1
|
Yin Q, Zhang Y, Xie X, Hou M, Chen X, Ding J. Navigating the future of gastric cancer treatment: a review on the impact of antibody-drug conjugates. Cell Death Discov 2025; 11:144. [PMID: 40188055 PMCID: PMC11972320 DOI: 10.1038/s41420-025-02429-5] [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: 11/05/2024] [Revised: 03/07/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
Gastric cancer, marked by its high incidence and poor prognosis, demands the urgent development of novel and effective treatment strategies, especially for patients ineligible for surgery or those who have had limited success with chemotherapy, radiotherapy and targeted therapies. Recently, antibody-drug conjugates (ADCs) have become a key area of investigation due to their high specificity and potent antitumor effects. These therapies combine monoclonal antibodies, designed to bind to tumor-specific antigens, with cytotoxic agents that selectively target and destroy malignant cells. ADCs have generated significant interest in clinical trials as a promising approach to improve both treatment efficacy and patient outcomes in gastric cancer. However, their clinical application is not without challenges and limitations that must be addressed. This review discusses the recent progress in the use of ADCs for gastric cancer treatment.
Collapse
Affiliation(s)
- Qingling Yin
- GuiZhou University Medical College, Guiyang, 550025, Guizhou, China
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Yanlong Zhang
- GuiZhou University Medical College, Guiyang, 550025, Guizhou, China
| | - Xueqing Xie
- GuiZhou University Medical College, Guiyang, 550025, Guizhou, China
| | - Meijun Hou
- Graduate School, Zunyi Medical University, Zunyi, Guizhou, 563006, China
| | - Xunsheng Chen
- Department of Gastrointestinal Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, Guiyang, China
| | - Jie Ding
- Department of Gastrointestinal Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, Guiyang, China.
| |
Collapse
|
2
|
Azadi SS, Safaralizadeh R, Amini M, Hosseinpour Feizi MA, Abdolzadeh S, Najafi S, Bahojb Mahdavi SZ, Yari A, Narimanzadeh L, Baradaran B, Mokhtarzadeh AA. Investigating the effect of LncRNA DLGAP1-AS2 suppression on chemosensitivity of gastric cancer to chemotherapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7891-7903. [PMID: 38748228 DOI: 10.1007/s00210-024-03130-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 04/29/2024] [Indexed: 10/04/2024]
Abstract
Gastric cancer, as the fifth most frequent disease and the fourth foremost cause of cancer-related death worldwide, remains a main clinical challenge due to its poor prognosis, limited treatment choices, and ability to metastasize. Combining siRNAs to suppress lncRNA with chemotherapeutic medications is a novel treatment approach that eventually increases the therapeutic efficacy of the drug while lessening its adverse effects. This study was performed with the purpose of examining the impact of inhibiting DLGAP1-AS2 expression on gastric cancer cells' drug chemosensitivity. AGS cells were cultured as the study cell line and were transfected with an optimum dose of DLGAP1-AS2 siRNA and then treated with oxaliplatin. Cell viability was examined using the MTT technique. Apoptosis and cell cycle were evaluated using Annexin V/PI staining and flow cytometry. Later, the scratch test was conducted to investigate the ability of cells to migrate, and the inhibition of the stemness of AGS cells was further investigated through the colony formation method. Finally, the qRT-PCR technique was used to assess the expression of Bax, Bcl-2, Caspase-3, p53, MMP-2, and CD44 genes. The MTT test indicated the effect of gene therapy with siRNA and oxaliplatin in combination reduced the chemotherapy drug dose to 29.92 µM and increased AGS cells' sensitivity to oxaliplatin. Also, the combination therapy caused a significant increase in apoptosis. However, it reduced the stemness feature, the rate of cell viability, proliferation, and metastasis compared to the effect of each treatment alone; the results also showed the arrest of the cell cycle in the Sub G1 phase after the combined treatment and a further reduction in the number and size of the formed colonies. Suppressing the expression of lncRNA DLGAP1-AS2 by siRNA followed by treatment with oxaliplatin can be utilized as an effective and new therapeutic technique for gastric cancer therapy.
Collapse
Affiliation(s)
- Seyedeh Samin Azadi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Safaralizadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Samin Abdolzadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - AmirHossein Yari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Narimanzadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
3
|
Abdullah Y. An Overview of Current Biomarkers, the Therapeutic Implications, and the Emerging Role of hERG1 Expression in Gastric Cancer: A Literature Review. Cureus 2023; 15:e47501. [PMID: 37877107 PMCID: PMC10591113 DOI: 10.7759/cureus.47501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2023] [Indexed: 10/26/2023] Open
Abstract
Gastric cancer remains one of the most commonly diagnosed cancers in the world. It carries a high mortality rate, with cases being more prevalent in the developing world, and has been linked to diet and Helicobacter pylori infection. It is a highly heterogeneous disease, with most cases being of a sporadic nature. Most patients present at an advanced stage due to the asymptomatic nature of the early stages of the disease. A multidisciplinary approach is often best implemented to help decide how to best manage individual cases. However, the overall clinical outcome and survival of patients with advanced gastric cancer remain poor. Recent therapeutic advancements focus on the identification of molecular biomarkers associated with gastric cancer that have predictive, diagnostic, and prognostic implications. This enables the development of specific targeted therapies that have shown efficacy in numerous trials, either as monotherapy or in combination with standard chemotherapy. Despite this, tumour heterogeneity and treatment resistance are still issues leading to poor survival outcomes. An emerging approach is focusing efforts on the bidirectional crosstalk between tumour cells and the microenvironment through targeting ion channels. A key player in this is human ether-á-go-go-related gene 1 (hERG1). This voltage-gated potassium ion channel has been shown to have predictive, diagnostic, and prognostic significance, enabling the stratification of high-risk individuals. In addition, targeting hERG1 in combination with chemotherapy has been shown to potentiate tumour regression. This comprehensive literature review will aim to consolidate our understanding of current biomarkers in gastric cancer. The relevance of hERG1 in gastric cancer as a useful novel biomarker and the potential therapeutic implications as targeted therapy will be explored. This offers a new and personalised approach to helping to manage patients with gastric cancer.
