1
|
Shi Y, Wang J, Yuan Q, Chen Y, Zhao M, Li X, Wang Z, Zhou H, Zhu F, Wei B, Jiang Y, Zhao J, Qiao Y, Dong Z, Liu K. DDX5 promotes esophageal squamous cell carcinoma growth through sustaining VAV3 mRNA stability. Oncogene 2024; 43:3240-3254. [PMID: 39289531 DOI: 10.1038/s41388-024-03162-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 09/19/2024]
Abstract
Novel therapeutic targets and their inhibitors for esophageal squamous cell carcinoma (ESCC) prevention and therapy are urgently needed. This study aimed to investigate the function of DEAD-box helicase 5 (DDX5) in ESCC progression and to identify a promising inhibitor of DDX5. We verified that DDX5 was highly expressed in ESCC and played an oncogenic role, binding with vav guanine nucleotide exchange factor 3 (VAV3) mRNA and facilitating VAV3 mRNA N6-methyladenosine (m6A) modification by interacting with the m6A methyltransferase 3 (METTL3). M6A-modified VAV3 mRNA was identified by insulin-like growth factor 1 (IGF2BP1), increasing mRNA stability. Methylnissolin-3-β-D-O-glucoside (MD) inhibited ESCC progression through the DDX5-VAV3 axis. Our findings suggest that DDX5 promotes ESCC progression. MD inhibits ESCC progression by targeting DDX5.
Collapse
Affiliation(s)
- Yunshu Shi
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
- Department of Molecule and Pathology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Junyong Wang
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Qiang Yuan
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Yingying Chen
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Miao Zhao
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiaoyu Li
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zitong Wang
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Hao Zhou
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Fangli Zhu
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Bing Wei
- Department of Molecule and Pathology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Yanan Jiang
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan, China
| | - Jimin Zhao
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| | - Yan Qiao
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
| | - Zigang Dong
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan, China.
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China.
| | - Kangdong Liu
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China.
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China.
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China.
| |
Collapse
|
2
|
Jin S, Wang Q, Wei Y, Ju Y, Wang K. Patients With Gastric Cancer Who Undergo HIPEC Treatment and Have a Peritoneal Cancer Index of Less Than 12 Can Benefit From Surgery. Am J Ther 2024:00045391-990000000-00208. [PMID: 39167429 DOI: 10.1097/mjt.0000000000001798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Affiliation(s)
- Shiyang Jin
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | | | | | | | | |
Collapse
|
3
|
Wang Q, Shen K, Fei B, Wei M, Ge X, Xie Z. Development and validation of a nomogram to predict cancer-specific survival of elderly patients with unresected gastric cancer who received chemotherapy. Sci Rep 2024; 14:9008. [PMID: 38637579 PMCID: PMC11026516 DOI: 10.1038/s41598-024-59516-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 04/11/2024] [Indexed: 04/20/2024] Open
Abstract
This investigation aimed to explore the prognostic factors in elderly patients with unresected gastric cancer (GC) who have received chemotherapy and to develop a nomogram for predicting their cancer-specific survival (CSS). Elderly gastric cancer patients who have received chemotherapy but no surgery in the Surveillance, Epidemiology, and End Results Database between 2004 and 2015 were included in this study. Cox analyses were conducted to identify prognostic factors, leading to the formulation of a nomogram. The nomogram was validated using receiver operating characteristic (ROC) and calibration curves. The findings elucidated six prognostic factors encompassing grade, histology, M stage, radiotherapy, tumor size, and T stage, culminating in the development of a nomogram. The ROC curve indicated that the area under curve of the nomogram used to predict CSS for 3, 4, and 5 years in the training queue as 0.689, 0.708, and 0.731, and in the validation queue, as 0.666, 0.693, and 0.708. The calibration curve indicated a high degree of consistency between actual and predicted CSS for 3, 4, and 5 years. This nomogram created to predict the CSS of elderly patients with unresected GC who have received chemotherapy could significantly enhance treatment accuracy.
Collapse
Affiliation(s)
- Qi Wang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Kexin Shen
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Bingyuan Fei
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Mengqiang Wei
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xinbin Ge
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhongshi Xie
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
4
|
Xu P, Liu K, Huang S, Lv J, Yan Z, Ge H, Cheng Q, Chen Z, Ji P, Qian Y, Li B, Xu H, Yang L, Xu Z, Zhang D. N 6-methyladenosine-modified MIB1 promotes stemness properties and peritoneal metastasis of gastric cancer cells by ubiquitinating DDX3X. Gastric Cancer 2024; 27:275-291. [PMID: 38252226 DOI: 10.1007/s10120-023-01463-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Peritoneal metastasis (PM), one of the most typical forms of metastasis in advanced gastric cancer (GC), indicates a poor prognosis. Exploring the potential molecular mechanism of PM is urgently necessary, as it has not been well studied. E3 ubiquitin ligase has been widely established to exert a biological function in various cancers, but its mechanism of action in GC with PM remains unknown. METHODS The effect of MIB1 on PM of GC was confirmed in vitro and in vivo. Co-immunoprecipitation (Co-IP) and mass spectrometry demonstrated the association between MIB1 and DDX3X. Western blot, flow cytometry and immunofluorescence determined that DDX3X was ubiquitylated by MIB1 and promoted stemness. We further confirmed that METTL3 promoted the up-regulation of MIB1 by RNA immunoprecipitation (RIP), luciferase reporter assay and other experiments. RESULTS We observed that the E3 ubiquitin ligase Mind bomb 1 (MIB1) was highly expressed in PMs, and patients with PM with high MIB1 expression showed a worse prognosis than those with low MIB1 expression. Mechanistically, our study demonstrated that the E3 ubiquitin ligase MIB1 promoted epithelial-mesenchymal transition (EMT) progression and stemness in GC cells by degrading DDX3X. In addition, METTL3 mediated m6A modification to stabilize MIB1, which required the m6A reader IGF2BP2. CONCLUSIONS Our study elucidated the specific molecular mechanism by which MIB1 promotes PM of GC, and suggested that targeting the METTL3-MIB1-DDX3X axis may be a promising therapeutic strategy for GC with PM.
Collapse
Affiliation(s)
- Peng Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Kanghui Liu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shansong Huang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jialun Lv
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zhengyuan Yan
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Department of Surgery, Nanjing Lishui People's Hospital, Nanjing, 211200, China
| | - Han Ge
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Quan Cheng
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zetian Chen
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Peicheng Ji
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yawei Qian
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Bowen Li
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Hao Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Li Yang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zekuan Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Diancai Zhang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
5
|
Wen L, Xu K, Huang M, Pan Q. Identification of oxeiptosis-associated lncRNAs and prognosis-related signature to predict the immune status in gastric cancer. Medicine (Baltimore) 2024; 103:e37189. [PMID: 38363905 PMCID: PMC10869064 DOI: 10.1097/md.0000000000037189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 01/17/2024] [Indexed: 02/18/2024] Open
Abstract
As a novel form of cell death, oxeiptosis is mainly caused by oxidative stress and has been defined to contribute to the cellular death program in cancer. However, the precise involvement of oxeiptosis-related long non-coding RNAs (lncRNAs) within gastric cancer (GC) remains elusive. Thus, our study was aimed to elucidate the pivotal effect of hub oxeiptosis-related lncRNAs on GC by comprehensively analyzing lncRNA and gene expression data obtained from The Cancer Genome Atlas (TCGA) database. Subsequently, we constructed a risk signature (risk-sig) using lncRNAs and further evaluated its prognostic significance. We successfully identified thirteen lncRNAs closely related with oxeiptosis that exhibited significant relevance to the prognosis of GC, forming the foundation of our meticulously constructed risk-sig. Notably, our clinical analyses unveiled a strong correlation between the risk-sig and crucial clinical parameters including overall survival (OS), gender, TNM stage, grade, M stage, and N stage among GC patients. Intriguingly, the diagnostic accuracy of this risk-sig surpassed that of conventional clinicopathological characteristics, underscoring its potential as a highly informative prognostic tool. In-depth mechanistic investigations further illuminated a robust association between this risk-sig and fundamental biological processes such as tumor stemness, immune cell infiltration, and immune subtypes. These findings provide valuable insights into the complex interplay between oxeiptosis-related lncRNAs and the intricate molecular landscape of GC. Ultimately, leveraging the risk scores derived from our comprehensive analysis, we successfully developed a nomogram that enables accurate prediction of GC prognosis. Collectively, our study established a solid foundation for the integration of thirteen hub oxeiptosis-related lncRNAs into a clinically applicable risk-sig, potentially revolutionizing prognostic assessment in GC and facilitating the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Li Wen
- Nursing Department, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Kaili Xu
- Department of Operating Room, The First People’s Hospital of Linping District, Hangzhou, China
| | - Min Huang
- Department of Medicine, Zhejiang Rehabilitation Hospital, Hangzhou, China
| | - Qin Pan
- Department of Operating Room, The First People’s Hospital of Linping District, Hangzhou, China
| |
Collapse
|
6
|
Shi Y, Yuan Q, Chen Y, Li X, Zhou Y, Zhou H, Peng F, Jiang Y, Qiao Y, Zhao J, Zhang C, Wang J, Liu K, Dong Z. Corynoline inhibits esophageal squamous cell carcinoma growth via targeting Pim-3. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155235. [PMID: 38128397 DOI: 10.1016/j.phymed.2023.155235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/09/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is an aggressive and deadly malignancy characterized by late-stage diagnosis, therapy resistance, and a poor 5-year survival rate. Finding novel therapeutic targets and their inhibitors for ESCC prevention and therapy is urgently needed. METHODS We investigated the proviral integration site for maloney murine leukemia virus 3 (Pim-3) protein levels using immunohistochemistry. Using Methyl Thiazolyl Tetrazolium and clone formation assay, we verified the function of Pim-3 in cell proliferation. The binding and inhibition of Pim-3 by corynoline were verified by computer docking, pull-down assay, cellular thermal shift assay, and kinase assay. Cell proliferation, Western blot, and a patient-derived xenograft tumor model were performed to elucidate the mechanism of corynoline inhibiting ESCC growth. RESULTS Pim-3 was highly expressed in ESCC and played an oncogenic role. The augmentation of Pim-3 enhanced cell proliferation and tumor development by phosphorylating mitogen-activated protein kinase 1 (MAPK1) at T185 and Y187. The deletion of Pim-3 induced apoptosis with upregulated cleaved caspase-9 and lower Bcl2 associated agonist of cell death (BAD) phosphorylation at S112. Additionally, binding assays demonstrated corynoline directly bound with Pim-3, inhibiting its activity, and suppressing ESCC growth. CONCLUSIONS Our findings suggest that Pim-3 promotes ESCC progression. Corynoline inhibits ESCC progression through targeting Pim-3.
