1
|
Nakayama I. Therapeutic strategy for scirrhous type gastric cancer. Jpn J Clin Oncol 2025:hyaf081. [PMID: 40403741 DOI: 10.1093/jjco/hyaf081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 05/09/2025] [Indexed: 05/24/2025] Open
Abstract
Scirrhous-type gastric cancer (SGC) is a rare but well-recognized subset of resectable gastric cancer (GC), accounting for ⁓10% of cases. Despite its long history of clinical recognition dating back to the pre-1900s, SGC remains one of the most challenging GC subtypes to treat. Traditionally, SGC has been clinically defined as Borrmann type 4 GC, with histological classifications such as signet ring cell carcinoma or diffuse-type histology serving as alternative diagnostic criteria. Therapeutic advancements for SGC have largely focused on locally advanced or oligometastatic disease, yet no SGC-specific treatment has been established. The phase III JCOG0501 trial failed to demonstrate a survival benefit of neoadjuvant S-1 plus cisplatin for Borrmann type 4 and large type 3 GC. Recent developments in biomarker-driven therapies may redefine SGC by molecular subtypes, with CLDN18.2-targeted therapy emerging as a potential option for some SGC cases. However, as the landscape of medical oncology evolves, SGC may not remain a distinct therapeutic entity. The focus should shift toward understanding the intrinsic biology of SGC. Treatment development for SGC is expected to continue advancing, becoming increasingly stratified based on molecular abnormalities while maintaining a commitment to addressing unmet needs, such as early-onset GC and GC with symptomatic peritoneal dissemination.
Collapse
Affiliation(s)
- Izuma Nakayama
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwanoha 6-5-1, Kashiwa, 277-8577, Japan
| |
Collapse
|
2
|
Queiroz FR, Braga LDC, Melo CPDS, Gomes MDS, do Amaral LR, Salles PGDO. Cluster classification of a Brazilian gastric cancer cohort reveals remarkable populational differences in normal p53 rate. EINSTEIN-SAO PAULO 2024; 22:eAO0508. [PMID: 39356938 PMCID: PMC11461015 DOI: 10.31744/einstein_journal/2024ao0508] [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: 03/10/2023] [Accepted: 02/09/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Queiroz et al. showed that the application of cluster methodology for classifying gastric cancer is suitable and efficient within a Brazilian cohort, which is known for its population heterogeneity. The study highlighted the potential utilization of this method within public health services due to its low-cost, presenting a viable means to improve the diagnosis and prognosis of gastric cancer. BACKGROUND Our Brazilian cohort with gastric cancer has a distinct distribution between mutated and normal p53. BACKGROUND New genetic marker-based classifications improve gastric cancer diagnosis accuracy. BACKGROUND Machine learning integration enhances predictive value in gastric cancer diagnosis. BACKGROUND Molecular biomarkers complement clinical decisions, advancing personalized medicine. OBJECTIVE Gastric adenocarcinoma remains an aggressive disease with a poor prognosis, as evidenced by a 5-year survival rate of approximately 31%. The histological classifications already proposed do not accurately reflect the high biological heterogeneity of this neoplasm, particularly in diverse populations, and new classification systems using genetic markers have recently been proposed. Following these newly proposed models, we aimed to assess the cluster distribution in a Brazilian cohort. Furthermore, we evaluated whether the inclusion of other clinical and histological parameters could enhance the predictive value. METHODS We used a previously described four-immunohistochemistry/EBER-ISH marker to classify a cohort of 30 Brazilian patients with gastric adenocarcinoma into five different clusters and compared the distribution with other genetically diverse populations. Furthermore, we used artificial intelligence methods to evaluate whether other clinical and pathological parameters could improve the results of the methodology. RESULTS Disclosing the genetic variability between populations, we observed a more balanced distribution of the aberrant/normal p53 ratio (0.6) between patients negative for the other markers tested, unlike previous studies with Asian and North American populations. In addition, decision tree analysis reinforced the efficiency of these new classifications, as the stratification accuracy was not altered with or without additional data. CONCLUSION Our study underscores the importance of local research in characterizing diverse populations and highlights the complementary role of molecular biomarkers in personalized medicine for gastric adenocarcinoma, enhancing diagnostic accuracy and potentially improving survival rates.
