1
|
Yang C, Xue R, Qin C, Huang L, Nie R, Luo Y, Xu S, Tang K, Chen J, Jia L, Tan Q. Celastrol Induces Ferroptosis by Regulating CERKL to Exert Anti-Gastric Cancer Effect. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2025; 53:931-949. [PMID: 40374372 DOI: 10.1142/s0192415x25500351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
Abstract
Gastric cancer is a significant global health issue. Celastrol, a natural compound, has shown antitumor potential, but its molecular mechanism in gastric cancer remains unclear. In this study, we treated HGC-27 cells with celastrol and employed CCK8, colony formation, and Transwell assays, revealing its inhibitory effect on cell proliferation and migration. Flow cytometry assay results showed that celastrol could elevate the level of reactive oxygen species (ROS) in HGC-27 cells. By using the iron ion and malondialdehyde (MDA) detection kits, it was found that celastrol promoted the accumulation of iron ions (Fe[Formula: see text] in HGC-27 cells, increased the MDA content, and simultaneously decreased the glutathione (GSH) content. Additionally, Western blot analysis indicated that celastrol exerts an inhibitory effect on the expression of ferroptosis-marker proteins GPX4 and SLC7A11. PCR array and further experiments identified CERKL as a key factor, whose downregulation by celastrol was associated with enhanced ferroptosis. In vivo, celastrol inhibited tumor growth without affecting body weight or organ histology. Our findings suggest that celastrol may inhibit gastric cancer via CERKL-regulated ferroptosis, providing a potential therapeutic strategy.
Collapse
Affiliation(s)
- Chang Yang
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Rui Xue
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Chuling Qin
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Lingyue Huang
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Rongrong Nie
- Rehabilitation Department, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Yuqin Luo
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Siyuan Xu
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Ke Tang
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Jianning Chen
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Lulu Jia
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Qinyou Tan
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin 541001, Guangxi, China
| |
Collapse
|
2
|
Ma H, Suo L, Zhao J, Ma R, Wang Q, Liu J, Qiao J, Wu J, An J, Liu Y, Xing Y, Wang H, Su Z. Prognostic biomarkers based on GUF1, EFTUD2 and GSPT1 targets affecting migration of gastric cancer cells. Transl Cancer Res 2024; 13:4827-4845. [PMID: 39430826 PMCID: PMC11483415 DOI: 10.21037/tcr-24-125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/10/2024] [Indexed: 10/22/2024]
Abstract
Background Eukaryotic elongation factor 1 alpha 2 (eEF1A2) is a protein coding gene which is involved in tumor development and progression in several types of human cancer, but little is known about the function of eEF1A2 proteins in gastric cancer (GC). This study aimed to investigate the effects of GUF1, EFTUD2 and GSPT1 on the migration of GC cells. Methods The Oncomine and The Cancer Genome Atlas (TCGA) databases were used to evaluate the expression of GUF1, EFTUD2, GSPT1 and GSPT2 in GC and the association of eEF1A2 family with individual clinical characteristics. Kaplan-Meier (K-M) Plotter hinted the prognostic value of GUF1, EFTUD2, GSPT1 and GSPT2. GSE62254 and GSE66222 datasets were used to validate the expression of GUF1, EFTUD2, GSPT1. AGS cell line and GES line were also used for validating the function of GUF1, EFTUD2, GSPT1. RNA interference (RNAi) of GUF1, EFTUD2 and GSPT1 had been used to query those genes expression pattern and dissect the proliferation and migration in GC cell lines. Results GUF1, EFTUD2 and GSPT1 were significantly up-regulated in GC cell lines. High expression of GUF1, EFTUD2 and GSPT1 was correlated with cell proliferation and migration induced in GC cells. GUF1, EFTUD2 and GSPT1 may be potential novel oncogenes that helps to maintain the survival of GC cells. Conclusions This study identified that high levels of GUF1, EFTUD2 and GSPT1 expression are predictive biomarkers for a poor prognosis in GC.
Collapse
Affiliation(s)
- Haixiu Ma
- Research Center for High Altitude Medicine, Key Laboratory for High Altitude Medicine, Ministry of Education, Qinghai University, Xining, China
| | - Lina Suo
- Qinghai University Medical College, Xining, China
| | - Jing Zhao
- Research Center for High Altitude Medicine, Key Laboratory for High Altitude Medicine, Ministry of Education, Qinghai University, Xining, China
| | - Ronghua Ma
- Research Center for High Altitude Medicine, Key Laboratory for High Altitude Medicine, Ministry of Education, Qinghai University, Xining, China
| | - Qi Wang
- Research Center for High Altitude Medicine, Key Laboratory for High Altitude Medicine, Ministry of Education, Qinghai University, Xining, China
| | - Jun Liu
- Research Center for High Altitude Medicine, Key Laboratory for High Altitude Medicine, Ministry of Education, Qinghai University, Xining, China
| | - Jinwan Qiao
- Research Center for High Altitude Medicine, Key Laboratory for High Altitude Medicine, Ministry of Education, Qinghai University, Xining, China
| | - Juan Wu
- Research Center for High Altitude Medicine, Key Laboratory for High Altitude Medicine, Ministry of Education, Qinghai University, Xining, China
| | - Juan An
- Qinghai University Medical College, Xining, China
| | - Yan Liu
- Qinghai University Medical College, Xining, China
| | - Yonghua Xing
- Qinghai University Medical College, Xining, China
| | - Haiyan Wang
- Qinghai University Medical College, Xining, China
| | - Zhanhai Su
- Qinghai University Medical College, Xining, China
| |
Collapse
|
3
|
Jiang YK, Li W, Qiu YY, Yue M. Advances in targeted therapy for human epidermal growth factor receptor 2 positive in advanced gastric cancer. World J Gastrointest Oncol 2024; 16:2318-2334. [PMID: 38994153 PMCID: PMC11236256 DOI: 10.4251/wjgo.v16.i6.2318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 04/04/2024] [Accepted: 04/18/2024] [Indexed: 06/13/2024] Open
Abstract
Emerging therapeutic methods represented by targeted therapy are effective supplements to traditional first-line chemoradiotherapy resistance. Human epidermal growth factor receptor 2 (HER2) is one of the most important targets in targeted therapy for gastric cancer. Trastuzumab combined with chemotherapy has been used as the first-line treatment for advanced gastric cancer. The safety and efficacy of pertuzumab and margetuximab in the treatment of gastric cancer have been verified. However, monoclonal antibodies, due to their large molecular weight, inability to penetrate the blood-brain barrier, and drug resistance, lead to decreased therapeutic efficacy, so it is necessary to explore the efficacy of other HER2-targeting therapies in gastric cancer. Small-molecule tyrosine kinase inhibitors, such as lapatinib and pyrrotinib, have the advantages of small molecular weight, penetrating the blood-brain barrier and high oral bioavailability, and are expected to become the drugs of choice for perioperative treatment and neoadjuvant therapy of gastric cancer after validation by large-scale clinical trials in the future. Antibo-drug conjugate, such as T-DM1 and T-DXd, can overcome the resistance of monoclonal antibodies despite their different mechanisms of tumor killing, and are a supplement for the treatment of patients who have failed the treatment of monoclonal antibodies such as trastuzumab. Therefore, after more detailed stratification of gastric cancer patients, various gastric cancer drugs targeting HER2 are expected to play a more significant role.
Collapse
Affiliation(s)
- Ya-Kun Jiang
- Department of Gastroenterology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, Shandong Province, China
| | - Wei Li
- Health Management Center, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, Shandong Province, China
| | - Ying-Yang Qiu
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Meng Yue
- Department of Gastroenterology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, Shandong Province, China
| |
Collapse
|
4
|
Liang Y, Zhang L. Influence of reminiscence therapy on mental health and quality of life in elderly patients with unresectable, metastatic gastrointestinal cancer. Braz J Med Biol Res 2024; 57:e13344. [PMID: 38808887 PMCID: PMC11136486 DOI: 10.1590/1414-431x2024e13344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 04/09/2024] [Indexed: 05/30/2024] Open
Abstract
Reminiscence therapy (RT) attenuates psychological disorders in cancer patients. This study aimed to evaluate the effect of RT on anxiety, depression, spiritual well-being, and quality of life in elderly patients with unresectable, metastatic gastrointestinal cancer. A total of 222 elderly patients with unresectable, metastatic gastrointestinal cancer were randomized into RT group (RT plus usual care, n=112) or control group (usual care, n=110) with a 6-month intervention. Hospital Anxiety and Depression Scale for Anxiety (HADS-A) and Depression (HADS-D), Functional Assessment of Chronic Illness Therapy-Spiritual Well-Being Scale (FACIT-Sp), and Quality of Life Questionnaire-Core 30 (QLQ-C30) were evaluated at month (M)0, M1, M3, and M6. Concerning the primary outcome, HADS-A score at M6 decreased in the RT group compared to the control group (P=0.005). As to secondary outcomes, the RT group showed decreased HADS-A scores at M3, anxiety rate at M3, HADS-D scores at M3 and M6, depression rate at M6, as well as greater FACIT-Sp scores at M1, M3, and M6 vs the control group (all P<0.050). Additionally, QLQ-C30 global health score was elevated at M1 (P=0.046) and M6 (P=0.005), functions score was greater at M6 (P=0.038), and symptoms score was lower at M3 (P=0.019) in the RT group than in the control group. Subgroup analysis revealed that the addition of RT was more effective for patients with anxiety or depression at baseline. In summary, RT alleviated anxiety and depression, and improved the spiritual well-being and quality of life within 6 months in elderly patients with unresectable, metastatic gastrointestinal cancer.
Collapse
Affiliation(s)
- Yu Liang
- Department of Gastrointestinal Surgery, Cancer Hospital, Harbin Medical University, Harbin, China
| | - Limin Zhang
- Department of Gastrointestinal Surgery, Cancer Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
5
|
Fazaeli H, Sheikholeslami A, Ghasemian F, Amini E, Sheykhhasan M. The Emerging Role of LncRNA FENDRR in Multiple Cancers: A Review. Curr Mol Med 2023; 23:606-629. [PMID: 35579154 DOI: 10.2174/1566524022666220509122505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 11/22/2022]
Abstract
Long noncoding RNAs (lncRNAs) are prominent as crucial regulators of tumor establishment and are repeatedly dysregulated in multiple cancers. Therefore, lncRNAs have been identified to play an essential function in carcinogenesis and progression of cancer at genetic and epigenetic levels. FENDRR (fetal-lethal noncoding developmental regulatory RNA) as a LncRNA is a hallmark of various malignancies. FENDRR is crucial for multiple organs' development, such as the lung and heart. The effects of FENDRR under signaling pathways in different cancers have been identified. In addition, it has been verified that FENDRR can affect the development and progression of various cancers. In addition, FENDRR expression has been associated with epigenetic regulation of target genes participating in tumor immunity. Furthermore, FENDRR downregulation was observed in various types of cancers, including colorectal cancer, gastric cancer, pancreatic cancer, cholangiocarcinoma, liver cancer, gallbladder cancer, lung cancer, breast cancer, endometrial cancer, prostate cancer, chronic myeloid leukemia, osteosarcoma, and cutaneous malignant melanoma cells. Here, we review the biological functions and molecular mechanisms of FENDRR in several cancers, and we will discuss its potential as a cancer biomarker and as a probable option for cancer treatment.
Collapse
Affiliation(s)
- Hoda Fazaeli
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research (ACECR), Qom Branch, Qom, Iran
| | - Azar Sheikholeslami
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research (ACECR), Qom Branch, Qom, Iran
| | - Fatemeh Ghasemian
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Elaheh Amini
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mohsen Sheykhhasan
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research (ACECR), Qom Branch, Qom, Iran
- Department of Molecular Medicine and Genetics, Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
6
|
Wang Y, Guo Y, Zhuang T, Xu T, Ji M. SP1-Induced Upregulation of lncRNA LINC00659 Promotes Tumour Progression in Gastric Cancer by Regulating miR-370/AQP3 Axis. Front Endocrinol (Lausanne) 2022; 13:936037. [PMID: 35957833 PMCID: PMC9361049 DOI: 10.3389/fendo.2022.936037] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Growing evidence demonstrates that long noncoding RNAs (lncRNAs) play critical roles in various human tumors. LncRNA LINC00659 (LINC00659) is a newly identified lncRNA and its roles in tumors remain largely unclear. In this study, we elucidated the potential functions and molecular mechanisms of LINC00659 on the biological behaviors of gastric cancer (GC), and also explored its clinical significance. We firstly demonstrated that LINC00659 levels were distinctly up-regulated in both GC specimens and cells using bioinformatics analysis and RT-PCR. The results of ChIP assays and luciferase reporter assays confirmed that upregulation of LINC00659 was activated by SP1 in GC. Clinical assays revealed that higher levels of LINC00659 were associated with TNM stage, lymphatic metastasis, and poorer prognosis. Moreover, LINC00659 was confirmed to be an independent prognostic marker for the patients with GC using multivariate assays. Lost-of-function assays indicated that knockdown of LINC00659 suppressed the proliferation, metastasis, and EMT progress of GC cells in vitro. Mechanistic investigation indicated that LINC00659 served as a competing endogenous RNA (ceRNA) for miR-370, thereby resulting in the upregulation of leading to the depression of its endogenous target gene AQP3. Overall, our present study revealed that the LINC00659/miR-370/AQP3 axis contributes to GC progression, which may provide clues for the exploration of cancer biomarkers and therapeutic targets for GC.