Collapse
Affiliation(s)
- Yunshu Shi
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan 450052, China
| | - Qiang Yuan
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan 450052, China
| | - Yingying Chen
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Xiaoyu Li
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Yujuan Zhou
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Hao Zhou
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Feng Peng
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Yanan Jiang
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan 450052, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan 450000, China; Center for Basic Medical Research, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Yan Qiao
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Jimin Zhao
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan 450000, China; Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan 450000, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan 450000, China
| | - Chi Zhang
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Junyong Wang
- Center for Basic Medical Research, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450000, China.
| | - Kangdong Liu
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan 450052, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan 450000, China; Center for Basic Medical Research, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450000, China; Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan 450000, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan 450000, China.
| | - Zigang Dong
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan 450052, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan 450000, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan 450000, China.
| |
Collapse
|
7
|
Yang J, Shu G, Chen T, Dong A, Dong C, Li W, Sun X, Zhou Y, Li D, Zhou J. ESM1 Interacts with c-Met to Promote Gastric Cancer Peritoneal Metastasis by Inducing Angiogenesis. Cancers (Basel) 2023; 16:194. [PMID: 38201620 PMCID: PMC10778290 DOI: 10.3390/cancers16010194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
The peritoneum is the most common metastatic site of advanced gastric cancer and is associated with extremely poor prognosis. Endothelial-specific molecule 1 (ESM1) was found to be significantly associated with gastric cancer peritoneal metastasis (GCPM); however, the biological functions and molecular mechanisms of ESM1 in regulating GCPM remain unclear. Herein, we demonstrated that ESM1 expression was significantly upregulated in gastric cancer tissues and positively correlated with platelet endothelial cell adhesion molecule-1 (CD31) levels. Moreover, clinical validation, in in vitro and in vivo experiments, confirmed that ESM1 promoted gastric cancer angiogenesis, eventually promoting gastric cancer peritoneal metastasis. Mechanistically, ESM1 promoted tumor angiogenesis by binding to c-Met on the vascular endothelial cell membrane. In addition, our results confirmed that ESM1 upregulated VEGFA, HIF1α, and MMP9 expression and induced angiogenesis by activating the MAPK/ERK pathway. In conclusion, our findings identified the role of ESM1 in gastric cancer angiogenesis and GCPM, thus providing insights into the diagnosis and treatment of advanced gastric cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Dongbao Li
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; (J.Y.); (G.S.); (T.C.); (A.D.); (C.D.); (W.L.); (X.S.); (Y.Z.)
| | - Jin Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; (J.Y.); (G.S.); (T.C.); (A.D.); (C.D.); (W.L.); (X.S.); (Y.Z.)
| |
Collapse
|
8
|
Tan Z, Ko JMY, Yu VZ, Lam KO, Kwong DLW, Wong IYH, Chan FSY, Wong CLY, Chan KK, Law TT, Choy FSF, Ng HY, Law SYK, Lung ML. Multigene Profiling of Circulating Tumor Cells in Esophageal Squamous Cell Carcinoma Identifies Prognostic Cancer Driver Genes Associated with Epithelial-Mesenchymal-Transition Progression and Chemoresistance. Cancers (Basel) 2023; 15:5329. [PMID: 38001588 PMCID: PMC10670643 DOI: 10.3390/cancers15225329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/15/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
We investigated the clinical significance of CTCs in cancer progression by detecting multiple cancer driver genes associated with epithelial-to-mesenchymal transition (EMT) at the transcript level. The 10-gene panel, comprising CCND1, ECT2, EpCAM, FSCN1, KRT5, KRT18, MET, TFRC, TWIST1, and VEGFC, was established for characterizing CTCs from mouse ESCC xenograft models and clinical ESCC peripheral blood (PB) samples. Correlations between gene expression in CTCs from PB samples (n = 77) and clinicopathological features in ESCC patients (n = 55) were examined. The presence of CTCs at baseline was significantly correlated with tumor size (p = 0.031). The CTC-high patients were significantly correlated with advanced cancer stages (p = 0.013) and distant metastasis (p = 0.029). High mRNA levels of TWIST1 (Hazard Ratio (HR) = 5.44, p = 0.007), VEGFC (HR = 6.67, p < 0.001), TFRC (HR = 2.63, p = 0.034), and EpCAM (HR = 2.53, p = 0.041) at baseline were significantly associated with a shorter overall survival (OS) in ESCC patients. This study also revealed that TWIST1 facilitates EMT and enhances malignant potential by promoting tumor migration, invasion, and cisplatin chemoresistance through the TWIST1-TGFBI-ZEB1 axis in ESCC, highlighting the prognostic and therapeutic potential of TWIST1 in clinical ESCC treatment.
Collapse
Affiliation(s)
- Zhen Tan
- Department of Clinical Oncology, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Josephine Mun-Yee Ko
- Department of Clinical Oncology, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Valen Zhuoyou Yu
- Department of Clinical Oncology, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Ka-On Lam
- Department of Clinical Oncology, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Dora Lai-Wan Kwong
- Department of Clinical Oncology, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Ian Yu-Hong Wong
- Department of Surgery, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Fion Siu-Yin Chan
- Department of Surgery, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Claudia Lai-Yin Wong
- Department of Surgery, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Kwan-Kit Chan
- Department of Surgery, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Tsz-Ting Law
- Department of Surgery, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Faith Sin-Fai Choy
- Department of Clinical Oncology, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Hoi-Yan Ng
- Department of Clinical Oncology, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Simon Ying-Kit Law
- Department of Surgery, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Maria Li Lung
- Department of Clinical Oncology, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| |
Collapse
|
9
|
Lv H, Jia X, Yang H, Zhu X, Zhao Z, Jiang X. Prediction of the mechanism for the combination of diallyl trisulfide and cisplatin against gastric cancer: a network pharmacology study and pharmacological evaluation. Front Pharmacol 2023; 14:1269895. [PMID: 37964870 PMCID: PMC10642245 DOI: 10.3389/fphar.2023.1269895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/16/2023] [Indexed: 11/16/2023] Open
Abstract
Background: In this research, we aimed to explore the efficacy of diallyl trisulfide (DATS) combined with cisplatin (DDP) for gastric cancer treatment and its underlying mechanism based on network pharmacology. Methods: First, the pharmacological mechanism by which DATS combined with DDP acts against gastric cancer was predicted using network pharmacology. The TTD, GeneCards, and OMIM databases were used to extract drug and disease targets. The David Bioinformatics Resources 6.8 database was used to conduct GO and KEGG analyses. We investigated the efficacy of DATS combined with DDP against gastric cancer in SGC7901 cells and a xenograft model. Furthermore, the specific mechanism of DATS combined with DDP, inferred by network pharmacology, was identified by Western blotting and immunohistochemistry. Results: The combination of DDP and DATS significantly increased cytotoxicity and cell apoptosis compared to the DATS or DDP treatment group in vitro. In addition, continuous intraperitoneal injection of DATS markedly improved the tumor inhibitory effect of DDP in the SGC-7901 tumor-bearing mouse model. Furthermore, network pharmacology and experimental validation studies revealed that the combination of DATS and DDP synergistically enhanced antitumor activity by regulating endoplasmic reticulum stress and inhibiting STAT3/PKC-δ and MAPK signaling pathways. Conclusion: Our study showed that the combination of DATS and DDP could exert outstanding therapeutic effects in gastric cancer. Moreover, network pharmacology coupled with experimental validation revealed the molecular mechanisms of combination therapy for gastric cancer. This study offers a new adjuvant strategy based on DATS and DDP for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Huaiyou Lv
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, China
- Department of Pharmacy, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China
| | - Xiumei Jia
- Department of Pharmacy, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China
| | - Huatian Yang
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, China
| | - Xiaosong Zhu
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, China
- Department of Infection Management, Linyi People’s Hospital, Linyi, Shandong, China
| | - Zhongxi Zhao
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, China
| | - Xiaoyan Jiang
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, China
| |
Collapse
|
10
|
Long J, Zhu B, Tian T, Ren L, Tao Y, Zhu H, Li D, Xu Y. Activation of UBEC2 by transcription factor MYBL2 affects DNA damage and promotes gastric cancer progression and cisplatin resistance. Open Med (Wars) 2023; 18:20230757. [PMID: 37840753 PMCID: PMC10571520 DOI: 10.1515/med-2023-0757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 10/17/2023] Open
Abstract
Ubiquitin-conjugating enzyme E2 C (UBE2C) plays a carcinogenic role in gastric cancer (GC); yet, its role in cisplatin (DDP) resistance in GC is enigmatic. This study sought to probe into the impact of UBE2C on DDP resistance in GC and its concrete molecular mechanism in GC progression. Bioinformatics analysis was used to analyze differentially expressed mRNAs and predict upstream regulatory molecules in GC. Real-time quantitative reverse transcriptase polymerase chain reaction and western blot were used to detect the expression of UBE2C and MYB proto-oncogene like 2 (MYBL2). Dual luciferase and chromatin immunoprecipitation (ChIP) assays were used to verify the binding relationship. Cell counting kit-8 was used to detect cell viability and calculate IC50 values. Flow cytometry was used to detect the cell cycle. Comet assay was used to detect DNA damage. Western blot was used to detect the expression of DNA loss-related proteins (γ-H2AX, ATM/p-ATM). The knockdown of highly expressed UBE2C in GC cell lines could reduce cell viability, induce G2/M arrest, induce apoptosis, and promote DNA damage and DDP sensitivity. Bioinformatics analysis predicted that the substantially upregulated MYBL2 was an upstream transcription factor in UBE2C. The binding relationship between the UBE2C promoter region and MYBL2 was verified by dual luciferase and ChIP. Overexpression of UBE2C in the rescue experiment was found to reverse the inhibited GC progression and promoted DDP sensitivity brought by the knockdown of MYBL2. In conclusion, the MYBL2/UBE2C regulatory axis may be a potential way to overcome DDP resistance in GC.