Collapse
Affiliation(s)
- Fábio Ribeiro Queiroz
- Instituto Mário PennaBelo HorizonteMGBrazilInstituto Mário Penna, Belo Horizonte, MG, Brazil.
| | | | | | - Matheus de Souza Gomes
- Universidade Federal de UberlândiaLaboratório de Bioinformática e Análises MolecularesPatos de MinasMGBrazilLaboratório de Bioinformática e Análises Moleculares, Universidade Federal de Uberlândia, Patos de Minas, MG, Brazil.
| | - Laurence Rodrigues do Amaral
- Universidade Federal de UberlândiaLaboratório de Bioinformática e Análises MolecularesPatos de MinasMGBrazilLaboratório de Bioinformática e Análises Moleculares, Universidade Federal de Uberlândia, Patos de Minas, MG, Brazil.
| | | |
Collapse
|
3
|
Zhang Y, Yin XL, Ji M, Chen Y, Chai Z. Decoupling the dynamic mechanism revealed by FGFR2 mutation-induced population shift. J Biomol Struct Dyn 2024; 42:1940-1951. [PMID: 37254996 DOI: 10.1080/07391102.2023.2217924] [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: 03/01/2023] [Accepted: 04/08/2023] [Indexed: 06/01/2023]
Abstract
The fibroblast growth factor receptor 2 (FGFR2) is a key component in cellular signaling networks, and its dysfunctional activation has been implicated in various diseases including cancer and developmental disorders. Mutations at the activation loop (A-loop) have been suggested to trigger an increased basal kinase activity. However, the molecular mechanism underlying this highly dynamic process has not been fully understood due to the limitation of static structural information. Here, we conducted multiple, large-scale Gaussian accelerated molecular dynamics simulations of five (K659E, K659N, K659M, K659Q, and K659T) FGFR2 mutants at the A-loop, and comprehensively analyzed the dynamic molecular basis of FGFR2 activation. The results quantified the population shift of each system, revealing that all mutants had a higher proportion of active-like states. Using Markov state models, we extracted the representative structure of different conformational states and identified key residues related to the increased kinase activity. Furthermore, community network analysis showed enhanced information connections in the mutants, highlighting the long-range allosteric communication between the A-loop and the hinge region. Our findings may provide insights into the dynamic mechanism for FGFR2 dysfunctional activation and allosteric drug discovery.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yuxiang Zhang
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Lan Yin
- Department of Radiotherapy, Shanghai 411 Hospital, China RongTong Medical Healthcare Group Co. Ltd, Shanghai, China
| | - Mingfei Ji
- Department of Urology, The Second Affiliated Hospital of Navy Medical University, Shanghai, China
| | - Yi Chen
- Department of Ultrasound interventional, Eastern Hepatobiliary Surgery Hospital, Navy Medical University, Shanghai, China
| | - Zongtao Chai
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Hepatic Surgery, Shanghai Geriatric Medical Center, Shanghai, China
| |
Collapse
|
4
|
Gan L, Yang C, Zhao L, Wang S, Ye Y, Gao Z. L3MBTL3 Is a Potential Prognostic Biomarker and Correlates with Immune Infiltrations in Gastric Cancer. Cancers (Basel) 2023; 16:128. [PMID: 38201555 PMCID: PMC10778146 DOI: 10.3390/cancers16010128] [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: 10/10/2023] [Revised: 11/23/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Recent research has linked lethal (3) malignant brain tumor-like 3 (L3MBTL3) to cancer aggressiveness and a dismal prognosis, but its function in gastric cancer (GC) is unclear. This research investigated the association between L3MBTL3 expression and clinicopathological characteristics of GC cases, as well as its prognostic value and biological function based on large-scale databases and clinical samples. The results showed that L3MBTL3 expression was upregulated in malignant GC tissues, which was associated with a shortened survival time and poor clinicopathological characteristics, including TNM staging. A functional enrichment analysis including GO/KEGG and GSEA illustrated the enrichment of different L3MBTL3-associated pathways involved in carcinogenesis and immune response. In addition, the correlations between L3MBTL3 and tumor-infiltrating immune cells were determined based on the TIMER database; the results showed that L3MBTL3 was associated with the immune infiltration of macrophages and their polarization from M1 to M2. Furthermore, our findings suggested a possible function for L3MBTL3 in the regulation of the tumor immune microenvironment of GC. In summary, L3MBTL3 has diagnostic potential, and it also offers new insights into the development of aggressiveness and prognosis in GC.