Collapse
Affiliation(s)
- Yahya Abdullah
- Internal Medicine, Countess of Chester Hospital NHS Foundation Trust, Chester, GBR
| |
Collapse
|
4
|
Jiang Q, Miao J, Wu F, Li F, Zhang J, Jing X, Cai S, Ma X, Wang X, Zhao L, Huang C. RNF6 promotes gastric cancer progression by regulating CCNA1/CREBBP transcription. Cell Cycle 2023; 22:2018-2037. [PMID: 37904524 PMCID: PMC10761170 DOI: 10.1080/15384101.2023.2275899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 11/01/2023] Open
Abstract
Ring finger protein 6 (RNF6) is a member of the E3 ubiquitin ligase family. Previous studies have reported the involvement of RNF6 as a ubiquitin ligase in the progression of gastric cancer (GC). However, this study found that RNF6 has a clear localization in the nucleus of GC, indicating a role other than ubiquitin ligase. Further chromatin immunoprecipitation sequencing (ChIP-seq) analysis revealed that RNF6 has DNA binding and transcriptional regulatory effects and is involved in important pathways such as tumor cell cycle and apoptosis. Cyclin A1 (CCNA1) and CREB binding protein (CREBBP) are downstream targets for RNF6 transcription regulation in GC. RNF6 binds to the promoter region of CCNA1/CREBBP and is actively regulating their expression in GC cells. Silencing CCNA1/CREBBP partially reversed the promoting effect of RNF6 overexpression on the biological function of GC cells. Our study suggests that RNF6 promotes the progression of GC by regulating CCNA1/CREBBP transcription.
Collapse
Affiliation(s)
- Qiuyu Jiang
- Department of Cell Biology and Genetics/Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Jiyu Miao
- Department of Hematology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Fei Wu
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Fang Li
- Department of Cell Biology and Genetics/Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Jinyuan Zhang
- Department of Cell Biology and Genetics/Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Xintao Jing
- Department of Cell Biology and Genetics/Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Shuang Cai
- Department of Cell Biology and Genetics/Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Xiaoping Ma
- Department of Cell Biology and Genetics/Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Xiaofei Wang
- Department of Cell Biology and Genetics/Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Lingyu Zhao
- Department of Cell Biology and Genetics/Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Chen Huang
- Department of Cell Biology and Genetics/Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| |
Collapse
|
5
|
Sinnarasan VSP, Paul D, Das R, Venkatesan A. Gastric Cancer Biomarker Candidates Identified by Machine Learning and Integrative Bioinformatics: Toward Personalized Medicine. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2023. [PMID: 37229622 DOI: 10.1089/omi.2023.0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Gastric cancer (GC) is among the leading causes of cancer-related deaths worldwide. The discovery of robust diagnostic biomarkers for GC remains a challenge. This study sought to identify biomarker candidates for GC by integrating machine learning (ML) and bioinformatics approaches. Transcriptome profiles of patients with GC were analyzed to identify differentially expressed genes between the tumor and adjacent normal tissues. Subsequently, we constructed protein-protein interaction networks so as to find the significant hub genes. Along with the bioinformatics integration of ML methods such as support vector machine, the recursive feature elimination was used to select the most informative genes. The analysis unraveled 160 significant genes, with 88 upregulated and 72 downregulated, 10 hub genes, and 12 features from the variable selection method. The integrated analyses found that EXO1, DTL, KIF14, and TRIP13 genes are significant and poised as potential diagnostic biomarkers in relation to GC. The receiver operating characteristic curve analysis found KIF14 and TRIP13 are strongly associated with diagnosis of GC. We suggest KIF14 and TRIP13 are considered as biomarker candidates that might potentially inform future research on diagnosis, prognosis, or therapeutic targets for GC. These findings collectively offer new future possibilities for precision/personalized medicine research and development for patients with GC.
Collapse
Affiliation(s)
| | - Dahrii Paul
- Department for Bioinformatics, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Rajesh Das
- Department for Bioinformatics, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Amouda Venkatesan
- Department for Bioinformatics, School of Life Sciences, Pondicherry University, Puducherry, India
| |
Collapse
|
6
|
Zhu Y, Zhou M, Kong W, Li C. Antibody-drug conjugates: the clinical development in gastric cancer. Front Oncol 2023; 13:1211947. [PMID: 37305567 PMCID: PMC10250015 DOI: 10.3389/fonc.2023.1211947] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/17/2023] [Indexed: 06/13/2023] Open
Abstract
Gastric cancer (GC) is a prevalent malignant tumor of the digestive system worldwide, ranking among the top five in terms of incidence and mortality. However, the clinical efficacy of conventional treatments for gastric cancer remains limited, with a median overall survival of approximately eight months for advanced cases. In recent years, researchers have increasingly focused on antibody-drug conjugates (ADCs) as a promising approach. ADCs are potent chemical drugs that selectively target cancer cells by binding to specific cell surface receptors with antibodies. Notably, ADCs have demonstrated promising results in clinical studies and have made significant strides in the treatment of gastric cancer. Currently, several ADCs are under investigation in clinical trials for gastric cancer patients, targeting various receptors such as EGFR, HER-2, HER-3, CLDN18.2, Mucin 1, among others. This review offers a comprehensive exploration of ADC drug characteristics and provides an overview of the research progress in ADC-based therapies for gastric cancer.