Collapse
Affiliation(s)
- Jiegen Long
- Department of General Surgery, Affiliated Banan Hospital of Chongqing Medical University, Chongqing, 401320, China
| | - Bin Zhu
- Department of General Surgery, Affiliated Banan Hospital of Chongqing Medical University, Chongqing, 401320, China
| | - Tao Tian
- Department of General Surgery, Affiliated Banan Hospital of Chongqing Medical University, Chongqing, 401320, China
| | - Linfei Ren
- Department of General Surgery, Affiliated Banan Hospital of Chongqing Medical University, Chongqing, 401320, China
| | - Yong Tao
- Department of General Surgery, Affiliated Banan Hospital of Chongqing Medical University, Chongqing, 401320, China
| | - Haitao Zhu
- Department of General Surgery, Affiliated Banan Hospital of Chongqing Medical University, Chongqing, 401320, China
| | - Dengwei Li
- Department of General Surgery, Affiliated Banan Hospital of Chongqing Medical University, Chongqing, 401320, China
| | - Yonghong Xu
- Department of General Surgery, Affiliated Banan Hospital of Chongqing Medical University, No. 659, Yunan Road, Longzhouwan Street, Banan District, Chongqing, 401320, China
| |
Collapse
|
11
|
Klempner SJ, Lee KW, Shitara K, Metges JP, Lonardi S, Ilson DH, Fazio N, Kim TY, Bai LY, Moran D, Yang J, Arozullah A, Park JW, Raizer JJ, Bang YJ, Shah MA. ILUSTRO: Phase II Multicohort Trial of Zolbetuximab in Patients with Advanced or Metastatic Claudin 18.2-Positive Gastric or Gastroesophageal Junction Adenocarcinoma. Clin Cancer Res 2023; 29:3882-3891. [PMID: 37490286 PMCID: PMC10543966 DOI: 10.1158/1078-0432.ccr-23-0204] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/20/2023] [Accepted: 07/20/2023] [Indexed: 07/26/2023]
Abstract
PURPOSE Zolbetuximab, an IgG1 monoclonal antibody, binds to claudin 18.2 (CLDN18.2) and mediates tumor cell death through antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity. We sought to examine zolbetuximab combinations in CLDN18.2-positive HER2-negative gastric/gastroesophageal junction (G/GEJ) adenocarcinoma. PATIENTS AND METHODS This phase II study assessed efficacy and safety of zolbetuximab, alone or with modified FOLFOX6 (mFOLFOX6) or pembrolizumab, in CLDN18.2-positive advanced/metastatic G/GEJ adenocarcinoma. Patients received zolbetuximab as monotherapy in third/later-line (Cohort 1A, n = 30), with mFOLFOX6 in first-line (Cohort 2, n = 21), or with pembrolizumab in third/later-line (Cohort 3A, n = 3) treatment. The primary endpoint for Cohort 1A was objective response rate (ORR). Key secondary endpoints were ORR (Cohorts 2 and 3A), overall survival (OS; Cohort 1A), and progression-free survival (PFS) and safety (all cohorts). RESULTS ORR was 0% in Cohorts 1A and 3A, and 71.4% [95% confidence interval (CI), 47.82-88.72] in Cohort 2. Median PFS was 1.54 months (95% CI, 1.31-2.56) in Cohort 1A, 2.96 months (95% CI, 1.48-4.44) in Cohort 3A, and 17.8 months (95% CI, 8.05-25.69) in Cohort 2. Median OS in Cohort 1A was 5.62 months (95% CI, 2.27-11.53). Gastrointestinal adverse events occurred across cohorts [nausea, 63%-90% (grade ≥ 3, 4.8%-6.7%) and vomiting, 33%-67% (grade ≥ 3, 6.7%-9.5%)]. CONCLUSIONS Zolbetuximab plus mFOLFOX6 demonstrated promising efficacy in previously untreated patients with CLDN18.2-positive G/GEJ adenocarcinoma. These data support the first-line development of zolbetuximab in patients whose tumors are CLDN18.2-positive. Across cohorts, zolbetuximab treatment was tolerable with no new safety signals.
Collapse
Affiliation(s)
- Samuel J. Klempner
- Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Keun-Wook Lee
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | | | - Sara Lonardi
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - David H. Ilson
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nicola Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | - Tae Yong Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Li-Yuan Bai
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, and College of Medicine, China Medical University, Taichung, Taiwan
| | | | | | | | | | - Jeffrey J. Raizer
- Clinical Sciences, Oncology, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts
| | - Yung-Jue Bang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Manish A. Shah
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, New York
| |
Collapse
|
12
|
Lin G, Liu Z, Shang‐Guan Z, Zeng G, Lin J, Wu J, Chen Q, Xie J, Li P, Huang C, Zheng C. Comparison of the efficacy between immunochemotherapy and chemotherapy in gastric cancer accompanied with synchronous liver metastases: A real-world retrospective study. Cancer Med 2023; 12:12221-12233. [PMID: 37062073 PMCID: PMC10278523 DOI: 10.1002/cam4.5917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/20/2023] [Accepted: 03/27/2023] [Indexed: 04/17/2023] Open
Abstract
BACKGROUND Few studies have investigated the efficacy of comprehensive therapies, including immunotherapy, for gastric cancer with synchronous liver metastases (GCLM). We retrospectively compared the effect of immunochemotherapy and chemotherapy alone as conversion therapies on the oncological outcomes of patients with GCLM. METHODS The clinicopathological data of 100 patients with GCLM from February 2017 to October 2021 at our institution were retrospectively analyzed. Patients were divided into immunochemotherapy (n = 33) and chemotherapy-alone (n = 67) groups. RESULTS Baseline clinicopathological data did not differ significantly between the two groups. The immunochemotherapy group had a higher overall response rate (59.4% vs. 44.0%, p = 0.029) and disease control rate (71.9% vs. 49.2%, p = 0.036) than the chemotherapy group. The immunochemotherapy group showed better tumor regression in the gastric mass, metastatic lymph nodes, and liver lesions than the chemotherapy group. Ten (30.3%) patients in the immunochemotherapy group and 13 (19.4%) patients in the chemotherapy group underwent surgery after conversion therapy. However, the difference was not statistically significant. The overall survival (OS) and progression-free survival (PFS) rates were better in the immunochemotherapy group than in the chemotherapy group. Treatment-related adverse events occurred in 24 (72.7%) and 47 (70.1%) patients in the immunochemotherapy and chemotherapy groups, respectively. CONCLUSIONS As a conversion therapy for GCLM, immunotherapy yielded better primary and metastatic tumor regression and survival benefits, with no increase in adverse events compared to chemotherapy.
Collapse
Affiliation(s)
- Guang‐Tan Lin
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
| | - Zhi‐Yu Liu
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
| | - Zhi‐Xin Shang‐Guan
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
| | - Gui‐Rong Zeng
- Fujian Medical University Diagnostic Pathology CenterFujian Medical UniversityFuzhouChina
| | - Jian‐Xian Lin
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
- Key Laboratory of Ministry of Education of Gastrointestinal CancerFujian Medical UniversityFuzhouChina
| | - Ju Wu
- Department of General SurgeryAffiliated Zhongshan Hospital of Dalian UniversityDalianChina
| | - Qi‐Yue Chen
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
| | - Jian‐Wei Xie
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
| | - Ping Li
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
- Key Laboratory of Ministry of Education of Gastrointestinal CancerFujian Medical UniversityFuzhouChina
| | - Chang‐Ming Huang
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
- Key Laboratory of Ministry of Education of Gastrointestinal CancerFujian Medical UniversityFuzhouChina
| | - Chao‐Hui Zheng
- Department of Gastric SurgeryFujian Medical University Union HospitalFuzhouChina
- Department of General SurgeryFujian Medical University Union HospitalFuzhouChina
- Key Laboratory of Ministry of Education of Gastrointestinal CancerFujian Medical UniversityFuzhouChina
| |
Collapse
|
13
|
Shah MA, Altorki N, Patel P, Harrison S, Bass A, Abrams JA. Improving outcomes in patients with oesophageal cancer. Nat Rev Clin Oncol 2023; 20:390-407. [PMID: 37085570 DOI: 10.1038/s41571-023-00757-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 04/23/2023]
Abstract
The care of patients with oesophageal cancer or of individuals who have an elevated risk of oesophageal cancer has changed dramatically. The epidemiology of squamous cell and adenocarcinoma of the oesophagus has diverged over the past several decades, with a marked increase in incidence only for oesophageal adenocarcinoma. Only in the past decade, however, have molecular features that distinguish these two forms of the disease been identified. This advance has the potential to improve screening for oesophageal cancers through the development of novel minimally invasive diagnostic technologies predicated on cancer-specific genomic or epigenetic alterations. Surgical techniques have also evolved towards less invasive approaches associated with less morbidity, without compromising oncological outcomes. With improvements in multidisciplinary care, advances in radiotherapy and new tools to detect minimal residual disease, certain patients may no longer even require surgical tumour resection. However, perhaps the most anticipated advance in the treatment of patients with oesophageal cancer is the advent of immune-checkpoint inhibitors, which harness and enhance the host immune response against cancer. In this Review, we discuss all these advances in the management of oesophageal cancer, representing only the beginning of a transformation in our quest to improve patient outcomes.
Collapse
Affiliation(s)
- Manish A Shah
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - Nasser Altorki
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Pretish Patel
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sebron Harrison
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Adam Bass
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Julian A Abrams
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
14
|
Yang J, Xu P, Chen Z, Zhang X, Xia Y, Fang L, Xie L, Li B, Xu Z. N6-methyadenosine modified SUV39H2 regulates homologous recombination through epigenetic repression of DUSP6 in gastric cancer. Cancer Lett 2023; 558:216092. [PMID: 36806557 DOI: 10.1016/j.canlet.2023.216092] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/02/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023]
Abstract
Despite many advances in treatment over the past few years, the poor 5-year survival rate and high recurrence rate of gastric cancer (GC) remain unsatisfactory. As the most abundant epigenetic modification in the eukaryotic mRNA, N6-methyladenosine (m6A) methylation participates in tumor progression and tissue development. During tumor progression, DNA damage repair mechanisms can be reprogrammed to give new growth advantages on tumor clones whose genomic integrity is disturbed. Here we detected the elevated SUV39H2 expression in GC tissues and cell lines. Functionally, SUV39H2 promoted GC proliferation and inhibited apoptosis in vitro and in vivo. Mechanistically, METTL3-mediated m6A modification promotes mRNA stability of SUV39H2 in an IGF2BP2 dependent manner, resulting in upregulated mRNA expression of SUV39H2. As a histone methyltransferase, SUV39H2 was verified to increase the phosphorylation level of ATM through transcriptional repression of DUSP6, thereby promoting HRR and ultimately inhibiting GC chemosensitivity to cisplatin. Collectively, these results indicate the specific mechanism of m6A-modified SUV39H2 as a histone methyltransferase promoting HRR to inhibit the chemosensitivity of GC. SUV39H2 is expected to become a key target in the precision targeted therapy of GC.
Collapse
Affiliation(s)
- Jing Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Penghui Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Zetian Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Xing Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Yiwen Xia
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Lang Fang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Li Xie
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Bowen Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, China.
| |
Collapse
|
15
|
Roshani M, Baniebrahimi G, Mousavi M, Zare N, Sadeghi R, Salarinia R, Sheida A, Molavizadeh D, Sadeghi S, Moammer F, Zolfaghari MR, Mirzaei H. Exosomal long non-coding RNAs: novel molecules in gastrointestinal cancers' progression and diagnosis. Front Oncol 2022; 12:1014949. [PMID: 36591473 PMCID: PMC9795196 DOI: 10.3389/fonc.2022.1014949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/10/2022] [Indexed: 12/15/2022] Open
Abstract
Gastrointestinal (GI) cancers arise in the GI tract and accessory organs, including the mouth, esophagus, stomach, liver, biliary tract, pancreas, small intestine, large intestine, and rectum. GI cancers are a major cause of cancer-related morbidity and mortality worldwide. Exosomes act as mediators of cell-to-cell communication, with pleiotropic activity in the regulation of homeostasis, and can be markers for diseases. Non-coding RNAs (ncRNAs), such as long non-coding RNAs (lncRNAs), can be transported by exosomes derived from tumor cells or non-tumor cells. They can be taken by recipient cells to alter their function or remodel the tumor microenvironment. Moreover, due to their uniquely low immunogenicity and excellent stability, exosomes can be used as natural carriers for therapeutic ncRNAs in vivo. Exosomal lncRNAs have a crucial role in regulating several cancer processes, including angiogenesis, proliferation, drug resistance, metastasis, and immunomodulation. Exosomal lncRNA levels frequently alter according to the onset and progression of cancer. Exosomal lncRNAs can therefore be employed as biomarkers for the diagnosis and prognosis of cancer. Exosomal lncRNAs can also monitor the patient's response to chemotherapy while also serving as potential targets for cancer treatment. Here, we discuss the role of exosomal lncRNAs in the biology and possible future treatment of GI cancer.