Collapse
Affiliation(s)
- Lin Gan
- Department of Gastroenterological Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China;
| | - Changjiang Yang
- Department of Gastroenterological Surgery, Peking University People’s Hospital, Beijing 100044, China; (C.Y.); (L.Z.); (S.W.)
- Laboratory of Surgical Oncology, Peking University People’s Hospital, Beijing 100044, China
| | - Long Zhao
- Department of Gastroenterological Surgery, Peking University People’s Hospital, Beijing 100044, China; (C.Y.); (L.Z.); (S.W.)
- Laboratory of Surgical Oncology, Peking University People’s Hospital, Beijing 100044, China
| | - Shan Wang
- Department of Gastroenterological Surgery, Peking University People’s Hospital, Beijing 100044, China; (C.Y.); (L.Z.); (S.W.)
- Laboratory of Surgical Oncology, Peking University People’s Hospital, Beijing 100044, China
| | - Yingjiang Ye
- Department of Gastroenterological Surgery, Peking University People’s Hospital, Beijing 100044, China; (C.Y.); (L.Z.); (S.W.)
- Laboratory of Surgical Oncology, Peking University People’s Hospital, Beijing 100044, China
| | - Zhidong Gao
- Department of Gastroenterological Surgery, Peking University People’s Hospital, Beijing 100044, China; (C.Y.); (L.Z.); (S.W.)
- Laboratory of Surgical Oncology, Peking University People’s Hospital, Beijing 100044, China
| |
Collapse
|
5
|
Zhou H, Liu H, Li J, Wang J, Fu X, Li Y, Mao S, Du J. Postoperative circulating tumor DNA detection and CBLB mutations are prognostic biomarkers for gastric cancer. Genes Genomics 2023; 45:1037-1046. [PMID: 37306927 PMCID: PMC10349697 DOI: 10.1007/s13258-023-01412-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 05/30/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Several studies have demonstrated that circulating tumor DNA (ctDNA) can be used to predict the postoperative recurrence of several cancers. However, there are few studies on the use of ctDNA as a prognosis tool for gastric cancer (GC) patients. OBJECTIVE This study aims to determine whether ctDNA could be used as a prognostic biomarker in GC patients through multigene-panel sequencing. METHODS Using next-generation sequencing (NGS) Multigene Panels, the mutational signatures associated with the prognosis of GC patients were identified. We calculated the survival probability with Kaplan-Meier and used the Log-rank test to compare survival curves between ctDNA-positive and ctDNA-negative groups. Potential application of radiology combined with tumor plasma biomarker analysis of ctDNA in GC patients was carried out. RESULTS Disease progression is more likely in ctDNA-positive patients as characterized clinically by a generally higher T stage and a poorer therapeutic response (P < 0.05). ctDNA-positive patients also had worse overall-survival (OS: P = 0.203) and progression-free survival (PFS: P = 0.037). The combined analysis of ctDNA, radiological, and serum biomarkers in four patients indicated that ctDNA monitoring can be a good complement to radiological and plasma tumor markers for GC patients. Kaplan-Meier analysis using a cohort of GC patients in the TCGA database showed that patients with CBLB mutations had shorter OS and PFS than wild-type patients (OS: P = 0.0036; PFS: P = 0.0027). CONCLUSIONS This study confirmed the utility and feasibility of ctDNA in the prognosis monitoring of gastric cancer.
Collapse
Affiliation(s)
- Hekai Zhou
- Research Center for Clinical and Translational Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, and the 6th Affiliated Hospital of Shenzhen University Medical School, Guangdong, 518052, Shenzhen, China
| | - Houcong Liu
- Research Center for Clinical and Translational Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, and the 6th Affiliated Hospital of Shenzhen University Medical School, Guangdong, 518052, Shenzhen, China
| | - Jun Li
- HaploX Biotechnology Co., Ltd., Shenzhen, 518057, Guangdong, China
| | - Jidong Wang
- Research Center for Clinical and Translational Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, and the 6th Affiliated Hospital of Shenzhen University Medical School, Guangdong, 518052, Shenzhen, China
| | - Xiaohong Fu
- Department of Oncology, Huazhong University of Science and Technology Union Shenzhen Hospital, and the 6th Affiliated Hospital of Shenzhen University Medical School, Guangdong, 518052, Shenzhen, China
| | - Yingqiang Li
- HaploX Biotechnology Co., Ltd., Shenzhen, 518057, Guangdong, China
| | - Shaolong Mao
- HaploX Biotechnology Co., Ltd., Shenzhen, 518057, Guangdong, China
| | - Jihui Du
- Research Center for Clinical and Translational Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, and the 6th Affiliated Hospital of Shenzhen University Medical School, Guangdong, 518052, Shenzhen, China.
