training, minimizing nasogastric tube retention time, and optimizing perioperative blood transfusion strategies. For high-risk patients, such as the elderly, those undergoing prolonged surgeries, experiencing excessive intraoperative blood loss, undergoing total gastrectomy, or receiving open surgery, close postoperative monitoring is essential. Early recognition of pneumonia signs and timely intervention can improve patient outcomes and reduce complications.
Collapse
Affiliation(s)
- Siyue Fan
- Department of General Surgery, Zhongshan Hospital of Xiamen University, Xiamen, 361004, China
- Nursing College, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Hongzhan Jiang
- School of Nursing, Beijing University of Chinese Medicine, Beijing, China
| | - Qiuqin Xu
- Xiamen Hospital of Traditional Chinese Medicine, Xiamen, China
| | - Jiali Shen
- Nursing Department, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Huihui Lin
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Liping Yang
- Nursing College, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Doudou Yu
- Nursing College, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Nengtong Zheng
- Nursing College, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Lijuan Chen
- Department of General Surgery, Zhongshan Hospital of Xiamen University, Xiamen, 361004, China.
- Nursing College, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| |
Collapse
|
14
|
Ling T, Zuo Z, Huang M, Ma J, Wu L. Stacking classifiers based on integrated machine learning model: fusion of CT radiomics and clinical biomarkers to predict lymph node metastasis in locally advanced gastric cancer patients after neoadjuvant chemotherapy. BMC Cancer 2025; 25:834. [PMID: 40329193 PMCID: PMC12057267 DOI: 10.1186/s12885-025-14259-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: 02/20/2025] [Accepted: 05/02/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND The early prediction of lymph node positivity (LN+) after neoadjuvant chemotherapy (NAC) is crucial for optimizing individualized treatment strategies. This study aimed to integrate radiomic features and clinical biomarkers through machine learning (ML) approaches to enhance prediction accuracy by focusing on patients with locally advanced gastric cancer (LAGC). METHODS We retrospectively enrolled 277 patients with LAGC and randomly divided them into training (n = 193) and validation (n = 84) sets at a 7:3 ratio. In total, 1,130 radiomics features were extracted from pre-treatment portal venous phase computed tomography scans. These features were linearly combined to develop a radiomics score (rad score) through feature engineering. Then, using the rad score and clinical biomarkers as input features, we applied simple statistical strategies (relying on a single ML model) and integrated statistical strategies (including classification model integration techniques, such as hard voting, soft voting, and stacking) to predict LN+ post-NAC. The diagnostic performance of the model was assessed using receiver operating characteristic curves with corresponding areas under the curve (AUC). RESULTS Of all ML models, the stacking classifier, an integrated statistical strategy, exhibited the best performance, achieving an AUC of 0.859 for predicting LN+ in patients with LAGC. This predictive model was transformed into a publicly available online risk calculator. CONCLUSIONS We developed a stacking classifier that integrates radiomics and clinical biomarkers to predict LN+ in patients with LAGC undergoing surgical resection, providing personalized treatment insights.
Collapse
Affiliation(s)
- Tong Ling
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Zhichao Zuo
- School of Mathematics and Computational Science, Xiangtan University, Xiangtan, Hunan province, China
| | - Mingwei Huang
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Jie Ma
- Department of Medical Imaging, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Liucheng Wu
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China.
| |
Collapse
|
15
|
Zhong Q, Weng CM, Jiang MC, Sun YQ, Li BL, Zhao W, Zhang HX, Zhang ZQ, Ma YB, Wu SC, Ye W, Wu J, Du H, Zheng CH, Li P, Chen QY, Huang CM, Xie JW. Patterns of Survival and Recurrence in Poor Responders to Neoadjuvant Therapy for Gastric Cancer: A Real-World Multicenter Study. Ann Surg Oncol 2025:10.1245/s10434-025-17396-5. [PMID: 40329137 DOI: 10.1245/s10434-025-17396-5] [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: 11/01/2024] [Accepted: 04/13/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Neoadjuvant therapy (NAT) is increasingly used in locally advanced gastric cancer (LAGC), but a significant proportion of patients respond poorly, causing adverse outcomes. Few studies have specifically examined the prognosis of this subgroup. This study aimed to analyze survival and recurrence in poor responders to guide follow-up and treatment strategies. METHODS This multicenter retrospective study included patients with LAGC who received NAT. Tumor regression was graded following the Becker system, defining TRG 2-3 as poor response. Outcomes were assessed for overall survival (OS), recurrence-free survival (RFS), and recurrence patterns. RESULTS 648 patients were included: 341 with TRG 2 and 307 with TRG 3. In the entire cohort, the 3-year OS and RFS were 54.6% and 55.2%, respectively. Recurrence occurred in 299 patients, with the following recurrence patterns: distant metastasis (26.1%, n = 78), peritoneal metastasis (21.1%, n = 63), locoregional recurrence (18.7%, n = 56), and multiple-site recurrence (18.4%, n = 55). Liver metastasis was significantly higher in the TRG 3 group than in the TRG 2 group (14.1% versus 5.3%, P = 0.010). ypN+ was the most significant independent risk factor for recurrence (OR = 2.73, 95% CI 1.83-4.08, P < 0.001); an increasing number of positive lymph nodes led to higher 3-year cumulative mortality in patients. Despite poor response to NAT, completing over four adjuvant chemotherapy cycles was associated with improved survival outcomes. CONCLUSION Poor NAT responders in LAGC have high recurrence rates, particularly in the first year post-surgery, with ypN+ status being the strongest predictor of recurrence. Completing over four cycles of AC was associated with survival improvement in this group.
Collapse
Affiliation(s)
- Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Cai-Ming Weng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Mei-Chen Jiang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yu-Qin Sun
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Bao-Long Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Department of General Surgery, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Wei Zhao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- General Hospital of Ningxia Medical University, Yinchuan, China
| | - Hao-Xiang Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Zhi-Quan Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Yu-Bin Ma
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qinghai University, Xining, China
| | - Shi-Chao Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Gastrointestinal Surgery Unit 2, Putian First Hospital of Fujian Medical University, Putian, China
| | - Wen Ye
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Department of Gastrointestinal Surgery, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Ju Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - He Du
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qinghai University, Xining, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
16
|
Dat TQ, Thong DQ, Nguyen DT, Hai NV, Vuong NL, Bac NH, Long VD. Laparoscopic versus open distal gastrectomy with d2 lymphadenectomy in treatment of locally T4A gastric cancer: the protocol of a randomized controlled trial. BMC Surg 2025; 25:193. [PMID: 40316937 PMCID: PMC12049068 DOI: 10.1186/s12893-025-02933-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) remains one of the leading causes of cancer-related mortality worldwide. While laparoscopic gastrectomy (LG) has been widely adopted for early and locally advanced gastric cancer (AGC), its safety and oncological efficacy in T4a GC remain unclear. To date, no randomized controlled trials have specifically examined the role of LG in the treatment of T4a GC. This study aims to provide robust evidence comparing the short- and long-term outcomes of laparoscopic distal gastrectomy (LDG) versus open distal gastrectomy (ODG) in resectable T4a GC. METHODS This is a phase III, randomized controlled, non-inferiority trial. Patients with clinical T4a GC (cT4aN0-3M0) suitable for distal gastrectomy with D2 dissection will be randomly assigned in a 1:1 ratio to undergo either LDG or ODG. A total 240 patients (120 each group) are required to statistically show non-inferiority of the LDG with respect to the primary end-point, 3-years disease-free survival (DFS). Secondary endpoints include morbiity, mortality, postoperative recovery, and quality of life. DISCUSSION This study is the first prospective randomized trial specifically designed to compare laparoscopic and open approaches for T4a GC. By standardizing surgical techniques and ensuring experienced surgeons perform the procedures, this trial aims to establish whether LDG can provide equivalent oncological outcomes while reducing perioperative morbidity and enhancing postoperative recovery. The findings will provide high-quality evidence to inform future guidelines and clinical decision-making in the management of T4a gastric cancer. TRIAL REGISTRATION This study is registered at ClinicalTrials.gov (NCT04384757), version 6. Registration Date: 08/05/2020.
Collapse
Affiliation(s)
- Tran Quang Dat
- Gastro-intestinal Surgery Department, University Medical Center, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Dang Quang Thong
- Gastro-intestinal Surgery Department, University Medical Center, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Doan Thuy Nguyen
- Gastro-intestinal Surgery Department, University Medical Center, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Nguyen Viet Hai
- Gastro-intestinal Surgery Department, University Medical Center, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Nguyen Lam Vuong
- Department of Medical Statistics and Informatics, Faculty of Public Health, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Nguyen Hoang Bac
- Gastro-intestinal Surgery Department, University Medical Center, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Viet Nam
- Department of General Surgery, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Vo Duy Long
- Gastro-intestinal Surgery Department, University Medical Center, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Viet Nam.
- Department of General Surgery, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam.
| |
Collapse
|
17
|
Zhang W, Wang J, Shi W, Qiu H, Ke S, Tian Y, Gong Y, Zhang C, Chen J, Wu Y, Zhao W, Chen Y. First-Line Apatinib Combined with Tislelizumab and Chemotherapy for Patients with Advanced Gastric and Gastroesophageal Junction Adenocarcinoma with Poor Prognosis. Mol Cancer Ther 2025; 24:720-727. [PMID: 39895587 DOI: 10.1158/1535-7163.mct-24-0143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/22/2024] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
In this prospective, open-label, exploratory study (RENMIN-213), patients with previously untreated, HER2-negative, advanced G/GEJ adenocarcinoma with signet ring cell carcinoma or peritoneal metastasis were enrolled to receive eight cycles of apatinib, tislelizumab, and chemotherapy every 3 weeks, with a maintenance therapy with apatinib plus tislelizumab for a maximum of 1 year. Homogeneous patients receiving immune checkpoint inhibitors combined with chemotherapy at the same time were deemed as the control group for efficacy. The primary endpoint was progression-free survival. Secondary endpoints were objective response rate, disease control rate, duration of response, overall survival, biomarkers, health-related quality of life, and safety. A total of 33 patients [median (range) age, 60 (32-72) years; 21 (63.6%) male; 11 (33.3%) signet ring cell carcinoma; 30 (90.9%) peritoneal metastasis] were enrolled and deemed evaluable for efficacy analysis. Of these patients, 32 (97%) were without disease progression, of whom 1 (3.0%) patient had complete response and 18 (54.6%) had partial response. Six patients (18.2%) were able to undergo surgery after treatment. After propensity score matching, the median progression-free survival was 10.17 months (95% confidence interval, 7.13-13.21) in the apatinib combined with tislelizumab and chemotherapy group, as compared with 6.0 months in the immune checkpoint inhibitor combined with chemotherapy group. The median duration of response increased from 4.5 to 8.7 months. The objective response rate increased from 33.3% to 54.6%, and the disease control rate increased from 87.9% to 97.0%. Treatment-related grade 3 or higher adverse events for evaluable patients occurred in 11 patients (33.3%), and patients' health-related quality of life improved after treatment.
Collapse
Affiliation(s)
- Wenjie Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jing Wang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Shi
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hu Qiu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shaobo Ke
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuanyuan Tian
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Gong
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Caiyutian Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiamei Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yong Wu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wensi Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yongshun Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
18
|
Fu Y, Qin C, Li M, Zhang X, Gai Y, Ruan W, Lan X. Comparative Evaluation of 68Ga-FAPI-04 PET for Initial N and M Staging in Gastric Cancer: A Study Against Histopathology and Contrast-Enhanced CT. Clin Nucl Med 2025; 50:394-403. [PMID: 40179292 DOI: 10.1097/rlu.0000000000005795] [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: 10/20/2024] [Accepted: 01/23/2025] [Indexed: 04/05/2025]
Abstract
PURPOSE To evaluate the efficiency of 68Ga-FAPI-04 PET (PET/MRI or PET/CT) for N and M staging in gastric carcinoma and compare outcomes with histopathology and contrast-enhanced computed tomography (CECT). PATIENTS AND METHODS Patients with gastric carcinoma who had undergone 68Ga-FAPI-04 PET/MRI or PET/CT before treatment were retrospectively enrolled. Histopathology post lymphadenectomy was the gold standard for N staging, while histopathology and follow-up data were the reference for overall outcomes. The diagnostic efficiency of 68Ga-FAPI-04 PET for detecting regional lymph node involvement and distant metastases was compared to that of CECT. RESULTS Sixty-two patients were enrolled. In 18 patients who underwent 68Ga-FAPI-04 PET/MRI and lymphadenectomy, 532 lymph nodes were dissected. 68Ga-FAPI-04 PET/MRI showed similar sensitivity, specificity, and accuracy compared to CECT (28.3% vs. 23.2%, 99.8% vs. 99.3%, and 86.5% vs. 85.2%, all P > 0.05). Fifty-five patients had regional lymph node metastasis, 68Ga-FAPI-04 PET exhibited comparable diagnostic efficiency to CECT, with sensitivity of 83.6% versus 87.3%, specificity of 100% versus 85.7%, accuracy of 85.5% versus 87.1% (all P > 0.05). Excluding 3 patients with only abdominal CECT, 32 out of 59 patients had distant metastasis, with no significant differences in sensitivity, specificity, and accuracy between 68Ga-FAPI-04 PET and CECT based on patient (100% vs. 87.5%, 92.6% vs. 96.3%, and 96.6% vs. 91.5%, all P >0.05). Notably, 68Ga-FAPI-04 PET outperformed CECT in detecting peritoneal, distant lymph nodes, bone, liver, and ovarian metastases by visualizing more lesions or greater lesion extent. CONCLUSIONS 68Ga-FAPI-04 PET exhibits comparable diagnostic performance to CECT for patient-based N staging and M staging of gastric cancer. However, it surpasses CECT in visualizing distant metastases.