Collapse
Affiliation(s)
- Mohammad Roshani
- Internal Medicine and Gastroenterology, Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Baniebrahimi
- Department of Pediatric Dentistry, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahboubeh Mousavi
- Department of Anatomy, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Noushid Zare
- Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Reza Sadeghi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Salarinia
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Sciences, Bojnurd, Iran
| | - Amirhossein Sheida
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran,Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Danial Molavizadeh
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran,Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Sara Sadeghi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran,Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Farzaneh Moammer
- Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran,*Correspondence: Farzaneh Moammer, ; Mohammad Reza Zolfaghari, ; Hamed Mirzaei, ;
| | - Mohammad Reza Zolfaghari
- Department of Microbiology, Qom Branch, Islamic Azad University, Qom, Iran,*Correspondence: Farzaneh Moammer, ; Mohammad Reza Zolfaghari, ; Hamed Mirzaei, ;
| | - Hamed Mirzaei
- Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran,*Correspondence: Farzaneh Moammer, ; Mohammad Reza Zolfaghari, ; Hamed Mirzaei, ;
| |
Collapse
|
16
|
Shitara K, Kawazoe A, Hirakawa A, Nakanishi Y, Furuki S, Fukuda M, Ueno Y, Raizer J, Arozullah A. Phase 1 trial of zolbetuximab in Japanese patients with CLDN18.2+ gastric or gastroesophageal junction adenocarcinoma. Cancer Sci 2022; 114:1606-1615. [PMID: 36478334 PMCID: PMC10067400 DOI: 10.1111/cas.15684] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/22/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
Zolbetuximab is a chimeric monoclonal antibody that targets claudin-18.2, a candidate biomarker in patients with advanced gastric/gastroesophageal cancer. This nonrandomized phase 1 study (NCT03528629) enrolled previously treated Japanese patients with claudin-18.2-positive locally advanced/metastatic gastric/gastroesophageal cancer in two parts: Safety (Arms A and B, n = 3 each) and Expansion (n = 12). Patients received intravenous zolbetuximab 800 mg/m2 on cycle 1, day 1 followed by 600 mg/m2 every 3 weeks (Q3W; Safety Part Arm A and Expansion) or 1000 mg/m2 Q3W (Safety Part Arm B). For the Safety Part, the primary endpoint was safety (i.e., dose-limiting toxicities [DLTs]) and a secondary endpoint was objective response rate (ORR) by investigator. For the Expansion Part, the primary endpoint was ORR by investigator and secondary endpoints included ORR by central review and safety. Additional secondary endpoints for both the Safety and Expansion Parts were disease control rate (DCR), overall survival (OS), progression-free survival (PFS), duration of response, pharmacokinetics, and immunogenicity. In 18 patients, no DLTs (Safety Part) or drug-related treatment-emergent adverse events (TEAEs) grade ≥3 were observed. Most TEAEs were gastrointestinal. In 17 patients with measurable lesions, best overall response was stable disease (64.7%) or progressive disease (35.3%). The DCR was 64.7% (95% confidence interval 38.3-85.8). In Arm A and Expansion combined (n = 15), median OS was 4.4 months (2.6-11.4) and median PFS was 2.6 months (0.9-2.8). In Arm B (n = 3), median OS was 6.4 months (2.9-6.8) and median PFS was 1.7 months (1.2-2.1). Zolbetuximab exhibited no new safety signals with limited single-agent activity in Japanese patients.
Collapse
Affiliation(s)
- Kohei Shitara
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Akihito Kawazoe
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | | | | | | | | | | | - Jeffrey Raizer
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | - Ahsan Arozullah
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| |
Collapse
|
17
|
Nagata Y, Yamamoto S, Kato K. Immune checkpoint inhibitors in esophageal cancer: Clinical development and perspectives. Hum Vaccin Immunother 2022; 18:2143177. [PMID: 36375821 PMCID: PMC9746438 DOI: 10.1080/21645515.2022.2143177] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/11/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022] Open
Abstract
Esophageal cancer is the sixth most common cause of cancer-related mortality worldwide. The standard treatment for unresectable esophageal cancer is systemic chemotherapy. However, the survival benefit is limited, with a median overall survival of less than 10 months. The advent of immune checkpoint inhibitors (ICIs), including programmed cell death-1 antibodies, has revolutionized the treatment paradigm for esophageal cancer. Since demonstrating promising efficacy with manageable safety in several clinical trials, ICIs has finally reached the point where they can be used in various tumor stages in the clinical setting. ICIs are most promising treatments that can be expected to improve the prognosis in patients with esophageal cancer now and in the future. This review outlines the mechanisms, results of clinical trials, and prospects for future studies of ICIs in esophageal cancer. It also discusses clinical questions and challenges in the therapeutic development of ICIs.
Collapse
Affiliation(s)
- Yusuke Nagata
- Department of Gastroenterology, Nagano Municipal Hospital, Nagano, Japan
| | - Shun Yamamoto
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ken Kato
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
18
|
Zhao XY, Liu X, Li WH, Qiu LX, Huang MZ, Wang CC, Chen ZY, Zhang W, Feng WJ, Guo WJ, Zhu X. Randomized phase II study of TX followed by XELOX versus the reverse sequence for chemo-naive patients with metastatic gastric cancer. Front Oncol 2022; 12:911160. [PMID: 36387112 PMCID: PMC9643736 DOI: 10.3389/fonc.2022.911160] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 09/07/2022] [Indexed: 10/19/2024] Open
Abstract
UNLABELLED This research found that the clinical outcomes (PFS, ORR, OS) of the non-platinum-based doublet regimen (docetaxel capecitabine combination) were similar to those of the platinum-based (oxaliplatin capecitabine combination) when used as first line therapy for MGC patients. BACKGROUND Docetaxel, platinum and fluorouracil are the three most important drugs in the treatment of MGC. This study was to compare clinical outcomes of the docetaxel capecitabine combination and the oxaliplatin capecitabine combination as first-line therapy in MGC patients. METHODS In this phase II trial, MGC patients were randomly assigned and treated with either TX (capecitabine 1000 mg/m2/twice daily/1-14 days and docetaxel 60/75 mg/m2 on the 1st day) (because of toxicity, the dose of docetaxel was reduced to 60 mg/m2) or XELOX (capecitabine the same dose with TX and oxaliplatin 130 mg/m2 on the 1st day) as first-line therapy. After progression, patients were crossover to the other group as second-line treatment. RESULTS Total 134 MGC patients were randomized (69 in TX, 65 in XELOX). There was no significant difference between the PFS of the two groups (TX vs XELOX, 4.6 months vs 5.1 months, p=0.359), and the SFS (9.3 months vs 7.5 months, p=0.705), OS (13.1 months vs 9.6 months, p=0.261), and ORR (46.4% vs 46.2%) were also similar. Among patients with ascites, the TX group had significantly longer PFS and OS than the XELOX group. A total of 85 patients (48 in TX, 37 in XELOX) received second-line treatment, with overall survival of second-line chemotherapy (OS2) of 8.0 m and 5.3 m (p=0.046), respectively. Grade 3 to 4 treatment-related adverse events of first line treatment occurred more in TX group than that in XELOX group(60.6% vs 55.4%). CONCLUSION TX regimen is an alternative choice of first-line treatment for MGC patients. We still need to explore the large number of cohort to confirm this results.
Collapse
Affiliation(s)
- Xiao-Yin Zhao
- Department of Gastrointestinal Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xin Liu
- Department of Gastrointestinal Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Hua Li
- Department of Gastrointestinal Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Li-Xin Qiu
- Department of Gastrointestinal Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ming-Zhu Huang
- Department of Gastrointestinal Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen-Chen Wang
- Department of Gastrointestinal Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhi-Yu Chen
- Department of Gastrointestinal Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen Zhang
- Department of Gastrointestinal Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wan-Jing Feng
- Department of Gastrointestinal Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei-Jian Guo
- Department of Gastrointestinal Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaodong Zhu
- Department of Gastrointestinal Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Ning Y, Deng C, Li C, Peng W, Yan C, Ran J, Chen W, Liu Y, Xia J, Ye L, Wei Z, Xiang T. PCDH20 inhibits esophageal squamous cell carcinoma proliferation and migration by suppression of the mitogen-activated protein kinase 9/AKT/β-catenin pathway. Front Oncol 2022; 12:937716. [PMID: 36248995 PMCID: PMC9555239 DOI: 10.3389/fonc.2022.937716] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/02/2022] [Indexed: 11/13/2022] Open
Abstract
Aberrant protocadherins (PCDHs) expression trigger tumor invasion and metastasis. PCDH20 anti-tumor functions in various tumor have been identified. Tumor suppression is due to Wnt/β-catenin pathway antagonism and may be suppressed caused by PCDH20 downregulation through promotor methylation, whereas PCDH20 effects and regulation mechanism in esophageal squamous cell carcinoma (ESCC) remains elusive. We analyzed PCDH20 effects on ESCC and underlying action mechanisms for PCDH20. We test PCDH20 expression in ESCC tissues and cells by semi-quantitative PCR (RT-PCR) and q-PCR (real-time quantitative polymerase chain reaction). MSP (methylation-specific PCR) was carried out to assess the methylation of PCDH20 in ESCC cells and tissues. Anti-tumor effects of PCDH20 in vitro were assessed by clone formation assay, CCK8 assay, Transwell assay, and flow cytometry. Nude mice tumorigenicity was used to assess PCDH20 anti-tumor effect in vivo. Online database, qPCR, and Western blotting were used to identify the downregulation of MAP3K9 by PCDH20, associated with AKT/β-catenin signaling inactivation. We found that PCDH20 expression was dramatically attenuated in esophageal cancer tissues and cells, maybe due to promotor methylation, and ectopic PCDH20 expression suppressed ESCC malignant biological phenotypes. PCDH20 exerted anti-tumor effects by MAP3K9 downregulation, which suppressed AKT/β-catenin signaling in ESCC cells.ConclusionPCDH20 was a tumor suppressor gene, which antagonized AKT/β-catenin signaling pathway in ESCC by decreasing MAP3K9.