| |
Collapse
|
6
|
Shinozuka T, Kanda M, Kodera Y. Site-specific protein biomarkers in gastric cancer: a comprehensive review of novel biomarkers and clinical applications. Expert Rev Mol Diagn 2023; 23:701-712. [PMID: 37395000 DOI: 10.1080/14737159.2023.2232298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
INTRODUCTION Gastric cancer (GC) is the fifth most common cancer and the fourth leading cause of cancer-related death worldwide, thus representing a significant global health burden. Early detection and monitoring of GC are essential to improve patient outcomes. While traditional cancer biomarkers such as carcinoembryonic antigen, carbohydrate antigen (CA) 19-9, and CA 72-4 are widely used, their limited sensitivity and specificity necessitate the exploration of alternative biomarkers. AREAS COVERED This review comprehensively analyzes the landscape of GC protein biomarkers identified from 2019 to 2022, with a focus on tissue, blood, urine, saliva, gastric juice, ascites, and exhaled breath as sample sources. We address the potential clinical applications of these biomarkers in early diagnosis, monitoring recurrence, and predicting survival and therapeutic response of GC patients. EXPERT OPINION The discovery of novel protein biomarkers holds great promise for improving the clinical management of GC. However, further validation in large, diverse cohorts is needed to establish the clinical utility of these biomarkers. Integrating these biomarkers with existing diagnostic and monitoring approaches will likely lead to improved personalized treatment plans and patient outcomes.
Collapse
Affiliation(s)
- Takahiro Shinozuka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
7
|
Ooki A, Osumi H, Chin K, Watanabe M, Yamaguchi K. Potent molecular-targeted therapies for advanced esophageal squamous cell carcinoma. Ther Adv Med Oncol 2023; 15:17588359221138377. [PMID: 36872946 PMCID: PMC9978325 DOI: 10.1177/17588359221138377] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 10/21/2022] [Indexed: 01/15/2023] Open
Abstract
Esophageal cancer (EC) remains a public health concern with a high mortality and disease burden worldwide. Esophageal squamous cell carcinoma (ESCC) is a predominant histological subtype of EC that has unique etiology, molecular profiles, and clinicopathological features. Although systemic chemotherapy, including cytotoxic agents and immune checkpoint inhibitors, is the main therapeutic option for recurrent or metastatic ESCC patients, the clinical benefits are limited with poor prognosis. Personalized molecular-targeted therapies have been hampered due to the lack of robust treatment efficacy in clinical trials. Therefore, there is an urgent need to develop effective therapeutic strategies. In this review, we summarize the molecular profiles of ESCC based on the findings of pivotal comprehensive molecular analyses, highlighting potent therapeutic targets for establishing future precision medicine for ESCC patients, with the most recent results of clinical trials.
Collapse
Affiliation(s)
- Akira Ooki
- Department of Gastroenterological Chemotherapy,
Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31
Ariake, Koto-ku, Tokyo 135-8550, Japan
| | - Hiroki Osumi
- Department of Gastroenterological Chemotherapy,
Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo,
Japan
| | - Keisho Chin
- Department of Gastroenterological Chemotherapy,
Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo,
Japan
| | - Masayuki Watanabe
- Department of Gastroenterological Surgery,
Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo,
Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy,
Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo,
Japan
| |
Collapse
|
8
|
Abstract
Predictive biomarkers are the mainstay of precision medicine. This review summarizes the advancements in tissue-based diagnostic biomarkers for gastric cancer, which is considered the leading cause of cancer-related deaths worldwide. A disease seen in the elderly, it is often diagnosed at an advanced stage, thereby limiting therapeutic options. In Western countries, neoadjuvant/perioperative (radio-)chemotherapy is administered, and adjuvant chemotherapy is administered in the East. The morpho-molecular classification of gastric cancer has opened novel avenues identifying Epstein-Barr-Virus (EBV)-positive, microsatellite instable, genomically stable and chromosomal instable gastric cancers. In chromosomal instable tumors, receptor tyrosine kinases (RKTs) (e.g., EGFR, FGFR2, HER2, and MET) are frequently overexpressed. Gastric cancers such as microsatellite instable and EBV-positive types often express immune checkpoint molecules, such as PD-L1 and VISTA. Genomically stable tumors show alterations in claudin 18.2. Next-generation sequencing is increasingly being used to search for druggable targets in advanced palliative settings. However, most tissue-based biomarkers of gastric cancer carry the risk of a sampling error due to intratumoral heterogeneity, and adequate tissue sampling is of paramount importance.