Collapse
Affiliation(s)
- Yingze Zhu
- School of Clinical Medicine, Affiliated Hospital, North China University of Science and Technology, Tangshan, China
| | - Miao Zhou
- Tangshan Central Hospital, Tangshan, China
| | - Wenyue Kong
- School of Clinical Medicine, Affiliated Hospital, North China University of Science and Technology, Tangshan, China
| | - Congling Li
- School of Clinical Medicine, Affiliated Hospital, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
7
|
Wang SY, Wang YX, Shen A, Jian R, An N, Yuan SQ. Construction and validation of a prognostic prediction model for gastric cancer using a series of genes related to lactate metabolism. Heliyon 2023; 9:e16157. [PMID: 37234661 PMCID: PMC10205640 DOI: 10.1016/j.heliyon.2023.e16157] [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: 02/16/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Background Gastric cancer (GC) is one of the most common clinical malignant tumors worldwide, with high morbidity and mortality. The commonly used tumor-node-metastasis (TNM) staging and some common biomarkers have a certain value in predicting the prognosis of GC patients, but they gradually fail to meet the clinical demands. Therefore, we aim to construct a prognostic prediction model for GC patients. Methods A total of 350 cases were included in the STAD (Stomach adenocarcinoma) entire cohort of TCGA (The Cancer Genome Atlas), including the STAD training cohort of TCGA (n = 176) and the STAD testing cohort of TCGA (n = 174). GSE15459 (n = 191), and GSE62254 (n = 300) were for external validation. Results Through differential expression analysis and univariate Cox regression analysis in the STAD training cohort of TCGA, we screened out five genes among 600 genes related to lactate metabolism for the construction of our prognostic prediction model. The internal and external validations showed the same result, that is, patients with higher risk score were associated with poor prognosis (all p < 0.05), and our model works well without regard of patients' age, gender, tumor grade, clinical stage or TNM stage, which supports the availability, validity and stability of our model. Gene function analysis, tumor-infiltrating immune cells analysis, tumor microenvironment analysis and clinical treatment exploration were performed to improve the practicability of the model, and hope to provide a new basis for more in-depth study of the molecular mechanism for GC and for clinicians to formulate more reasonable and individualized treatment plans. Conclusions We screened out and used five genes related to lactate metabolism to develop a prognostic prediction model for GC patients. The prediction performance of the model is confirmed by a series of bioinformatics and statistical analysis.
Collapse
Affiliation(s)
- Si-yu Wang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Yu-xin Wang
- The First Hospital of Jilin University, Changchun, 130000, China
| | - Ao Shen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Rui Jian
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Nan An
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Shu-qiang Yuan
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| |
Collapse
|
8
|
Liao T, Lu Y, Li W, Wang K, Zhang Y, Luo Z, Ju Y, Ouyang M. Construction and validation of a glycolysis-related lncRNA signature for prognosis prediction in Stomach Adenocarcinoma. Front Genet 2022; 13:794621. [PMID: 36313430 PMCID: PMC9614251 DOI: 10.3389/fgene.2022.794621] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 09/20/2022] [Indexed: 01/12/2024] Open
Abstract
Background: Glycolysis is closely related to the occurrence and progression of gastric cancer (GC). Currently, there is no systematic study on using the glycolysis-related long non-coding RNA (lncRNA) as a model for predicting the survival time in patients with GC. Therefore, it was essential to develop a signature for predicting the survival based on glycolysis-related lncRNA in patients with GC. Materials and methods: LncRNA expression profiles, containing 375 stomach adenocarcinoma (STAD) samples, were obtained from The Cancer Genome Atlas (TCGA) database. The co-expression network of lncRNA and glycolysis-related genes was used to identify the glycolysis-related lncRNAs. The Kaplan-Meier survival analysis and univariate Cox regression analysis were used to detect the glycolysis-related lncRNA with prognostic significance. Then, Bayesian Lasso-logistic and multivariate Cox regression analyses were performed to screen the glycolysis-related lncRNA with independent prognostic significance and to develop the risk model. Patients were assigned into the low- and high-risk cohorts according to their risk scores. A nomogram model was constructed based on clinical information and risk scores. Gene Set Enrichment Analysis (GSEA) was performed to visualize the functional and pathway enrichment analyses of the glycolysis-related lncRNA. Finally, the robustness of the results obtained was verified in an internal validation data set. Results: Seven glycolysis-related lncRNAs (AL353804.1, AC010719.1, TNFRSF10A-AS1, AC005586.1, AL355574.1, AC009948.1, and AL161785.1) were obtained to construct a risk model for prognosis prediction in the STAD patients using Lasso regression and multivariate Cox regression analyses. The risk score was identified as an independent prognostic factor for the patients with STAD [HR = 1.315, 95% CI (1.056-1.130), p < 0.001] via multivariate Cox regression analysis. Receiver operating characteristic (ROC) curves were drawn and the area under curve (AUC) values of 1-, 3-, and 5-year overall survival (OS) were calculated to be 0.691, 0.717, and 0.723 respectively. Similar results were obtained in the validation data set. In addition, seven glycolysis-related lncRNAs were significantly enriched in the classical tumor processes and pathways including cell adhesion, positive regulation of vascular endothelial growth factor, leukocyte transendothelial migration, and JAK_STAT signaling pathway. Conclusion: The prognostic prediction model constructed using seven glycolysis-related lncRNA could be used to predict the prognosis in patients with STAD, which might help clinicians in the clinical treatment for STAD.
Collapse
Affiliation(s)
- Tianyou Liao
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Yan Lu
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Wangji Li
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Kang Wang
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yanxiang Zhang
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Zhentao Luo
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Yongle Ju
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Manzhao Ouyang
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Amjadi O, Alizadeh-Navaei R, Rezapour M, Omrani-Nava V, Moosazadeh M, Azadeh H, Zaboli E, Ahmadi M, Hedayatizadeh-Omran A. Association between Serum Progastrin Biomarker Level and Gastric Cancer. Asian Pac J Cancer Prev 2022; 23:3595-3599. [PMID: 36308387 PMCID: PMC9924311 DOI: 10.31557/apjcp.2022.23.10.3595] [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: 11/24/2021] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND AND OBJECTIVE gastric cancer is the fifth most prevalent cancer and the fourth cause of death because of cancer. In Iran, northern and northwestern regions are considered gastric cancer hot spots. Identifying serum biomarkers could be helpful in early diagnosis of patients with gastric adenocarcinoma (GAC). Increase in progastrin level has been reported in different cancers. Given the diagnostic value of this biomarker, this study aimed to determine the diagnostic role of progastrin serum biomarker in patients with gastric cancer. METHODOLOGY In this case-control study, forty patients with gastric cancer who were diagnosed by endoscopy and pathologic findings and visited Mazandaran Comprehensive Cancer Center. The participants had received no treatment yet and entered this study. The participants in case group were compared with the control group including forty-two individuals with no history of gastrointestinal cancer in their first-degree relatives and visiting the lab for routine tests. Progastrin serum level was assessed using ELISA kit. The Kruskal-Wallis test and Mann Whitney test, both non-parametric) were used for statistical analysis and the relation between the variables was examined using Pearson's correlation coefficient at 95% confidence level in SPSS 16. FINDINGS In this study, progastrin serum level was significantly higher in patients with gastric cancer compared with normal participants (P = 0.035). Progastrin serum level had no significant relation with tumor clinicopathologic parameters (p-value > 0.05). CONCLUSION Increase in progastrin may be utilized as a predictive factor for gastric cancer.