Collapse
Affiliation(s)
- Yijiao Ning
- Gastrointestinal Surgical Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chaoqun Deng
- Gastrointestinal Surgical Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunhong Li
- Department of Oncology, Suining Central Hospital, Suining, China
- *Correspondence: Tingxiu Xiang, ; Chunhong Li,
| | - Weiyan Peng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chun Yan
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Ran
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Weihong Chen
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yujia Liu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiuyi Xia
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Ye
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhengqiang Wei
- Gastrointestinal Surgical Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tingxiu Xiang
- Gastrointestinal Surgical Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Tingxiu Xiang, ; Chunhong Li,
| |
Collapse
|
20
|
Parmar K, Subramanyam S, Attwood K, Appiah D, Fountzilas C, Mukherjee S. Anti PD-1/Anti PDL-1 Inhibitors in Advanced Gastroesophageal Cancers: A Systematic Review and Meta-Analysis of Phase 2/3 Randomized Controlled Trials. Pharmaceutics 2022; 14:1953. [PMID: 36145703 PMCID: PMC9501109 DOI: 10.3390/pharmaceutics14091953] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/17/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Importance: Immune checkpoint inhibitors (ICI) have revolutionized the treatment for gastroesophageal cancers (GEC). It is important to investigate the factors that influence the response to anti-PD-1/PD-L1 ICIs. Objective: To assess the benefits of PD-1/PD-L1 ICIs in advanced GEC and perform subgroup analysis to identify patient populations who would benefit from ICI. Data sources: PubMed, Embase, Scopus, and the Cochrane Library databases were systematically searched from database inception to September 2021 for all relevant articles. We also reviewed abstracts and presentations from all major conference proceedings including relevant meetings of the American Society of Clinical Oncology (ASCO), and the European Society for Medical Oncology (ESMO) during the last four years (2018 to 2021) and reviewed citation lists. Study selection, data extraction, and synthesis: Full articles and presentations were further assessed if the information suggested that the study was a phase 2/3 randomized controlled trial (RCT) comparing PD-1/PD-L1 inhibitor either alone, or in combination with standard therapy vs. standard therapy in advanced GEC. The full text of the resulting studies/presentations and extracted data were reviewed independently according to PRISMA guidelines. Main outcomes and measures: The main outcomes were OS, PFS, and treatment-related adverse events (TRAEs). Results: A total of 168 studies were assessed for eligibility, and 17 RCTs with 12,312 patients met the inclusion criteria. There was an OS benefit in the overall population with ICIs (HR 0.78; 95% CI 0.73−0.83 p < 0.001). Immunotherapy showed better OS benefit in males (HR 0.77 95% CI 0.72−0.83; p < 0.001) than females (HR 0.89; 95% CI 0.80−0.99 p < 0.03), esophageal primary tumors (HR 0.70 95% CI 0.64−0.76 p < 0.001) vs. gastric cancer (HR 0.84 95% CI 0.74−0.94 p 0.002) or GEJ cancer (HR 0.84 95% CI 0.72−0.98 p 0.024) and in squamous cell carcinoma (HR 0.71 95% CI 0.66−0.77 p < 0.001) vs. adenocarcinoma (HR 0.85 95% CI 0.78−0.93 p < 0.001). PD-L1 positive patients seemed to benefit more (HR 0.74 95% CI 0.67−0.82 p < 0.001) compared to PD-L1 negative patients (HR 0.86 95% CI 0.74−1.00 p < 0.043), and Asians showed OS benefit (HR 0.76 95% CI 0.67−0.87 p < 0.001) compared to their White counterparts (HR 0.92 95% CI 0.74−1.14; p 0.424). Conclusions and relevance: ICIs improve survival in advanced GEC without significantly increasing the side effects. However, certain subgroups of patients such as males, Asians, and those with esophageal primary, PD-L1 positive tumors and squamous cell carcinoma benefit more from such treatments. Further translational research is needed to understand the mechanistic links and develop new biomarkers.
Collapse
Affiliation(s)
- Kanak Parmar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Sai Subramanyam
- Department of Internal Medicine, Appalachian Regional Healthcare, Harlan, KY 40831, USA
| | - Kristopher Attwood
- Department of Epidemiology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Duke Appiah
- Department of Public Health, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Christos Fountzilas
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Sarbajit Mukherjee
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
21
|
Sridharan S, Day F, Loh J, Lynam J, Smart J, Holt B, Mandaliya H, Bonaventura A, Kumar M, Martin J. Phase I trial of hypofractionated chemoradiotherapy in the palliative management of esophageal and gastro-esophageal cancer. Radiat Oncol 2022; 17:158. [PMID: 36104707 PMCID: PMC9472395 DOI: 10.1186/s13014-022-02127-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 09/05/2022] [Indexed: 12/24/2022] Open
Abstract
Background Many patients with incurable esophageal cancer (ECa) present with dysphagia as their predominant symptom. Currently there is no consensus on how best to initially manage this scenario with multiple therapeutic options available. We aimed to assess the safety and efficacy of using hypofractionated radiotherapy given over a progressively shorter timeframe with concurrent carboplatin and paclitaxel in the management of patients with ECa and dysphagia. Methods In this phase I trial we enrolled patients with histologically proven squamous cell carcinoma or adenocarcinoma of the esophagus or the gastro-esophageal junction with symptomatic dysphagia from local disease and not for curative treatment. Patients needed to be 18 years or older, have an ECOG performance status of 0–2 and be suitable to receive carboplatin and paclitaxel chemotherapy. Patients were placed in four progressively shorter radiation schedules culminating in 30 Gy in 10 fractions in a step wise manner, all with concurrent carboplatin AUC 2 and paclitaxel 50 mg/m2 chemotherapy delivered weekly with the radiation therapy. The primary endpoint was the development of the dose limiting toxicities (DLTs) esophageal perforation or febrile neutropenia. Secondary endpoints were relief of dysphagia, time to improvement of dysphagia, dysphagia progression free survival and overall survival. Results Eighteen patients were enrolled in the study between October 2014 and March 2019. There were no DLTs experienced during the trial. The most common grade 3 + acute toxicity experienced by patients were nausea and vomiting (both in 4/18 patients). The most common radiation specific acute toxicity experienced was esophagitis with 67% of patients experiencing grade 1–2 symptoms. All patients experienced improvement in dysphagia. The median time to dysphagia improvement was 3 weeks from the start of chemoradiotherapy (CTRT) (range 2–10 weeks). The median dysphagia free survival was 5.8 months with a median overall survival of 8.9 months. Conclusion Hypofractionated palliative CTRT with 30 Gy/10# of radiation therapy with concurrent weekly carboplatin and paclitaxel chemotherapy is well tolerated and provides a good response in improvement of dysphagia. Further studies need to be undertaken which provide both symptomatic improvement in the primary tumor but also control of the metastatic burden in these patients. Clinical Trial Registration: This trial was prospectively registered with www.anzctr.org.au Identifier: ACTRN12614000821695.
Collapse
|
22
|
Explore and Analyze the Composition and Characteristics of Intestinal Microbiota between Gastric Cancer Patients and Healthy People. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:5834293. [PMID: 36118097 PMCID: PMC9477631 DOI: 10.1155/2022/5834293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/30/2022] [Accepted: 07/20/2022] [Indexed: 11/25/2022]
Abstract
Gastric cancer is one of the most common malignant tumors in the world. As the intestine is downstream of the digestive tract, the occurrence of gastric cancer may have a certain significant impact on it. Therefore, it is particularly important to find out the intestinal bacteria closely related to gastric cancer, to identify the specific flora related to gastric cancer, and to maintain the stability of the core structure of intestinal microecology in patients with gastric cancer. Based on this, the fecal samples of gastric cancer patients and healthy people were collected, and the diversity and composition of intestinal flora in patients with gastric cancer were analyzed by 16S rRNA sequencing technology. We found that there was no significant difference in the diversity and abundance of intestinal flora between gastric cancer patients and healthy people. The relative abundance of Faecalibacterium, Bifidobacterium, and Subdoligranulum in the intestinal tract of patients with gastric cancer was significantly lower than that in healthy people, while the relative abundance of Enterococcus, Streptococcus, and Bacteroides was increased. This study found that there were six kinds of intestinal microflora closely related to the occurrence of gastric cancer, which provided a theoretical basis for further exploring the pathogenesis of gastric cancer.
Collapse
|
23
|
Xie Q, Luo Y, Peng X. Cost-effectiveness analysis of pembrolizumab for patients with advanced esophageal cancer at PD-L1 combined positive score ≥10. J Comp Eff Res 2022; 11:1095-1103. [PMID: 36043412 DOI: 10.2217/cer-2021-0165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Due to the high price of pembrolizumab, it is still unknown whether the use of pembrolizumab for advanced esophageal cancer would be a cost-effective option for patients whose PD-L1 combined positive score is ≥10. Methods: A Markov simulation model was performed based on clinical trial KEYNOTE-181. Incremental cost-effectiveness ratios were calculated to compare the two treatments. Results: The total costs were US$193,575.60 and $8789.24 for pembrolizumab and chemotherapy treatment, respectively. The pembrolizumab group produced 0.93 quality-adjusted life years (QALYs), while the chemotherapy group produced 0.58 QALYs. Thus, patients in the pembrolizumab group spent an additional US$184,786.36 and produced 0.35 QALYs more than the chemotherapy group, which resulted in an incremental cost-effectiveness ratio of US$527,961.03 per QALY. Conclusion: For patients with advanced esophageal cancer whose PD-L1 combined positive score is ≥10, pembrolizumab is not a cost-effective second-line therapy versus chemotherapy from the US payer perspective.
Collapse
Affiliation(s)
- Qian Xie
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, 610041, ChinaChina
| | - Yaxin Luo
- West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
24
|
Fu S, Li L, Li X, Wu Q, Wang X, Huang Y, Hu H, Cao D. Case report: Long-term partial response of apatinib plus paclitaxel as second-line therapy in a patient with metastatic gastric cancer. Front Pharmacol 2022; 13:888106. [PMID: 36034835 PMCID: PMC9400896 DOI: 10.3389/fphar.2022.888106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/28/2022] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer is the second most prevalent cancer and the second leading cause of cancer-related death in China. The prognosis of metastatic gastric cancer is poor with a median overall survival of 8–10 months. Apatinib, an oral small-molecule, selective vascular endothelial growth factor receptor-2 tyrosine kinase inhibitor, is approved as third-line or subsequent therapy for gastric cancer in China. Several recent small-scale studies and case reports showed that it may be great help in improvement of prognosis as second-line treatment in patients with advanced or metastatic gastric cancer. Here, we present a case of advanced gastric adenocarcinoma with multiple hepatic metastases who was treated with apatinib plus paclitaxel as second-line therapy, realized a long progression-free survival of 37 months. Until 29 January 2022, the disease remains an efficacy of partial response. We believe that the good outcome of this case is not an accident, because of the typically hyper-vascular of his liver metastases, the treatment toxicities of hypertension and proteinuria, all may be potential predictive biomarkers for anti-angiogenic treatments.