Collapse
Affiliation(s)
- Yao Wang
- Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yuanyuan Guo
- School of Medicine Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tianchi Zhuang
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Ting Xu
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Minghui Ji
- School of Nursing, Nanjing Medical University, Nanjing, China
| |
Collapse
|
7
|
Li L, Wang K, Liu Z, Lü Y, Wang C, Yi X, Guo J. Compound Kushen injection inhibits EMT of gastric cancer cells via the PI3K/AKT pathway. World J Surg Oncol 2022; 20:161. [PMID: 35590327 PMCID: PMC9121642 DOI: 10.1186/s12957-022-02609-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 04/22/2022] [Indexed: 12/02/2022] Open
Abstract
Background The effective components contained in compound Kushen injection (CKI) and the genes and signalling pathways related to gastric cancer (GC) were analyzed through the network pharmacology method of traditional Chinese medicine, and various possible mechanisms by which CKI affects the proliferation, differentiation, survival, and metastasis of GC cells were discussed. The PI3K/AKT signalling pathway is considered to be one of the most important pathways targeted by CKI in the regulation of GC cells. The implementation of related cell experiments also confirmed the information we revealed. Methods Effective drug components of Kushen and Baituling in CKI were identified from the Traditional Chinese Medicine Systems Pharmacology Database (TCMSP). Genes related to GC were identified using the GENECARD and OMIM databases. The common target genes related to the effective components of the drug and GC were identified using the intersection method and visualized using software. A protein–protein interaction network (PPI) was established using STRING online software to confirm the key genes. Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed to predict the key pathways of CKI in GC treatment. BGC-803 and MKN-28 GC cells were used to verify the signalling pathway. Cell proliferation, apoptosis, migration ability, and invasion ability were assessed using CCK8, flow cytometry, scratch, and transwell assays. Immunofluorescence assays and western blotting were used to detect the expression of related proteins. Results CKI regulated GC cells through 35 effective drug components of GC-related target genes. In total, 194 genes were common targets of CKI and GC. The most significant function of the enriched genes was DNA-binding transcription activator activity as demonstrated by GO enrichment analysis. The metabolic pathway with the highest enrichment was the PI3K/AKT signalling pathway as demonstrated by KEGG enrichment analysis. Our cell experimental evidence also shows that CKI inhibits GC cell growth and migration and induce GC cell apoptosis. In addition, CKI inhibits the EMT process in GC cells through the PI3K/AKT signalling pathway. Conclusion AKT1 is a key gene for CKI treatment of GC. CKI inhibited GC cell growth and migration and induced GC cell apoptosis. In addition, CKI regulated the EMT process in GC cells through the PI3K/AKT signalling pathway.
Collapse
Affiliation(s)
- Luo Li
- Quality Management Office, Zibo Central Hospital, Zibo, 255000, Shandong, China
| | - Keshan Wang
- Department of Intervention, The Fourth People's Hospital of ZiBo City, Zibo, 255000, Shandong, China
| | - Zhenguo Liu
- Department of Oncology, The People's Hospital of Gaoqing District, Zibo, 256300, Shandong, China
| | - Yajuan Lü
- Department of Radiotherapy, Shandong Qianfoshan Hospital, Jinan, 250014, Shandong, China
| | - Congcong Wang
- Department of Oncology, Maternal and Child Health Care Hospital of Zibo, Zibo, 255029, Shandong, China
| | - Xuefei Yi
- Department of Oncology, Maternal and Child Health Care Hospital of Zibo, Zibo, 255029, Shandong, China
| | - Jianping Guo
- Department of Oncology, Maternal and Child Health Care Hospital of Zibo, Zibo, 255029, Shandong, China.
| |
Collapse
|
8
|
Ribeiro HF, de Castro Sant' Anna C, de Jesus Oliveira Kato V, de Sousa Brasil RM, Bona AB, da Costa DF, Lima IK, Soares PC, Guimarães APA, de Assumpção PP, Burbano RR. CDC25B Inhibition by menadione: a potential new therapeutical approach. Anticancer Agents Med Chem 2022; 22:2927-2932. [PMID: 35440317 DOI: 10.2174/1871520622666220418131935] [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: 11/19/2021] [Revised: 01/28/2022] [Accepted: 02/24/2022] [Indexed: 12/24/2022]
Abstract
Gastric cancer (GC) is the fifth most common type of tumor and the third leading cause of cancer death worldwide. The evolution of gastric carcinogenesis is still poorly understood and, for this reason, preclinical research protocols were established that included the development of gastric cancer cell lines and the establishment of models of gastric carcinogenesis in non-human primate Sapajus apella. A comprehensive literature search was performed in relevant databases such as PubMed, ResearchGate and Google Scholar to identify studies related to the topic. After an in-depth study of these reports, significant data/data were collected and compiled under appropriate headings. The main result of the studies carried out by the group on GC is the demonstration of the MYC gene overexpression as a common phenomenon in stomach carcinogenesis. Furthermore, we revealed that reducing the expression of the CDC25B gene, regulated by the MYC protein, is a therapeutic strategy against stomach tumors. This review article reveals preclinical evidence that treatment with menadione in experimental models of gastric tumorigenesis, in vivo and in vitro, inhibits the action of the phosphatase CDC25B and, consequently, prevents cell proliferation, invasion and migration.
Collapse
|
9
|
Shen L, Guo J, Zhang Q, Pan H, Yuan Y, Bai Y, Liu T, Zhou Q, Zhao J, Shu Y, Huang X, Wang S, Wang J, Zhou A, Ye D, Sun T, Gao Y, Yang S, Wang Z, Li J, Wu YL. Tislelizumab in Chinese patients with advanced solid tumors: an open-label, non-comparative, phase 1/2 study. J Immunother Cancer 2021; 8:jitc-2019-000437. [PMID: 32561638 PMCID: PMC7304812 DOI: 10.1136/jitc-2019-000437] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Tislelizumab is an investigational, humanized, IgG4 monoclonal antibody with high affinity and binding specificity for programmed cell death-1 (PD-1) that was engineered to minimize binding to FcγR on macrophages in order to abrogate antibody-dependent phagocytosis, a mechanism of T-cell clearance and potential resistance to anti-PD-1 therapy. METHODS The purpose of this phase 1/2, open-label, non-comparative study was to examine the safety, tolerability, and antitumor activity of tislelizumab in adult (≥18 years) Chinese patients with histologically or cytologically confirmed advanced solid tumors with measurable disease. The phase 1 portion of the study consisted of a dose-verification study and a pharmacokinetic (PK) substudy; phase 2 was an indication-expansion study including 11 solid tumor cohorts. Patients previously treated with therapies targeting PD-1 or its ligand, programmed cell death ligand-1 were excluded. During dose-verification, dose-limiting toxicities (DLTs) were monitored; safety and tolerability were examined and the previously determined recommended phase 2 dose (RP2D) was verified. The primary endpoint of phase 2 was investigator-assessed objective response rate per Response Evaluation Criteria in Solid Tumors V.1.1. RESULTS As of December 1, 2018, 300 patients were treated with tislelizumab 200 mg intravenously once every 3 weeks (Q3W). Median duration of follow-up was 8.1 months (range 0.2-21.9). No DLTs were reported during the phase 1 dose-verification study and the RP2D was confirmed to be 200 mg intravenously Q3W. Most treatment-related adverse events (62%) were grade 1 or 2, with the most common being anemia (n=70; 23%) and increased aspartate aminotransferase (n=67; 22%). Of the 251 efficacy evaluable patients, 45 (18%) achieved a confirmed clinical response, including one patient from the PK substudy who achieved a complete response. Median duration of response was not reached for all except the nasopharyngeal carcinoma cohort (8.3 months). Antitumor responses were observed in multiple tumor types. CONCLUSIONS Tislelizumab was generally well tolerated among Chinese patients. Antitumor activity was observed in patients with multiple solid tumors. TRIAL REGISTRATION NUMBER CTR20160872.
Collapse
Affiliation(s)
- Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Ying Yuan
- The Cancer Institute Key Laboratory of Cancer Prevention and Intervention, Chinese National Ministry of Education, The Second Affiliated Hospital Zhejiang University School of Medicine, Zhejiang, China
| | - Yuxian Bai
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jun Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yongqian Shu
- Department of Medical Oncology, Jiangsu Province People's Hospital, Hangzhou, China
| | - Xiaoming Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Zhuhai, China
| | - Siyang Wang
- Department of Radiation Oncology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Jie Wang
- Cancer Institute and Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Aiping Zhou
- Cancer Institute and Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer, Shanghai, China
| | - Ting Sun
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yujuan Gao
- BeiGene (Beijing) Co., Ltd, Beijing, China
| | - Silu Yang
- BeiGene (Beijing) Co., Ltd, Beijing, China
| | | | - Jian Li
- BeiGene (Beijing) Co., Ltd, Beijing, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
10
|
Gourzoulidis G, Koulentaki M, Koumarianou A, Samadas E, Androulakis N, Xynogalos S, Papakotoulas P, Boukovinas I, Karamouzis M, Souglakos J, Chotzagiannoglou V, Beletsi A, Kourlaba G. Cost-effectiveness of trifluridine/tipiracil as a third-line treatment of metastatic gastric cancer, including adenocarcinoma of the gastrohesophageal junction, among patients previously treated in Greece. Expert Rev Pharmacoecon Outcomes Res 2021; 22:259-269. [PMID: 33900864 DOI: 10.1080/14737167.2021.1921576] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Objective: To evaluate the cost-effectiveness of trifluridine/tipiracil (FTD/TPI) compared with best supportive care (BSC) for the treatment of patients with metastatic gastric cancer(mGC), including gastroesophageal junction adenocarcinoma(GEJ), who have received at least two prior therapies for metastatic disease and are eligible for third-line treatment, in Greece.Methods: A partitioned survival model was locally adapted from a public payer perspective over a 10-year time horizon. Clinical, safety and utility data were extracted from literature. Resource consumption data obtained from a panel of local experts using a questionnaire developed for the study was combined with unit costs obtained from official sources. All costs reflect the year 2020 (€). Outcomes of the model were patients' life years (LYs) and quality-adjusted life years (QALYs), total costs and incremental cost-effectiveness ratio (ICER) per QALY and LY gained.Results: The total cost per patient was estimated to be €6,965 for FTD/TPI and €1,906 for BSC, while FTD/TPI was associated with 0.180 and 0.107 increments in LYs and QALYs, respectively, compared with BSC, resulting in an ICER of €47,144 per QALY gained and €28,112 per LY gained.Conclusion: FTD/TPI was estimated to be a cost-effective treatment option for eligible third line mGC patients, including GEJ in Greece.
Collapse
Affiliation(s)
| | | | - Anna Koumarianou
- Hematology Oncology Unit, Fourth Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | | - Nikolaos Androulakis
- Medical Oncology Unit, Pananio-Venizelio General Hospital of Heraklion, Heraklion, Greece
| | | | - Pavlos Papakotoulas
- First Department of Clinical Oncology, Theagenio Hospital, Thessaloniki, Greece
| | | | - Michalis Karamouzis
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - John Souglakos
- Department of Medical Oncology, University Hospital of Heraklion, Crete, Greece
| | | | | | | |
Collapse
|
11
|
Yang Y, Wang C, Dai C, Liu X, Li W, Huang M, Zhao X, Ji D, Li J, Guo W. Amplification and expression of c-MET correlate with poor prognosis of patients with gastric cancer and upregulate the expression of PDL1. Acta Biochim Biophys Sin (Shanghai) 2021; 53:547-557. [PMID: 33693450 DOI: 10.1093/abbs/gmab026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Indexed: 12/22/2022] Open
Abstract
The prognostic significance of c-MET in gastric cancer (GC) remains uncertain. In the present study, we examined the amplification, expression, and the prognostic value of c-MET, human epidermal growth factor receptor 2 (HER2), and programmed cell death 1 ligand 1 (PDL1), together with the correlations among them in a large cohort of Chinese samples. A total of 444 patients were included. The immunohistochemistry (IHC) and the dual-color silver in situ hybridization (SISH) were performed to examine their expression and amplification. Univariate and multivariate analyses were performed by the Cox proportional hazard regression model, and survival curves were estimated by the Kaplan-Meier method. The positivity determined by IHC of c-MET was 24.8%, and the MET amplification rate was 2.3%. The positivity rates of HER2 and PDL1 were 8% and 34.7%, respectively. PDL1 expression had a significantly positive association with c-MET expression. c-MET positivity played a significant prognostic role in disease-free survival (DFS) (P = 0.032). Patients with mesenchymal-epithelial transition (MET) amplification had significantly poorer prognosis on both DFS and overall survival (OS). Subgroup analysis showed that in HER2-negative patients, but not in HER2-positive patients, MET-positive patients had significantly worse DFS (P = 0.000) and OS (P = 0.006). c-MET regulated the expression of PDL1 through an AKT-dependent pathway. c-MET inhibitor enhanced the T-cell killing ability and increased the efficacy of PD1 antibody. c-MET was found to be an independent prognostic factor for DFS of GC patients. A combination of c-MET inhibitors and PD1 antibodies could enhance the killing capacity of T cells, providing a preliminary basis for the clinical research on the same combination in GC treatment.