Collapse
Affiliation(s)
- C. Röcken
- Department of Pathology, Christian-Albrechts-University, Arnold-Heller-Str. 3, Haus 14, Haus U33, 24105 Kiel, Germany
| |
Collapse
|
9
|
Li R, Yin B, Zeng D, Liu Z. A novobiocin derivative, XN4, triggers ferroptosis in gastric cancer cells via the activation of NOX4. PHARMACEUTICAL BIOLOGY 2022; 60:1449-1457. [PMID: 35938505 PMCID: PMC9361764 DOI: 10.1080/13880209.2022.2099431] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 06/28/2022] [Accepted: 07/04/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT A novobiocin derivative, XN4, has been shown to promote cell apoptosis in chronic myeloid leukaemia. OBJECTIVE This study explores the mechanism by which XN4 promotes ferroptosis of gastric cancer (GC) cells. MATERIALS AND METHODS Human GC SGC-7901 and BGC-823 cells were treated with different XN4 concentrations (0, 0.1, 0.5, 1.0, 5.0, and 10.0 μmol/L) to evaluate effects of XN4. Additionally, cells were pre-treated for 24 h with si-NOX4, for 1 h with the iron chelator deferoxamine mesylate (DFO) or for 1 h with the lipid peroxidation inhibitor liproxstatin-1 before being treated with XN4 to analyse the mechanism of XN4. RESULTS XN4 increased cell death (IC50 values of XN4 on SGC-7901 and BGC-823 cells: 1.592 ± 0.14 μmol/L and 2.022 ± 0.19 μmol/L) and Fe2+ levels in SGC-7901 and BGC-823 cells. These effects of 2.0 μmol/L XN4 were abolished by 100 μmol/L DFO treatment. XN4 enhanced transferrin and transferrin receptor expression to induce Fe2+ accumulation. XN4 decreased mitochondrial membrane potentials in GC cells, similar to erastin. Additionally, XN4 increased MDA, hydrogen peroxide, and ROS levels, but diminished total glutathione levels. Liproxstatin-1 (200 nmol/L) nullified the effects of XN4 (2.0 μmol/L) on MDA levels and cell death. Moreover, GPX4 levels decreased, but NOX4 and ferroptosis-related protein PTGS2 levels increased in GC cells following XN4 treatment, which was nullified by NOX4 knockdown. DISCUSSION AND CONCLUSIONS The pro-ferroptotic role of XN4 in GC might enable it to become a promising drug for GC treatment in the future despite the need for extensive research.