Collapse
Affiliation(s)
- Shengya Fu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, SC, China
- Second Department of Oncology, Sichuan Friendship Hospital, Chengdu, SC, China
| | - Linjuan Li
- Department of Abdominal Oncology, West China Hospital, Sichuan University/ West China School of Nursing, Sichuan University, Chengdu, SC, China
| | - Xiaofen Li
- Department of Abdominal Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, SC, China
| | - Qiang Wu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, SC, China
| | - Xiaohui Wang
- Second Department of Oncology, Sichuan Friendship Hospital, Chengdu, SC, China
| | - Yan Huang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, SC, China
| | - Haoyue Hu
- Department of Medical Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Medicine School of University of Electronic Science and Technology, Chengdu, SC, China
| | - Dan Cao
- Department of Abdominal Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, SC, China
- *Correspondence: Dan Cao,
| |
Collapse
|
25
|
Shah MA, Shitara K, Lordick F, Bang YJ, Tebbutt NC, Metges JP, Muro K, Lee KW, Shen L, Tjulandin S, Hays JL, Starling N, Xu RH, Sturtz K, Fontaine M, Oh C, Brooks EM, Xu B, Li W, Li CJ, Borodyansky L, Van Cutsem E. Randomized, Double-Blind, Placebo-Controlled Phase III Study of Paclitaxel ± Napabucasin in Pretreated Advanced Gastric or Gastroesophageal Junction Adenocarcinoma. Clin Cancer Res 2022; 28:OF1-OF9. [PMID: 35833783 PMCID: PMC9433958 DOI: 10.1158/1078-0432.ccr-21-4021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/22/2022] [Accepted: 05/24/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE To compare napabucasin (generator of reactive oxygen species) plus paclitaxel with paclitaxel only in patients with second-line advanced gastric or gastroesophageal junction (GEJ) adenocarcinoma. EXPERIMENTAL DESIGN In the double-blind, phase III BRIGHTER study (NCT02178956), patients were randomized (1:1) to napabucasin (480 mg orally twice daily) plus paclitaxel (80 mg/m2 i.v. weekly for 3 of 4 weeks) or placebo plus paclitaxel. The primary endpoint was overall survival (OS). Secondary endpoints included progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), and safety. RESULTS Overall, 714 patients were randomized (napabucasin plus paclitaxel, n = 357; placebo plus paclitaxel, n = 357). 72.1% were male, 74.6% had gastric adenocarcinoma, and 46.2% had peritoneal metastases. The study was unblinded following an interim analysis at 380 deaths. The final efficacy analysis was performed on 565 deaths (median follow-up, 6.8 months). No significant differences were observed between napabucasin plus paclitaxel and placebo plus paclitaxel for OS (6.93 vs. 7.36 months), PFS (3.55 vs. 3.68 months), ORR (16% vs. 18%), or DCR (55% vs. 58%). Grade ≥3 adverse events occurred in 69.5% and 59.7% of patients administered napabucasin plus paclitaxel and placebo plus paclitaxel, respectively, with grade ≥3 diarrhea reported in 16.2% and 1.4%, respectively. CONCLUSIONS Adding napabucasin to paclitaxel did not improve survival in patients with pretreated advanced gastric or GEJ adenocarcinoma. Consistent with previous reports, the safety profile of napabucasin was driven by manageable gastrointestinal events; grade ≥3 diarrhea occurred at a higher frequency with napabucasin plus paclitaxel versus placebo plus paclitaxel.
Collapse
Affiliation(s)
- Manish A. Shah
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine/New York-Presbyterian Hospital, New York, New York
| | - Kohei Shitara
- Department of Immunology, Nagoya University Graduate School of Medicine and Department of Gastrointestinal Oncology, National Cancer Center Hospital East and the Department of Immunology, Nagoya University Graduate School of Medicine, Tokyo, Japan
| | - Florian Lordick
- Department of Oncology, University Cancer Center Leipzig, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany
| | - Yung-Jue Bang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Niall C. Tebbutt
- Department of Medical Oncology, Austin Health, Heidelberg, Victoria, Australia
| | - Jean-Phillippe Metges
- Department of Medical Oncology, CHRU de Brest-Hopital Morvan, Arpego Network Brest, Bretagne, France
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Sergei Tjulandin
- Department of Clinical Pharmacology and Chemotherapy, N.N. Blokhin Russian Cancer Research Centre, Moscow, Russia
| | - John L. Hays
- Department of Internal Medicine, The Ohio State University, James Cancer Hospital, Columbus, Ohio
| | - Naureen Starling
- Gastrointestinal Unit, The Royal Marsden, London & Surrey, United Kingdom
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Keren Sturtz
- Western States Cancer Research NCORP, Denver, Colorado
| | | | - Cindy Oh
- Sumitomo Pharma Oncology, Inc., Cambridge, Massachusetts
| | | | - Bo Xu
- Sumitomo Pharma Oncology, Inc., Cambridge, Massachusetts
| | - Wei Li
- Sumitomo Pharma Oncology, Inc., Cambridge, Massachusetts
| | - Chiang J. Li
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- 1Globe Health Institute, Boston, Massachusetts
| | | | - Eric Van Cutsem
- Department of Gastroenterology/Digestive Oncology, University Hospitals Gasthuisberg Leuven and KU Leuven, Leuven, Belgium
| |
Collapse
|
26
|
Koutroumpakis E, Agrawal N, Palaskas NL, Abe JI, Iliescu C, Yusuf SW, Deswal A. Myocardial Dysfunction in Patients with Cancer. Heart Fail Clin 2022; 18:361-374. [PMID: 35718412 DOI: 10.1016/j.hfc.2022.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Myocardial dysfunction in patients with cancer is a major cause of morbidity and mortality. Cancer therapy-related cardiotoxicities are an important contributor to the development of cardiomyopathy in this patient population. Furthermore, cardiac AL amyloidosis, cardiac malignancies/metastases, accelerated atherosclerosis, stress cardiomyopathy, systemic and pulmonary hypertension are also linked to the development of myocardial dysfunction. Herein, we summarize current knowledge on the mechanisms of myocardial dysfunction in the setting of cancer and cancer-related therapies. Additionally, we briefly outline key recommendations on the surveillance and management of cancer therapy-related myocardial dysfunction based on the consensus of experts in the field of cardio-oncology.
Collapse
Affiliation(s)
- Efstratios Koutroumpakis
- Department of Cardiology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1451, Houston, TX 77030, USA
| | - Nikhil Agrawal
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA
| | - Nicolas L Palaskas
- Department of Cardiology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1451, Houston, TX 77030, USA
| | - Jun-Ichi Abe
- Department of Cardiology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1451, Houston, TX 77030, USA
| | - Cezar Iliescu
- Department of Cardiology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1451, Houston, TX 77030, USA
| | - Syed Wamique Yusuf
- Department of Cardiology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1451, Houston, TX 77030, USA
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1451, Houston, TX 77030, USA.
| |
Collapse
|
27
|
Neutrophil-lymphocyte ratio combined with plasma fibrinogen is a useful predictor for the diagnosis of liver metastases from gastric cancer. Asian J Surg 2022; 45:2378-2379. [DOI: 10.1016/j.asjsur.2022.05.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/13/2022] [Indexed: 11/21/2022] Open
|
28
|
Therapeutic Advances and Challenges in the Management of HER2-Positive Gastroesophageal Cancers. Diseases 2022; 10:diseases10020023. [PMID: 35645244 PMCID: PMC9149813 DOI: 10.3390/diseases10020023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/10/2022] [Accepted: 04/16/2022] [Indexed: 11/17/2022] Open
Abstract
Gastroesophageal cancer is one of the most common cancers in the world, with a high rate of mortality. While there has been significant progress over the past decade, particularly with the addition of anti-HER2 therapies to platinum-based chemotherapy agents in the advanced setting, the prognosis remains poor and the treatment options for this disease entity remain limited. In this review, we discuss the current therapeutic landscape for HER2-positive gastroesphageal cancer and the seminal clinical trials that have shaped our approach to this disease entity. In addition, we highlight some of the challenges to the understanding and management of this disease, specifically discussing the breadth of molecular diversity and intratumoral heterogeneity of HER2 expression that impact the clinical efficacy and prognosis. Furthermore, we discuss the potential role of next-generation sequencing (NGS) and circulating-tumor DNA (ctDNA) as complementary tools to immunohistochemistry (IHC) and fluorescent in-situ hybridization (FISH) to guiding clinical decision making. Finally, we highlight promising clinical trials of new treatment regimens that will likely reshape the therapeutic approach to this disease entity.
Collapse
|
29
|
USP49 mediates tumor progression and poor prognosis through a YAP1-dependent feedback loop in gastric cancer. Oncogene 2022; 41:2555-2570. [PMID: 35318441 DOI: 10.1038/s41388-022-02267-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 02/12/2022] [Accepted: 03/01/2022] [Indexed: 11/08/2022]
Abstract
The importance of the Hippo-Yes-associated protein 1 (YAP1) pathway in gastric carcinogenesis and metastasis has attracted considerable research attention; however, the regulatory network of YAP1 in gastric cancer (GC) is not completely understood. In this study, ubiquitin-specific peptidase 49 (USP49) was identified as a novel deubiquitinase of YAP1, knockdown of USP49 inhibited the proliferation, metastasis, chemoresistance, and peritoneal metastasis of GC cells. Overexpression of USP49 showed opposing biological effects. Moreover, USP49 was transcriptionally activated by the YAP1/TEAD4 complex, which formed a positive feedback loop with YAP1 to promote the malignant progression of GC cells. Finally, we collected tissue samples and clinical follow-up information from 482 GC patients. The results showed that USP49 expression was high in GC cells and positively correlated with the expression of YAP1 and its target genes, connective tissue growth factor (CTGF) and cysteine-rich angiogenic inducer 61 (CYR61). Survival and Cox regression analysis showed that high USP49 expression was associated with poor prognosis and was an independent prognostic factor. Moreover, patients with high USP49 and YAP1 expression had extremely short overall survival. The findings of this study reveal that the aberrant activation of the USP49/YAP1 positive feedback loop plays a critical role in the malignant progression of GC, thus providing potential novel prognostic factors and therapeutic targets for GC.