Collapse
Affiliation(s)
- Ya’nan Yang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chenchen Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Congqi Dai
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xinyang Liu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Wenhua Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Mingzhu Huang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiaoying Zhao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Dongmei Ji
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jin Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Medical Oncology, Tongji University Shanghai East Hospital, Shanghai 200120, China
| | - Weijian Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
12
|
Zhang Y, Huang F, Xu N, Wang J, Li D, Yin L. Overexpression of serum extracellular vesicle microRNA-215-5p is associated with early tumor recurrence and poor prognosis of gastric cancer. Clinics (Sao Paulo) 2021; 76:e2081. [PMID: 33978071 PMCID: PMC8075109 DOI: 10.6061/clinics/2021/e2081] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 10/15/2020] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES Extracellular vesicle microRNAs (EV-miRNAs) have been demonstrated to be reliable candidate biomarkers for clinical applications. However, the clinical application potential of serum EV-miR-215-5p for gastric cancer (GC) remains poorly understood. The goal of our study was to determine the efficacy of serum EV-miR-215-5p in predicting the prognosis of GC. METHODS Blood samples were collected from 118 patients with GC, 60 patients with benign gastric disease and BGD and 70 healthy controls. The relative levels of serum EV-miR-215-5p were measured using quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS Compared to patients with BGD and normal controls, GC patients exhibited remarkably higher serum EV-miR-215-5p level, especially those with early tumor recurrence (ETR). Receiver operating characteristic (ROC) curve analysis showed that serum EV-miR-215-5p was able to distinguish GC patients from BGD patients or healthy controls and GC patients with ETR from those without ETR. In addition, increased serum EV-miR-215-5p levels were notably correlated with invasive depth, TNM stage, and lymph node metastasis. Moreover, serum EV-miR-215-5p levels were greatly decreased after surgical treatment, but increased at the time of ETR. Survival analysis showed that patients with higher serum EV-miR-215-5p had shorter survival. Furthermore, serum EV-miR-215-5p was an independent risk factor for GC. CONCLUSIONS Serum EV-miR-215-5p might be a novel biomarker for predicting ETR and prognosis of GC.
Collapse
Affiliation(s)
- Yunfei Zhang
- Department of Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Fengchang Huang
- Department of Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Ning Xu
- Department of Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Jin Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Dan Li
- Department of Acupuncture, Yunnan Hospital of Integrated Traditional Chinese and Western Medicine, Kunming 650200, Yunnan, China
| | - Liang Yin
- Department of Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
- *Corresponding author. E-mail:
| |
Collapse
|
13
|
Feng Y, Wu M, Hu S, Peng X, Chen F. LncRNA DDX11-AS1: a novel oncogene in human cancer. Hum Cell 2020; 33:946-953. [PMID: 32772230 DOI: 10.1007/s13577-020-00409-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 07/25/2020] [Indexed: 12/20/2022]
Abstract
Long noncoding RNA (lncRNA) is a newly identified type of noncoding RNA with a length of more than 200 nucleotides. The latest research shows that lncRNAs play important roles in the occurrence and development of human tumours by acting both as carcinogenic genes and as tumour suppressor genes. LncRNAs plays a role in various biological processes, such as cell growth, apoptosis, migration and invasion. The newly discovered lncRNA DDX11-AS1 is abnormally highly expressed in various malignant tumours, such as hepatocellular carcinoma, colorectal cancer, osteosarcoma, bladder cancer, NSCLC and gastric cancer. DDX11-AS1 mainly regulates the expression of related genes through direct or indirect ways to perform its functions in carcinogenicity. These results indicate that DDX11-AS1 may be a marker or therapeutic target of tumours. This review summarizes the biological function and mechanism of DDX11-AS1 in the process of tumour development.
Collapse
Affiliation(s)
- Yubin Feng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Hefei, Anhui, China
| | - Maomao Wu
- Department of Pharmacy, Anhui Chest Hospital, Hefei, Anhui Province, China
| | - Shuang Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Hefei, Anhui, China
| | - Xiaoqing Peng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China. .,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Hefei, Anhui, China.
| | - Feihu Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China. .,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Hefei, Anhui, China.
| |
Collapse
|
14
|
Accordino G, Lettieri S, Bortolotto C, Benvenuti S, Gallotti A, Gattoni E, Agustoni F, Pozzi E, Rinaldi P, Primiceri C, Morbini P, Lancia A, Stella GM. From Interconnection between Genes and Microenvironment to Novel Immunotherapeutic Approaches in Upper Gastro-Intestinal Cancers-A Multidisciplinary Perspective. Cancers (Basel) 2020; 12:cancers12082105. [PMID: 32751137 PMCID: PMC7465773 DOI: 10.3390/cancers12082105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 02/07/2023] Open
Abstract
Despite the progress during the last decade, patients with advanced gastric and esophageal cancers still have poor prognosis. Finding optimal therapeutic strategies represents an unmet need in this field. Several prognostic and predictive factors have been evaluated and may guide clinicians in choosing a tailored treatment. Data from large studies investigating the role of immunotherapy in gastrointestinal cancers are promising but further investigations are necessary to better select those patients who can mostly benefit from these novel therapies. This review will focus on the treatment of metastatic esophageal and gastric cancer. We will review the standard of care and the role of novel therapies such as immunotherapies and CAR-T. Moreover, we will focus on the analysis of potential predictive biomarkers such as Modify as: Microsatellite Instability (MSI) and PD-L1, which may lead to treatment personalization and improved treatment outcomes. A multidisciplinary point of view is mandatory to generate an integrated approach to properly exploit these novel antiproliferative agents.
Collapse
Affiliation(s)
- Giulia Accordino
- Department of Medical Sciences and Infective Diseases, Unit of Respiratory Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo Foundation and University of Pavia Medical School, 27000 Pavia, Italy; (G.A.); (S.L.)
| | - Sara Lettieri
- Department of Medical Sciences and Infective Diseases, Unit of Respiratory Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo Foundation and University of Pavia Medical School, 27000 Pavia, Italy; (G.A.); (S.L.)
| | - Chandra Bortolotto
- Department of Intensive Medicine, Unit of Radiology, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, 27000 Pavia, Italy; (C.B.); (A.G.)
| | - Silvia Benvenuti
- Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia (FPO)-IRCCS-Str. Prov.le 142, km. 3,95, 10060 Candiolo (TO), Italy;
| | - Anna Gallotti
- Department of Intensive Medicine, Unit of Radiology, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, 27000 Pavia, Italy; (C.B.); (A.G.)
| | - Elisabetta Gattoni
- Department of Oncology, Azienda Sanitaria Locale (ASL) AL, 27000 Casale Monferrato (AL), Italy;
| | - Francesco Agustoni
- Department of Medical Sciences and Infective Diseases, Unit of Oncology, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, 27000 Pavia, Italy; (F.A.); (E.P.)
| | - Emma Pozzi
- Department of Medical Sciences and Infective Diseases, Unit of Oncology, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, 27000 Pavia, Italy; (F.A.); (E.P.)
| | - Pietro Rinaldi
- Department of Intensive Medicine, Unit of Thoracic Surgery, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, 27000 Pavia, Italy; (P.R.); (C.P.)
| | - Cristiano Primiceri
- Department of Intensive Medicine, Unit of Thoracic Surgery, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, 27000 Pavia, Italy; (P.R.); (C.P.)
| | - Patrizia Morbini
- Department of Diagnostic Medicine, Unit of Pathology, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, 27000 Pavia, Italy;
| | - Andrea Lancia
- Department of Medical Sciences and Infective Diseases, Unit of Radiation Therapy, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, 27000 Pavia, Italy;
| | - Giulia Maria Stella
- Department of Medical Sciences and Infective Diseases, Unit of Respiratory Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo Foundation and University of Pavia Medical School, 27000 Pavia, Italy; (G.A.); (S.L.)
- Correspondence: ; Tel.: +39-0382503369; Fax: +39-0382502719
| |
Collapse
|
15
|
Xia T, Pan Z, Zhang J. CircSMC3 regulates gastric cancer tumorigenesis by targeting miR-4720-3p/TJP1 axis. Cancer Med 2020; 9:4299-4309. [PMID: 32314520 PMCID: PMC7300406 DOI: 10.1002/cam4.3057] [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: 02/11/2020] [Revised: 03/17/2020] [Accepted: 03/23/2020] [Indexed: 12/12/2022] Open
Abstract
Circular RNAs (circRNAs) are identified to play an evident role in many human cancers, such as gastric cancer. However, the potential mechanisms underlying the circRNA-induced pathogenesis in gastric cancers are still elusive. The present study is designed to unfold the mechanism by which circRNAs involve in gastric cancer progression. Using circRNAs microarray, we detected the dysregulated circRNAs and identified an upregulated circRNA, circSMC3 (hsa_circ_0000260), in gastric cancer tissues. Patients with high circSMC3 expression levels had a poor overall survival via Kaplan-Meier survival analysis implied that gastric cancer. Functionally, loss of circSMC3 abolished the proliferation and motility of gastric cancer cells. Mechanically, circSMC3 decreased miR-4720-3p expression by acting as a miRNA sponge, and tight junction protein 1 (TJP1) 3'UTR was identified to be the target of miR-4720-3p, contributing to a circSMC3/miR-4720-3p/TJP1 axis. Thus, our results indicate that circSMC3 promotes gastric cancer cell proliferation and motility through miR-4720-3p/TJP1.
Collapse
Affiliation(s)
- Tianfang Xia
- Department of General Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian City, Huaiyin District, Jiangsu Province, China
| | - Zhenguo Pan
- Department of Gastroenterology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian City, Jiangsu Province, China
| | - Jie Zhang
- Department of General Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian City, Huaiyin District, Jiangsu Province, China
| |
Collapse
|
16
|
Montenegro RC, Howarth A, Ceroni A, Fedele V, Farran B, Mesquita FP, Frejno M, Berger BT, Heinzlmeir S, Sailem HZ, Tesch R, Ebner D, Knapp S, Burbano R, Kuster B, Müller S. Identification of molecular targets for the targeted treatment of gastric cancer using dasatinib. Oncotarget 2020; 11:535-549. [PMID: 32082487 PMCID: PMC7007292 DOI: 10.18632/oncotarget.27462] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 01/13/2020] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) remains the third leading cause of cancer-related death despite several improvements in targeted therapy. There is therefore an urgent need to investigate new treatment strategies, including the identification of novel biomarkers for patient stratification. In this study, we evaluated the effect of FDA-approved kinase inhibitors on GC. Through a combination of cell growth, migration and invasion assays, we identified dasatinib as an efficient inhibitor of GC proliferation. Mass-spectrometry-based selectivity profiling and subsequent knockdown experiments identified members of the SRC family of kinases including SRC, FRK, LYN and YES, as well as other kinases such as DDR1, ABL2, SIK2, RIPK2, EPHA2, and EPHB2 as dasatinib targets. The expression levels of the identified kinases were investigated on RNA and protein level in 200 classified tumor samples from patients, who had undergone gastrectomy, but had received no treatment. Levels of FRK, DDR1 and SRC expression on both mRNA and protein level were significantly higher in metastatic patient samples regardless of the tumor stage, while expression levels of SIK2 correlated with tumor size. Collectively, our data suggest dasatinib for treatment of GC based on its unique property, inhibiting a small number of key kinases (SRC, FRK, DDR1 and SIK2), highly expressed in GC patients.