Collapse
Affiliation(s)
- Rongrong Li
- Department of Medical Oncology-Gastroenterology and Urology, Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Bin Yin
- Department of Gastric Pancreatic Surgery, Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Deyu Zeng
- Department of Medical Oncology-Gastroenterology and Urology, Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhenyang Liu
- Department of Medical Oncology-Gastroenterology and Urology, Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
10
|
Yang C, Song D, Zhao F, Wu J, Zhang B, Ren H, Sun Q, Qin S. Comprehensive analysis of the prognostic value and immune infiltration of FGFR family members in gastric cancer. Front Oncol 2022; 12:936952. [PMID: 36147913 PMCID: PMC9487308 DOI: 10.3389/fonc.2022.936952] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Background Fibroblast growth factor receptors (FGFRs) modulate numerous cellular processes in tumor cells and tumor microenvironment. However, the effect of FGFRs on tumor prognosis and tumor-infiltrating lymphocytes in gastric cancer (GC) remains controversial. Methods The expression of four different types of FGFRs was analyzed via GEPIA, TCGA-STAD, and GTEX databases and our 27 pairs of GC tumor samples and the adjacent normal tissue. Furthermore, the Kaplan–Meier plot and the TCGA database were utilized to assess the association of FGFRs with clinical prognosis. The R software was used to evaluate FGFRs co-expression genes with GO/KEGG Pathway Enrichment Analysis. In vitro and in vivo functional analyses and immunoblotting were performed to verify FGFR4 overexpression consequence. Moreover, the correlation between FGFRs and cancer immune infiltrates was analyzed by TIMER and TCGA databases. And the efficacy of anti-PD-1 mAb treatment was examined in NOG mouse models with overexpressed FGFR1 or FGFR4. Results The expression of FGFRs was considerably elevated in STAD than in the normal gastric tissues and was significantly correlated with poor OS and PFS. ROC curve showed the accuracy of the FGFRs in tumor diagnosis, among which FGFR4 had the highest ROC value. Besides, univariate and multivariate analysis revealed that FGFR4 was an independent prognostic factor for GC patients. According to a GO/KEGG analysis, the FGFRs were implicated in the ERK/MAPK, PI3K-AKT and extracellular matrix (ECM) receptor signaling pathways. In vivo and in vitro studies revealed that overexpression of FGFR4 stimulated GC cell proliferation, invasion, and migration. In addition, FGFR1 expression was positively correlated with infiltrating levels of CD8+ T-cells, CD4+ T-cells, macrophages, and dendritic cells in STAD. In contrast, FGFR4 expression was negatively correlated with tumor-infiltrating lymphocytes. Interestingly, overexpression of FGFR1 in the NOG mouse model improved the immunotherapeutic impact of GC, while overexpression of FGFR4 impaired the effect. When combined with an FGFR4 inhibitor, the anti-tumor effect of anti-PD-1 treatment increased significantly in a GC xenograft mouse model with overexpressed FGFR4. Conclusions FGFRs has critical function in GC and associated with immune cell infiltration, which might be a potential prognosis biomarker and predictor of response to immunotherapy in GC.
Collapse
Affiliation(s)
- Chengcheng Yang
- Department of Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Dingli Song
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Fengyu Zhao
- Department of Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jie Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Boxiang Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Hong Ren
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Qi Sun
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Sida Qin, ; Qi Sun,
| | - Sida Qin
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Sida Qin, ; Qi Sun,
| |
Collapse
|
11
|
Paolino G, Esposito I, Hong S, Basturk O, Mattiolo P, Kaneko T, Veronese N, Scarpa A, Adsay V, Luchini C. Intraductal tubulopapillary neoplasm (ITPN) of the pancreas: a distinct entity among pancreatic tumors. Histopathology 2022; 81:297-309. [PMID: 35583805 PMCID: PMC9544156 DOI: 10.1111/his.14698] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 05/03/2022] [Accepted: 05/16/2022] [Indexed: 11/30/2022]
Abstract
AIMS Intraductal tubulopapillary neoplasm (ITPN) of the pancreas is a recently recognized pancreatic tumor entity. Here we aimed to determine the most important features with a systematic review coupled with an integrated statistical approach. METHODS AND RESULTS PubMed, SCOPUS, and Embase were searched for studies reporting data on pancreatic ITPN. The clinicopathological, immunohistochemical, and molecular data were summarized. Then a comprehensive survival analysis and a comparative analysis of the molecular alterations of ITPN with those of pancreatic ductal adenocarcinoma (PDAC) and intraductal papillary mucinous neoplasm (IPMN) from reference cohorts (including the International Cancer Genome Consortium- ICGC dataset and The Cancer Genome Atlas, TCGA program) were conducted. The core findings of 128 patients were as follows: (i) Clinicopathological parameters: pancreatic head is the most common site; presence of an associated adenocarcinoma was reported in 60% of cases, but with rare nodal metastasis. (ii) Immunohistochemistry: MUC1 (>90%) and MUC6 (70%) were the most frequently expressed mucins. ITPN lacked the intestinal marker MUC2; unlike IPMN, it did not express MUC5AC. (iii) Molecular landscape: Compared with PDAC/IPMN, the classic pancreatic drivers KRAS, TP53, CDKN2A, SMAD4, GNAS, and RNF43 were less altered in ITPN (P < 0.001), whereas MCL amplifications, FGFR2 fusions, and PI3KCA mutations were commonly altered (P < 0.001). (iv) Survival analysis: ITPN with a "pure" branch duct involvement showed the lowest risk of recurrence. CONCLUSION ITPN is a distinct pancreatic neoplasm with specific clinicopathological and molecular characteristics. Its recognition is fundamental for its clinical/prognostic implications and for the enrichment of potential targets for precision oncology.