Collapse
Affiliation(s)
- Yiru Fu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Chunxia Qin
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Mengting Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xiao Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Weiwei Ruan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
19
|
You Y, Liang Y, Chen L, Li Z, Gao B, Wang X, Yuan M, Xue Y, Liu Y, Gao J. Radiomics analysis of dual-energy CT-derived iodine maps for differentiating between T1/2 and T3/4a in gastric cancer: A multicenter study. Eur J Radiol 2025; 186:112054. [PMID: 40121898 DOI: 10.1016/j.ejrad.2025.112054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 02/23/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
OBJECTIVE To investigate the value of radiomic analysis of dual-energy CT (DECT)-derived iodine maps (IMs) for the differentiation between T1/2 and T3/4a stage tumors in gastric cancer (GC). METHODS A total of 263 patients who received upfront surgery and were pathologically confirmed with gastric adenocarcinoma were enrolled in this study. Dual-phase enhanced CT scans with gemstone spectral imaging (GSI) mode were performed within two weeks before surgery. 151 patients were retrospectively collected for the training (n = 105) and validation (n = 46) cohorts, and 112 patients were prospectively collected for the external test1 (n = 68) and external test2 (n = 44) cohorts. According to the postoperative pathological T stage, patients were classified into T1/2 and T3/4a stage groups. Clinical characteristics were recorded and quantitative iodine concentration (IC) of tumors was measured. Radiomics features were extracted from the venous phase (VP) IMs by three-dimensional region of interest (3D-ROI) segmentation. Feature selection was performed using the least absolute shrinkage and selection operator. Four machine learning algorithms, including random forest, logistic regression, naive Bayes, and support vector machine, were used to construct radiomics models. Finally, the most valuable clinical characteristics, DECT parameters, and the best radiomics model were combined to build a nomogram. The diagnostic performance of nomogram was evaluated by the area under receiver operating characteristic curve (AUC), calibration curve, and decision curve. RESULTS The nomogram combined tumor clinical T stage (cT), tumor thickness, venous-phase iodine concentration (ICVP), normalized arterial-phase iodine concentration (nICAP), and Radscore (derived from logistic regression model). This integrated model demonstrated favorable performance in the differentiation between T1/2 and T3/4a stage tumors in GC, with AUCs of 0.892 (95 %CI: 0.829-0.956), 0.846 (95 %CI: 0.734-0.958), 0.894 (95 %CI: 0.818-0.970) and 0.821 (95 %CI: 0.689-0.952) observed for the training, validation, external test 1, and external test 2 cohorts, respectively. Hosmer-Lemeshow test showed a good fit (all P > 0.05). Decision curves confirmed that the nomogram provided more net clinical benefit than the default simple strategy over a wide range of threshold probabilities. CONCLUSION We have developed and validated a multidimensional personalized nomogram that integrates a radiomics model based on DECT-derived IMs, DECT quantitative parameters, and traditional clinical features. The proposed model demonstrated favorable performance in preoperative identification of T3/4a stage tumors in GC.
Collapse
Affiliation(s)
- Yaru You
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Imaging Diagnosis and Treatment for Digestive System Tumor, Zhengzhou 450052, China
| | - Yan Liang
- Department of Radiology, Sanmenxia Central Hospital, Sanmenxia 472100, China
| | - Lihong Chen
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Zhanzhan Li
- Department of Radiology, Sanmenxia Central Hospital, Sanmenxia 472100, China
| | - Beijun Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Imaging Diagnosis and Treatment for Digestive System Tumor, Zhengzhou 450052, China
| | - Xiangxiang Wang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Imaging Diagnosis and Treatment for Digestive System Tumor, Zhengzhou 450052, China
| | - Mengchen Yuan
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Imaging Diagnosis and Treatment for Digestive System Tumor, Zhengzhou 450052, China
| | - Yunjing Xue
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, China.
| | - Yiyang Liu
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Imaging Diagnosis and Treatment for Digestive System Tumor, Zhengzhou 450052, China.
| | - Jianbo Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Imaging Diagnosis and Treatment for Digestive System Tumor, Zhengzhou 450052, China.
| |
Collapse
|
20
|
Cao Z, Wang Z, Yang L, Li T, Tao X, Niu X. Reshaping the immune microenvironment and reversing immunosenescence by natural products: Prospects for immunotherapy in gastric cancer. Semin Cancer Biol 2025; 110:1-16. [PMID: 39923925 DOI: 10.1016/j.semcancer.2025.02.002] [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: 12/10/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025]
Abstract
Gastric cancer (GC) represents a global health-care challenge. Recent progress in immunotherapy has elicited attracted considerable attention as a viable treatment option through modulating the host immune system and unleashing pre-existing immunity, which has profoundly revolutionized oncology, especially GC. Nonetheless, low clinical response and intrinsic and acquired resistance remain persistently challenging. The microenvironment of GC comprising multifarious stromal cell types has remarkable immunosuppressive elements that may impact the efficacy of immunotherapy. Immunosenescence is increasingly regarded as a factor that contributes to cancer development, remodels the tumor microenvironment and affects the efficacy of immunotherapy. Natural products are at the forefront of traditional medicine. Senotherapeutics is a class of drugs and natural products capable of delaying, preventing, or reversing the senescence process (i.e., senolytics) or suppressing senescence-associated secretory phenotype (i.e., senomorphics). Emerging evidence supports that natural products can improve the efficacy of existing immunotherapy and expand their indications in GC mainly based upon remodeling the immunosuppressive microenvironment and reversing immunosenescence. The review provides an integrated review of previously reported and ongoing clinical trials with immunotherapeutic regimens in GC and discusses current challenges. Next, we focus on natural compounds that exert anti-GC functions and possess immunomodulatory properties. More attention is paid to the potential of these natural compounds in modulating the immune microenvironment and immunosenescence. Lastly, we discuss the nanomedicine that can overcome the deficiencies of natural products. Altogether, our review suggests the enormous potential of natural compounds in GC immunotherapy, and provides an important direction for future research.
Collapse
Affiliation(s)
- Zhipeng Cao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, China
| | - Zhilin Wang
- Department of Pain Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Li Yang
- Department of Anesthesiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Tian Li
- Tianjin Key Laboratory of Acute Abdomen Disease-Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China.
| | - Xueshu Tao
- Department of Pain Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Xing Niu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
21
|
Hou YJ, Sang ZT, Li Q, Feng QX, Wu J, Nickel MD, Hsu YC, Wang WZ, Wu CJ, Xu H, Liu XS. Advanced Multiparametric MRI Strategies for Tumor Restaging After Neoadjuvant Therapy in Locally Advanced Gastric Cancer. Ann Surg Oncol 2025; 32:3382-3391. [PMID: 39900717 DOI: 10.1245/s10434-025-16972-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND This study tested the diagnostic accuracy of multiparametric magnetic resonance imaging (mpMRI) in restaging locally advanced gastric cancer after neoadjuvant therapy (NAT) using pathologic T stage (ypT) and pathologic N stage (ypN) as the reference standard. METHODS Between August 2022 and September 2023, the study enrolled a prospective cohort of 70 gastric cancer patients who underwent NAT and subsequent surgical resection. MRI procedures, including DLSB T2-weighted imaging (T2WI), ZOOMit diffusion-weighted imaging (DWI), and XD-VIBE dynamic contrast-enhanced imaging (DCE), were performed after NAT and before surgery. Four abdominal radiologists independently assigned radiologic T stage (yrT) and radiologic N stage (yrN) based on individual and combined sequences. Inter-reader agreement was quantified using Kendall's coefficient. Diagnostic accuracy was determined by comparing MRI assessments and pathologic outcomes, with pairwise comparisons analyzed via the McNemar test. Subgroup analysis evaluated the performance in identifying good responders to NAT. RESULTS Inter-reader agreement was almost perfect for T restaging and substantial for N restaging. Diagnostic accuracy for T restaging was 0.432 using DLSB-T2WI, 0.586 using ZOOMit DWI, 0.557 using XD-VIBE DCE, and 0.586 using mpMRI. The accuracy demonstrated by DWI, DCE and mpMRI was superior to that of T2WI (all P < 0.05). For N restaging, the accuracy of the mpMRI protocol was 0.443. Notably, mpMRI achieved an AUC of 0.879 (95% confidence interval 0.835-0.915) for differentiating ypT0-1 tumors. CONCLUSIONS Advanced mpMRI strategies can serve as a valuable tool for restaging gastric cancer after NAT. Accurately differentiating good responders to neoadjuvant therapy through mpMRI holds significant clinical implications for personalized treatment strategies and improved patient outcomes.
Collapse
Affiliation(s)
- Ya-Jun Hou
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zi-Tong Sang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiong Li
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiu-Xia Feng
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Wu
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | - Yi-Cheng Hsu
- MR Research Collaboration Team, Siemens Healthineers Ltd, Shanghai, China
| | - Wei-Zhi Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chen-Jiang Wu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Hao Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Xi-Sheng Liu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
22
|
Liu Q, Meng C, Cao S, Liu X, Tian Y, Li Z, Zhong H, Sun Y, Yu J, Zhou Y. Comparison of short- and long-term outcomes of robotic versus laparoscopic gastrectomy for locally advanced gastric cancer after neoadjuvant therapy: a high-volume center retrospective study with propensity score matching. Surg Endosc 2025; 39:2814-2827. [PMID: 40064692 DOI: 10.1007/s00464-025-11626-3] [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: 10/28/2024] [Accepted: 02/18/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND Although neoadjuvant therapy (NAT) for advanced gastric cancer (AGC) can benefit patient survival, few studies have compared the short- and long-term outcomes of robotic and laparoscopic gastrectomy for AGC after NAT. METHODS The clinical data of 321 AGC patients who received NATs and who underwent robotic gastrectomy (RG, n = 109) or laparoscopic gastrectomy (LG, n = 212) between May 2017 and September 2022 were collected and analyzed retrospectively at our center. After propensity score matching (PSM) for 1:1 matching to eliminate bias, both groups had 106 cases. Short-term clinical outcomes and long-term survival-related indicators were compared between the two groups of patients. RESULTS A total of 212 patients were included in the groups after matching. There were fewer overall complications (13.2% vs. 28.3%, P = 0.007) and surgical complications (8.5% vs. 17.9%, P = 0.043) in the RG group than in the LG group. Compared with the LG group, the RG group had more harvested overall lymph nodes (35.25 ± 4.99 vs. 31.45 ± 6.31, P < 0.001) and more suprapancreatic lymph nodes (13.12 ± 4.38 vs. 10.05 ± 4.13, P < 0.001). Patients in the RG group had significantly shorter surgery times (217.62 ± 47.49 vs. 267.25 ± 70.68, P < 0.001) and less blood loss (46.51 ± 27.02 vs. 70.75 ± 37.25, P < 0.001) than patients in the LG group. The RG group had significantly faster bowel function recovery, earlier liquid diet, and shorter hospital stay. Compared with LG, RG had a better 3-year RFS (75.5% vs. 62.3%, P = 0.017). CONCLUSION Compared with laparoscopic surgery, robotic surgery significantly increased the number of lymph node dissected, reduced intraoperative blood loss, and postoperative surgical complications rate. Although RG did not statistically improve 3-year overall survival, there was a significant improvement in RFS and could be an alternative surgical method for GC patients after NAC.
Collapse
Affiliation(s)
- Qi Liu
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Cheng Meng
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Shougen Cao
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Xiaodong Liu
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Yulong Tian
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Zequn Li
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Hao Zhong
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Yuqi Sun
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Junjian Yu
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China
| | - Yanbing Zhou
- Department of General Surgery, Affiliated Hospital of Qingdao University, 16# Jiangsu Road, Qingdao, 266000, Shandong Province, People's Republic of China.