Collapse
Affiliation(s)
| | - Alison Howarth
- Novo Nordisk Research Centre Oxford (NNRCO), Discovery Technologies and Genomics, Oxford, UK
| | - Alessandro Ceroni
- Novo Nordisk Research Centre Oxford (NNRCO), Discovery Technologies and Genomics, Oxford, UK
| | - Vita Fedele
- Novo Nordisk Research Centre Oxford (NNRCO), Discovery Technologies and Genomics, Oxford, UK
| | - Batoul Farran
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Felipe Pantoja Mesquita
- Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Martin Frejno
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Benedict-Tilman Berger
- Structural Genomics Consortium, Buchmann Institute for Life Sciences, Goethe-University Frankfurt, Frankfurt, Germany.,Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| | - Stephanie Heinzlmeir
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Heba Z Sailem
- Institute of Biomedical Engineering, Department of Engineering, University of Oxford, Oxford, UK.,Big Data Institute, University of Oxford, Li Ka Shing Centre for Health Information and Discovery, Old Road Campus Research Building, Oxford, UK
| | - Roberta Tesch
- Structural Genomics Consortium, Buchmann Institute for Life Sciences, Goethe-University Frankfurt, Frankfurt, Germany.,Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| | - Daniel Ebner
- Novo Nordisk Research Centre Oxford (NNRCO), Discovery Technologies and Genomics, Oxford, UK
| | - Stefan Knapp
- Structural Genomics Consortium, Buchmann Institute for Life Sciences, Goethe-University Frankfurt, Frankfurt, Germany.,Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt, Frankfurt, Germany
| | | | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), Technische Universität München, Freising, Germany
| | - Susanne Müller
- Structural Genomics Consortium, Buchmann Institute for Life Sciences, Goethe-University Frankfurt, Frankfurt, Germany
| |
Collapse
|
17
|
Huang Z, Wei P. Compound Kushen Injection for gastric cancer: A protocol of systematic review and meta-analysis. Medicine (Baltimore) 2019; 98:e17927. [PMID: 31702676 PMCID: PMC6855605 DOI: 10.1097/md.0000000000017927] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 10/15/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND AND AIMS In recent years, the clinical research about Compound Kushen Injection (CKI) treatment of Gastric cancer (GC) has been increased, but the conclusion is different. The aim of our study is to objective comment the efficacy and adverse effects of CKI treatment of GC. METHODS We will retrieve the Randomized controlled trials from the following 6 electronic databases on their inception to April 2019: PubMed, Embase, Cochrane Library, China National Knowledge Infrastructure, Wangfang and Chinese Biomedical Literature Database. Study selection and data collection will be performed independently by 2 reviewers. Cochrane Risk of Bias tool will be used to assess the risk of bias of included studies. The outcomes included overall response rate, complete response rate, 3-year progression-free survival rate, 3-year overall survival rate, and different types of treatment-related adverse events. We calculated the risk ratios as well as their 95% confidence intervals of these outcomes and pooled the results using RevMan 5.2 software and Stata 14.0 software. RESULTS The results will be published in a peer-reviewed journal. CONCLUSION The results of this review will be widely disseminated through peer-reviewed publications and conference presentations. This evidence may also provide helpful evidence for clinical practice and health policy-makers for the treatment of GC. PROSPERO REGISTRATION NUMBER CRD42019133770.
Collapse
|
18
|
High Expression of TTYH3 is Related to Poor Clinical Outcomes in Human Gastric Cancer. J Clin Med 2019; 8:jcm8111762. [PMID: 31652813 PMCID: PMC6912211 DOI: 10.3390/jcm8111762] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/13/2022] Open
Abstract
Ion channels play important roles in regulating various cellular processes and malignant transformation. Expressions of some chloride channels have been suggested to be associated with patient survival in gastric cancer (GC). However, little is known about the expression and function of TTYH3, a gene encoding a chloride ion channel, in cancer progression. Here, we comprehensively analyzed the expression of TTYH3 and its clinical outcome in GC using publicly available cancer gene expression and patient survival data through various databases. We examined the differences of TTYH3 expression between cancers and their normal tissues using the Oncomine, UALCAN, and GEO (Gene Expression Omnibus) databases. TTYH3 expression was investigated from immunohistochemistry images using the Human Protein Atlas database. Copy number alterations and mutations of TTYH3 were analyzed using cBioPortal. The co-expression profile of TTYH3 in GC was revealed using Oncomine. The gene ontology and pathway analyses were done using those co-expressed genes via the Enrichr tool to explore the predicted signaling pathways in GC. TTYH3 mRNA and protein levels in GC were significantly greater than those in normal tissue. Kaplan–Meier analysis revealed the upregulation of TTYH3 expression, which was significantly correlated with worse patient survival. Collectively, our data suggest that TTYH3 might be a potential prognostic marker for GC patients.
Collapse
|
19
|
Wang Z, Yu K, Hu Y, Su F, Gao Z, Hu T, Yang Y, Cao X, Qian F. Schisantherin A induces cell apoptosis through ROS/JNK signaling pathway in human gastric cancer cells. Biochem Pharmacol 2019; 173:113673. [PMID: 31629709 DOI: 10.1016/j.bcp.2019.113673] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/15/2019] [Indexed: 02/07/2023]
Abstract
Gastric cancer is one of the most lethal cancers with unmet clinical treatment and low 5-year survival rate. Schisantherin A is a major compound derived from Fructusschisandrae while its anti-tumor role remains nearly unknown. Here, we reported that schisantherin A had an anti-proliferation effect on gastric cancer cell lines MKN45 and SGC-7901. Schisantherin A induced cell cycle arrest at G2/M phase and cell apoptosis, and inhibited cell migration in gastric cancer MKN45 and SGC7901 cells. Meanwhile, upregulation of cleaved caspase-9, cleaved caspase-3 and cleaved PARP were accompanied with the loss of mitochondrial membrane potential (MMP). Moreover, schisantherin A induced ROS-dependent JNK phosphorylation with higher ROS production. The JNK inhibitor and ROS scavenger NAC rescued the cell apoptosis and cycle inhibition elicited by schisantherin A. Furthermore, the expression level of antioxidant factor Nrf2 was suppressed by schisantherin A. These findings suggest that schisantherin A possesses an anti-tumor activity via activation of ROS/JNK with Nrf2 inhibition, indicating that schisantherin A is a promising chemotherapeutic candidate for gastric cancer.
Collapse
Affiliation(s)
- Zishu Wang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, Anhui Province 233004, PR China
| | - Kaikai Yu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Yudong Hu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Fang Su
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, Anhui Province 233004, PR China
| | - Zhenyuan Gao
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, Anhui Province 233004, PR China
| | - Ting Hu
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, Anhui Province 233004, PR China
| | - Yang Yang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, Anhui Province 233004, PR China
| | - Xiangliao Cao
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, Anhui Province 233004, PR China
| | - Feng Qian
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, Anhui Province 233004, PR China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou 221004, PR China.
| |
Collapse
|
20
|
Li Y, Zhu G, Ma Y, Qu H. lncRNA CCAT1 contributes to the growth and invasion of gastric cancer via targeting miR-219-1. J Cell Biochem 2019; 120:19457-19468. [PMID: 31478245 DOI: 10.1002/jcb.29239] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 12/05/2017] [Indexed: 01/17/2023]
Abstract
Gastric cancer (GC) is one of the most malignant tumors that seriously threaten human health. Increased reports have indicated that long noncoding RNAs (lncRNAs) are associated with GC. This study aims to investigate the regulatory role of colon cancer-associated transcript-1 (CCAT1) in GC. The results exhibited the fact that CCAT1 was expressed higher in 57 GC tissue samples than in 57 paired adjacent normal tissue samples. The expression of CCAT1 was also increased in GC cell lines (MKN45, Hs746T, and SGC-7901) compared with the gastric epithelial cell line GES-1. Besides this, decreased cell proliferation with increased cell apoptosis was detected in SGC-7902 cells transfected with CCAT1 short hairpin RNA (shRNA). At the same time, a lower cell invasion ability was measured in SCG-7901 cells transfected with CCAT1 shRNA.In addition, miR-219-1 was predicted and convinced a direct target of CCAT1. The expression of miR-219-1 was decreased in GC tissues and GC cell lines. Further studies demonstrated that the roles of CCAT1 in cell proliferation, apoptosis, and invasion were inhibited by miR-219-1. Finally, in vivo experiment indicated that tumor growth of GC was suppressed through knockdown of CCAT1. In conclusion, these results suggested that CAT1 promotes the tumorigenesis and progression of GC by negatively regulating miR-219-1.
Collapse
Affiliation(s)
- Yanfeng Li
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Guanyu Zhu
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yan Ma
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongyan Qu
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
21
|
Wang H, Luo YH, Shen GN, Piao XJ, Xu WT, Zhang Y, Wang JR, Feng YC, Li JQ, Zhang Y, Zhang T, Wang SN, Xue H, Wang HX, Wang CY, Jin CH. Two novel 1,4‑naphthoquinone derivatives induce human gastric cancer cell apoptosis and cell cycle arrest by regulating reactive oxygen species‑mediated MAPK/Akt/STAT3 signaling pathways. Mol Med Rep 2019; 20:2571-2582. [PMID: 31322207 PMCID: PMC6691246 DOI: 10.3892/mmr.2019.10500] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 04/12/2019] [Indexed: 12/15/2022] Open
Abstract
1,4-Naphthoquinone derivatives have superior anticancer effects, but their use has been severely limited in clinical practice due to adverse side effects. To reduce the side effects and extend the anticancer effects of 1,4-naphthoquinone derivatives, 2-(butane-1-sulfinyl)-1,4-naphthoquinone (BQ) and 2-(octane-1-sulfinyl)-1,4-naphthoquinone (OQ) were synthesized, and their anticancer activities were investigated. The anti-proliferation effects, determined by MTT assays, showed that BQ and OQ significantly inhibited the viability of gastric cancer cells and had no significant cytotoxic effect on normal cell lines. The apoptotic effect was determined by flow cytometry, and the results showed that BQ and OQ induced cell apoptosis by regulating the mitochondrial pathway and cell cycle arrest at the G2/M phase via inhibition of the Akt signaling pathway in AGS cells. Furthermore, BQ and OQ significantly increased the levels of reactive oxygen species (ROS) and this effect was blocked by the ROS scavenger NAC in AGS cells. BQ and OQ induced apoptosis by upregulating the protein expression of p38 and JNK and downregulating the levels of ERK and STAT3. Furthermore, expression levels of these proteins were also blocked after NAC treatment. These results demonstrated that BQ and OQ induced apoptosis and cell cycle arrest at the G2/M phase in AGS cells by stimulating ROS generation, which caused subsequent activation of MAPK, Akt and STAT3 signaling pathways. Thus, BQ and OQ may serve as potential therapeutic agents for the treatment of human gastric cancer.
Collapse
Affiliation(s)
- Hao Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Ying-Hua Luo
- Department of Grass Science, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Gui-Nan Shen
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Xian-Ji Piao
- Department of Gynaecology and Obstetrics, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, Heilongjiang 163316, P.R. China
| | - Wan-Ting Xu
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yi Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Jia-Ru Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yu-Chao Feng
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Jin-Qian Li
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yu Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Tong Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Shi-Nong Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Hui Xue
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Hong-Xing Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Chang-Yuan Wang
- Department of Food Science and Engineering, College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Cheng-Hao Jin
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| |
Collapse
|
22
|
Oue N, Sentani K, Sakamoto N, Uraoka N, Yasui W. Molecular carcinogenesis of gastric cancer: Lauren classification, mucin phenotype expression, and cancer stem cells. Int J Clin Oncol 2019; 24:771-778. [PMID: 30980196 DOI: 10.1007/s10147-019-01443-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 04/02/2019] [Indexed: 12/13/2022]
Abstract
Gastric cancer (GC), one of the most common human cancers, is a heterogeneous disease with different phenotypes, prognoses, and responses to treatment. Understanding the pathogenesis of GC at the molecular level is important for prognosis prediction and determining treatments. Microsatellite instability (MSI), silencing of MLH1, MGMT, and CDKN2A genes by DNA hypermethylation, KRAS mutation, APC mutation, and ERBB2 amplification are frequently found in intestinal type GC. Inactivation of CDH1 and RARB by DNA hypermethylation, and amplification of FGFR and MET, are frequently detected in diffuse type GC. In addition, BST2 and PCDHB9 genes are overexpressed in intestinal type GC. Both genes are associated with GC progression. GC can be divided into gastric/intestinal mucin phenotypes according to mucin expression. MSI, alterations of TP73, CDH1 mutation, and DNA methylation of MLH are detected frequently in the gastric mucin phenotype. TP53 mutation, deletion of APC, and DNA methylation of MGMT are detected frequently in the intestinal mucin phenotype. FKTN is overexpressed in the intestinal mucin phenotype, and IQGAP3 is overexpressed in the gastric mucin phenotype. These genes are involved in GC progression. To characterize cancer stem cells, a useful method is spheroid colony formation. KIFC1 and KIF11 genes show more than twofold higher expression in spheroid-forming cells than that in parental cells. Both KIF genes are overexpressed in GC, and knockdown of these genes inhibits spheroid formation. Alterations of these molecules may be useful to understand gastric carcinogenesis. Specific inhibitors of these molecules may also be promising anticancer drugs.