Collapse
Affiliation(s)
- Gaetano Paolino
- Department of Diagnostics and Public Health, Section of PathologyUniversity and Hospital Trust of VeronaVeronaItaly
| | - Irene Esposito
- Institute of PathologyUniversity Hospital of DuesseldorfDuesseldorfGermany
| | - Seung‐Mo Hong
- Department of Pathology, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Olca Basturk
- Department of PathologyMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Paola Mattiolo
- Department of Diagnostics and Public Health, Section of PathologyUniversity and Hospital Trust of VeronaVeronaItaly
| | - Takuma Kaneko
- Department of Hepato‐Biliary‐Pancreatic MedicineNTT Medical CenterTokyoJapan
| | - Nicola Veronese
- Department of Internal MedicineUniversity of PalermoPalermoItaly
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of PathologyUniversity and Hospital Trust of VeronaVeronaItaly
- ARC‐Net Research CenterUniversity and Hospital Trust of VeronaVeronaItaly
| | - Volkan Adsay
- Department of PathologyKoç University Hospital and Koç University Research Center for Translational Medicine (KUTTAM)IstanbulTurkey
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of PathologyUniversity and Hospital Trust of VeronaVeronaItaly
- ARC‐Net Research CenterUniversity and Hospital Trust of VeronaVeronaItaly
| |
Collapse
|
12
|
Yu X, He S, Shen J, Huang Q, Yang P, Huang L, Pu D, Wang L, Li L, Liu J, Liu Z, Zhu L. Tumor vessel normalization and immunotherapy in gastric cancer. Ther Adv Med Oncol 2022; 14:17588359221110176. [PMID: 35872968 PMCID: PMC9297465 DOI: 10.1177/17588359221110176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 06/09/2022] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is a common malignant tumor, and patients with GC have a low survival rate due to limited effective treatment methods. Angiogenesis and immune evasion are two key processes in GC progression, and they act synergistically to promote tumor progression. Tumor vascular normalization has been shown to improve the efficacy of cancer immunotherapy, which in turn may be improved through enhanced immune stimulation. Therefore, it may be interesting to identify synergies between immunomodulatory agents and anti-angiogenic therapies in GC. This strategy aims to normalize the tumor microenvironment through the action of the anti-vascular endothelial growth factor while stimulating the immune response through immunotherapy and prolonging the survival of GC patients.
Collapse
Affiliation(s)
- Xianzhe Yu
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Shan He
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Jian Shen
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Qiushi Huang
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Peng Yang
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Lin Huang
- West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Dan Pu
- West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Li Wang
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Lu Li
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Jinghua Liu
- Department of Hepatobiliary Surgery, Linyi People's Hospital, Linyi, Shandong 276000, People's Republic of China
| | - Zelong Liu
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lingling Zhu
- Lung Cancer Center, West China Hospital of Sichuan University, No. 37, Guo Xue Xiang, Wuhou District, Chengdu, Sichuan 610041, People's Republic of China
| |
Collapse
|
13
|
Diffuse gastric cancer: Emerging mechanisms of tumor initiation and progression. Biochim Biophys Acta Rev Cancer 2022; 1877:188719. [PMID: 35307354 DOI: 10.1016/j.bbcan.2022.188719] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 02/07/2023]
Abstract
Gastric cancer is globally the fourth leading cause of cancer-related deaths. Patients with diffuse-type gastric cancer (DGC) particularly have a poor prognosis that only marginally improved over the last decades, as conventional chemotherapies are frequently ineffective and specific therapies are unavailable. Early-stage DGC is characterized by intramucosal lesions of discohesive cells, which can be present for many years before the emergence of advanced DGC consisting of highly proliferative and invasive cells. The mechanisms underlying the key steps of DGC development and transition to aggressive tumors are starting to emerge. Novel mouse- and organoid models for DGC, together with multi-omic analyses of DGC tumors, revealed contributions of both tumor cell-intrinsic alterations and gradual changes in the tumor microenvironment to DGC progression. In this review, we will discuss how these recent findings are leading towards an understanding of the cellular and molecular mechanisms responsible for DGC initiation and malignancy, which may provide opportunities for targeted therapies.