Collapse
Affiliation(s)
- Omolbanin Amjadi
- Gastrointestinal Cancer Research Center, Non-communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Reza Alizadeh-Navaei
- Gastrointestinal Cancer Research Center, Non-communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Mahsa Rezapour
- Gastrointestinal Cancer Research Center, Non-communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Versa Omrani-Nava
- Gastrointestinal Cancer Research Center, Non-communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Mahmood Moosazadeh
- Gastrointestinal Cancer Research Center, Non-communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Hossein Azadeh
- Department of Internal Medicine, Rheumatology Division, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Ehsan Zaboli
- Gastrointestinal Cancer Research Center, Non-communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Mohadeseh Ahmadi
- Gastrointestinal Cancer Research Center, Non-communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Akbar Hedayatizadeh-Omran
- Gastrointestinal Cancer Research Center, Non-communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran. ,For Correspondence:
| |
Collapse
|
10
|
Mazidimoradi A, Momenimovahed Z, Salehiniya H. Barriers and Facilitators Associated with Delays in the Diagnosis and Treatment of Gastric Cancer: a Systematic Review. J Gastrointest Cancer 2022; 53:782-796. [PMID: 34499307 DOI: 10.1007/s12029-021-00673-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND The present study was conducted to identify barriers and facilitators of early diagnosis and treatment of gastric cancer. METHODS Comprehensive search was conducted on 2021 in various databases, including Medline, Web of science, and Scopus. Keywords such as gastric cancer, screening programs, endoscopy, barriers, facilitators, and factor were used for the search, as single or in combination. Also a manual search was done in valid scientific journals to find related full-text articles. The search results were entered into the Endonote-X8 software, which automatically removes duplicate articles. Then, the title and the abstract and finally, the text of the articles were studied. Articles that addressed barriers and facilitators of early diagnosis and treatment of gastric cancer were included. RESULTS In according to the results of 22 included articles, delay time in the diagnosis and treatment of gastric cancer were high, and factors such as age, sex, race and ethnicity, economic and social status, access to diagnostic services, implementation of screening programs, type and accuracy of screening methods, use of insurance services, error in care services, and presence of gastrointestinal symptoms were considered to be contributing factors in this regard. CONCLUSIONS It seems that to reduce delay in the diagnosis and treatment of gastric cancer, factors such as implementing screening programs using acceptable methods with high sensitivity and accuracy with a high level of participation, increasing insurance coverage and reducing the share of people in payments, increasing people's access to diagnostic services, educating people about the symptoms and risks of gastric cancer, undertaking proper follow-up in patients and suspects cases identified in screening, as well as increasing patients' access to medical services through financial and insurance support are significantly important.
Collapse
Affiliation(s)
| | | | - Hamid Salehiniya
- Social Determinants of Health Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
11
|
Sarma RJ, Subbarayan S, Zohmingthanga J, Chenkual S, Zomuana T, Lalruatfela ST, Pautu JL, Maitra A, Kumar NS. Transcriptome analysis reveals SALL4 as a prognostic key gene in gastric adenocarcinoma. J Egypt Natl Canc Inst 2022; 34:11. [PMID: 35284980 DOI: 10.1186/s43046-022-00108-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 01/28/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Stomach adenocarcinoma (STAD) dominates 80-90% of gastric cancer (GC). Over the years, it has been realized that the identification of the genes responsible for gastric carcinogenesis is essential to understand the biomarker discovery. METHODS This study aims to identify candidate genes for biomarker discovery in STAD. RNA-Seq was performed on three paired tumor-normal and one unpaired tumor samples from four GC patients and investigated for differentially expressed genes (DEGs) using DESeq2. Gene set enrichment analysis were performed. The DEGs were compared with two STAD microarray datasets available on Gene Expression Omnibus (GEO) database. Survival study (OS) were performed using KM-Plotter on the common genes between all the datasets. RESULTS Totally, 148 DEGs were identified, wherein 55 genes were upregulated and 93 genes were downregulated with |log2foldchange| > 1 and Benjamini-Hochberg (BH) Adjusted P value < 0.01. Cell adhesion molecule (CAM) Pathway was found to be the most significant among the upregulated genes. Gastric acid secretion and mineral absorption pathways were the most significant pathways among the downregulated genes. Comparison with two GEO datasets followed by OS analysis revealed two upregulating genes, APOC1 and SALL4 with prognostic significance. CONCLUSION Upregulation of APOC1 is associated with marginal overall survival (OS) and SALL4 over-expression was associated with the poor OS using KM-Plotter during 5 years data period. Our study suggests that SALL4 could be a promising biomarker candidate in STAD.
Collapse
Affiliation(s)
- Ranjan Jyoti Sarma
- Department of Biotechnology, Mizoram University, Aizawl, Mizoram, 796 004, India
| | | | | | - Saia Chenkual
- Department of Surgery, Civil Hospital Aizawl, Aizawl, Mizoram, 796 001, India
| | - Thomas Zomuana
- Department of Surgery, Civil Hospital Aizawl, Aizawl, Mizoram, 796 001, India
| | | | - Jeremy L Pautu
- Department of Medical Oncology, Mizoram State Cancer Institute, Aizawl, Mizoram, 796017, India
| | - Arindam Maitra
- National Institute of Biomedical Genomics, Kalyani, West Bengal, 741251, India.