Collapse
|
30
|
Tan D, Zhang Y. Silencing of Nudix type 5 represses proliferation and invasion and enhances chemosensitivity of gastric carcinoma cells by affecting the AKT/GSK-3β/β-catenin pathway. Toxicol Appl Pharmacol 2022; 441:115968. [PMID: 35247377 DOI: 10.1016/j.taap.2022.115968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/22/2022] [Accepted: 02/27/2022] [Indexed: 01/01/2023]
Abstract
Nudix type 5 (NUDT5) has been recently identified as a new cancer-associated protein that is involved in numerous cancers. To date, the relationship between NUDT5 and gastric carcinoma has not been addressed. In the current research, we focused on exploring the potential relevance of NUDT5 in gastric carcinoma. The initial analysis of NUDT5 expression in gastric carcinoma by TCGA data revealed a clear increase in NUDT5 expression in tumor versus normal tissue. The increased expression of NUDT5 was also validated in the clinical specimens of gastric carcinoma by immunoblotting detection. Moreover, high NUDT5 levels predicted a poorer overall survival in gastric carcinoma patients. A series of cellular functional assays demonstrated that gastric carcinoma cells with silenced NUDT5 exhibited decreased proliferative and invasive ability, increased cell cycle arrest at the G0/G1 phase, and enhanced chemosensitivity. In-depth research showed that the silencing of NUDT5 led to a reduction in the activation of AKT and β-catenin. The reactivation of AKT blocked the repressive effect of NUDT5 silencing on β-catenin activation. The forced expression of β-catenin also reversed NUDT5-silencing-mediated anticancer effects. A Xenograft tumor assay confirmed the anticancer role of NUDT5 in gastric carcinoma in vivo. In short, these findings reveal elevated NUDT5 levels in gastric carcinoma and demonstrate that the inhibition of NUDT5 displays promising anticancer effects by affecting the AKT/β-catenin pathway. Thus, our work unveils a vital role of NUDT5 in gastric carcinoma and indicates it as a viable candidate target for anticancer drug discovery.
Collapse
Affiliation(s)
- Dong Tan
- Department of General Surgery, No. 215 Hospital of Shaanxi Nuclear Industry, No. 52 Weiyang West Road, Xianyang, Shaanxi 712000, PR China
| | - Yafei Zhang
- Department of Endoscopy, No. 215 Hospital of Shaanxi Nuclear Industry, No. 52 Weiyang West Road, Xianyang, Shaanxi 712000, China.
| |
Collapse
|
31
|
Li D, Li Q. MicroRNA-200b-3p restrains gastric cancer cell proliferation, migration, and invasion via C-X-C motif chemokine ligand 12/CXC chemokine receptor 7 axis. Bioengineered 2022; 13:6509-6520. [PMID: 35226830 PMCID: PMC8974025 DOI: 10.1080/21655979.2022.2034585] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
This study was conducted to investigate the impact of microRNA (miR)-200b-3p on viability, migration, and invasion of gastric cancer (GC) cells and its mechanism. Quantitative real-time PCR (qRT-PCR) was conducted to measure miR-200b-3p expression in GC tissues and cells; besides, the relationship between miR-200b-3p expression and overall survival time (OS) was analyzed with OncomiR database; cell counting kit-8 (CCK-8), colony formation assay, flow cytometry, scratch healing assay, and Transwell assay were performed to detect the proliferation, cell cycle progression, migration, and invasion of GC cells; a lung metastasis model in nude mice was used to examine the effect of miR-200b-3p on the metastasis of GC cells in vivo; the interplay between miR-200b-3p and C-X-C motif chemokine ligand 12 (CXCL12) mRNA 3’ UTR was predicted by bioinformatics and verified with a dual-luciferase reporter gene assay; besides, the expression of CXCL12 and CXC chemokine receptor 7 (CXCR7) was probed by Western blot. It was found that miR-200b-3p expression was down-regulated in GC tissues, which was remarkably associated with the lymph node metastasis and decrease of differentiation of GC; transfection with miR-200b-3p mimics restrained the growth, migration, and invasion of GC cells in vitro, induced cell cycle arrest, and inhibited CXCL12 and CXCR7 expression levels; transfection of miR-200b-3p inhibitors worked oppositely in vitro and promoted lung metastasis in vivo. CXCL12 was confirmed as the downstream target of miR-200b-3p and was negatively modulated by miR-200b-3p. In conclusion, miR-200b-3p inhibited GC progression via regulating CXCL12/CXCR7 axis.
Collapse
Affiliation(s)
- Dinuo Li
- Department of General Gastropathy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Qiang Li
- Department of Gastrosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
32
|
Mei X, Li H, Zhou X, Cheng M, Cui K. The Emerging Role of Tissue-Resident Memory CD8 + T Lymphocytes in Human Digestive Tract Cancers. Front Oncol 2022; 11:819505. [PMID: 35096624 PMCID: PMC8795735 DOI: 10.3389/fonc.2021.819505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 12/23/2021] [Indexed: 12/15/2022] Open
Abstract
Malignant digestive tract tumors are a great threat to human public health. In addition to surgery, immunotherapy brings hope for the treatment of these tumors. Tissue-resident memory CD8+ T (Trm) cells are a focus of tumor immunology research and treatment due to their powerful cytotoxic effects, ability to directly kill epithelial-derived tumor cells, and overall impact on maintaining mucosal homeostasis and antitumor function in the digestive tract. They are a group of noncirculating immune cells expressing adhesion and migration molecules such as CD69, CD103, and CD49a that primarily reside on the barrier epithelium of nonlymphoid organs and respond rapidly to both viral and bacterial infection and tumorigenesis. This review highlights new research exploring the role of CD8+ Trm cells in a variety of digestive tract malignant tumors, including esophageal cancer, gastric cancer, colorectal cancer, and hepatocellular carcinoma. A summary of CD8+ Trm cell phenotypes and characteristics, tissue distribution, and antitumor functions in different tumor environments is provided, illustrating how these cells may be used in immunotherapies against digestive tract tumors.
Collapse
Affiliation(s)
- Xinyu Mei
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huan Li
- Department of Thoracic Surgery, Anhui Provincial Hospital Affiliated With Anhui Medical University, Hefei, China
| | - Xinpeng Zhou
- Department of Thoracic Surgery, Anhui Provincial Hospital, Wannan Medical College, Hefei, China
| | - Min Cheng
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China.,Cancer Immunotherapy Center, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kele Cui
- Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China.,Cancer Immunotherapy Center, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Department of Clinical Laboratory, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
33
|
NF- κB-Related Metabolic Gene Signature Predicts the Prognosis and Immunotherapy Response in Gastric Cancer. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5092505. [PMID: 35036435 PMCID: PMC8753254 DOI: 10.1155/2022/5092505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/01/2021] [Indexed: 11/23/2022]
Abstract
Background Sufficient evidence indicated the crucial role of NF-κB family played in gastric cancer (GC). The novel discovery that NF-κB could regulate cancer metabolism and immune evasion greatly increased its attraction in cancer research. However, the correlation among NF-κB, metabolism, and cancer immunity in GC still requires further improvement. Methods TCGA, hTFtarget, and MSigDB databases were employed to identify NF-κB-related metabolic genes (NFMGs). Based on NFMGs, we used consensus clustering to divide GC patients into two subtypes. GSVA was employed to analyze the enriched pathway. ESTIMATE, CIBERSORT, ssGSEA, and MCPcounter algorithms were applied to evaluate immune infiltration in GC. The tumor immune dysfunction and exclusion (TIDE) algorithm was used to predict patients' response to immunotherapy. We also established a NFMG-related risk score by using the LASSO regression model and assessed its efficacy in TCGA and GSE62254 datasets. Results We used 27 NFMGs to conduct an unsupervised clustering on GC samples and classified them into two clusters. Cluster 1 was characterized by high active metabolism, tumor mutant burden, and microsatellite instability, while cluster 2 was featured with high immune infiltration. Compared to cluster 2, cluster 1 had a better prognosis and higher response to immunotherapy. In addition, we constructed a 12-NFMG (ADCY3, AHCY, CHDH, GUCY1A2, ITPA, MTHFD2, NRP1, POLA1, POLR1A, POLR3A, POLR3K, and SRM) risk score. Followed analysis indicated that this risk score acted as an effectively prognostic factor in GC. Conclusion Our data suggested that GC subtypes classified by NFMGs may effectively guide prognosis and immunotherapy. Further study of these NFMGs will deepen our understanding of NF-κB-mediated cancer metabolism and immunity.
Collapse
|
34
|
Wu Q, Liu F, Ge M, Laster KV, Wei L, Du R, Jiang M, Zhang J, Zhi Y, Jin G, Zhao S, Kim DJ, Dong Z, Liu K. BRD4 drives esophageal squamous cell carcinoma growth by promoting RCC2 expression. Oncogene 2022; 41:347-360. [PMID: 34750516 DOI: 10.1038/s41388-021-02099-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 10/18/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022]
Abstract
The low survival rate of esophageal squamous cell carcinoma patients is primarily attributed to technical limitations and a lack of insight regarding the molecular mechanisms contributing to its progression. Alterations in epigenetic modulators are critical to cancer development and prognosis. BRD4, a chromatin reader protein, plays an essential role in regulating oncogene expression. Here, we investigated the contributing role of BRD4 and its related mechanisms in the context of ESCC tumor progression. Our observations showed that BRD4 transcript and protein expression levels are significantly increased in ESCC patient tissues. Genetic or pharmacological inhibition of BRD4 suppressed ESCC cell proliferation in vitro and in vivo. Proteomic and transcriptomic analyses were subsequently used to deduce the potential targets of BRD4. Mechanistic studies showed that RCC2 is a downstream target of BRD4. Inhibition of either BRD4 or RCC2 resulted in decreased ESCC cell proliferation. The BRD4-TP73 interaction facilitated the binding of BRD4 complex to the promoter region of RCC2, and subsequently modulated RCC2 transcription. Furthermore, targeting BRD4 with inhibitors significantly decreased tumor volume in ESCC PDX models, indicating that BRD4 expression may contribute to tumor progression. Collectively, these findings suggest that BRD4 inhibition could be a promising strategy to treat ESCC by downregulating RCC2.