Collapse
Affiliation(s)
- Naohide Oue
- Department of Molecular Pathology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Kazuhiro Sentani
- Department of Molecular Pathology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Naoya Sakamoto
- Department of Molecular Pathology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Naohiro Uraoka
- Department of Molecular Pathology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Wataru Yasui
- Department of Molecular Pathology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
23
|
Moehler M, Ryu MH, Dvorkin M, Lee KW, Coşkun HŞ, Wong R, Chung HC, Poltoratsky A, Tsuji A, Yen CJ, Muntean AS, Le Sourd S, Vaccaro GM, Overton L, Boku N, Wainberg ZA, Patel M, Sharma M, Xiong H, Conti I, Taieb J, Bang YJ. Maintenance avelumab versus continuation of first-line chemotherapy in gastric cancer: JAVELIN Gastric 100 study design. Future Oncol 2019; 15:567-577. [PMID: 30379568 DOI: 10.2217/fon-2018-0668] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/12/2018] [Indexed: 12/12/2022] Open
Abstract
Avelumab is a human anti-PD-L1 IgG1 monoclonal antibody that has shown antitumor activity in early phase studies in advanced/metastatic gastric/gastroesophageal junction cancer, including as first-line maintenance therapy. Here, we describe the design of JAVELIN Gastric 100 (NCT02625610), an open-label, Phase III trial. A total of 499 patients with locally advanced/metastatic HER2- gastric/gastroesophageal junction cancer adenocarcinoma, who had achieved at least stable disease following 12 weeks of first-line oxaliplatin/fluoropyrimidine chemotherapy, have been randomized 1:1 to receive avelumab maintenance therapy or continue chemotherapy. The primary objective is to demonstrate superior overall survival in all randomized patients or in the PD-L1+ population. Secondary objectives are to demonstrate superiority for progression-free survival and objective response rate, compare quality of life measures, and determine safety.
Collapse
Affiliation(s)
- Markus Moehler
- Department of Internal Medicine, Johannes-Gutenberg University, Mainz, Germany
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Mikhail Dvorkin
- Department of Oncology, BHI of Omsk Region Clinical Oncology Dispensary, Omsk, Russian Federation
| | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea
| | - Hasan Ş Coşkun
- Department of Medical Oncology, Akdeniz University Medical Faculty, Antalya, Turkey
| | - Rachel Wong
- Department of Medical Oncology, Eastern Health, Box Hill Hospital, Melbourne, VIC, Australia
- Monash University, Faculty of Medicine, Nursing and Health Sciences, Melbourne, VIC, Australia
| | - Hyun C Chung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Yonsei University Health System, Seoul, South Korea
| | - Artem Poltoratsky
- Department of Oncology, FBI "Scientific Research Institute of Oncology n. a. N. N. Petrov", Russian Federation
| | - Akihito Tsuji
- Department of Clinical Oncology, Kagawa University Hospital, Kagawa, Japan
| | - Chia Jui Yen
- Department of Internal Medicine, National Cheng Kung University Hospital, Taiwan, PR China
| | - Alina S Muntean
- Department of Research, Oncology Institute "Prof Dr Ion Chiricuta", Cluj Napoca, Romania
| | - Samuel Le Sourd
- Department of Medical Oncology, Centre Eugene-Marquis, Oncologie Médicale, Rennes, France
| | - Gina M Vaccaro
- Division of Hematology and Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Lindsay Overton
- Oncology & Hematology & Clinical Oncology Research, Confluence Health, Wenatchee, WA 98801, USA
| | - Narikazu Boku
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Zev A Wainberg
- Division of Hematology/Oncology, Department of Medicine, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - Manish Patel
- Department of Medical Oncology, Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota, FL 34236, USA
| | - Maitreyi Sharma
- Global Clinical Development Immuno-Oncology, EMD Serono, Billerica, MA 01821, USA
| | - Huiling Xiong
- Global Clinical Development Immuno-Oncology, EMD Serono, Billerica, MA 01821, USA
| | - Ilaria Conti
- Global Clinical Development Immuno-Oncology, EMD Serono, Billerica, MA 01821, USA
| | - Julien Taieb
- Sorbonne Paris Cité, Paris Descartes University, Department of Gastroenterology & Digestive Oncology, Georges Pompidou European Hospital, Paris, France
| | - Yung-Jue Bang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
24
|
He Z, Wang X, Huang C, Gao Y, Yang C, Zeng P, Chen Z. The FENDRR/miR-214-3P/TET2 axis affects cell malignant activity via RASSF1A methylation in gastric cancer. Am J Transl Res 2018; 10:3211-3223. [PMID: 30416662 PMCID: PMC6220211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/22/2018] [Indexed: 06/09/2023]
Abstract
To explore the effect of fetal-lethal non-coding developmental regulatory RNA (FENDRR) in the initiation and progression of gastric cancer (GC). We detected the levels of FENDRR, microRNA-214-3p (miR-214-3p), and ten-eleven-translocation (TET2) in GC tissues and GC cell lines. In addition, we evaluated the location of FENDRR in GC cell lines by fluorescence in situ hybridization (FISH). Cell proliferation and apoptosis were measured by CCK-8 and Hoechst staining assays. Methylation-specific PCR assay (MSP) was used to evaluate the methylation status of ras-association domain family 1A (RASSF1A). We also observed the direct binding of miR-214-3p on FENDRR by dual-luciferase activity assay in GC cells. FENDRR and TET2 expressions were significantly down-regulated and miR-214-3p was up-regulated in gastric cancer tissues compared to adjacent unaffected tissues. In addition, RASSF1A was hypermethylated in gastric cancer tissues compared to adjacent tissues. The expressions of all the three indicators were influenced by differentiation of tumor, TNM stage of tumors, and lymph node metastasis in patients with GC. A gastric cancer cell line with low FENDRR expression compared to a high FENDRR expressing cell line showed again increased miR-214-3p expression, decreased TET2 and RASSF1A expressions, and RASSF1A hypermethylation, resulting in decreased apoptosis and increased proliferation. Furthermore, we observed a negative correlation between FENDRR and miR-214-3p in GC. The FENDRR/miR-214-3P/TET2 axis plays a critical role in GC progress via methylation of RASSF1A.
Collapse
Affiliation(s)
- Zhaocai He
- Department of General Surgery, Xiangya Hospital, Central South UniversityChangsha, China
- Department of General Surgery, Heping Hospital Affiliated to Changzhi Medical CollegeChangzhi, China
| | - Xin Wang
- Department of General Surgery, Xiangya Hospital, Central South UniversityChangsha, China
| | - Changhao Huang
- Department of General Surgery, Xiangya Hospital, Central South UniversityChangsha, China
| | - Yu Gao
- Department of General Surgery, Xiangya Hospital, Central South UniversityChangsha, China
| | - Chen Yang
- Department of General Surgery, Xiangya Hospital, Central South UniversityChangsha, China
| | - Pengwei Zeng
- Department of General Surgery, Xiangya Hospital, Central South UniversityChangsha, China
| | - Zihua Chen
- Department of General Surgery, Xiangya Hospital, Central South UniversityChangsha, China
| |
Collapse
|
25
|
Yang X, Wang M, Lin B, Yao D, Li J, Tang X, Li S, Liu Y, Xie R, Yu S. miR-487a promotes progression of gastric cancer by targeting TIA1. Biochimie 2018; 154:119-126. [PMID: 30144499 DOI: 10.1016/j.biochi.2018.08.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 08/17/2018] [Indexed: 12/12/2022]
Abstract
Gastric cancer (GC) is one of the most common malignancies as well as the third leading cause for cancer-related death. Molecular basis of GC are essential and critical for its therapeutic treatment, but still remain poorly understood. T-cell intracellular antigen-1 (TIA1) extensively involves in cancer progression, whereas its role and regulation mechanism in GC have not been revealed. In the present study, we found that TIA-1 protein level was down-regulated in GC tissues and TIA1 inhibited proliferation and promoted apoptosis of GC cells. Then, we used bioinformatics to predict miR-487a as the upstream regulator of TIA1 and we also observed an inverse correlation between miR-487a level and TIA-1 protein level in GC tissues. Next, we demonstrated that miR-487a directly targeted TIA1 via binding to its 3'-untranslated region. Furthermore, we investigated the role of miR-487a-TIA1 pathway in the growth of GC cells both in vitro and in vivo. The repression of TIA-1 by miR-487a promoted cell proliferation and suppressed cell apoptosis in vitro, and the knockdown of miR-487a had the opposite effects. Finally, we demonstrated that miR-487a promoted the development of gastric tumor growth in xenograft mice by targeting TIA-1. These effects could be partially reversed by restoring the expression of TIA-1. Overall, our results reveal that TIA1 is a tumor suppressor gene and is directly regulated by miR-487a in GC, which may offer new therapeutic targets for GC treatment.
Collapse
Affiliation(s)
- Xuefeng Yang
- Department of Gastrointestinal Surgery, Affiliated Hospital, Zunyi Medical University, Zunyi, China
| | - Mingda Wang
- Department of Cell Biology, Zunyi Medical University, Zunyi, China; Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
| | - Bohao Lin
- Department of Cell Biology, Zunyi Medical University, Zunyi, China; Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
| | - Dongjie Yao
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| | - Jin Li
- Research Center for Medicine and Biology, Zunyi Medical University, Zunyi, China
| | - Xianchun Tang
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| | - Sanhua Li
- Research Center for Medicine and Biology, Zunyi Medical University, Zunyi, China
| | - Yun Liu
- Research Center for Medicine and Biology, Zunyi Medical University, Zunyi, China
| | - Rui Xie
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical University, Zunyi, China
| | - Shouyang Yu
- Department of Cell Biology, Zunyi Medical University, Zunyi, China; Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
26
|
Wang J, Su Z, Lu S, Fu W, Liu Z, Jiang X, Tai S. LncRNA HOXA-AS2 and its molecular mechanisms in human cancer. Clin Chim Acta 2018; 485:229-233. [PMID: 29981289 DOI: 10.1016/j.cca.2018.07.004] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 06/28/2018] [Accepted: 07/03/2018] [Indexed: 12/21/2022]
Abstract
Long non-coding RNAs (lncRNAs), a novel class of noncoding RNAs, are commonly defined as RNA molecules more than 200 nucleotides in length. Emerging research indicated that lncRNA played a vital role in human tumorigenesis and progression by serving as tumor oncogenes or suppressors. LncRNA has been shown to get involved in participate various biological processes, such as cell growth, anti-apoptosis, migration and invasion. LncRNA HOXA cluster antisense RNA2 (HOXA-AS2) is a novel cancer-related lncRNA. It was recently found to exhibit aberrant expression in a variety of malignancies, including breast cancer, gastric cancer, gallbladder carcinoma, hepatocellular carcinoma and pancreatic cancer. The oncogenicity of lncRNA HOXA-AS2 mainly inhibits or promotes the expression of related genes through direct or indirect pathways, suggesting that HOXA-AS2 likely represents a feasible biomarker or therapeutic target in human cancers. In this review, we summarize current evidences concerning the biological functions and mechanisms of HOXA-AS2 during tumor development.
Collapse
Affiliation(s)
- Jicai Wang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, No.246 XueFu Avenue, Harbin 150086, China
| | - Zhilei Su
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, No.246 XueFu Avenue, Harbin 150086, China
| | - Shounan Lu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, No.246 XueFu Avenue, Harbin 150086, China
| | - Wen Fu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, No.246 XueFu Avenue, Harbin 150086, China
| | - Zhifa Liu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, No.246 XueFu Avenue, Harbin 150086, China
| | - Xingming Jiang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, No.246 XueFu Avenue, Harbin 150086, China.
| | - Sheng Tai
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, No.246 XueFu Avenue, Harbin 150086, China.
| |
Collapse
|
27
|
Wang J, Li H, Xie D, Li L, Wang J, Peng L, Zhou Y. The reactivation of P53 by saRNA affects the biological behavior in vitro in gastric cancer cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:3157-3164. [PMID: 31938445 PMCID: PMC6958090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/11/2018] [Indexed: 06/10/2023]
Abstract
This study sought to verify the reactivation effect of dsP53-285 that can up-regulate P53 expression in vitro. In addition, we explored the reactivation effect that dsP53-285 has on the biological behavior of gastric cancer cells. The specific small activating RNA (saRNA), dsP53-285, targeting the P53 gene promoter was synthesized. Also, a double strained control RNA (dsControl) was synthesized as a negative control, and then siP53 was synthesized to exclude the off-target effect. Both BGC-823 and MGC-803 cells were transfected with the corresponding microRNA, or just treated with lipofectamine2000. RT-qPCR and Western blot were adopted to detect P53 mRNA or the protein content of each group. CCK-8 was adopted to detect the proliferation of each group. The migration ability was assessed using the scratch-wound assay. The results of RT-qPCR and Western blot showed that dsP53-285 caused a significant up-regulation of the P53 gene (P<0.01), and the expression level of the P21 gene changed with the reactivation. The CCK-8 showed that, compared to the control group, the proliferation ability of the dsP53-285 group was inhibited significantly (P<0.01). The reactivation effect was in a time-course manner. The wound scratch assay proved that, compared to the control group, the migration ability of dsP53-285 group was inhibited significantly (P<0.01). This phenomenon provides a theoretical basis for the carcinostatic activity of small activating RNA (saRNA) and might indicate a new targeted treatment option for gastric cancer.