| | | |
Collapse
|
12
|
Jin J, Tu J, Ren J, Cai Y, Chen W, Zhang L, Zhang Q, Zhu G. Comprehensive Analysis to Identify MAGEA3 Expression Correlated With Immune Infiltrates and Lymph Node Metastasis in Gastric Cancer. Front Oncol 2022; 11:784925. [PMID: 34970496 PMCID: PMC8712941 DOI: 10.3389/fonc.2021.784925] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/23/2021] [Indexed: 11/13/2022] Open
Abstract
Gastric cancer (GC) is an aggressive malignant tumor and causes a significant number of deaths every year. With the coming of the age of cancer immunotherapy, search for a new target in gastric cancer may benefit more advanced patients. Melanoma-associated antigen-A3 (MAGEA3), one of the members of the cancer-testis antigen (CTA) family, was considered an important part of cancer immunotherapy. We evaluate the potential role of MAGEA3 in GC through the TCGA database. The result revealed that MAGEA3 is upregulated in GC and linked to poor OS and lymph node metastasis. MAGEA3 was also correlated with immune checkpoints, TMB, and affected the tumor immune microenvironment and the prognosis of GC through CIBERSORT, TIMER, and Kaplan-Meier plotter database analysis. In addition, GSEA-identified MAGEA3 is involved in the immune regulation of GC. Moreover, the protein-protein interaction (PPI) networks of MAGEA3 were constructed through STRING database and MAGEA3-correlated miRNAs were screened based on the joint analysis of multiple databases. In terms of experimental verification, we constructed pET21a (+)/MAGEA3 restructuring plasmids and transformed to Escherichia coli Rosetta. MAGEA3 protein was used as an antigen after being expressed and purified and can effectively detect the specific IgG in 93 GC patients' serum specimens with 44.08% sensitivity and 92.54% specificity. Through further analysis, the positive rate of MAGEA3 was related to the stage and transfer number of lymph nodes. These results indicated that MAGEA3 is a novel biomarker and correlated with lymph node metastasis and immune infiltrates in GC, which could be a new target for immunotherapy.
Collapse
Affiliation(s)
- Jinji Jin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianxin Tu
- Department of Rheumatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiahuan Ren
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiqi Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenjing Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lifang Zhang
- Department of Medical Microbiology and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Qiyu Zhang
- Department of Hepato-Bilio-Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guanbao Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
13
|
Relationship of prognostic factors in stomach cancer with Helicobacter pylori: A retrospective study. Acta Gastroenterol Belg 2022; 85:35-45. [PMID: 35304992 DOI: 10.51821/85.1.7352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background and study aims The prognostic value of H. pylori, which infects more than half of the human population living in the world and plays a role in gastric cancer pathogenesis, is controversial. Our aim is to investigate the relationship between H. pylori and prognostic factors in gastric cancer. Patients and methods The data of 110 patients (38 females and 72 males) that underwent surgeries due to gastric cancer between 2014 and 2017 were retrospectively analyzed. The relationships between survival (disease-free and overall) and factors such as p53, HER2/neu, Ki-67, neutrophil and platelet lymphocyte ratio (NLR / PLR), histopathological and demographic characteristics were examined. In addition, the results of H. pylori positive and negative groups were compared. Results Sixty-one (55%) patients were H. pylori negative and 49 (45%) were positive. In multivariate analysis, TNM stage, lymph node capsule invasion and NLR were determined as independent prognostic factors in both disease-free and overall survival. Age>62 and PLR>14.3 were determined as independent predictive factors of poor prognosis in overall survival. In univariate analysis, tumor diameter of >4.3 cm, lymphovascular and perineural invasion, and diffuse p53 expression were determined as predictive factors of poor prognosis in disease-free and overall survival. The effectiveness of these markers in prognosis was not different between H. pylori negative and positive groups. Conclusion While age, tumor diameter, TNM stage, lymph node capsule invasion, perineural and lymphovascular invasion, diffuse p53, PLR, and NLR were determined as prognostic factors in gastric cancer, these factors were not affected by the presence of H. pylori.
Collapse
|
14
|
Huang S, Ma L, Lan B, Liu N, Nong W, Huang Z. Comprehensive analysis of prognostic genes in gastric cancer. Aging (Albany NY) 2021; 13:23637-23651. [PMID: 34686626 PMCID: PMC8580339 DOI: 10.18632/aging.203638] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 10/03/2021] [Indexed: 12/16/2022]
Abstract
Background: Gastric cancer is associated with high mortality, and effective methods for predicting prognosis are lacking. We aimed to identify potential prognostic markers associated with the development of gastric cancer through bioinformatic analyses. Methods: Gastric cancer-associated gene expression profiles were obtained from The Cancer Genome Atlas and Gene Expression Omnibus databases. The key genes involved in the development of gastric cancer were obtained by differential expression analysis, coexpression analysis, and short time-series expression miner (STEM) analysis. The potential prognostic value of differentially expressed genes was further evaluated using a Cox regression model and risk scores. Hierarchical clustering was applied to validate the impact of key genes on the overall survival of gastric cancer patients. Results: A total of 1381 genes were consistently dysregulated in the development of gastric cancer. Among them, 186 genes affected the overall survival of gastric cancer patients. The following genes had areas under the receiver operating characteristic curve greater than 0.9 in both datasets and were therefore considered key genes: ADAM12, CEP55, LRFN4, INHBA, ADH1B, DPT, FAM107A, and LOC100506388. LRFN4, DPT, and LOC100506388 were identified as potential prognostic genes for gastric cancer through a nomogram. Overexpression of LRFN4 and LOC100506388 was associated with a higher risk of gastric cancer. Finally, we found that tumors were infiltrated with high levels of Th2 cells and mast cells, and the infiltration levels were associated with overall survival in gastric cancer patients. Conclusions: We found that key dysregulated genes may have a prognostic value for the development of gastric cancer.