Collapse
|
14
|
Ooki A, Yamaguchi K. The dawn of precision medicine in diffuse-type gastric cancer. Ther Adv Med Oncol 2022; 14:17588359221083049. [PMID: 35281349 PMCID: PMC8908406 DOI: 10.1177/17588359221083049] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 01/31/2022] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide. The histology- and morphology-based Lauren classification of GC has been widely used for over 50 years in clinical practice. The Lauren classification divides GC into intestinal and diffuse types, which have distinct etiology, molecular profiles, and clinicopathological features. Diffuse-type GC (DGC) accounts for approximately 30% of GCs. Tumor cells lack adhesion and infiltrate the stroma as single cells or small subgroups, leading to easy dissemination in the abdominal cavity. Clinically, DGC has aggressive traits with a high risk of recurrence and metastasis, which results in unfavorable prognosis. Although systemic chemotherapy is the main therapeutic approach for recurrent or metastatic GC patients, clinical benefits are limited for patients with DGC. Therefore, it is urgent to develop effective therapeutic strategies for DGC patients. Considerable research studies have characterized the molecular and genomic landscape of DGC, of which tight junction protein claudin-18 isoform 2 (CLDN18.2) and fibroblast growing factors receptor-2 isoform IIIb (FGFR2-IIIb) are the most attractive targets because of their close association with DGC. Recently, the impressive results of two phase II FAST and FIGHT trials demonstrate proof-of-concept, suggesting that anti-CLDN18.2 antibody (zolbetuximab) and FGFR2-IIIb antibody (bemarituzumab) are promising approaches for patients with CLDN18.2-positive and FGFR2-IIIb-positive GC, respectively. In this review, we summarize the clinicopathological features and molecular profiles of DGC and highlight a potential therapeutic target based on the findings of pivotal clinical trials.
Collapse
Affiliation(s)
- Akira Ooki
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
15
|
Schrumpf T, Behrens HM, Haag J, Krüger S, Röcken C. FGFR2 overexpression and compromised survival in diffuse-type gastric cancer in a large central European cohort. PLoS One 2022; 17:e0264011. [PMID: 35167603 PMCID: PMC8846517 DOI: 10.1371/journal.pone.0264011] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 02/01/2022] [Indexed: 12/24/2022] Open
Abstract
The significance of fibroblast growth factor receptor 2 (FGFR2) in gastric cancer (GC) has been studied predominantly in Asian patient cohorts. Data on White patients are scarce. Here, we aimed to independently validate the expression and putative tumor biological significance of FGFR2 in a large non-Asian GC cohort. Immunohistochemistry (IHC) was performed on large-area tissue sections from 493 patients with GC and evaluated using the HScore. GCs with moderate and strong FGFR2 expression were studied for Fgfr2 amplification using chromogenic in situ hybridization (CISH). Median overall survival was determined using the Kaplan–Meier method. The majority [240 (99.1%)] of FGFR2-positive GCs showed a variable combination of staining intensities with marked intratumoral heterogeneity, including weak [198 (40.2%) cases], moderate [145 (29.4%)], and strong [108 (21.9%)] staining in diverse combinations. 250 (50.9%) GCs expressed no FGFR2. Fgfr2 gene amplification was found in 40% of selected cases with high protein expression and was also heterogeneous at the cell level. FGFR2 protein expression did not correlate with patient survival in the entire cohort However, using different cutoff values, a negative correlation between FGFR2-expression and patient outcome was found for diffuse-type GC. FGFR2 expression was associated with a lower tumor grade and intestinal phenotype (p≤0.0001). FGFR2–positive diffuse-type GCs classify a small subset of patients with a poor tumor specific survival (5.29±1.3 vs. 14.67±1.9 months; p = 0.004).
Collapse
Affiliation(s)
- Thorben Schrumpf
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Hans-Michael Behrens
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jochen Haag
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sandra Krüger
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Christoph Röcken
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany
- * E-mail:
| |
Collapse
|