- Gastrointestinal Tumor Translational Medicine Research Institute of Qingdao University, Qingdao, Shandong Province, China.
| |
Collapse
|
23
|
Chen W, Cai Z, Li J, Li Z, Xu Z, Wu J, Liu F, Zhao H, Xu Y. Five-year survival outcome of laparoscopic D2 lymphadenectomy plus complete mesogastrium excision for patients with stage II/III gastric cancer without invasion of the squamocolumnar junction. Surgery 2025; 181:109290. [PMID: 40068362 DOI: 10.1016/j.surg.2025.109290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/30/2024] [Accepted: 02/01/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Gastric cancer patients without invasion of the squamocolumnar junction have a low probability of extragastric mesenteric metastasis, and the survival benefit of conducting D2 lymphadenectomy plus complete mesogastrium excision is unclear in this group of patients. METHODS This was a retrospective analysis of patients with stage II/III gastric cancer without invasion of the squamocolumnar junction who underwent laparoscopic radical surgery for gastric cancer at the First Hospital of Putian City, Fujian Province, from January 2014 to May 2019. Five-year disease-free survival was the primary outcome, and 5-year overall survival and the recurrence pattern were the secondary outcomes. Morbidity, mortality, and surgery-related outcomes within 30 days of surgery were also analyzed. RESULTS This study included 366 patients, 133 in the D2 lymphadenectomy group and 233 in the D2 lymphadenectomy plus complete mesogastrium excision group. There was no statistically significant difference in 5-year disease-free survival and overall survival in the D2 lymphadenectomy plus complete mesogastrium excision group compared with the D2 lymphadenectomy group (P > .05). Regarding recurrence pattern, the D2 lymphadenectomy group had a higher rate of local recurrence (12.3% and 38.3%, P = .001). In subgroup analysis, patients with stage III and T3-4aN1-3 gastric cancer in the D2 lymphadenectomy plus complete mesogastrium excision group had significantly better 5-year disease-free survival compared with the D2 lymphadenectomy group (P < .05). CONCLUSION Laparoscopic D2 lymphadenectomy plus complete mesogastrium excision not only decreased the local recurrence rate of stage II/III gastric cancer without invasion of the squamocolumnar junction, but it also improved the 5-year disease-free survival of patients with stage III and T3-4aN1-3 gastric cancer without invasion of the squamocolumnar junction.
Collapse
Affiliation(s)
- Weixiang Chen
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China; Gastrointestinal Surgery Unit 1, The First Hospital of Putian City, Putian, Fujian, China
| | - Zhiming Cai
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Junpeng Li
- Gastrointestinal Surgery Unit 1, The First Hospital of Putian City, Putian, Fujian, China
| | - Zhixiong Li
- Gastrointestinal Surgery Unit 1, The First Hospital of Putian City, Putian, Fujian, China
| | - Zhengnan Xu
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China; Gastrointestinal Surgery Unit 1, The First Hospital of Putian City, Putian, Fujian, China
| | - Jihuang Wu
- Gastrointestinal Surgery Unit 1, The First Hospital of Putian City, Putian, Fujian, China
| | - Feng Liu
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China; Gastrointestinal Surgery Unit 1, The First Hospital of Putian City, Putian, Fujian, China
| | - Hongrui Zhao
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China; Gastrointestinal Surgery Unit 1, The First Hospital of Putian City, Putian, Fujian, China
| | - Yanchang Xu
- Gastrointestinal Surgery Unit 1, The First Hospital of Putian City, Putian, Fujian, China.
| |
Collapse
|
24
|
Weng CM, Zhong Q, Sun YQ, Liu ZY, Ma YB, Zhang ZQ, Zhang HX, Zhu JY, Ye W, Wu J, Du H, Zheng CH, Li P, Chen QY, Huang CM, Xie JW. A novel ypN-TRG staging system for gastric cancer patients after neoadjuvant therapy based on the metro-ticket paradigm: a multicenter and large sample retrospective analysis. Gastric Cancer 2025; 28:465-477. [PMID: 39918688 DOI: 10.1007/s10120-025-01586-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/15/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Conventional ypT category evaluates the depth of invasion after neoadjuvant therapy (NAT) for gastric cancer (GC) and has limited prognostic value. Tumor regression grade (TRG) measures the extent of tumor response to treatment, and when combined with the ypN category, it may enhance the prediction of patient outcomes. This study aims to develop a new staging system by integrating TRG and ypN category to better evaluate the prognosis of GC patients receiving NAT. METHODS This retrospective analysis included 962 patients who underwent radical gastrectomy after NAT, with 513 in the development cohort (from one center) and 449 in the external validation cohort (from five centers). The ypN-TRG staging system was established by calculating the distance from the origin on a cartesian plane incorporating the ypN (x-axis) stage and TRG (y-axis) grade, and five sub-stages were delineated. RESULTS In the development cohort, 3-year overall survival rates according to ypN-TRG stage I, IIA, IIB, IIIA, IIB were 87.6%, 80.2%, 70.7%, 47.3%, 21.5%, p < 0.01. Compared with ypTNM, the ypN-TRG staging system performed better in terms of the prognostic discrimination power (C-index), goodness-of-fit (AIC, BIC), model improvement (NRI, IDI), and model stability (time-AUC). Multivariate Cox regression analysis confirmed the superiority of ypN-TRG over ypTNM staging. In the external validation cohort, ypN-TRG staging was a better predictor of OS and DFS in patients with GC. CONCLUSIONS The ypN-TRG staging system is superior to the AJCC eighth edition ypTNM staging system in accurately assessing the prognosis of patients with GC after NAT.
Collapse
Affiliation(s)
- Cai-Ming Weng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Yu-Qin Sun
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, 363000, Fujian, China
| | - Zhi-Yu Liu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Yu-Bin Ma
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qinghai University, Xining, 810000, Qinghai, China
| | - Zhi-Quan Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Hao-Xiang Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Ji-Yun Zhu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Hepatopancreatobiliary Surgery Department, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Wen Ye
- Department of Gastrointestinal Surgery, Longyan First Hospital Affiliated to, Fujian Medical University, Longyan, 364000, Fujian, China
| | - Ju Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116000, Liaoning, China
| | - He Du
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qinghai University, Xining, 810000, Qinghai, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No.29 Xin-Quan Road, Fuzhou, 350001, Fujian, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
25
|
Kunisaki C, Sato S, Kasahara K, Sato T, Yago A, Tamura Y, Kondo H, Oshi M, Kosaka T, Akiyama H, Endo I. Impact of histologically poorly cohesive phenotype as a prognostic factor in patients with pStage II/III gastric cancer undergoing adjuvant chemotherapy. Gastric Cancer 2025; 28:478-492. [PMID: 40019627 DOI: 10.1007/s10120-025-01599-6] [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: 10/02/2024] [Accepted: 02/06/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Few studies have focussed on using histological phenotype to evaluate prognostic factors after adjuvant chemotherapy (AC) in patients with pStage II/III gastric cancer. We evaluated the impact of histological type based on the World Health Organization classification. METHODS Overall, 348 patients with pStage II/III advanced gastric cancer undergoing R0 gastrectomy without neoadjuvant chemotherapy were included. Of these, 143 underwent AC. Univariate and multivariate analyses for prognostic factors of relapse-free survival (RFS) and overall survival (OS) were performed in all patients and patients receiving AC. Moreover, long-term survivals were compared by histological phenotype between patients with and without AC. RESULTS Multivariate analysis in all patients revealed that histologically poorly cohesive carcinoma with not otherwise specified and signet ring cell subtype (PCC-NOS/SRC) and pN 2/3 independently and adversely affected RFS. Alternatively, male sex, poor PS and pN 2/3 adversely affected OS. In multivariate analysis of patients receiving AC, longer tumour size (≥ 80 mm), histologically PCC-NOS/SRC phenotype and pN 3 independently influenced RFS. Multivariate analysis in this population for OS revealed that pN 3 and poor postoperative immune-nutritional status significantly induced worse OS. Comparison of RFS and OS by histological phenotype between patients with and without AC showed that AC had no efficacy for improving long-term survivals in histologically PCC phenotype. CONCLUSIONS Histologically PCC phenotype by WHO classification, particularly PCC-NOS/SRC phenotypes, showed poor long-term RFS even after AC in patients with pStage II and III gastric cancer. An effective chemotherapeutic regimen needs to be developed for this specific subtype of gastric cancer.
Collapse
Affiliation(s)
- Chikara Kunisaki
- Department of Surgery, Gastroenterological Center, Yokohama City University, 4-57, Urafune-chi, Minami-ku, Yokohama, 232-0024, Japan.
| | - Sho Sato
- Department of Surgery, Gastroenterological Center, Yokohama City University, 4-57, Urafune-chi, Minami-ku, Yokohama, 232-0024, Japan
| | - Kohei Kasahara
- Department of Surgery, Gastroenterological Center, Yokohama City University, 4-57, Urafune-chi, Minami-ku, Yokohama, 232-0024, Japan
| | - Tsutomu Sato
- Department of Surgery, Gastroenterological Center, Yokohama City University, 4-57, Urafune-chi, Minami-ku, Yokohama, 232-0024, Japan
| | - Akikazu Yago
- Department of Surgery, Gastroenterological Center, Yokohama City University, 4-57, Urafune-chi, Minami-ku, Yokohama, 232-0024, Japan
| | - Yuko Tamura
- Department of Surgery, Gastroenterological Center, Yokohama City University, 4-57, Urafune-chi, Minami-ku, Yokohama, 232-0024, Japan
| | - Hiroki Kondo
- Department of Surgery, Gastroenterological Center, Yokohama City University, 4-57, Urafune-chi, Minami-ku, Yokohama, 232-0024, Japan
| | - Masanori Oshi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan, 236-0004
| | - Takashi Kosaka
- Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan, 236-0004
| | - Hirotoshi Akiyama
- Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan, 236-0004
| | - Itaru Endo
- Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan, 236-0004
| |
Collapse
|
26
|
Wang B, Han X, Zhang Z, Song H, Song Y, Liu R, Li Z, Liu S. Longitudinal CT Radiomics to Predict Progression-free Survival in Patients with Locally Advanced Gastric Cancer After Neoadjuvant Chemotherapy. Acad Radiol 2025; 32:2618-2629. [PMID: 39732617 DOI: 10.1016/j.acra.2024.11.068] [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: 09/20/2024] [Revised: 11/24/2024] [Accepted: 11/28/2024] [Indexed: 12/30/2024]
Abstract
RATIONALE AND OBJECTIVES To develop and validate a radiomics signature, utilizing baseline and restaging CT, for preoperatively predicting progression-free survival (PFS) after neoadjuvant chemotherapy (NAC) in locally advanced gastric cancer (LAGC). METHODS A total of 316 patients with LAGC who received NAC followed by gastrectomy were retrospectively included in this single-center study; these patients were split into two cohorts, one for training (n = 243) and the other for validation (n = 73), based on the different districts of our hospital. A total of 1316 radiomics features were extracted from the volume of interest of the gastric-cancer lesion on venous phase CT images. Four radiomics signatures were built for predicting PFS based on baseline CT (Pre-Rad), restaging CT (Post-Rad), delta radiomics (Delta-Rad) and multi-time radiomics (PrePost-Rad), respectively. Then the PrePost-Rad was combined with clinical factors to establish a nomogram (Rad-clinical model). Kaplan-Meier survival curves with log-rank tests were used to assess the prognostic usefulness of the Rad-clinical model. RESULTS All baseline characteristics were not statistically different between the two cohorts. The PrePost-Rad achieved improved predictive value by a C-index of 0.724 (95% CI: 0.639-0.809) in the validation cohort [Pre-Rad: 0.715 (0.632-0.798); Post-Rad: 0.632 (0.538-0.725), Delta-Rad: 0.549 (0.447-0.651)]. In terms of clinical benefit, calibration capability, and prediction efficacy, the Rad-clinical model performed well for PFS prediction, with a C-index of 0.754 (95% CI: 0.707-0.800) and 0.719 (95% CI: 0.639-0.800) in the training and validation cohorts, respectively, superior to the clinical model (cN stage and CA199) but comparable to the PrePost-Rad. Moreover, the Rad-clinical model could accurately classify gastric-cancer patients after NAC into three PFS risk groups in both training and validation cohorts. The risk stratification also performed well in most subgroups (good responders, poor responders, ypTNM Ⅱ, and ypTNM Ⅲ/Ⅳ). CONCLUSIONS The Rad-clinical model integrating longitudinal radiomics score and clinical factors performed well in preoperatively predicting PFS of LAGC patients after NAC and surgery.