Collapse
Affiliation(s)
- Jing Wang
- Department of Human Anatomy, Qingdao University Medical CollegeShandong, China
| | - Han Li
- Department of General Surgery, Affiliated Hospital of Qingdao UniversityShandong, China
| | - Detian Xie
- Affiliated Hospital of Shandong University of Traditional Chinese MedicineShandong, China
| | - Leping Li
- Department of General Surgery, Shandong Provincial Hospital Affiliated to Shandong UniversityShandong, China
| | - Jinshen Wang
- Department of General Surgery, Shandong Provincial Hospital Affiliated to Shandong UniversityShandong, China
| | - Lipan Peng
- Department of General Surgery, Shandong Provincial Hospital Affiliated to Shandong UniversityShandong, China
| | - Yanbing Zhou
- Department of General Surgery, Affiliated Hospital of Qingdao UniversityShandong, China
| |
Collapse
|
28
|
You X, Ma M, Hou G, Hu Y, Shi X. Gene expression and prognosis of NOX family members in gastric cancer. Onco Targets Ther 2018; 11:3065-3074. [PMID: 29872318 PMCID: PMC5975617 DOI: 10.2147/ott.s161287] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
INTRODUCTION Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOX) are frequently deregulated in several human malignancies, including gastric cancer (GC). NOX-derived reactive oxygen species have been reported to contribute to gastric carcinogenesis and cancer progression. However, the expression and prognostic role of individual NOX in GC patients remain elusive. METHODS AND MATERIALS We investigated genetic alteration and mRNA expression of NOX family in GC patients via the cBioPortal, Human Protein Atlas, and Oncomine databases. Furthermore, we evaluated prognostic value of distinct NOX in GC patients through "The Kaplan-Meier plotter" database. RESULTS Our analysis demonstrated that mRNA deregulation of NOX genes was common alteration in GC patients. Compared with normal tissues, NOX1/2/4 mRNA expression levels in GC tissues were higher, while NOX5 and DUOX1/2 expression levels were lower. Importantly, our results indicated that high mRNA expression of NOX2 was associated with better overall survival whereas NOX4 and DUOX1 were correlated with worse overall survival in all GC patients, particularly in intestinal-type GC patients. In addition, our data also shed light on the diverse roles of individual NOX members in GC patients with different clinicopathological features, including human epidermal growth factor receptor 2 status, clinical stages, pathological grades, and different choices of treatments of GC patients. CONCLUSION These findings suggest that individual NOX family genes, especially NOX2/4, and DUOX1, are potential prognostic markers in GC and implicate that the use of NOX inhibitor targeting NOX4 and DUOX1 may be an effective strategy for GC therapy.
Collapse
Affiliation(s)
- Xin You
- The First Department of Chemotherapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Mingzhe Ma
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Guoxin Hou
- Department of Oncology, The First Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yumin Hu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xi Shi
- The First Department of Chemotherapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
29
|
Taieb J, Moehler M, Boku N, Ajani JA, Yañez Ruiz E, Ryu MH, Guenther S, Chand V, Bang YJ. Evolution of checkpoint inhibitors for the treatment of metastatic gastric cancers: Current status and future perspectives. Cancer Treat Rev 2018; 66:104-113. [PMID: 29730461 DOI: 10.1016/j.ctrv.2018.04.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/17/2018] [Accepted: 04/19/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Standard treatment options for patients with advanced gastric or gastroesophageal junction cancer (GC/GEJC) are associated with limited efficacy and some toxicity. Recently, immunotherapy with antibodies that inhibit the programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) interaction has emerged as a new treatment option. This manuscript reviews early-phase and late-phase trials of immunotherapy in advanced GC/GEJC. METHODS Searches for studies of immunotherapy in GC/GEJC were performed using PubMed, ClinicalTrials.gov, and abstract databases for select annual congresses. Findings were interpreted based on expert opinion. RESULTS Monotherapy with anti-PD-1/PD-L1 antibodies, including pembrolizumab, nivolumab, avelumab, durvalumab, and atezolizumab, has shown interesting objective response rates (ORRs; 7-26%) across varying GC/GEJC populations, with ORRs potentially higher in PD-L1 + vs PD-L1 - tumors. Safety profiles compare favorably with chemotherapy, with grade ≥3 treatment-related adverse events occurring in 5-17%. Based on a large phase 2 study, pembrolizumab was approved in the United States for third-line treatment of patients with PD-L1 + GC/GEJC. In a phase 3 trial, third-line or later nivolumab increased overall survival vs placebo in an Asian population, leading to regulatory approval in Japan, although other completed phase 3 trials did not show superiority for pembrolizumab or avelumab monotherapy vs chemotherapy. Other trials in advanced GC/GEJC are assessing various anti-PD-1/PD-L1-based strategies, including administration in first-line and later-line settings and as combination (with chemotherapy or agents targeting other immune checkpoint proteins, eg, CTLA-4, LAG-3, and IDO) or switch-maintenance regimens. CONCLUSIONS Anti-PD-1/PD-L1 antibodies have shown encouraging clinical activity in advanced GC/GEJC. Results from ongoing phase 3 trials are needed to further evaluate the potential roles of these agents within the continuum of care.
Collapse
Affiliation(s)
- Julien Taieb
- Sorbonne Paris Cité, Paris Decartes University, Hôpital Européen Georges-Pompidou, Paris, France.
| | | | | | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | | | - Min-Hee Ryu
- University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea.
| | | | | | - Yung-Jue Bang
- Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
30
|
Wang F, Sun Y. Overexpression of Myosin Phosphatase Target Subunit 1 (MYPT1) Inhibits Tumor Progression and Metastasis of Gastric Cancer. Med Sci Monit 2018; 24:2508-2517. [PMID: 29687789 PMCID: PMC5937360 DOI: 10.12659/msm.906852] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Myosin phosphatase target subunit 1 (MYPT1) serves as a subgroup of myosin phosphatases, and is frequently low-expressed in human cancers. However, little is known about the effects of MYPT1 in gastric cancer (GC). Material/Methods In our study, MYPT1 expression was detected by quantitative real-time reverse transcription PCR (qRT-PCR) in GC tissues, different advanced pathological stages of GC tissues, and preoperative and postoperative patients. Kaplan-Meier analysis was used to measure the overall survival of GC patients. MYPT1 expression was analyzed by qRT-PCR and Western blot assays in GES-1 cells and GC cells. Cell proliferation, cycle, and migration and invasion abilities were detected by CCK-8, flow cytometry, and Transwell assays. E-cadherin, TIMP-2, MMP-2, MMP-9 RhoA, and p-RhoA expressions were assessed by qRT-PCR and Western blot assays in treated SNU-5 cells. Results Our results indicated that MYPT1 was down-regulated in GC tissues and cells, and is related to clinical stages and overall survival of GC. Functional research demonstrated that overexpression of MYPT1 can inhibit cell proliferation, cell cycle progression, and migration and invasion of GC cells. Many studies on mechanisms reported that overexpression of MYPT1 dramatically improved the expression levels of cell cycle-related genes (Cyclin D1 and c-myc), significantly increased epithelial marker (E-cadherin) expression, and decreased invasion-associated genes (TIMP-2 and MMP-2) expressions in SNU-5 cells. In addition, we found that MYPT1 suppressed RhoA phosphorylation. Conclusions We verified that MYPT1 inhibits GC cell proliferation and metastasis by regulating RhoA phosphorylation.
Collapse
Affiliation(s)
- Fengyong Wang
- Department of Gastrointestinal and Pancreatic Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China (mainland)
| | - Yuanshui Sun
- Department of Gastrointestinal and Pancreatic Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
31
|
Xu C, Liu Y, Jiang D, Li Q, Ge X, Zhang Y, Huang J, Su J, Ji Y, Hou J, Lu S, Hou Y, Liu T. Poor efficacy response to trastuzumab therapy in advanced gastric cancer with homogeneous HER2 positive and non-intestinal type. Oncotarget 2018; 8:33185-33196. [PMID: 28388541 PMCID: PMC5464860 DOI: 10.18632/oncotarget.16567] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/16/2017] [Indexed: 12/20/2022] Open
Abstract
Introduction Factors affecting trastuzumab efficacy in advanced gastric cancer (GC) are largely unknown. Heterogeneity is a notable feature of HER2 in GC. Whether the heterogeneity influences trastuzumab efficacy is still unknown. Results The HER2homogeneous group and HER2heterogeneous group showed no statistical difference in RR (46.4% vs 55.0%, P = 0.558), PFS (5.80 vs 6.30 months, P = 0.804) and OS (16.00 vs 16.00 months, P = 0.787). The Laurenintestinal group and Laurennon-intestinal group demonstrated no discrepancy in PFS (6.00 vs 6.00 months, P = 0.912) and OS (16.50 vs 14.00 months, P = 0.227). However, by combining HER2 heterogeneity and Lauren classification, PFS and OS of HER2homogeneous/Laurennon-intestinal subgroup was the shortest among the 4 subgroups (P = 0.012 and P = 0.037), which was much shorter than the other patients (PFS:3.00 vs 6.30 months, P = 0.003; OS: 4.50 vs 16.50 months, P = 0.004). Univariate and multivariate analysis showed that HER2 heterogeneity combined with Lauren classification was an independent prognostic factor in both PFS (P = 0.031 and P = 0.002) and OS (P = 0.039 and P = 0.013). Materials and Methods 48 patients with HER2 positive advanced GCs accepting trastuzumab treatment were retrospectively analyzed. Based on HER2 heterogeneity, the patients were divided into a HER2homogeneous group and a HER2heterogeneous group. Response rate (RR), progression free survival (PFS), and overall survival (OS) were compared. Main clinicopathological factors including Lauren classification were subjected to subgroup analysis. Conclusions HER2 heterogeneity alone may not correlate with trastuzumab efficacy in HER2 positive advanced GCs. HER2 heterogeneity combined with Lauren classification may help to identify a subgroup with poor response to trastuzumab which is homogeneous HER2 positive and non-intestinal type.
Collapse
Affiliation(s)
- Chen Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Pathology, School of Basic Medical Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yalan Liu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Pathology, School of Basic Medical Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dongxian Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Pathology, School of Basic Medical Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qian Li
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaowen Ge
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Pathology, School of Basic Medical Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying Zhang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Pathology, School of Basic Medical Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Pathology, School of Basic Medical Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jieakesu Su
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Pathology, School of Basic Medical Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Pathology, School of Basic Medical Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jun Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Pathology, School of Basic Medical Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shaohua Lu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Pathology, School of Basic Medical Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Pathology, School of Basic Medical Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianshu Liu
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
32
|
Guo J, Zhang CD, An JX, Xiao YY, Shao S, Zhou NM, Dai DQ. Expression of miR-634 in gastric carcinoma and its effects on proliferation, migration, and invasion of gastric cancer cells. Cancer Med 2018; 7:776-787. [PMID: 29464926 PMCID: PMC5852365 DOI: 10.1002/cam4.1204] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 08/24/2017] [Accepted: 08/25/2017] [Indexed: 12/18/2022] Open
Abstract
This study aims to observe the expression of microRNA (miR)‐634 in different gastric cancer cell lines and tissues, and to study the effects of miR‐634 on the proliferation, migration, and invasion of the gastric cancer cells. The miR‐634 mimics and miR‐634 inhibitors were transfected by lentivirus into human gastric cancer SGC‐7901 and MGC‐803 cells, and the miR‐634 cells without transfection were used as the control group (NC group). The expression of miR‐634 in the transfected cells was detected by qRT‐PCR. Cell viability was measured by the CCK8 assay. The migration and invasion ability of the cells were detected by scratch assays and Transwell® chamber assays, respectively, and the luciferase assay verified the binding of miR‐634 to the target gene JAG1. The expression level of miR‐634 in gastric cancer tissues and cell lines was significantly lower than that in normal adjacent tissues and control cells. The survival of cells was significantly decreased, and number of cells migrating and invading was decreased in the miR‐634 mimics group. However, in the miR‐634 inhibitor group, the opposite results were observed. Over‐expression of miR‐634 inhibited the proliferation, migration, and invasion of gastric cancer cell lines, and the miR‐634 target gene was JAG1.