Collapse
Affiliation(s)
- Shaohua Huang
- Department of General Surgery, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, China
| | - Liping Ma
- Department of Clinical Laboratory, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, China
| | - Biyang Lan
- Department of General Surgery, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, China
| | - Ning Liu
- Department of General Surgery, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, China
| | - Wenwei Nong
- Department of General Surgery, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, China
| | - Zhihu Huang
- Department of Clinical Laboratory, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
15
|
Relationship of prognostic factors in stomach cancer with helicobacter pylori: a retrospective study. Acta Gastroenterol Belg 2021; 84:607-617. [PMID: 34965043 DOI: 10.51821/84.4.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND STUDY AIMS The prognostic value of H. pylori, which infects more than half of the human population living in the world and plays a role in gastric cancer pathogenesis, is controversial. Our aim is to investigate the relationship between H. pylori and prognostic factors in gastric cancer. PATIENTS AND METHODS The data of 110 patients (38 females and 72 males) that underwent surgeries due to gastric cancer between 2014 and 2017 were retrospectively analyzed. The relationships between survival (disease-free and overall) and factors such as p53, HER2/neu, Ki-67, neutrophil and platelet lymphocyte ratio (NLR / PLR), histopathological and demographic characteristics were examined. In addition, the results of H. pylori positive and negative groups were compared. RESULTS Sixty-one (55%) patients were H. pylori negative and 49 (45%) were positive. In multivariate analysis, TNM stage, lymph node capsule invasion and NLR were determined as independent prognostic factors in both disease-free and overall survival. Age>62 and PLR>14.3 were determined as independent predictive factors of poor prognosis in overall survival. In univariate analysis, tumor diameter of >4.3 cm, lymphovascular and perineural invasion, and diffuse p53 expression were determined as predictive factors of poor prognosis in disease-free and overall survival. The effectiveness of these markers in prognosis was not different between H. pylori negative and positive groups. CONCLUSION While age, tumor diameter, TNM stage, lymph node capsule invasion, perineural and lymphovascular invasion, diffuse p53, PLR, and NLR were determined as prognostic factors in gastric cancer, these factors were not affected by the presence of H. pylori.
Collapse
|
16
|
Alikhani M, Saberi S, Esmaeili M, Michel V, Tashakoripour M, Abdirad A, Aghakhani A, Eybpoosh S, Vosough M, Mohagheghi MA, Eshagh Hosseini M, Touati E, Mohammadi M. Mitochondrial DNA Copy Number Variations and Serum Pepsinogen Levels for Risk Assessment in Gastric Cancer. IRANIAN BIOMEDICAL JOURNAL 2021; 25:323-33. [PMID: 34425651 PMCID: PMC8487685 DOI: 10.52547/ibj.25.5.323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/13/2021] [Indexed: 01/14/2023]
Abstract
Background Variations in mitochondrial DNA copy number (mtDNA-CN) of peripheral blood leukocytes (PBLs), as a potential biomarker for gastric cancer (GC) screening has currently been subject to controversy. Herein, we have assessed its efficiency in GC screening, in parallel and in combination with serum pepsinogen (sPG) I/II ratio, as an established indicator of gastric atrophy. Methods The study population included GC (n = 53) and non-GC (n = 207) dyspeptic patients. The non-GC group was histologically categorized into CG (n = 104) and NM (n = 103) subgroups. The MtDNA-CN of PBLs was measured by quantitative real-time PCR. The sPG I and II levels and anti-H. pylori serum IgG were measured by ELISA. Results The mtDNA-CN was found significantly higher in GC vs. non-GC (OR = 3.0; 95% CI = 1.4, 6.4) subjects. Conversely, GC patients had significantly lower sPG I/II ratio than the non-GC (OR = 3.2; CI = 1.4, 7.2) subjects. The combination of these two biomarkers yielded a dramatic amplification of the odds of GC risk in double-positive (high mtDNA-CN-low sPGI/II) subjects, in reference to double-negatives (low mtDNA-CN-high sPGI/II), when assessed against non-GC (OR = 27.1; CI = 5.0, 147.3), CG (OR = 13.1; CI = 2.4, 72.6), or NM (OR = 49.5; CI = 7.9, 311.6) groups. Conclusion The combination of these two biomarkers, namely mtDNA-CN in PBLs and serum PG I/II ratio, drastically enhanced the efficiency of GC risk assessment, which calls for further validations.
Collapse
Affiliation(s)
- Mehdi Alikhani
- HPGC Research Group, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Samaneh Saberi
- HPGC Research Group, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Maryam Esmaeili
- HPGC Research Group, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Valérie Michel
- Institut Pasteur, Unit of Helicobacter Pathogenesis, CNRS UMR2001, 25-28 Rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Mohammad Tashakoripour
- Gastroenterology Department, Amiralam Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Afshin Abdirad
- Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Arezoo Aghakhani
- Clinical Research Dept., Pasteur Institute of Iran, Tehran, Iran
| | - Sana Eybpoosh
- Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Mahmoud Eshagh Hosseini
- Gastroenterology Department, Amiralam Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Eliette Touati
- Institut Pasteur, Unit of Helicobacter Pathogenesis, CNRS UMR2001, 25-28 Rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Marjan Mohammadi
- HPGC Research Group, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
17
|
Afshar J, Mehrzad J, Mehrad-Majd H, Goshayeshi L, Saeidi J. Prognostic Significance of Tripartite Motif Containing 16 Expression in Patients with Gastric Cancer. Asian Pac J Cancer Prev 2021; 22:2445-2451. [PMID: 34452557 PMCID: PMC8629450 DOI: 10.31557/apjcp.2021.22.8.2445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 08/19/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND AND AIM Tripartite Motif Containing 16 (TRIM16) is a member of the TRIM protein family which is known to play a suppressor role in development of numerous tumor types. However, a positive correlation between TRIM16 expression and gastric cancer (GC) progress has created a controversial situation that need to be fully delineated. The aim of this study was to assess the expression level of TRIM16 mRNA and its relationship with β-catenin, CyclinD, and BCL2 expression in Iranian GC patients and to investigate its possible association with patients' overall survival. Materials and Methods: The expression level of TRIM16 of fresh primary tumor and adjacent normal tissues in 40 GC patients was evaluated by real-time quantitative PCR method. Moreover, patients were subdivided into high or low expression subgroups based on the TRIM16 expression levels. The relationship between TRIM16 expression level, β-catenin, Cyclin D, BCL2, some clinicopathological data and prognosis of GC patients was also analyzed. RESULTS qPCR analysis showed a lower level of TRIM16 in GC tissues (fold change=0.351) in comparison to their matched adjacent noncancerous tissues (p <0.001). Contrary to this, the expression levels of β-catenin, Cyclin D, and BCL2 genes were up-regulated in cancerous samples. This may explain the tumor suppressive function of TRIM16 in GC; as reduction in TRIM16 expression leads to the accumulation of mRNAs from β-catenin, Cyclin D, and BCL2 genes and eventually cancer progression. We did not observe any significant correlation between TRIM16 expression and patients' overall survival. Univariate Cox regression analysis indicated that anemia, weight loss, bleeding, stomach ache, and smoking are statistically associated with overall survival; while, multivariate analysis did not support any correlation. Conclusions: In sum, this study suggests a tumor suppressive role for TRIM16 in gastric cancer and proposes it as a potential candidate for GC prognosis. .