Collapse
Affiliation(s)
- Bo Wang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China (B.W., X.H., Z.Z., Z.L., S.L.)
| | - Xiaomeng Han
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China (B.W., X.H., Z.Z., Z.L., S.L.)
| | - Zaixian Zhang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China (B.W., X.H., Z.Z., Z.L., S.L.)
| | - Hongzheng Song
- Department of Radiology, Qingdao Municipal Hospital, Shandong Province, Qingdao, Shandong Province, China (H.S.)
| | - Yaolin Song
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China (Y.S.)
| | - Ruiqing Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China (R.L.)
| | - Zhiming Li
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China (B.W., X.H., Z.Z., Z.L., S.L.)
| | - Shunli Liu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China (B.W., X.H., Z.Z., Z.L., S.L.).
| |
Collapse
|
27
|
Sundar R, Nakayama I, Markar SR, Shitara K, van Laarhoven HWM, Janjigian YY, Smyth EC. Gastric cancer. Lancet 2025:S0140-6736(25)00052-2. [PMID: 40319897 DOI: 10.1016/s0140-6736(25)00052-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 11/13/2024] [Accepted: 01/09/2025] [Indexed: 05/07/2025]
Abstract
Gastric cancer remains a major health challenge worldwide, with nearly 1 million new cases annually contributing to more than 650 000 deaths. Epidemiologically, gastric cancer shows substantial geographical variation in incidence, with higher rates in Asia, South America, and eastern Europe, and a rapid increase in early-onset cases among people younger than 50 years. Key risk factors for gastric cancer include Helicobacter pylori infection, diet, obesity, smoking, and genetic predisposition. Early detection through comprehensive diagnostic procedures is crucial for optimising treatment outcomes. Standard treatment approaches for locally advanced gastric cancer include surgical resection, particularly D2 lymphadenectomy, complemented by chemotherapy and radiotherapy. There is increasing implementation of minimally invasive surgical techniques for operable disease and integration of immune checkpoint inhibitors and targeted therapies for advanced stages. Emerging therapies, such as novel targeted treatments and next-generation immunotherapies, show promise in improving survival and quality of life. Future directions in the management of gastric cancer focus on precision medicine, continued advancement in immunotherapy, novel early detection methods, and a multidisciplinary approach to care. These strategies aim to enhance the overall effectiveness of treatment and prognosis worldwide.
Collapse
Affiliation(s)
- Raghav Sundar
- Department of Medicine, Section of Medical Oncology, Yale School of Medicine, New Haven, CT, USA; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Izuma Nakayama
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Sheraz R Markar
- Surgical Intervention Trials Unit, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hanneke W M van Laarhoven
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands; Department of Medical Oncology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Yelena Y Janjigian
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| | - Elizabeth C Smyth
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK.
| |
Collapse
|
28
|
Bahar F, Ibis B, Cakir Colak S, Banga A, Song J, Chang YC, Chi KY, Chang Y, Chiang CH, Chiang CH. Cardiovascular and thromboembolic outcomes with immune checkpoint inhibitors in gastroesophageal cancer: a propensity score-matched cohort study. Gastric Cancer 2025; 28:550-555. [PMID: 39838247 DOI: 10.1007/s10120-025-01582-1] [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: 11/06/2024] [Accepted: 01/06/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have been associated with an increased risk of cardiovascular and thromboembolic events. However, the incidence of cardiovascular and thromboembolic events associated with ICIs in gastroesophageal cancers is unknown. METHODS We performed a propensity score-matched cohort study using the TriNetX Analytics Network database, which comprises de-identified data from over 130 participating healthcare institutions. Patients who received ICI and chemotherapy were compared with those who received only chemotherapy. The primary outcomes were cardiovascular events including pericarditis, myocarditis, heart failure, myocardial infarction, ischemic stroke, atrial fibrillation, conduction disorders as well as venous thromboembolism (VTE) within 1-year of ICI or chemotherapy. We matched the cohorts based on predetermined variables including demographics, metastatic disease, chemotherapy, underlying comorbidities, and the use of cardiovascular and lipid-lowering medications. RESULTS We identified 1,448 patients who received ICI and chemotherapy and 11,966 patients who received chemotherapy only. After matching, 1,425 patients remained in each cohort. The mean age was 63.1 ± 12.7 years in the ICI and chemotherapy cohort and 62.9 ± 12.1 years in the chemotherapy-only cohort. ICI was associated with a higher incidence of pericarditis (45.6 vs. 30.9 cases per 1000 patient-years; HR 1.51 [95% CI 1.03-2.22]) and VTE (102.5 vs. 75.1 cases per 1000 patient-years; HR 1.40 [95% CI 1.09-1.80]). The incidence of other cardiovascular outcomes were similar between the two cohorts. CONCLUSION In this cohort study, the use of ICI and chemotherapy was associated with an approximately 40-50% increased risk of pericarditis and VTE than patients on chemotherapy only.
Collapse
Affiliation(s)
- Furkan Bahar
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, 330 Mt Auburn St, Cambridge, MA, MA 02138, USA
| | - Betul Ibis
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, 330 Mt Auburn St, Cambridge, MA, MA 02138, USA
| | - Sena Cakir Colak
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, 330 Mt Auburn St, Cambridge, MA, MA 02138, USA
| | - Akshat Banga
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, 330 Mt Auburn St, Cambridge, MA, MA 02138, USA
| | - Junmin Song
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yu-Cheng Chang
- Department of Medicine, Danbury Hospital, Danbury, CT, USA
| | - Kuan-Yu Chi
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yu Chang
- Section of Neurosurgery, Department of Surgery, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cho-Hung Chiang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Cho-Han Chiang
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, 330 Mt Auburn St, Cambridge, MA, MA 02138, USA.
| |
Collapse
|
29
|
Zhang J, Su C, Zhang Y, Gao R, Lu X, Liang J, Liu H, Tian S, Zhang Y, Ye Z. Spectral CT-based nomogram for preoperative prediction of Lauren classification in locally advanced gastric cancer: a prospective study. Eur Radiol 2025; 35:2794-2805. [PMID: 39532722 DOI: 10.1007/s00330-024-11163-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: 04/17/2024] [Revised: 08/24/2024] [Accepted: 09/26/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES To develop a nomogram based on clinical features and spectral quantitative parameters to preoperatively predict the Lauren classification for locally advanced gastric cancer (LAGC). METHODS Patients diagnosed with LAGC by postoperative pathology who underwent abdominal triple-phase enhanced spectral computed tomography (CT) were prospectively enrolled in this study between June 2023 and December 2023. All the patients were categorized into intestinal- and diffuse-type groups according to the Lauren classification. Traditional characteristics, including demographic information, serum tumor markers, gastroscopic pathology, and image semantic features, were collected. Spectral quantitative parameters, including iodine concentration (IC), effective atomic number (Zeff), and slope of the energy spectrum curve from 40 keV to 70 keV (λ), were measured three times for each patient by two blinded radiologists in arterial/venous/delayed phases (AP/VP/DP). Differences in traditional features and spectral quantitative parameters between the two groups were compared using univariable analysis. Independent predictors of the Lauren classification of LAGC were screened using multivariable logistic regression analysis. Receiver operating characteristic (ROC) curve analysis was used to assess the discriminating capability. Ultimately, the nomogram, including clinical features and spectral CT quantitative parameters, was developed. RESULTS Gender, nIC in AP (APnIC), and λ in DP (λd) were independent predictors for Lauren classification. The nomogram based on these indicators produced the best performance with an area under the curve of 0.841 (95% confidence interval: 0.749-0.932), specificity of 85.3%, accuracy of 76.4%, and sensitivity of 68.4%. CONCLUSION The nomogram based on clinical features and spectral CT quantitative parameters exhibits great potential in the preoperative and non-invasive assessment of Lauren classification for LAGC. KEY POINTS Question Can the proposed nomogram, integrating clinical features and spectral quantitative parameters, preoperatively predict the Lauren classification in locally advanced gastric cancer (LAGC)? Findings The nomogram, based on gender, arterial phase normalized iodine concentration, and slope of the energy spectrum curve in the delayed phase showed satisfactory predictive ability. Clinical relevance The established nomogram could contribute to guiding individualized treatment strategies and risk stratification in patients by predicting the Lauren classification for LAGC before surgery.
Collapse
Affiliation(s)
- Juan Zhang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy; Tianjin Key Laboratory of Digestive Cancer; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, China, Tianjin, China
- Department of Radiology, The Second Affiliated Hospital of Shandong First Medical University, Shandong, China
| | - Chao Su
- Department of General Surgery, The Second Affiliated Hospital of Shandong First Medical University, Shandong, China
| | - Yuyang Zhang
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Rongji Gao
- Department of Radiology, The Second Affiliated Hospital of Shandong First Medical University, Shandong, China
| | | | - Jing Liang
- Graduate School, Tianjin Medical University, Tianjin, China
| | | | | | - Yitao Zhang
- Department of Radiology, The Second Affiliated Hospital of Shandong First Medical University, Shandong, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy; Tianjin Key Laboratory of Digestive Cancer; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, China, Tianjin, China.
| |
Collapse
|
30
|
Shang CY, Sun XP, Dong XS, Wang YS, Chen X, Qiao HQ. Biomarker-Based Models Utilizing the Albumin-Fibrinogen Ratio Effectively Predict Peritoneal Metastasis in Patients with Gastric Cancer: A Retrospective Study. Curr Med Sci 2025:10.1007/s11596-025-00052-0. [PMID: 40304936 DOI: 10.1007/s11596-025-00052-0] [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: 01/05/2024] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 05/02/2025]
Abstract
OBJECTIVE Peritoneal carcinomatosis (PC) is a common pattern of recurrence in gastric cancer patients and is associated with a poor prognosis. This study aimed to evaluate the predictive value of the albumin-fibrinogen ratio (AFR) for PC in patients with gastric cancer and to develop two preoperative prediction models. METHODS A total of 745 gastric cancer patients were included in this study. Preoperative AFR, along with other serum markers and clinical tumor characteristics, was assessed. Univariate and multivariate logistic regression analyses were performed to determine the odds ratios (ORs) and 95% confidence intervals (CIs) of the independent variables. Propensity score matching (PSM) was used to control for potential confounders, and one-way ANOVA was conducted to evaluate differences in distribution between groups. Two prediction models incorporating the independent predictive indicators were constructed and validated via receiver operating characteristic (ROC) curves. RESULTS Poorly differentiated type (OR 2.679; P = 0.001), nondiffuse morphological type (OR 2.123; P = 0.040), BMI < 23.550 kg/m2 (OR 4.635; P = 0.001), AFR < 11.275 (OR 2.895; P = 0.003) and CA199 ≥ 73.615 U/mL (OR 2.040; P = 0.037) were identified as independent risk factors for PC in patients with gastric cancer. After PSM, the AFR remained the only inflammatory marker that was independently associated with PC (P = 0.003). AFR demonstrated consistent robustness in predicting PC across multiple sample sets. Among all the independent risk factors, the AFR had the highest area under the curve (AUC) for ROC analysis (AUC 0.648; 95% CI 0.580-0.715). Two combination models incorporating the AFR demonstrated enhanced predictive ability: Combination Model 1 (AUC 0.759; 95% CI 0.699-0.820) and Combination Model 2 (AUC 0.801; 95% CI 0.744-0.859). CONCLUSIONS The preoperative AFR serves as a useful indicator for predicting PC. Two reliable prediction models based on the AFR have been developed.