Collapse
Affiliation(s)
- Jiao Guo
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Chun-Dong Zhang
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Jia-Xiang An
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Yun-Yun Xiao
- Department of Obstetrics and Gynecology, the Shengjing Affiliated Hospital of China Medical University, Shenyang, 110004, China
| | - Shuai Shao
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Nuo-Ming Zhou
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Dong-Qiu Dai
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.,Cancer Center, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| |
Collapse
|
33
|
Xu B, Huang C, Yang X, Li X, Li L, Ding Y. Significance and prognostic role of human epidermal growth factor receptor 2 and RAB1A expression in gastric cancer. Oncol Lett 2018; 15:5185-5192. [PMID: 29552156 DOI: 10.3892/ol.2018.7992] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Accepted: 01/24/2018] [Indexed: 12/29/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER-2) has an important clinical role in various cancers. However, the prognostic impact of HER-2 in gastric cancer (GC) is controversial. RAB1A is an important small molecule in the mechanistic target of rapamycin signalling pathway, which is one of the downstream signalling pathways of the epidermal growth factor receptor family. In recent years, the aberrant expression of RAB1A has been reported in a number of tumours, but its regulation in GC has not been extensively examined. Therefore, the present study investigated the expression pattern and prognostic significance of HER-2 and RAB1A in gastric adenocarcinoma (CAG). A comprehensive analysis was performed to examine the expression level of HER-2 and RAB1A in 280 cases of paired paraffin-embedded GAC tissues and an additional 120 archived GAC tissue samples. HER-2 and RAB1A protein expression was assessed by immunohistochemistry and cases with a 2+ score for HER-2 expression levels were subjected to fluorescence in situ hybridization to determine the HER-2 amplification status. Furthermore, HER-2 and RAB1A mRNA expression was quantified by reverse transcription-quantitative polymerase chain reaction. The comparison of continuous data between two groups was performed using a paired-samples t-test. Clinical correlations were determined using Pearson's Chi-square and Fisher's exact tests. Kaplan-Meier survival curves were used to estimate overall survival (OS). Cox proportional hazards models were used to determine associations between HER-2 and RAB1A expression and outcomes. Regression analyses were performed to detect the correlation between the mRNA levels of HER-2 and RAB1A in GAC tissues. It was observed that RAB1A was significantly overexpressed in GAC tissues compared with normal tissues (P<0.001). Approximately 12.86% of the 280 GAC patients had HER-2 amplification. Additionally, RAB1A expression was significantly associated with a short OS (P<0.001) but there were no significant differences in survival between the HER-2 high-expression group and the HER-2 low-expression group. Additionally, the co-expression of HER-2 and RAB1A indicated poorer OS than the overexpression of each protein (P=0.001), and the two factors were significantly positively correlated in GAC (P=0.012). These findings may be used to further explore the molecular mechanisms and regulatory associations among signalling pathways in GC.
Collapse
Affiliation(s)
- Bihong Xu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Chunyu Huang
- Department of Endoscopy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Xianzi Yang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Xiangzhao Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Liang Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yanqing Ding
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
34
|
Okita A, Imai H, Takahashi M, Takahashi H, Umegaki S, Kawamura Y, Hiraide S, Ouchi K, Sato Y, Okada Y, Komine K, Saijo K, Takahashi S, Takahashi M, Shirota H, Ohori H, Gamoh M, Ishioka C. Efficacy and Safety of Trastuzumab in Combination with S-1 and Cisplatin Therapy for Japanese Patients with HER2-Positive Advanced Gastric Cancer: Retrospective Analysis. TOHOKU J EXP MED 2018; 245:123-129. [DOI: 10.1620/tjem.245.123] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Akira Okita
- Department of Medical Oncology, Tohoku University Hospital
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University
| | - Hiroo Imai
- Department of Medical Oncology, Tohoku University Hospital
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University
| | - Masahiro Takahashi
- Department of Medical Oncology, Tohoku University Hospital
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University
| | | | - Sho Umegaki
- Department of Medical Oncology, Tohoku University Hospital
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University
| | - Yoshifumi Kawamura
- Department of Medical Oncology, Tohoku University Hospital
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University
| | - Sakura Hiraide
- Department of Medical Oncology, Tohoku University Hospital
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University
| | - Kota Ouchi
- Department of Medical Oncology, Tohoku University Hospital
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University
| | - Yuko Sato
- Department of Medical Oncology, Tohoku University Hospital
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University
| | - Yoshinari Okada
- Department of Medical Oncology, Tohoku University Hospital
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University
| | - Keigo Komine
- Department of Medical Oncology, Tohoku University Hospital
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University
| | - Ken Saijo
- Department of Medical Oncology, Tohoku University Hospital
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University
| | - Shin Takahashi
- Department of Medical Oncology, Tohoku University Hospital
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University
| | - Masanobu Takahashi
- Department of Medical Oncology, Tohoku University Hospital
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University
| | - Hidekazu Shirota
- Department of Medical Oncology, Tohoku University Hospital
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University
| | - Hisatsugu Ohori
- Department of Medical Oncology, Ishinomaki Red Cross Hospital
| | - Makio Gamoh
- Department of Medical Oncology, Osaki Citizen Hospital
| | - Chikashi Ishioka
- Department of Medical Oncology, Tohoku University Hospital
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University
| |
Collapse
|
35
|
Farran B, Müller S, Montenegro RC. Gastric cancer management: Kinases as a target therapy. Clin Exp Pharmacol Physiol 2018; 44:613-622. [PMID: 28271563 DOI: 10.1111/1440-1681.12743] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/19/2017] [Accepted: 02/20/2017] [Indexed: 12/16/2022]
Abstract
The molecular diagnostics revolution has reshaped the practice of oncology by facilitating the identification of genetic, epigenetic and proteomic modifications correlated with cancer, thus delineating 'oncomaps' for various cancer types. These advances have enhanced our understanding of gastric cancer, one of the most fatal diseases worldwide, and culminated in the approval of novel molecular therapies such as trastuzumab. Gastric tumours display recurrent aberrations in key kinase oncogenes such as Her2, epidermal growth factor receptor (EGFR), PI3K, mTOR or c-Met, suggesting that these receptors are amenable to inhibition using specific drug agents. In this review, we examine the mutational landscape of gastric cancer, the use of kinase inhibitors as targeted therapies in gastric tumours and the clinical trials underway for novel inhibitors, highlighting successes, failures and future directions.
Collapse
Affiliation(s)
- Batoul Farran
- Department of Structural and Molecular Biology, University College London, London, UK
| | - Susanne Müller
- Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe-University, Frankfurt am Main, DE, Germany
| | - Raquel C Montenegro
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
36
|
Yuza K, Nagahashi M, Watanabe S, Takabe K, Wakai T. Hypermutation and microsatellite instability in gastrointestinal cancers. Oncotarget 2017; 8:112103-112115. [PMID: 29340115 PMCID: PMC5762383 DOI: 10.18632/oncotarget.22783] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/13/2017] [Indexed: 02/07/2023] Open
Abstract
Recent progress in cancer genome analysis using next-generation sequencing has revealed a high mutation burden in some tumors. The particularly high rate of somatic mutation in these tumors correlates with the generation of neo-antigens capable of eliciting an immune response. Identification of hypermutated tumors is therefore clinically valuable for selecting patients suitable for immunotherapy treatment. There are several known causes of hypermutation in tumors, such as ultraviolet light in melanoma, tobacco smoke in lung cancer, and excessive APOBEC (apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like) activity in breast and gastric cancer. In gastrointestinal cancers, one of the leading causes of hypermutation is a defect in DNA mismatch repair, which results in microsatellite instability (MSI). This review will focus on the frequency, characteristics and genomic signature of hypermutated gastrointestinal cancers with MSI. Detection of tumor hypermutation in cancer is expected to not only predict the clinical benefit of immune checkpoint inhibitor treatment, but also to provide better surgical strategies for the patients with hypermutated tumors. Thus, in an era of precision medicine, identification of hypermutation and MSI will play an important role directing surgical and chemotherapeutic treatment.
Collapse
Affiliation(s)
- Kizuki Yuza
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Masayuki Nagahashi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Satoshi Watanabe
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Kazuaki Takabe
- Breast Surgery, Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
- Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY 14203, USA
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| |
Collapse
|
37
|
Mukai S, Oue N, Oshima T, Imai T, Sekino Y, Honma R, Sakamoto N, Sentani K, Kuniyasu H, Egi H, Tanabe K, Yoshida K, Ohdan H, Yasui W. Overexpression of PCDHB9
promotes peritoneal metastasis and correlates with poor prognosis in patients with gastric cancer. J Pathol 2017; 243:100-110. [DOI: 10.1002/path.4931] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/31/2017] [Accepted: 06/16/2017] [Indexed: 01/12/2023]
Affiliation(s)
- Shoichiro Mukai
- Department of Molecular Pathology; Hiroshima University Graduate School of Biomedical Sciences; Hiroshima Japan
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences; Hiroshima University; Hiroshima Japan
| | - Naohide Oue
- Department of Molecular Pathology; Hiroshima University Graduate School of Biomedical Sciences; Hiroshima Japan
| | - Takashi Oshima
- Department of Surgery; Yokohama City University; Yokohama Japan
| | - Takeharu Imai
- Department of Molecular Pathology; Hiroshima University Graduate School of Biomedical Sciences; Hiroshima Japan
- Department of Surgical Oncology, Graduate School of Medicine; Gifu University; Gifu Japan
| | - Yohei Sekino
- Department of Molecular Pathology; Hiroshima University Graduate School of Biomedical Sciences; Hiroshima Japan
| | - Ririno Honma
- Department of Molecular Pathology; Hiroshima University Graduate School of Biomedical Sciences; Hiroshima Japan
| | - Naoya Sakamoto
- Department of Molecular Pathology; Hiroshima University Graduate School of Biomedical Sciences; Hiroshima Japan
| | - Kazuhiro Sentani
- Department of Molecular Pathology; Hiroshima University Graduate School of Biomedical Sciences; Hiroshima Japan
| | - Hiroki Kuniyasu
- Department of Molecular Pathology; Nara Medical University; Nara Japan
| | - Hiroyuki Egi
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences; Hiroshima University; Hiroshima Japan
| | - Kazuaki Tanabe
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences; Hiroshima University; Hiroshima Japan
| | - Kazuhiro Yoshida
- Department of Surgical Oncology, Graduate School of Medicine; Gifu University; Gifu Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences; Hiroshima University; Hiroshima Japan
| | - Wataru Yasui
- Department of Molecular Pathology; Hiroshima University Graduate School of Biomedical Sciences; Hiroshima Japan
| |
Collapse
|
38
|
Bacigalupo ML, Carabias P, Troncoso MF. Contribution of galectin-1, a glycan-binding protein, to gastrointestinal tumor progression. World J Gastroenterol 2017; 23:5266-5281. [PMID: 28839427 PMCID: PMC5550776 DOI: 10.3748/wjg.v23.i29.5266] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/04/2017] [Accepted: 06/18/2017] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal cancer is a group of tumors that affect multiple sites of the digestive system, including the stomach, liver, colon and pancreas. These cancers are very aggressive and rapidly metastasize, thus identifying effective targets is crucial for treatment. Galectin-1 (Gal-1) belongs to a family of glycan-binding proteins, or lectins, with the ability to cross-link specific glycoconjugates. A variety of biological activities have been attributed to Gal-1 at different steps of tumor progression. Herein, we summarize the current literature regarding the roles of Gal-1 in gastrointestinal malignancies. Accumulating evidence shows that Gal-1 is drastically up-regulated in human gastric cancer, hepatocellular carcinoma, colorectal cancer and pancreatic ductal adenocarcinoma tissues, both in tumor epithelial and tumor-associated stromal cells. Moreover, Gal-1 makes a crucial contribution to the pathogenesis of gastrointestinal malignancies, favoring tumor development, aggressiveness, metastasis, immunosuppression and angiogenesis. We also highlight that alterations in Gal-1-specific glycoepitopes may be relevant for gastrointestinal cancer progression. Despite the findings obtained so far, further functional studies are still required. Elucidating the precise molecular mechanisms modulated by Gal-1 underlying gastrointestinal tumor progression, might lead to the development of novel Gal-1-based diagnostic methods and/or therapies.