Collapse
Affiliation(s)
- Jalil Afshar
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran.
| | - Jamshid Mehrzad
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran.
| | - Hassan Mehrad-Majd
- Cancer Molecular Pathology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Ladan Goshayeshi
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Jafar Saeidi
- Department of Physiology, School of Basic Science, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran.
| |
Collapse
|
18
|
Ghidini M, Petrillo A, Botticelli A, Trapani D, Parisi A, La Salvia A, Sajjadi E, Piciotti R, Fusco N, Khakoo S. How to Best Exploit Immunotherapeutics in Advanced Gastric Cancer: Between Biomarkers and Novel Cell-Based Approaches. J Clin Med 2021; 10:1412. [PMID: 33915839 PMCID: PMC8037391 DOI: 10.3390/jcm10071412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Despite extensive research efforts, advanced gastric cancer still has a dismal prognosis with conventional treatment options. Immune checkpoint inhibitors have revolutionized the treatment landscape for many solid tumors. Amongst gastric cancer subtypes, tumors with microsatellite instability and Epstein Barr Virus positive tumors provide the strongest rationale for responding to immunotherapy. Various predictive biomarkers such as mismatch repair status, programmed death ligand 1 expression, tumor mutational burden, assessment of tumor infiltrating lymphocytes and circulating biomarkers have been evaluated. However, results have been inconsistent due to different methodologies and thresholds used. Clinical implementation therefore remains a challenge. The role of immune checkpoint inhibitors in gastric cancer is emerging with data from monotherapy in the heavily pre-treated population already available and studies in earlier disease settings with different combinatorial approaches in progress. Immune checkpoint inhibitor combinations with chemotherapy (CT), anti-angiogenics, tyrosine kinase inhibitors, anti-Her2 directed therapy, poly (ADP-ribose) polymerase inhibitors or dual checkpoint inhibitor strategies are being explored. Moreover, novel strategies including vaccines and CAR T cell therapy are also being trialed. Here we provide an update on predictive biomarkers for response to immunotherapy with an overview of their strengths and limitations. We discuss clinical trials that have been reported and trials in progress whilst providing an account of future steps needed to improve outcome in this lethal disease.
Collapse
Affiliation(s)
- Michele Ghidini
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | | | - Andrea Botticelli
- Department of Clinical and Molecular Medicine, Sapienza University, 00189 Rome, Italy;
- Medical Oncology (B), Policlinico Umberto I, 00161 Rome, Italy
| | - Dario Trapani
- Division of Early Drug Development for innovative therapies, European Institute of Oncology, IRCCS, 20141 Milan, Italy;
| | - Alessandro Parisi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
- Medical Oncology Unit, St. Salvatore Hospital, 67100 L’Aquila, Italy
| | - Anna La Salvia
- Department of Oncology, University Hospital 12 De Octubre, 28041 Madrid, Spain;
| | - Elham Sajjadi
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (E.S.); (R.P.); (N.F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Roberto Piciotti
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (E.S.); (R.P.); (N.F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Nicola Fusco
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (E.S.); (R.P.); (N.F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Shelize Khakoo
- Department of Medicine, Royal Marsden Hospital, London and Surrey, Sutton SM25PT, UK;
| |
Collapse
|
19
|
Hong H, Baatar D, Hwang SG. Anticancer Activities of Ginsenosides, the Main Active Components of Ginseng. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:8858006. [PMID: 33623532 PMCID: PMC7875636 DOI: 10.1155/2021/8858006] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 01/20/2021] [Accepted: 01/27/2021] [Indexed: 12/11/2022]
Abstract
Cancer incidence rate has been increasing drastically in recent years. One of the many cancer treatment methods is chemotherapy. Traditional medicine, in the form of complementary and alternative therapy, is actively used to treat cancer, and many herbs and active ingredients of such therapies are being intensely studied to integrate them into modern medicine. Ginseng is traditionally used as a nourishing tonic and for treating various diseases in Asian countries. The therapeutic potential of ginseng in modern medicine has been studied extensively; the main bioactive component of ginseng is ginsenosides, which have gathered attention, particularly for their prospects in the treatment of fatal diseases such as cancer. Ginsenosides displayed their anticancer and antimetastatic properties not only via restricting cancer cell proliferation, viability, invasion, and migration but also by promoting apoptosis, cell cycle arrest, and autophagy in several cancers, such as breast, brain, liver, gastric, and lung cancer. Additionally, ginsenosides can work synergistically with already existing cancer therapies. Thus, ginsenosides may be used alone or in combination with other pharmaceutical agents in new therapeutic strategies for cancer. To date however, there is little systematic summary available for the anticancer effects and therapeutic potential of ginsenosides. Therefore, we have reviewed and discussed all available literature in order to facilitate further research of ginsenosides in this manuscript.