Collapse
Affiliation(s)
- Chun-Yang Shang
- Department of Gastrosplenic Surgery, Harbin Medical University, Harbin, 150000, China
| | - Xue-Pu Sun
- Department of Gastrosplenic Surgery, Harbin Medical University, Harbin, 150000, China
| | - Xue-Song Dong
- Department of Gastrosplenic Surgery, Harbin Medical University, Harbin, 150000, China
| | - Yang-Shuai Wang
- Department of Gastrosplenic Surgery, Harbin Medical University, Harbin, 150000, China
| | - Xiao Chen
- Department of Gastrosplenic Surgery, Harbin Medical University, Harbin, 150000, China
| | - Hai-Quan Qiao
- Department of Gastrosplenic Surgery, Harbin Medical University, Harbin, 150000, China.
| |
Collapse
|
31
|
Ramadhar A, Kagura J, Muchengeti M, Gaskill C, Khamisa N. Exploring the gastric cancer care pathway in South Africa. Afr J Prim Health Care Fam Med 2025; 17:e1-e8. [PMID: 40336421 DOI: 10.4102/phcfm.v17i1.4774] [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: 09/30/2024] [Revised: 01/20/2025] [Accepted: 01/26/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) diagnosis and care data in South Africa (SA) is sparse, and SA has a high GC mortality rate. Mapping the GC care pathway is needed to explore its efficacy in association with the SA GC burden and mortality. AIM The study aims to map the GC care pathway in SA from diagnosis to management by healthcare professionals (HCPs) involved in the GC patient journey and explore barriers and facilitators to the effective flow of the GC care pathway. SETTING Interviews conducted with South African HCPs were the data source used in this article for analysis. General physicians (GP) were the first contact point with chain-referral sampling sourcing other clinicians. METHODS Interviews were conducted via Microsoft Teams (MS Teams) and Google Meet with qualitative analyses via MAXQDA. RESULTS Themes identified were GC care pathway processes, public versus private healthcare system differences and care pathway challenges. Multidisciplinary team (MDT) care is practised for GC in SA, starting with the GP or nurse followed by gastroenterologist (GI), surgeon and pathologist. Thereafter, nurses, dieticians and palliative care specialists are involved. Healthcare sector differences are diagnosis time, GC staging, HCP and treatment access. Challenges include low GC index of suspicion by primary care clinicians (PCC) and Helicobacter pylori (H. pylori) detection. CONCLUSION A MDT approach for optimal treatment and patient care may be the best method for prolonged life.Contribution: A South African national consensus for GC care via a MDT, emphasising early diagnosis to aid in a robust treatment plan for improved patient outcomes is warranted.
Collapse
Affiliation(s)
- Anishka Ramadhar
- Division of Epidemiology and Biostatistics, School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg.
| | | | | | | | | |
Collapse
|
32
|
Aoyama T, Ooki A, Oba K, Nishikawa K, Kawabata R, Honda M, Maeda H, Kanda M, Sugiyama K, Makiyama A, Segami K, Takahashi M, Shindo Y, Namikawa T, Oshima T, Katayama A, Shiosakai K, Sakamoto J. A multicenter randomized open-label phase 2 study investigating optimal antiemetic therapy for patients with advanced/recurrent gastric cancer treated with trastuzumab deruxtecan: the EN-hance study. Int J Clin Oncol 2025:10.1007/s10147-025-02748-8. [PMID: 40293658 DOI: 10.1007/s10147-025-02748-8] [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: 10/15/2024] [Accepted: 03/14/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Trastuzumab deruxtecan (T-DXd) has been approved for the treatment of human epidermal growth factor receptor-2 (HER2)-positive gastric cancer and other indications in several countries and is considered moderately or highly emetogenic. The management of nausea and vomiting associated with T-DXd treatment has not been fully evaluated and the effectiveness of conventional prophylaxis remains unknown. METHODS This open-label, randomized, multicenter, phase 2 study aimed to investigate the optimal antiemetic therapy for Japanese patients with gastric cancer undergoing T-DXd treatment. Patients were randomized to a doublet regimen group (dexamethasone and palonosetron) or triplet regimen group (aprepitant, dexamethasone, and palonosetron) at a ratio of one to one, stratified by sex, gastrectomy status, and study institution. Both antiemetic treatments were administered from day 1 before T-DXd administration, and emetic events and nausea were observed for 21 days. The primary endpoint was the antiemetic complete response (CR) rate to assess control for emetic events based on voluntary patient-reported outcomes (PROs) during cycle 1 (1-21 days). RESULTS Of the 60 enrolled patients, 58 were eligible for inclusion in this analysis (29 patients in each regimen group). The overall CR rates for the doublet and triplet regimens were 41.4% (12/29 patients) and 37.9% (11/29 patients), respectively, and neither regimen met the pre-specified threshold (> 18/29 patients). The CR rate in the acute phase (0-24 h) was 86.2% (25/29 patients) for both regimens, and the CR rates in the delayed phase (2-21 days) were 41.4% (12/29 patients) and 37.9% (11/29 patients) for the doublet and triplet regimens, respectively. CONCLUSIONS Given that the primary endpoint was not met, further research is needed to better characterize nausea and vomiting with T-DXd to tailor an anti-emetic regimen that suits the needs of the patients.
Collapse
Affiliation(s)
- Toru Aoyama
- Department of Surgery, Yokohama City University Hospital, Yokohama, Japan.
- Department of Gastric Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan.
| | - Akira Ooki
- The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto, Japan
| | - Koji Oba
- The University of Tokyo Graduate School of Medicine, Bunkyō, Japan
| | | | | | | | | | - Mitsuro Kanda
- Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | - Kenki Segami
- Saiseikai Yokohama-Shi Nanbu Hospital, Yokohama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Xu L, Li M, Dong X, Wang Z, Tong Y, Feng T, Xu S, Shang H, Zhao B, Lin J, Cao Z, Zheng Y. The value of deep learning and radiomics models in predicting preoperative serosal invasion in gastric cancer: a dual-center study. Abdom Radiol (NY) 2025:10.1007/s00261-025-04949-1. [PMID: 40285792 DOI: 10.1007/s00261-025-04949-1] [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: 02/18/2025] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025]
Abstract
PURPOSE To establish and validate a model based on deep learning (DL), integrating radiomic features with relevant clinical features to generate nomogram, for predicting preoperative serosal invasion in gastric cancer (GC). METHODS This retrospective study included 335 patients from dual centers. T staging (T1-3 or T4) was used to assess serosal invasion. Radiomic features were extracted from primary GC lesions in the venous phase CT, and DL features from 8 transfer learning models were combined to create the Hand-crafted Radiomics and Deep Learning Radiomics (HCR-DLR) model. The Clinical (CL) model was built using clinical features, and both were combined into the Clinical and Radiomics Combined (CRC) model. In total, 15 predictive models were developed using 5 machine learning algorithms. The best-performing models were visualized as nomograms. RESULTS The total of 14 radiomic features, 13 DL features, and 2 clinical features were considered valuable through dimensionality reduction and selection. Among the constructed models: CRC model (AUC, training cohort: 0.9212; internal test cohort: 0.8743; external test cohort: 0.8853) than HCR-DLR model (AUC, training cohort: 0.8607; internal test cohort: 0.8543; external test cohort: 0.8824) and CL model (AUC, training cohort: 0.7632; internal test cohort: 0.7219; external test cohort: 0.7294) showed better performance. A nomogram based on the logistic CL model was drawn to facilitate the usage and showed its excellent predictive performance. CONCLUSION The predictive performance of the CRC Model, which integrates clinical features, radiomic features, and DL features, exhibits robust predictive capability and can serve as a simple, non-invasive, and practical tool for predicting the serosal invasion status of GC.
Collapse
Affiliation(s)
- Lihang Xu
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Mingyu Li
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Xianling Dong
- Hebei International Research Center for Medical-Engineering, Chengde Medical University, Chengde, China
| | - Zhongxiao Wang
- Hebei International Research Center for Medical-Engineering, Chengde Medical University, Chengde, China
| | - Ying Tong
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Tao Feng
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Shuangyan Xu
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Hui Shang
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Bin Zhao
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Jianpeng Lin
- Hebei International Research Center for Medical-Engineering, Chengde Medical University, Chengde, China
| | - Zhendong Cao
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China.
| | - Yi Zheng
- Radiology department, Chengde Central Hospital, Chengde, China.
| |
Collapse
|
34
|
Lin X, Xu S, Wang Y, Ma F, Qu J, Wu Y, Li J. Sequential dual-energy CT for longitudinal assessment of pathologic response to neoadjuvant immuno-chemotherapy in locally advanced gastric cancer. Eur Radiol 2025:10.1007/s00330-025-11601-5. [PMID: 40278873 DOI: 10.1007/s00330-025-11601-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/17/2025] [Accepted: 03/23/2025] [Indexed: 04/26/2025]
Abstract
OBJECTIVES To longitudinally evaluate pathologic response outcomes after neoadjuvant immuno-chemotherapy (NICT) for patients with locally advanced gastric cancer (LAGC) using pre- and post-treatment dual-energy CT (DECT). MATERIALS AND METHODS Between Jan 2021 and Dec 2023, 115 patients who underwent NICT plus gastrectomy and triple-phase enhanced DECT scans before and after NICT were retrospectively enrolled. Pathologic tumor regression grade (TRG) was the reference standard, patients were labelled as responders (TRG = 0 + 1) and non-responders (TRG = 2 + 3) accordingly. A two-dimensional free-hand region of interest method was adopted to obtain the iodine concentration (IC) values. Pre- and post-NICT IC and normalized IC (nIC) were measured at arterial/venous/delay phase (AP/VP/DP), respectively; their changes [ΔIC (%)] defined as (IC_post-IC_pre)⁄IC_pre × 100% were calculated. Pre- and post-NICT imaging parameters and their changes were compared between different response groups. Non-responders' associated parameters were selected using multivariable logistic regression analysis. Their performances were analyzed by the area under the receiver operating characteristic curve (AUC). Their associations with patient survival were explored by using Kaplan-Meier survival analysis. RESULTS ICDP-pre, ΔICAP, thickness-post with cut-off value of > 2.306 mg/mL, ≤ 26.70%, > 18.5 mm, respectively, indicates non-responders with equivalent AUC being 0.616 (95% CI: 0.521-0.705), 0.625 (95% CI: 0.529-0.713), and 0.660 (95% CI: 0.565-0.745). Their combination demonstrated an improved AUC of 0.774 (95% CI: 0.686-0.846) and was associated with patient disease-free survival (DFS) with a hazard ratio being 2.239 (95% CI: 1.004-4.991) (p = 0.026). CONCLUSION Pre- and post-NICT DECT-based quantifications are useful for longitudinal assessment of pathologic response outcomes after NICT in LAGC. ICDP-pre, ΔICAP, and thickness-post are equally useful, their combination demonstrated incremental benefit. KEY POINTS Question Accurate evaluation of the efficacy of NICT in patients with LAGC remains challenging due to the lack of effective biomarkers. Findings Sequential DECT-based ICDP-pre, ΔICAP, and tumor thickness-post were predictive of TRG status. Their combination demonstrated enhanced performance and was associated with patient DFS. Clinical relevance DECT represents a promising imaging technique with added advantages for longitudinal assessment of pathologic response to NICT in LAGC, potentially facilitating more personalized treatment strategies among this population.
Collapse
Affiliation(s)
- Xiaoxiao Lin
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Shuning Xu
- Department of Gastrointestinal Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Yi Wang
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Fei Ma
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Jinrong Qu
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Yue Wu
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Jing Li
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China.
| |
Collapse
|
35
|
Wang XY, Yang FH, Yuan ZY, Wang ZJ, Zhang HF, Xu ZH. Downregulation of huntingtin-associated protein 1 predicts poor prognosis in gastric cancer. World J Clin Oncol 2025; 16:102958. [PMID: 40290688 PMCID: PMC12019257 DOI: 10.5306/wjco.v16.i4.102958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/18/2025] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Highly expressed in the gastrointestinal mucosa, huntingtin-associated protein 1 (HAP1) is closely associated with tumor development and prognosis. AIM To investigate the clinical utility of HAP1 expression in gastric cancer (GC). METHODS We randomly selected 124 GC patients had not undergone preoperative radiotherapy or chemotherapy, they were diagnosed at the Central Hospital of Wuhan between May 2013 and October 2018. Immunohistochemistry was used to detect HAP1 expression in paraffin-embedded GC tissues, as well as metastatic lymph nodes. Their clinical data were collected and all participants were follow up for 5 years. Western blotting and quantitative polymerase chain reaction were used to detect HAP1 levels in 20 matched pairs of fresh GC tissues. RESULTS HAP1 protein and mRNA levels were lower in fresh GC tissues than in normal mucosal tissues (P < 0.001, respectively). Immunohistochemistry also revealed lower HAP1 expression in GC tissues and metastatic lymph nodes than in normal mucosal tissues (P < 0.05). HAP1 expression in GC was closely associated with differentiation, lymph node metastasis, lymph node ratio, remote metastasis, clinical stage, tumor location, and survival time (P < 0.05). Furthermore, HAP1 expression independently predicted GC (P < 0.05) and was more accurate in advanced GC than in early GC (P < 0.05). CONCLUSION HAP1 is an important prognostic biomarker for GC, with low HAP1 expression positively correlating with poor overall survival, especially in advanced clinical stages.