Collapse
|
39
|
Klameth L, Rath B, Hamilton G. In vitro Cytotoxic Activities of the Oral Platinum(IV) Prodrug Oxoplatin and HSP90 Inhibitor Ganetespib against a Panel of Gastric Cancer Cell Lines. J Cancer 2017; 8:1733-1743. [PMID: 28819369 PMCID: PMC5556635 DOI: 10.7150/jca.17816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 04/01/2017] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer exhibits a poor prognosis and is the third most common cause of cancer death worldwide. Chemotherapy of metastatic gastric cancer is based on combinations of platinum drugs and fluoropyrimidines, with added agents. Oxoplatin is a stable oral platinum(IV) prodrug which is converted to a highly active tetrachlorido(IV) complex under acidic conditions. In the present work, we studied the cytotoxic effects of oxoplatin against a panel of four gastric cancer cell lines in vitro. Furthermore, the role of HSP90 in chemoresistance of these lines was investigated using the specific inhibitor ganetespib. The KATO-III, MKN-1, MKN-28, MKN-45 lines were used in MTT chemosensitivity, cell cycle and apoptosis assays. KATO-III is a signet ring diffuse cell type, MKN-1 an adenosquamous primary, MKN-28 a well-differentiated intestinal type and the MKN-45 a poorly differentiated, diffuse type gastric carcinoma line. Cytotoxicity was tested in MTT assays and intracellular signal transduction with proteome profiler Western blot arrays. Interactions of platinum drugs and ganetespib were calculated with help of the Chou-Talalay method. The prodrug oxoplatin revealed low activity against the four gastric cancer cell lines, whereas the platinum tetrachlorido(IV) complex and cisplatin gave IC50 values of 1-3 µg/ml with increasing chemoresistance observed in the order of MKN-1, KATO-III, MKN-28 to MKN-45. With exception of KATO-III and MKN-28/oxoplatin, all other cell lines featured marked synergistic toxicity with clinically achievable concentrations of ganetespib. Oral administration of a platinum agent such as oxoplatin would be of great value for patients and care providers alike. These results suggest that the oncogene-stabilizing HSP90 chaperone represents an important mediator of chemoresistance in gastric cancer. Ganetespib reduced the phosphorylation of p53, Akt1/2/3 and PRAS40, as well as of WNK1, a kinase which regulates intracellular chloride concentrations. Intracellular chloride was reported to control proliferation of gastric cancer cell lines. Expression of MUC1 was not downregulated in contrast to the expression of CAIX, a prognostic marker in gastric cancer. In conclusion, the HSP90 inhibitor ganetespib synergizes with platinum anticancer drugs and modulates intracellular signal transduction in direction of a less proliferative and aggressive phenotype.
Collapse
Affiliation(s)
- Lukas Klameth
- Department for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Barbara Rath
- Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Gerhard Hamilton
- Department of Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
40
|
Sokolova O, Naumann M. NF-κB Signaling in Gastric Cancer. Toxins (Basel) 2017; 9:toxins9040119. [PMID: 28350359 PMCID: PMC5408193 DOI: 10.3390/toxins9040119] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/14/2017] [Accepted: 03/22/2017] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is a leading cause of cancer death worldwide. Diet, obesity, smoking and chronic infections, especially with Helicobacter pylori, contribute to stomach cancer development. H. pylori possesses a variety of virulence factors including encoded factors from the cytotoxin-associated gene pathogenicity island (cagPAI) or vacuolating cytotoxin A (VacA). Most of the cagPAI-encoded products form a type 4 secretion system (T4SS), a pilus-like macromolecular transporter, which translocates CagA into the cytoplasm of the host cell. Only H. pylori strains carrying the cagPAI induce the transcription factor NF-κB, but CagA and VacA are dispensable for direct NF-κB activation. NF-κB-driven gene products include cytokines/chemokines, growth factors, anti-apoptotic factors, angiogenesis regulators and metalloproteinases. Many of the genes transcribed by NF-κB promote gastric carcinogenesis. Since it has been shown that chemotherapy-caused cellular stress could elicit activation of the survival factor NF-κB, which leads to acquisition of chemoresistance, the NF-κB system is recommended for therapeutic targeting. Research is motivated for further search of predisposing conditions, diagnostic markers and efficient drugs to improve significantly the overall survival of patients. In this review, we provide an overview about mechanisms and consequences of NF-κB activation in gastric mucosa in order to understand the role of NF-κB in gastric carcinogenesis.
Collapse
Affiliation(s)
- Olga Sokolova
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, Magdeburg 39120, Germany.
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, Magdeburg 39120, Germany.
| |
Collapse
|
41
|
Xia J, Wang H, Li S, Wu Q, Sun L, Huang H, Zeng M. Ion channels or aquaporins as novel molecular targets in gastric cancer. Mol Cancer 2017; 16:54. [PMID: 28264681 PMCID: PMC5338097 DOI: 10.1186/s12943-017-0622-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 02/22/2017] [Indexed: 12/21/2022] Open
Abstract
Gastric cancer (GC) is a common disease with few effective treatment choices and poor prognosis, and has the second-highest mortality rates among all cancers worldwide. Dysregulation and/or malfunction of ion channels or aquaporins (AQPs) are common in various human cancers. Furthermore, ion channels are involved in numerous important aspects of the tumor aggressive phonotype, such as proliferation, cell cycle, apoptosis, motility, migration, and invasion. Indeed, by localizing in the plasma membrane, ion channels or AQPs can sense and respond to extracellular environment changes; thus, they play a crucial role in cell signaling and cancer progression. These findings have expanded a new area of pharmaceutical exploration for various types of cancer, including GC. The involvement of multiple ion channels, such as voltage-gated potassium and sodium channels, intracellular chloride channels, ‘transient receptor potential’ channels, and AQPs, which have been shown to facilitate the pathogenesis of other tumors, also plays a role in GC. In this review, an overview of ion channel and aquaporin expression and function in carcinogenesis of GC is presented. Studies of ion channels or AQPs will advance our understanding of the molecular genesis of GC and may identify novel and effective targets for the clinical application of GC.
Collapse
Affiliation(s)
- Jianling Xia
- Cancer Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Hospital of the University of Electronic Science and Technology of China, The Western First Round Road, Section 2#32, Chengdu, 610072, China.,Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hongqiang Wang
- Department of Oncology, Zhoushan Hospital, Zhoushan, 316000, China.,Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shi Li
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, China
| | - Qinghui Wu
- Department of Urology, Hainan Provincial People's Hospital, Haikou, 570311, China
| | - Li Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hongxiang Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ming Zeng
- Cancer Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Hospital of the University of Electronic Science and Technology of China, The Western First Round Road, Section 2#32, Chengdu, 610072, China.
| |
Collapse
|
42
|
Okamoto M, Maeda K, Yanagitani A, Tanaka K. Case of pseudo-Meigs' syndrome caused by gastric cancer-related metastatic ovarian tumor with prolonged survival. World J Gastrointest Oncol 2016; 8:801-804. [PMID: 27895818 PMCID: PMC5108982 DOI: 10.4251/wjgo.v8.i11.801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 08/25/2016] [Accepted: 09/18/2016] [Indexed: 02/05/2023] Open
Abstract
A 48-year-old woman presented with bilateral enlarged ovaries, ascites, bilateral pleural effusion, and advanced gastric cancer. Pleural fluid cytology did not reveal malignant cells. Oophorectomy, performed as a palliative procedure, was followed by rapid resolution of the pleural effusion and ascites. The patient was diagnosed with pseudo-Meigs’ syndrome, and underwent chemotherapy followed by partial gastrectomy. At the last follow-up, 84 mo following oophorectomy, she was alive, and free of disease recurrence, despite not receiving any further treatment. Pseudo-Meigs’ syndrome should be considered in patients with bilateral ovarian tumors, ascites and pleural effusion, and treatment such as oophorectomy may result in symptomatic improvement and better prognosis in similar patients.
Collapse
|
43
|
Zhang XY, Zhang PY. Gastric cancer: somatic genetics as a guide to therapy. J Med Genet 2016; 54:305-312. [PMID: 27609016 DOI: 10.1136/jmedgenet-2016-104171] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 08/11/2016] [Indexed: 12/28/2022]
Abstract
Gastric cancer is the leading cause of cancer-related mortality across the world, with poor prognosis and a median overall survival of ≤12 months for advanced stage gastric cancer. Environmental, genetic and other predisposing factors contribute to the development of gastric cancer and a predominant factor was found to be infection of Helicobacter pylori Advances in understanding the deranged signalling pathways that are critical for normal cellular homeostasis helped in the development of novel drugs that target specific proteins and pathways to curtail the growth of gastric cancer. Genetic studies revealed several single nucleotide polymorphisms, chromosomal aberrations and epigenetic alterations that likely play a major role in elevating the susceptibility to develop gastric cancer. Methylation pattern of specific genes may likely prove to be a valid biomarker for early detection of gastric cancer, but much progress is needed to establish specific markers. Important developments have been made in targeting human epidermal growth factor receptor-2 and vascular endothelial growth factor receptor 2 for treating advanced gastro-oesophageal junction cancer, using specific monoclonal antibodies. Lack of efficacy with regard to targeting other signalling pathways including mesenchymal-epithelial transition/hepatocyte growth factor and mammalian target of rapamycin is probably due to suboptimal patient selection for these clinical trials, which is probably due to the lack of appropriate biomarkers, to decide on responsive patient population. Besides the development of antagonists for the cell growth-related signalling pathways, advances are also being made to tackle gastric cancer by immunotherapies, targeting immune check-points, which may hold promise for better treatment options in future.
Collapse
Affiliation(s)
- Xiao-Ying Zhang
- Nanjing University of Chinese Medicine, Information Institute, Nanjing, Jiangsu, China
| | - Pei-Ying Zhang
- Xuzhou Central Hospital, Xuzhou, Jiangsu Province, China.,The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou, Jiangsu Province, China.,Xuzhou Clinical School of Xuzhou Medical College, Xuzhou, Jiangsu Province, China.,Xuzhou Clinical Medical College of Nanjing University of Chinese Medicine, Xuzhou, Jiangsu Province, China
| |
Collapse
|
44
|
Deng R, Cui JY, Tao KX. Role of B7-H1 in gastrointestinal cancer. Shijie Huaren Xiaohua Zazhi 2016; 24:3135-3141. [DOI: 10.11569/wcjd.v24.i20.3135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Immune evasion is a hallmark of cancer. Although cancer cells have been shown to be able to be recognized by T cells, host immune system fails to develop effective antitumor activity and tumor control. B7-H1, an immune inhibitory molecule, plays an important role in the immune evasion process. B7-H1 inhibits T cell immunity during immune priming and effector phases and is also implicated in intrinsic proliferation, apoptosis and migration of tumor cells. Targeting B7-H1 using blockade antibodies has generated immense antitumor activity in preclinical tumor models. Durable response for a variety of tumor types was also documented in clinical trials. Thus, B7-H1 targeted immune therapy offers a new line of tumor treatment. Gastrointestinal cancer is one of the leading causes of tumor morbidity. However, traditional therapy has not been able to effectively improve survival. This review will focus on the role of B7-H1 in immune evasion and the latest progression of the B7-H1 blockade immune therapy in gastrointestinal cancer.
Collapse
|
45
|
Lian Y, Cai Z, Gong H, Xue S, Wu D, Wang K. HOTTIP: a critical oncogenic long non-coding RNA in human cancers. MOLECULAR BIOSYSTEMS 2016; 12:3247-3253. [PMID: 27546609 DOI: 10.1039/c6mb00475j] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We focus on the current knowledge of HOTTIP in various cancers and illustrate the corresponding mechanism and biological function of HOTTIP during tumor development.
Collapse
Affiliation(s)
- Yifan Lian
- Department of Oncology
- Second Affiliated Hospital
- Nanjing Medical University
- Nanjing 210000
- People's Republic of China
| | - Zeling Cai
- Department of General Surgery
- Second Affiliated Hospital
- Nanjing Medical University
- Nanjing
- People's Republic of China
| | - Huangbo Gong
- Department of General Surgery
- Second Affiliated Hospital
- Nanjing Medical University
- Nanjing
- People's Republic of China
| | - Songling Xue
- Department of Obstetrics and Gynecology
- The Second Affiliated Hospital of Southeast University
- Nanjing
- People's Republic of China
| | - Dongdong Wu
- Department of General Surgery
- Second Affiliated Hospital
- Nanjing Medical University
- Nanjing
- People's Republic of China
| | - Keming Wang
- Department of Oncology
- Second Affiliated Hospital
- Nanjing Medical University
- Nanjing 210000
- People's Republic of China
| |
Collapse
|