Collapse
Affiliation(s)
- Heeok Hong
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Republic of Korea
| | - Delgerzul Baatar
- Laboratory of Genetics, Institute of Biology, Mongolian Academy of Sciences, Peace Avenue 13330, Ulaanbaatar, Mongolia
| | - Seong Gu Hwang
- Department of Animal Life and Environmental Science, Hankyong National University, Anseong City 17579, Republic of Korea
| |
Collapse
|
20
|
Decreased expression of TRIM3 gene predicts a poor prognosis in gastric cancer. J Gastrointest Cancer 2021; 53:179-186. [PMID: 33415720 DOI: 10.1007/s12029-020-00563-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND/AIMS Tripartite motif-containing 3 (TRIM3) is a member of the TRIM protein family which is known to be involved in development of numerous tumor types. However, the prognostic role of TRIM3 in gastric cancer (GC) remained to be clarified. The aim of this study was to evaluate the expression pattern and prognostic significance of TRIM3 gene and its relationship with β-catenin, CyclinD, and BCL2 expression in patients with GC. METHODS A total of 40 fresh primary gastric cancer tumors and their matched adjacent noncancerous tissues were selected from the First Affiliated Hospital of Mashhad University. Real-time quantitative RT-PCR was performed to evaluate differences in TRIM3 expression in GC and normal tissues. The correlation between TRIM3 expression level and patients' overall survival, some clinicopathological variables, and β-catenin, CyclinD, and BCL-2 genes expression level were also studied. Moreover, patients were divided in two groups according to the TRIM3 expression levels: low and high. RESULTS Compared to noncancerous tissues, TRIM3 expression in GC tissues was significantly increased (fold change = 1.58). Kaplan-Meier survival analysis revealed a significant difference of patient survival according to TRIM3 expression status (P = 0.012). Low TRIM3 expression was associated with shorter overall survival and was an independent predictor for poor prognosis in GC patients (HR, 1.25; 95%CI, 1.02-1.60; P = 0.045). Expression of TRIM3 was negatively correlated with expression of β-catenin, BCL-2, and CyclinD as genes for proliferation, apoptosis, and the cell cycle in GC patients. CONCLUSIONS This study demonstrates that decreased level of TRIM3 mRNA expression is associated with poor prognosis in patients with gastric cancer. TRIM3 may play a protective role in gastric cancer by relieving the effects of cancer progressive genes and could be considered for further investigations as a prognostic biomarker.
Collapse
|
21
|
Prediction of gastric cancer risk: association between ZBTB20 genetic variance and gastric cancer risk in Chinese Han population. Biosci Rep 2020; 40:226430. [PMID: 32936247 PMCID: PMC7517264 DOI: 10.1042/bsr20202102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/19/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Gastric cancer (GC) is a complex multifactorial disease. Previous studies have revealed genetic variations associated with the risk of gastric cancer. The purpose of the present study was to determine the correlation between single-nucleotide polymorphisms (SNPs) of ZBTB20 and the risk of gastric cancer in Chinese Han population. Methods: We conducted a ‘case–control’ study involving 509 GC patients and 507 healthy individuals. We selected four SNPs of ZBTB20 (10934270 T/C, rs9288999 G/A, rs9841504 G/C and rs73230612 C/T), and used logistic regression to analyze the relationship between those SNPs and GC risk under different genetic models; multi-factor dimensionality reduction (MDR) was used to analyze the interaction of “SNP–SNP” in gastric cancer risk; ANOVA and univariate analysis were used to analyze the differences in clinical characteristics among different genotypes. Results: Our results showed that ZBTB20 rs9288999 is a protective factor for the risk of gastric cancer in multiple genetic models, of which the homozygous model is the most significant (OR = 0.48, P=0.0003); we also found that rs9288999 showed a significant correlation with reducing the risk of gastric cancer in different subgroups (BMI; age; gender; smoking or drinking status; adenocarcinoma); rs9841504 is associated with increased GC risk in the participants with BMI>24 kg/m2; rs9841504 and rs73230612 are certainly associated with clinical characteristics of platelet and carbohydrate antigen 242, respectively. Conclusion: Our results suggest that ZBTB20 rs9288999 may be important for reducing the risk of GC in the Chinese Han population.
Collapse
|
22
|
Zhou N, Qiao H, Zeng M, Yang L, Zhou Y, Guan Q. RETRACTED ARTICLE: Circ_002117 binds to microRNA-370 and promotes endoplasmic reticulum stress-induced apoptosis in gastric cancer. Cancer Cell Int 2020; 20:465. [PMID: 36514105 PMCID: PMC7519507 DOI: 10.1186/s12935-020-01493-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/29/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Mounting evidence implicates circular RNAs (circRNAs) in various biological processes during cancer progression. Gastric cancer is a main cause of cancer-related deaths worldwide. Herein, we aimed at investigating whether circ_002117 mediates gastric cancer progression through endoplasmic reticulum (ER) stress. METHODS Bioinformatics analysis detected differentially expressed circRNAs and their target miRNA candidates, and RT-qPCR was performed to detect expression of circ_002117, microRNA (miRNA)-370 and HERPUD1 in gastric cancer tissues and cells. Gastric cancer cells were transfected with plasmids and their proliferative ability and apoptosis were detected with gain- and loss-of-function assay. The ER of treated cells was observed under a transmission electron microscope. Dual-luciferase reporter gene assay and RIP were performed to detect the interaction between HEPRUD1, miR-370 and circ_002117-treated cells were injected into mice to establish xenograft tumor model. RESULTS Circ_002117 and HEPRUD1 were poorly expressed whereas miR-370 was highly expressed in clinical cancer tissues and cells. Circ_002117 was indicated to target and suppress miR-370 expression, while HERPUD1 was directly targeted by miR-370. Circ_002117 overexpression or miR-370 deficiency promoted ER stress-induced apoptosis and decreased proliferation of gastric cancer cells, which was reversed by silencing of HEPRUD1. Circ_002117 overexpression or miR-370 depletion significantly suppressed gastric cancer tumorigenesis in vivo. CONCLUSIONS Taken altogether, circ_002117 facilitated ER stress-induced apoptosis in gastric cancer by upregulating HERPUD1 through miR-370 inhibition.
Collapse
Affiliation(s)
- Nan Zhou
- grid.32566.340000 0000 8571 0482Department of the First Clinical Medical College, Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Hui Qiao
- grid.412643.6Department of Medical Oncology, The First Hospital of Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Miaomiao Zeng
- grid.412643.6Department of Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Lei Yang
- grid.412643.6Department of Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Yongning Zhou
- grid.412643.6Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000 People’s Republic of China ,grid.412643.6Key Laboratory for Gastrointestinal Disease of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Quanlin Guan
- grid.412643.6Key Laboratory for Gastrointestinal Disease of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000 People’s Republic of China ,grid.412643.6Department of Oncology Surgery, The First Hospital of Lanzhou University, No. 1, Donggang West Road, Chengguan District, Lanzhou, 730000 Gansu People’s Republic of China
| |
Collapse
|