Collapse
Affiliation(s)
- Xiang-Yang Wang
- Department of Gastrointestinal Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, Hubei Province, China
| | - Fu-Hai Yang
- Department of Gastrointestinal Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, Hubei Province, China
| | - Zhong-Yuan Yuan
- Department of Gastrointestinal Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, Hubei Province, China
| | - Zhong-Jing Wang
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, Hubei Province, China
| | - Hong-Feng Zhang
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, Hubei Province, China
| | - Zi-Hui Xu
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, Hubei Province, China
| |
Collapse
|
36
|
Lin KT, Muneer G, Huang PR, Chen CS, Chen YJ. Mass Spectrometry-Based Proteomics for Next-Generation Precision Oncology. MASS SPECTROMETRY REVIEWS 2025. [PMID: 40269546 DOI: 10.1002/mas.21932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025]
Abstract
Cancer is the leading cause of death worldwide characterized by patient heterogeneity and complex tumor microenvironment. While the genomics-based testing has transformed modern medicine, the challenge of diverse clinical outcomes highlights unmet needs for precision oncology. As functional molecules regulating cellular processes, proteins hold great promise as biomarkers and drug targets. Mass spectrometry (MS)-based clinical proteomics has illuminated the molecular features of cancers and facilitated discovery of biomarkers or therapeutic targets, paving the way for innovative strategies that enhance the precision of personalized treatment. In this article, we introduced the tools and current achievements of MS-based proteomics, choice of discovery and targeted MS from discovery to validation phases, profiling sensitivity from bulk samples to single-cell level and tissue to liquid biopsy specimens, current regulatory landscape of MS-based protein laboratory-developed tests (LDTs). The challenges, success and future perspectives in translating research MS assay into clinical applications are also discussed. With well-designed validation studies to demonstrate clinical benefits and meet the regulatory requirements for both analytical and clinical performance, the future of MS-based assays is promising with numerous opportunities to improve cancer diagnosis, treatment, and monitoring.
Collapse
Affiliation(s)
- Kuen-Tyng Lin
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Gul Muneer
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | | | - Ciao-Syuan Chen
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Yu-Ju Chen
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
37
|
Zhou J, Lin L, He C, Wang Z, Zhan Y, Sun S, He Q. Integrating serological and drainage fluid indicators: developing two predictive models for early detection of postoperative intra-abdominal infections in gastrointestinal tumor patients. Front Oncol 2025; 15:1566954. [PMID: 40330827 PMCID: PMC12052565 DOI: 10.3389/fonc.2025.1566954] [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: 01/26/2025] [Accepted: 04/01/2025] [Indexed: 05/08/2025] Open
Abstract
Purpose This study aimed to investigate the influencing factors of postoperative intra-abdominal infection (PIAI) in gastrointestinal cancer patients by combining biomarkers in serum and drainage fluid (DF). It also intended to construct the predictive models and explore their predictive value for PIAI, offering clinical guidance. Methods 383 patients from Institution A formed the development cohort, and 77 patients from Institution B formed the validation cohort. Independent predictors of PIAI were identified using LASSO and logistic regression analysis based on biomarkers in serum and DF, and the corresponding nomograms were constructed. The nomograms were evaluated for their performance using the calibration curve, area under the curve (AUC), decision curve analysis (DCA), and clinical impact curve (CIC). Results The prevalence of PIAI was 15.9% in the development cohort and 24.7% in the validation cohort. There were 5 indicators included in the nomogram on postoperative day (POD) 1, and 4 indicators on POD 3, including DF lactate dehydrogenase and C-reactive protein. The AUC values of the models in the development and validation cohorts were 0.731 and 0.958 on POD 1, and 0.834 and 0.951 on POD 3, respectively. The calibration curve, DCA, and CIC demonstrated the favorable clinical applicability of the models. Conclusions Two nomogram models including serum and DF biomarkers on POD 1 and POD 3 were developed and validated. These models can identify patients at risk of PIAI and have promise for clinical application.
Collapse
Affiliation(s)
- Junfeng Zhou
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Lin Lin
- Nursing Department, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Cankun He
- Department of General Surgery, Huian County Hospital, Hui’an, China
| | - Ziyi Wang
- Emergency Department, Zigong First People’s Hospital, Zigong, China
| | - Yuping Zhan
- Department of Infectious diseases, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Infectious diseases, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Sida Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Qingliang He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Gastrointestinal Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
38
|
Li X, Lei T, Fu L, Gao R, Cao N, Gu Y, Su H, Guo T, Che Y. Meta-analysis of the efficacy of applying reduced surgery for the treatment of asymptomatic unresectable advanced gastric cancer. BMC Gastroenterol 2025; 25:271. [PMID: 40251493 PMCID: PMC12007127 DOI: 10.1186/s12876-025-03849-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/03/2025] [Indexed: 04/20/2025] Open
Abstract
OBJECTIVES Systematic evaluation of the efficacy and safety of reduction surgery in asymptomatic unresectable advanced gastric cancer. MATERIALS AND METHODS PubMed, EMBASE, Cochrane Library and Web of Science were searched from database inception to 12 July 2024. The Cochrane Risk of Bias Assessment Tool and Newcastle-Ottawa Scale were used to evaluate the quality and analyze the bias of the randomized controlled and non-randomized controlled studies included in this study, and RevMan (Version 5.4) was used to perform the meta-analysis. RESULTS A total of 5 studies were finally included, including 1 randomized controlled study and 4 retrospective studies. The cumulative sample size was 1717 cases, including 701 cases in the reduced surgery group and 1016 cases in the non-surgical treatment group. The results of the Meta-analysis showed that the reduced surgery group did not offer a survival benefit compared with the non-surgical treatment group in terms of 1-year, 3-year, and 5-year survival rates. The reduced surgery group had a longer median survival time than the non-surgical group by 11.58 months. The incidence rate, morbidity rate, and mortality rate of the reduced surgery group were 5.5% and 6.5% higher than those of the non-surgical group, respectively. The incidence of perioperative complications and death rate in the reduced surgery group were 15% and 4%, respectively; about 3% of patients might have complications of the primary foci during non-surgical treatment and need palliative surgical resection. CONCLUSION Current evidence suggests that in asymptomatic patients with unresectable advanced gastric cancer, reduced surgery with resection of the primary site does not result in a long-term survival benefit. We look forward to more high-quality randomized controlled trials to provide more substantial evidence to support clinical practice.
Collapse
Affiliation(s)
- Xiong Li
- Department Cadre Ward of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
- Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
| | - Ting Lei
- The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Liangyin Fu
- Department Cadre Ward of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
- Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
| | - Ruiyu Gao
- Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
| | - Ning Cao
- Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
| | - Yuanhui Gu
- Department Cadre Ward of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
- Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
| | - He Su
- Department Cadre Ward of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
- Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
| | - Tiankang Guo
- Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
| | - Yang Che
- Department Cadre Ward of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China.
- Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China.
| |
Collapse
|
39
|
Chen J, Hu K, Lu J, Pan H, Huang Y, Yu Z, Gao T, Fan Y, Li C, Liu F. Optimizing surgical timing following neoadjuvant therapy for gastric Cancer: Insights from a multicenter retrospective analysis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:110063. [PMID: 40288218 DOI: 10.1016/j.ejso.2025.110063] [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: 12/22/2024] [Revised: 03/16/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Neoadjuvant therapy (NAT) is a cornerstone in the treatment of locally advanced gastric cancer, improving surgical outcomes and survival. However, the optimal timing for surgery following NAT remains controversial. This study evaluates the impact of the interval between NAT and surgery on overall survival (OS) and explores associated clinicopathological factors. METHODS A retrospective analysis of 893 patients undergoing NAT and curative surgery for gastric adenocarcinoma across three centers in China was conducted. Surgical intervals were categorized (∼28 days, 29-42 days, >42 days). Survival analyses employed restricted cubic spline (RCS) models, Kaplan-Meier methods, and Cox proportional hazards regression. RESULTS Patients operated on within 28 days post-NAT had the most favorable OS, while intervals longer than 28 days were independently associated with worse outcomes. RCS analysis revealed increased risks for intervals longer than 28 days. Prolonged intervals showed declining effectiveness in tumor regression. Stratified analyses indicated that patients with poor NAT response (TRG 3) particularly benefited from surgery within 4 weeks, while delays were detrimental. CONCLUSIONS Timely surgery, especially within 4 weeks post-NAT, optimizes survival outcomes, particularly in patients with limited NAT response. This study underscores the need for individualized surgical timing and calls for prospective multicenter validation.
Collapse
Affiliation(s)
- Jie Chen
- Second Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Keshu Hu
- Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jun Lu
- Second Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hongda Pan
- Second Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yakai Huang
- Second Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhenjia Yu
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tianyu Gao
- Department of General Surgery, The Second Hospital of Lanzhou University, 730030, China
| | - Yong Fan
- Department of General Surgery, The Second Hospital of Lanzhou University, 730030, China.
| | - Chen Li
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Fenglin Liu
- Second Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Medical Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
40
|
Duan LW, Li J, Liu SQ, Wang CC, Liu HK, Hu SL, Liu SL, Ma JL, Xu JY. Screening of optimal candidates for postoperative radiotherapy among patients with resectable thymic tumors: a multicenter retrospective study. Lung Cancer 2025; 203:108542. [PMID: 40273484 DOI: 10.1016/j.lungcan.2025.108542] [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: 12/22/2024] [Revised: 04/06/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025]
Abstract
OBJECTIVE The role of postoperative radiotherapy (PORT) in resectable thymic epithelial tumors remains debatable. This study aimed to identify patient populations in whom PORT would be beneficial and analyzed the prognostic factors that influence survival in patients with resectable thymic tumors. METHODS A retrospective analysis was conducted on 497 patients with resectable thymic epithelial tumors, including cases treated at our facility (January 2014 to December 2018) and cases from the SEER database (January 2014 to December 2015). Patients' data analyzed included age, gender, Masaoka-Koga stage, postoperative pathological type, and adjuvant therapy. Survival analysis was performed using the Kaplan-Meier method, while prognostic factors were evaluated using the Cox proportional hazards model. RESULTS No significant difference in survival outcomes was observed between patients with Masaoka-Koga stage I/IIA thymic tumors who underwent surgery alone compared with those who had surgery with adjuvant radiotherapy. However, patients with stage IIB and III/IVA tumors had significant survival benefits from PORT. In indolent histological subtypes (types A, AB, and B1), PORT did not provide a significant survival advantage compared to surgery alone. Conversely, patients with aggressive subtypes (types B2 and B3) and thymic carcinoma (type C) experienced improved long-term survival with PORT. Additionally, male patients, along with those with tumors ≥ 55 mm, age < 58 years, and incomplete resection, also benefited from PORT. Univariate and multivariate Cox regression analyses revealed several prognostic factors that affect survival, including age, gender, Masaoka-Koga stage, histological type, tumor size, PORT, postoperative chemotherapy, and recurrence. Independent prognostic factors identified were sex, Masaoka-Koga stage, histological grading, PORT, and postoperative chemotherapy. CONCLUSIONS PORT provides significant survival benefits in patients with Masaoka-Koga stage IIB and III/IVA thymic tumors, aggressive histological subtypes, and thymic carcinoma.
Collapse
Affiliation(s)
- Li-Wen Duan
- Harbin Medical University Cancer Hospital, China.
| | - Jian Li
- Harbin Medical University Cancer Hospital, China
| | | | | | - Hong-Kai Liu
- Harbin Medical University Cancer Hospital, China
| | - Song-Liu Hu
- Harbin Medical University Cancer Hospital, China
| | - Shi-Long Liu
- Harbin Medical University Cancer Hospital, China
| | - Jian-Li Ma
- Harbin Medical University Cancer Hospital, China
| | - Jian-Yu Xu
- Harbin Medical University Cancer Hospital, China.
| |
Collapse
|
41
|
He WZ, Yang YZ, Yin CX, Xian XY, Gu JM, Yi JH, Xue J, Zhao Y, Wang F, Hu WM, Xia LP. Evolution of HER2-low expression from primary to paired metastatic gastric cancer lesions. NPJ Precis Oncol 2025; 9:108. [PMID: 40234621 PMCID: PMC12000306 DOI: 10.1038/s41698-025-00881-1] [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: 11/04/2024] [Accepted: 03/15/2025] [Indexed: 04/17/2025] Open
Abstract
HER2-low expression has recently gained considerable attention as an actionable biomarker in gastric cancer. However, changes in HER2-low expression between primary and metastatic gastric cancers remain inadequately explored. This study included consecutive patients diagnosed with metastatic gastric cancer with both primary and metastatic tumors, between January 2014 and December 2023. HER2 status was evaluated in both primary and matched metastatic tumors. A total of 332 patients were enrolled, with HER2-negative, HER2-low, and HER2-positive statuses were observed in 226, 81, and 25 primary tumors, respectively, and in 175, 104, and 53 metastatic tumors, respectively. Among the 226 patients with HER2-negative primary tumors, 74 and 23 developed HER2-low and HER2-positive metastatic tumors, respectively. Conversion from HER2-negative primary to HER2-low metastatic gastric cancer was associated with metachronous and non-peritoneal metastasis. Overall, re-biopsy to evaluate HER2 status may be necessary, potentially broadening the patient population eligible for targeted HER2 therapy.
Collapse
Affiliation(s)
- Wen-Zhuo He
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Yuan-Zhong Yang
- Department of Pathology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Chen-Xi Yin
- Department of Intensive Care Unit, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Xin-Yi Xian
- Department of Pathology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Jia-Mei Gu
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Jia-Hong Yi
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Ju Xue
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Yue Zhao
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Fang Wang
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China.
| | - Wan-Ming Hu
- Department of Pathology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China.
| | - Liang-Ping Xia
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China.
| |
Collapse
|
42
|
Chen Y, Jia K, Xie Y, Yuan J, Liu D, Jiang L, Peng H, Zhong J, Li J, Zhang X, Shen L. The current landscape of gastric cancer and gastroesophageal junction cancer diagnosis and treatment in China: a comprehensive nationwide cohort analysis. J Hematol Oncol 2025; 18:42. [PMID: 40234884 PMCID: PMC12001465 DOI: 10.1186/s13045-025-01698-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: 12/20/2024] [Accepted: 04/07/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Gastric cancer is the fifth most common cancer globally and is associated with significant morbidity and mortality. Despite its alarming prevalence, limited comparative evidence exists on its treatment efficacy and prognosis across diverse China populations. METHODS To address this, our study used a large-scale dataset from the National Cancer Information Database, including data from 220,304 patients from 53 leading hospitals across 27 provinces in China. RESULTS From 2017 to 2023, early-stage (Stages I-II) gastric cancer diagnoses increased to 35.63% of all cancer cases. Our study evaluated the neoadjuvant treatment strategies, adjuvant post-operative therapy, first- and second-line management for progressive stages, alongside current gastric cancer treatment guidelines in China. Notably, immunotherapy accounted for 16.17% and 23.28% of first- and second-line treatments for late-stage gastric cancers, and 14.56% and 5.00% for neoadjuvant and adjuvant therapies, respectively. Analysis of survival rates revealed that the 1-, 2-, 3-, 4-, and 5-year survival rates were 74.07%, 54.89%, 44.21%, 37.97%, and 33.53%, respectively. The 5-year survival rates across stages I-IV were 85.07%, 49.34%, 35.56%, and 13.15%, respectively. CONCLUSIONS These findings offer critical insights into the current state of gastric cancer treatment in China and can inform future initiatives to improve therapeutic outcomes for patients with gastric cancer.
Collapse
Affiliation(s)
- Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| | - Keren Jia
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yi Xie
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Jiajia Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Dan Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Lei Jiang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Haoxin Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | | | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
43
|
Du XY, Xia RJ, Shen LW, Ma JG, Yao WQ, Xu W, Lin ZP, Ma LB, Niu GQ, Fan RF, Xu SM, Yan L. Quadruple therapy with immunotherapy and chemotherapy as first-line conversion treatment for unresectable advanced gastric adenocarcinoma: A case report. World J Gastrointest Oncol 2025; 17:102258. [PMID: 40235902 PMCID: PMC11995329 DOI: 10.4251/wjgo.v17.i4.102258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/20/2025] [Accepted: 02/24/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND The treatment of gastric cancer remains highly challenging, particularly in cases of unresectable locally advanced or metastatic disease. Although chemotherapy and immunotherapy have shown some efficacy in such patients, significant limitations persist in extending survival and enhancing safety. To address these challenges, we designed an innovative first-line quadruple conversion therapy regimen that integrates a programmed cell death protein 1 (PD-1) inhibitor with chemotherapy, and we successfully implemented this therapy regimen in the treatment of a patient with unresectable locally advanced gastric adenocarcinoma. CASE SUMMARY We report the case of a 55-year-old male who was diagnosed with unresectable locally advanced gastric adenocarcinoma and presented with intermittent epigastric pain and multiple lymph node metastases in the abdominal cavity, with the metastasis being notably large in size. The tumor tissue was negative for human epidermal growth factor receptor 2 by immunohistochemistry. Considering the patient's status, the multidisciplinary team decided to administer sintilimab in combination with albumin-bound paclitaxel (nab-paclitaxel), S-1, and oxaliplatin as a quadruple drug conversion therapy. After 4 cycles of conversion therapy, the patient's epigastric pain was significantly alleviated, his stool color normalized, the volume of the primary tumor and lymph node metastases was markedly reduced, and the tumor marker levels decreased to within the normal range. The patient subsequently underwent laparoscopic total gastrectomy with abdominal lymph node dissection, and postoperative pathological biopsy revealed a pathological complete response and R0 resection, after which the patient recovered to an excellent physical status. CONCLUSION To the best of our knowledge, this is the first reported case of unresectable locally advanced gastric adenocarcinoma successfully treated with quadruple therapy with a PD-1 inhibitor and chemotherapy as a first-line conversion regimen. This first-line conversion therapy with the quadruple regimen may be effective and safe for unresectable locally advanced gastric adenocarcinoma.
Collapse
Affiliation(s)
- Xiao-Yu Du
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Medicine, Northwest Minzu University, Lanzhou 730050, Gansu Province, China
| | - Ren-Jie Xia
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Medicine, Northwest Minzu University, Lanzhou 730050, Gansu Province, China
| | - Li-Wen Shen
- Department of Medical Support Center, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Jian-Guo Ma
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730030, Gansu Province, China
| | - Wei-Qing Yao
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Medicine, Northwest Minzu University, Lanzhou 730050, Gansu Province, China
| | - Wei Xu
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Zhi-Peng Lin
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Liang-Bin Ma
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Guo-Qiang Niu
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Rui-Fang Fan
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Shu-Mei Xu
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Long Yan
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| |
Collapse
|
44
|
Jiang XH, Yuan XH, Chen JM, Yu H, Chen XJ, Chen YH, Li SJ, Wen YE, Peng JS. Effects of a nurse-led individualized mHealth nutrition intervention for post-discharged gastric cancer patients following gastrectomy: A randomized controlled trial. Int J Nurs Stud 2025; 168:105092. [PMID: 40253832 DOI: 10.1016/j.ijnurstu.2025.105092] [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: 08/20/2024] [Revised: 04/08/2025] [Accepted: 04/13/2025] [Indexed: 04/22/2025]
Abstract
OBJECTIVE To evaluate the effects of a nurse-led individualized mHealth nutrition intervention on nutritional status, nutritional intake, cognitive beliefs related to nutritional behavior, weight loss, blood parameters, gastrointestinal symptoms, and quality of life in post-discharged gastric cancer patients following gastrectomy. DESIGN This was an assessor-blinded parallel-arm randomized controlled trial with a repeated-measures design. SETTING(S) The participants were recruited from inpatient gastric surgery units of two tertiary hospitals in Guangzhou, China. PARTICIPANTS A total of 108 patients with gastric cancer who underwent gastrectomy and were scheduled to be discharged to their homes were included. METHOD Participants were randomly allocated to either the intervention or the control group (n = 54 per group). The intervention group received the nurse-led individualized mHealth nutrition intervention in addition to the usual care, whereas the control group received only usual care. The intervention included face-to-face education before discharge, the use of an applet and phone consultations. Baseline data were collected on the day of discharge from the hospital (T0). The patients' nutritional status, cognitive beliefs related to nutritional behavior, nutritional intake, weight loss, blood parameters, gastrointestinal symptoms and quality of life were repeatedly measured at 4-week (T1) and 12-week (T2) after discharge. RESULTS Compared with the control group, the intervention group showed significant improvement in nutritional status (ꞵ₄ weeks = -1.08, 95 % CI -2.12 to 0.04, p = 0.042; ꞵ₁₂ weeks = -1.52, 95 % CI -2.57 to -0.47, p = 0.005). Improvements were also observed in energy and protein intake, weight loss, and cognitive beliefs related to nutritional behavior, including risk perception, outcome expectancy, self-efficacy, intention, and action plan (p < 0.05). However, no statistically significant differences were observed in coping plan, gastrointestinal symptoms, blood parameters, and quality of life (p > 0.05). CONCLUSIONS The nurse-led individualized mHealth nutrition intervention was effective for improving the cognitive beliefs related to nutritional behavior, energy and protein intake, and nutritional status, as well as reducing weight loss among post-discharged gastric cancer patients following gastrectomy. REGISTRATION NUMBER ChiCTR2200064808.
Collapse
Affiliation(s)
- Xiao-Han Jiang
- School of Nursing, Sun Yat-sen University, Guangzhou, Guangdong, China; Department of Gastric Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiu-Hong Yuan
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jia-Min Chen
- Guangdong Province Chaozhou Health School, China
| | - Hong Yu
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xi-Jie Chen
- Department of Gastric Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yong-He Chen
- Department of Gastric Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Si-Jia Li
- Department of Gastric Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yue-E Wen
- Department of Gastric Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun-Sheng Peng
- School of Nursing, Sun Yat-sen University, Guangzhou, Guangdong, China; Department of Gastric Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
45
|
Dai XR, Zhang MZ, Chen L, Guo XW, Li ZX, Yan KF, He QQ, Cheng HW. Diagnostic value of systemic immune-inflammation index and prognostic nutritional index combined with CEA in gastric cancer with lymph node metastasis. Front Endocrinol (Lausanne) 2025; 16:1522349. [PMID: 40297178 PMCID: PMC12034562 DOI: 10.3389/fendo.2025.1522349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/10/2025] [Indexed: 04/30/2025] Open
Abstract
Background Carcinoembryonic antigen (CEA), systemic immune-inflammation index(SII), and prognostic nutritional index (PNI) are diagnostic markers for cancer, but their combined significance in gastric cancer (GC) with lymph node metastasis remains unclear. The aim of this study was to evaluate the association between these serum biomarkers and lymph node metastasis in patients with GC. Methods Records of patients with GC were reviewed retrospectively. Univariate and multivariate logistic regression were performed to examine the association between tumor markers, serum biomarkers and lymph node metastasis in GC. Based on the results of multivariate regression, a nomogram was developed and verified. Results Of the 395 patients aged 68.5 ± 9.1 years, 192 (48.6%) were diagnosed with lymphatic node metastasis. After adjusting for confounding factors, CEA (Odd ratio (OR):2.21; 95%CI: 1.17-3.81) and SII (OR:1.02; 95%CI: 1.01-1.04) was identified as significant risk factors, while PNI (OR:0.90; 95%CI: 0.85~0.96) was a protective factor for lymph node metastasis. The established nomogram by incorporating CEA, SII, PNI, differentiation, and tumor diameter can effectively predict lymph node metastasis in GC. Conclusion CEA, SII, PNI, differentiation, and tumor diameter were significantly associated with lymph node metastasis in patients with GC, and the combination of CEA, SII, PNI, differentiation, and tumor diameter has a better diagnostic value than either index alone.
Collapse
Affiliation(s)
- Xiao-rong Dai
- Department of Gastroenterology, Taixing People’s Hospital, Taixing, China
| | - Min-zhe Zhang
- School of Public Health, Wuhan University, Wuhan, China
| | - Lei Chen
- Department of Gastroenterology, Taixing People’s Hospital, Taixing, China
| | - Xin-wei Guo
- Department of Gastroenterology, Taixing People’s Hospital, Taixing, China
| | - Zhen-xing Li
- Department of Gastroenterology, Taixing People’s Hospital, Taixing, China
| | - Kun-feng Yan
- Department of Gastroenterology, Taixing People’s Hospital, Taixing, China
| | - Qi-qiang He
- School of Public Health, Wuhan University, Wuhan, China
- Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, Wuhan University, Wuhan, China
| | - Hong-wei Cheng
- Department of Gastroenterology, Taixing People’s Hospital, Taixing, China
| |
Collapse
|
46
|
Wu Y, Liu Y, Yang Y, Yao MS, Yang W, Shi X, Yang L, Li D, Liu Y, Yin S, Lei C, Zhang M, Gee JC, Yang X, Wei W, Gu S. A concept-based interpretable model for the diagnosis of choroid neoplasias using multimodal data. Nat Commun 2025; 16:3504. [PMID: 40223097 PMCID: PMC11994757 DOI: 10.1038/s41467-025-58801-7] [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: 08/26/2024] [Accepted: 04/02/2025] [Indexed: 04/15/2025] Open
Abstract
Diagnosing rare diseases remains a critical challenge in clinical practice, often requiring specialist expertise. Despite the promising potential of machine learning, the scarcity of data on rare diseases and the need for interpretable, reliable artificial intelligence (AI) models complicates development. This study introduces a multimodal concept-based interpretable model tailored to distinguish uveal melanoma (0.4-0.6 per million in Asians) from hemangioma and metastatic carcinoma following the clinical practice. We collected a comprehensive dataset on Asians to date on choroid neoplasm imaging with radiological reports, encompassing over 750 patients from 2013 to 2019. Our model integrates domain expert insights from radiological reports and differentiates between three types of choroidal tumors, achieving an F1 score of 0.91. This performance not only matches senior ophthalmologists but also improves the diagnostic accuracy of less experienced clinicians by 42%. The results underscore the potential of interpretable AI to enhance rare disease diagnosis and pave the way for future advancements in medical AI.
Collapse
Affiliation(s)
- Yifan Wu
- University of Pennsylvania, Philadelphia, PA, USA
| | - Yang Liu
- University of Electronic Science and Technology of China, Chengdu, China
| | - Yue Yang
- University of Pennsylvania, Philadelphia, PA, USA
| | | | - Wenli Yang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology and Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xuehui Shi
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology and Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Lihong Yang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology and Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Dongjun Li
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology and Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yueming Liu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology and Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Shiyi Yin
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology and Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Chunyan Lei
- Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Meixia Zhang
- Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, China
| | - James C Gee
- University of Pennsylvania, Philadelphia, PA, USA
| | - Xuan Yang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
- Beijing Ophthalmology and Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| | - Wenbin Wei
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
- Beijing Ophthalmology and Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| | - Shi Gu
- University of Electronic Science and Technology of China, Chengdu, China.
- College of Computer Science and Technology, Zhejiang University, Hangzhou, China.
- State Key Laboratory of Brain Machine Intelligence, Zhejiang University, Hangzhou, China.
| |
Collapse
|
47
|
Li R, Sun X, Yu Z, Zhu X, Zhao X, Li P, Liu N. Defining Optimal Lymph Node Yield in Gastrectomy: A Real-World Cohort Analysis. World J Surg Oncol 2025; 23:141. [PMID: 40217295 PMCID: PMC11992801 DOI: 10.1186/s12957-025-03787-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 03/29/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) has a high global mortality and incidence rate. Lymph node (LN) invasion is crucial in TNM staging, and an accurate LN staging system is vital for treatment decisions. However, the appropriate number of examined LNs remains uncertain. METHODS We retrospectively analyzed consecutive GC patients who underwent gastrectomy at the First Medical Center of the Chinese PLA General Hospital from January 2010 to December 2023. A new statistical model based on the β-binomial distribution and maximum likelihood method in R software was employed to calculate false-negative probabilities. RESULTS A total of 6463 GC patients were included. For cT1 patients, even with only five LNs excised, the likelihood of encountering occult positive LNs remained below 5%. For cT2 patients, 17 nodes were needed to rule out occult nodal disease with 90% confidence. While for cT3 and cT4 patients, even after the removal of 35 LNs, the likelihood of overlooking a positive node was still above 20%. Considering surgical extent, 25 nodes were required for patients who underwent proximal gastrectomy or distal gastrectomy to rule out occult nodal disease with 90% confidence, whereas those who received entire gastrectomy needed 59 nodes to achieve the same level of confidence. CONCLUSION Our study establishes a novel quantitative framework linking LN harvest thresholds to false-negative metastasis risk in GC, derived from real-world clinicopathological data.
Collapse
Affiliation(s)
- Rui Li
- School of Medicine, Nankai University, Tianjin, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, China
| | - Xu Sun
- School of Medicine, Nankai University, Tianjin, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, China
| | - Zhiyuan Yu
- School of Medicine, Nankai University, Tianjin, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, China
| | - Xiangchao Zhu
- Department of Gastrointestinal Surgery, Zibo Central Hospital, Zibo, China
| | - Xudong Zhao
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, China
| | - Peiyu Li
- School of Medicine, Nankai University, Tianjin, China.
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China.
- Medical School of Chinese PLA, Beijing, China.
| | - Na Liu
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China.
- Medical School of Chinese PLA, Beijing, China.
| |
Collapse
|
48
|
Nasca V, Bergamo F, Foltran L, Antonuzzo L, Bencardino K, Dell'Aquila E, Corallo S, Spallanzani A, Brunetti O, Spada D, Tamberi S, Cella CA, Avallone A, Fornaro L, Di Donato S, Strippoli A, Puccini A, Tamburini E, Palermo F, Morano F, Pietrantonio F, Raimondi A. Adjuvant TRastuzumab deruxtecan plus fluoropyrimidine versus standard chemotherapy in HER2-positive gastric or gastroesophageal cancer patients with persistence of minimal residual disease in liquid biopsy after pre-operative chemotherapy and radical surgery: the multicentre, phase II randomized TRINITY trial. BMC Cancer 2025; 25:633. [PMID: 40200187 PMCID: PMC11980183 DOI: 10.1186/s12885-025-14063-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 04/01/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND The standard treatment for localized/locally advanced gastroesophageal adenocarcinoma (GEA) is radical surgery and peri-operative FLOT treatment (5-fluorouracil plus leucovorin, oxaliplatin, and docetaxel), but around half patients still experience disease relapse. In gastrointestinal cancers, the presence of circulating tumor DNA (ctDNA) after surgery is associated with a high risk of relapse, and the lack of ctDNA clearance after post-operative treatment is strongly associated with early relapse. Therefore, liquid biopsy may guide the selection of patients with micrometastatic disease after preoperative chemotherapy and surgery for non-cross resistant regimens in the post-operative setting. Trastuzumab deruxtecan (T-DXd) is approved in patients with HER2-positive advanced gastric or gastroesophageal adenocarcinoma after failure of at least one prior trastuzumab-based regimen. The DESTINY-Gastric01 and 02 trials showed remarkable activity and efficacy of T-DXd, thus supporting the investigation of this agent in early-stage disease to increase the chance of achieving disease eradication. Finally, the DESTINY-Gastric03 trial showed the safety profile and feasibility, with preliminary promising activity results of the combination of T-DXd with a fluoropyrimidine. TRIAL DESIGN TRINITY is an ongoing multicentre, randomized, open-label, interventional phase II study which will enroll approximately 46 patients with HER2-positive GEA, treated with pre-operative FLOT and radical surgery, and with the persistence of minimal residual disease detected by the Signatera™ assay in a liquid biopsy collected between 2 and 6 weeks after surgery. The trial is designed with an observational phase enrolling patients with HER2-positive GEA eligible for standard treatment with peri-operative FLOT and surgery. Eligible patients will be randomized on a 1:1 basis to the experimental treatment arm consisting of adjuvant T-DXd (6.4 mg/kg IV on day 1) plus either capecitabine (1000 mg/sqm BID orally on days 1-14) or 5-fluorouracil (600 mg/sqm continuous IV infusion on days 1-5) Q3 W for 6 cycles, or to the control arm with standard post-operative FLOT (at the same dose used during the last pre-operative cycle) for 4 cycles. Patients non-eligible for the interventional trial will continue the standard therapy and follow-up in the frame of the observational phase with collection of exploratory longitudinal liquid biopsies. The primary objective is ctDNA clearance at 1 year after randomization. Considering alpha- and beta-errors of 0.10 and 0.20 and hypothesizing a ctDNA clearance of 10% and 35% in the control and experimental arm, respectively, 23 patients per arm are required to prove the superiority of the experimental strategy. Secondary endpoints include disease-free survival, overall survival, metastases-free survival, patient-reported outcomes and safety. The trial also represents a translational platform, including extensive analysis of circulating, tissue, and immune biomarkers as exploratory endpoints. Enrollment is active and ongoing. TRIAL REGISTRATION TRINITY is registered at ClinicalTrials.gov (NCT06253650).
Collapse
Affiliation(s)
- Vincenzo Nasca
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Francesca Bergamo
- Medical Oncology 1, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Luisa Foltran
- Centro Riferimento Oncologico (CRO) Aviano, National Cancer Institute IRCCS, Aviano, Italy
| | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Katia Bencardino
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Salvatore Corallo
- SC Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia, Italy
| | - Andrea Spallanzani
- Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | | | | | - Stefano Tamberi
- Oncology Unit, Ospedale Santa Maria Delle Croci, Ravenna, Italy
| | - Chiara Alessandra Cella
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, Istituto Europeo Di Oncologia IRCCS, Milan, Italy
| | - Antonio Avallone
- Istituto Nazionale Dei Tumori Napoli IRCCS Pascale, Naples, Italy
| | - Lorenzo Fornaro
- U.O. Oncologia Medica 2 Universitaria, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | | | - Antonia Strippoli
- Comprehensive Cancer Center, Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Alberto Puccini
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Emiliano Tamburini
- Department of Oncology and Palliative Care, Ospedale Cardinale Panico, Tricase, Italy
| | - Federica Palermo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Federica Morano
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy.
| | - Alessandra Raimondi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| |
Collapse
|
49
|
Li M, Jiang S, Zhou S, Chen W, Xiao Y, Fu Y, Feng F, Xu G. Radiomics-based assessment of HER2 status and prognosis in gastric cancer: a retrospective dual-center CT study. Abdom Radiol (NY) 2025:10.1007/s00261-025-04912-0. [PMID: 40195138 DOI: 10.1007/s00261-025-04912-0] [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: 02/04/2025] [Revised: 03/01/2025] [Accepted: 03/23/2025] [Indexed: 04/09/2025]
Abstract
PURPOSE This research investigated the potential of CT-based radiomics analysis for predicting human epidermal growth factor receptor 2 (HER2) status and assessing the prognosis of patients with gastric cancer (GC). METHODS 431 patients with GC from two medical centers were included in this retrospective study, with patients allocated to a training cohort (n = 221), a testing cohort (n = 94), and an external validation cohort (n = 116). Radiomics features and clinical variables associated with HER2 status were identified, and the radiomics score was subsequently derived. A radiomics model was constructed using the radiomics score, and a nomogram was developed by integrating related variables. The predictive accuracy of models was assessed via receiver operating characteristic curves, with the area under the curve (AUC) being computed. Prognostic significance was assessed by exploring the association between nomogram-predicted HER2 status and overall survival (OS). RESULTS The radiomics model yielded AUCs of 0.801, 0.793, and 0.784 for the training, testing, and external validation cohorts, respectively. A nomogram that integrated sex, CA72-4 levels, and radiomics score exhibited enhanced predictive accuracy, achieving AUCs of 0.847, 0.836, and 0.828 across the cohorts. Decision curve analysis highlighted the clinical utility of the nomogram, illustrating its ability to deliver a higher net benefit. In addition, survival analysis indicated that individuals with nomogram-predicted HER2 positivity experienced significantly shorter OS, providing robust risk stratification and prognostic insights. CONCLUSION The CT-based radiomics nomogram demonstrated the ability to non-invasively predict preoperative HER2 status and stratify prognostic risk in this GC cohort.
Collapse
Affiliation(s)
- Manman Li
- The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng, China
| | - Shu Jiang
- The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng, China
| | - Siyu Zhou
- Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Wang Chen
- The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng, China
| | - Yong Xiao
- The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng, China
| | - Yigang Fu
- The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng, China
| | - Feng Feng
- Affiliated Tumor Hospital of Nantong University, Nantong, China.
| | - Guodong Xu
- The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng, China.
- the Third Affiliated Hospital of Soochow University, Changzhou, China.
| |
Collapse
|
50
|
Albano F, Severini FL, Calice G, Zoppoli P, Falco G, Notarangelo T. The role of the tumor microenvironment and inflammatory pathways in driving drug resistance in gastric cancer: A systematic review and meta-analysis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167821. [PMID: 40203956 DOI: 10.1016/j.bbadis.2025.167821] [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: 01/28/2025] [Revised: 03/14/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025]
Abstract
Tumor microenvironment (TME) plays a pivotal role in progression and low responsiveness to chemotherapy of gastric cancer (GC). The cascade of events that culminate with a sustained and chronic activation of inflammatory pathways underlies gastric tumorigenesis. Infiltrating immune cells enrolling in crosstalk with cancer cells that regulate inflammatory and immune status, generating an immunosuppressive TME that influences the response to therapy. Here we discuss the role of TME and the activation of inflammatory pathways to comprehend strategies to improve drug response. Furthermore, we provides systematic insight the role of TME cytotypes and related signatures reinforcing the critical roles of TAMs and Tregs, in promoting GC chemoresistance and tumor progression.
Collapse
Affiliation(s)
- Francesco Albano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Francesca Lospinoso Severini
- Laboratory of Preclinical and Translational Research, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, PZ, Rionero in Vulture, Italy
| | - Giovanni Calice
- Laboratory of Preclinical and Translational Research, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, PZ, Rionero in Vulture, Italy
| | - Pietro Zoppoli
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Geppino Falco
- Department of Biology, University of Naples Federico II, Naples, Italy; Biogem, Istituto di Biologia e Genetica Molecolare, AV, Ariano Irpino, Italy
| | - Tiziana Notarangelo
- Laboratory of Preclinical and Translational Research, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, PZ, Rionero in Vulture, Italy.
| |
Collapse
|