Collapse
Affiliation(s)
- Qiong Wu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Fangfang Liu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Mengmeng Ge
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | | | - Lixiao Wei
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Ruijuan Du
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Ming Jiang
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Jing Zhang
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Yafei Zhi
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Guoguo Jin
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China.,The Henan Luoyang Orthopedic Hospital, Zhengzhou, 450000, Henan, China
| | - Simin Zhao
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China.,Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Dong Joon Kim
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China.
| | - Zigang Dong
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China. .,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450000, Henan, China. .,Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Kangdong Liu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China. .,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450000, Henan, China. .,Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China. .,Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
35
|
Wang Q, Zhong S, Wu H, Wu Q. In vitro anti-cancer effect of marmesin by suppression of PI3K/Akt pathway in esophagus cancer cells. Esophagus 2022; 19:163-174. [PMID: 34398363 DOI: 10.1007/s10388-021-00872-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 08/10/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Marmesin, an important coumarin isolated from Broussonetia kazinoki, has been proposed to possess many pharmacological activities including anti-tumor activity. However, the anti-cancer effect of marmesin on esophageal cancer (EC) has not been characterized. The study aimed to explore the anti-cancer role of marmesin using EC cell lines in vitro. METHODS AND RESULTS Cell proliferation was evaluated by CCK-8 and Edu cell proliferation assays and apoptosis was detected by TUNEL assay. Western blot analysis was used to determine the expression of Ki67, proliferating cell nuclear antigen (PCNA), Bcl-2, Bax, phosphatidylinositol 3-kinase (PI3K), phosphoryrated-PI3K (p-PI3K), protein kinase B (Akt), and phosphoryrated-Akt (p-Akt). The mechanism of action of marmesin was analyzed using network pharmacology approach. Marmesin exhibited anti-proliferative effect against EC cells, which was further confirmed by the reduced expression of Ki67 and PCNA. Marmesin exerted pro-apoptotic activity on EC cells by downregulating Bcl-2 and upregulating Bax. According to the results from network pharmacology approach, we speculated that PI3K/Akt pathway may participate in the effect of marmesin on EC cells. Additionally, the PI3K/Akt pathway was suppressed by marmesin in EC cells. Moreover, forced expression of Akt reversed the inhibition of cell proliferation and induction of apoptosis induced by marmesin in EC cells. CONCLUSIONS Marmesin exerted anti-cancer activity in EC cells by inhibiting the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Qi Wang
- Department of Thoracic Surgery, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huan'an, 223000, China
| | - Sheng Zhong
- Department of Thoracic Surgery, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huan'an, 223000, China
| | - Hua Wu
- Department of Thoracic Surgery, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huan'an, 223000, China
| | - Qingquan Wu
- Department of Thoracic Surgery, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huan'an, 223000, China.
| |
Collapse
|
36
|
Lee J, Choi SH, Baek JH, Baek DW, Kim JG, Kang BW. Clinical Impact of Prognostic Nutrition Index for Advanced Gastric Cancer Patients with Peritoneal Metastases Treated Nivolumab Monotherapy. Chonnam Med J 2022; 58:24-28. [PMID: 35169556 PMCID: PMC8813651 DOI: 10.4068/cmj.2022.58.1.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/12/2021] [Accepted: 11/23/2021] [Indexed: 11/06/2022] Open
Abstract
Although nivolumab shows survival benefits for patients with advanced gastric cancer (AGC), predictive biomarkers for nivolumab treatment in AGC remain unclear, especially in the case of peritoneal metastases. This study investigated the clinical significance of the prognostic nutrition index (PNI), reflecting the host nutritional status and immunity, in AGC patients undergoing nivolumab monotherapy. This study retrospectively analyzed 53 AGC patients who received nivolumab between October 2017 and February 2021. Among them, 35 patients with peritoneal metastases were reviewed to investigate the relationship between the PNI and oncological outcomes. The PNI was calculated as 10×serum albumin level (g/dl)+0.005×total lymphocyte count (per mm3) at the first administration of nivolumab. With a median follow-up duration of 2.0 (0.3-13.5) months, the median overall survival (OS) was 2.0 months. The overall response and disease-control rates were 0.0% and 20.0%, respectively. Among the 35 patients, 13 patients were identified as a high-PNI group. In the univariate analysis, the high-PNI group showed a significantly longer PFS and OS than the low-PNI group. In the multivariate analysis, the high-PNI was independently associated with a longer PFS (p=0.021) and OS (p=0.022). The PNI can be useful for predicting PFS and OS in AGC patients with peritoneal metastases. However, further studies are required to validate these results in AGC and new strategies are needed to improve the outcome for AGC patients with peritoneal metastases.
Collapse
Affiliation(s)
- Jungmin Lee
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Kyungpook National University Cancer Research Institute, Daegu, Korea
| | - Soo Ho Choi
- Department of Internal Medicine, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Jin Ho Baek
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Kyungpook National University Cancer Research Institute, Daegu, Korea
| | - Dong Won Baek
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Kyungpook National University Cancer Research Institute, Daegu, Korea
| | - Jong Gwang Kim
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Kyungpook National University Cancer Research Institute, Daegu, Korea
| | - Byung Woog Kang
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Kyungpook National University Cancer Research Institute, Daegu, Korea
| |
Collapse
|
37
|
Jiang Z, Cai Z, Ma Q, Shen C, Yin Y, Yin X, Liu C, Chang C, Zhao Z, Mu M, Zhang B. Comparative efficacy and safety of anti-HGF/MET pathway agents plus chemotherapy versus chemotherapy alone as first-line treatment in advanced gastric cancer: a protocol for a systematic review and meta-analysis. BMJ Open 2021; 11:e049575. [PMID: 34952869 PMCID: PMC8712986 DOI: 10.1136/bmjopen-2021-049575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Phase I/II clinical trials suggested that the hepatocyte growth factor (HGF)/mesenchymal-epithelial transition (MET) pathway-targeted agents were active in suppression of gastric cancer (GC). Randomised controlled trials (RCTs) were undertaken assessing whether the addition of anti-HGF/MET agent (rilotumumab or onartuzumab) to chemotherapy improves survival outcomes of advanced GC, but conflict conclusions were reached. Therefore, we plan to perform this systematic review and meta-analysis to synthesise evidence concerning efficacy and safety of anti-HGF/MET agents combined with chemotherapy as the first-line treatment to advanced GC. METHODS AND ANALYSIS Systematic searches of the PubMed, Embase and the Cochrane Central Register of Controlled Trials will be performed with no language restriction from inception to 31 January 2022 to identify RCTs exploring the comparative efficacy and safety of anti-HGF/MET agents plus chemotherapy as first-line treatment in advanced GC. The primary outcome will be the time-to-event progression-free survival and overall survival, and the secondary outcomes will be disease control rate, overall adverse events rate and grade 3-5 adverse events rate. Statistical heterogeneity will be assessed by visual inspection of forest plots and measured using the I2 statistics. A fixed-effect model will be used when heterogeneity is low otherwise, a random-effect model will be chosen. Publication bias will be assessed by funnel plots; subgroup analysis and sensitivity analysis will be performed in the right context. For each outcome, we will perform data synthesis using Rev Man V.5.3 software, and compile 'summary of findings' tables using GRADEpro software. ETHICS AND DISSEMINATION There is no requirement for ethics approval because no individual data will be collected in this research. It is anticipated that the dissemination of results will take place at conferences and through publication in a peer-review journal, any adjustments from the protocol will be clearly documented and explained in its final report. PROSPERO REGISTRATION NUMBER CRD42020177404.
Collapse
Affiliation(s)
- Zhiyuan Jiang
- Department of Gastrointestinal Surgery, Sichuan University West China Hospital, Chengdu, China
| | - Zhaolun Cai
- Department of Gastrointestinal Surgery, Sichuan University West China Hospital, Chengdu, China
| | - Qin Ma
- Department of Gastrointestinal Surgery, Sichuan University West China Hospital, Chengdu, China
| | - Chaoyong Shen
- Department of Gastrointestinal Surgery, Sichuan University West China Hospital, Chengdu, China
| | - Yuan Yin
- Department of Gastrointestinal Surgery, Sichuan University West China Hospital, Chengdu, China
| | - Xiaonan Yin
- Department of Gastrointestinal Surgery, Sichuan University West China Hospital, Chengdu, China
| | - Chunyu Liu
- Evidence-Based Pharmacy Center, Sichuan University West China Second University Hospital, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, China
| | - Chen Chang
- Department of Abdominal Oncology, Cancer Center, Sichuan University West China Hospital, Chengdu, China
| | - Zhou Zhao
- Department of Gastrointestinal Surgery, Sichuan University West China Hospital, Chengdu, China
| | - Mingchun Mu
- Department of Gastrointestinal Surgery, Sichuan University West China Hospital, Chengdu, China
| | - Bo Zhang
- Department of Gastrointestinal Surgery, Sichuan University West China Hospital, Chengdu, China
- Sanya People's Hospital/ West China (Sanya) hospital, Sanya, China
| |
Collapse
|
38
|
Xu G, Fan L, Zhao S, OuYang C. Neuronal pentraxin II (NPTX2) hypermethylation promotes cell proliferation but inhibits cell cycle arrest and apoptosis in gastric cancer cells by suppressing the p53 signaling pathway. Bioengineered 2021; 12:1311-1323. [PMID: 33896384 PMCID: PMC8806217 DOI: 10.1080/21655979.2021.1915658] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer is a considerable health burden worldwide. DNA methylation, a major epigenetic phenomenon, is closely related to the pathogenesis of cancer. Neuronal pentraxin II (NPTX2) has been found to be hypermethylated in several cancers such as glioblastoma and pancreatic cancer. However, the roles of NPTX2 in gastric cancer have not been reported. To explore this issue, NPTX2 expression in gastric cancer cells was assessed by western blot and quantitative real-time polymerase chain reaction (qRT-PCR). The methylation analysis of NPTX2 was performed by qRT-PCR as well as methylation-specific PCR (MS-PCR). The effects of NPTX2 on gastric cancer cell proliferation, apoptosis and cell cycle were detected by colony formation, CCK-8 and flow cytometry assays, respectively. The interaction of NPTX2 with the p53 signaling pathway was evaluated by western blot. Our study found the down-regulated expression of NPTX2 in gastric cancer cells compared with human gastric mucosal cells. In addition, the hypermethylation of NPTX2 was observed in gastric cancer cells, which was correlated with the low expression of NPTX2. Moreover, NPTX2 inhibited gastric cancer cell proliferation, inhibited apoptosis and induced cell cycle arrest. Furthermore, NPTX2 enhanced the protein expression of p53, p21 and PTEN to activate the p53 signaling pathway. Therefore, NPTX2 hypermethylation caused the downregulation of NPTX2 expression, which could promote cell proliferation, inhibit apoptosis and cause cell cycle arrest in gastric cancer cells by suppressing the p53 signaling pathway. Therefore, NPTX2 may be crucial for the progression of gastric cancer.
Collapse
Affiliation(s)
- Guofeng Xu
- Department of Gastroenterology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| | - Linfeng Fan
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| | - Shufeng Zhao
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| | - Canhui OuYang
- Department of Gastroenterology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| |
Collapse
|
39
|
Shafabakhsh R, Arianfar F, Vosough M, Mirzaei HR, Mahjoubin-Tehran M, Khanbabaei H, Kowsari H, Shojaie L, Azar MEF, Hamblin MR, Mirzaei H. Autophagy and gastrointestinal cancers: the behind the scenes role of long non-coding RNAs in initiation, progression, and treatment resistance. Cancer Gene Ther 2021; 28:1229-1255. [PMID: 33432087 DOI: 10.1038/s41417-020-00272-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/06/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
Gastrointestinal (GI) cancers comprise a heterogeneous group of complex disorders that affect different organs, including esophagus, stomach, gallbladder, liver, biliary tract, pancreas, small intestine, colon, rectum, and anus. Recently, an explosion in nucleic acid-based technologies has led to the discovery of long non-coding RNAs (lncRNAs) that have been found to possess unique regulatory functions. This class of RNAs is >200 nucleotides in length, and is characterized by their lack of protein coding. LncRNAs exert regulatory effects in GI cancer development by affecting different functions such as the proliferation and metastasis of cancer cells, apoptosis, glycolysis and angiogenesis. Over the past few decades, considerable evidence has revealed the important role of autophagy in both GI cancer progression and suppression. In addition, recent studies have confirmed a significant correlation between lncRNAs and the regulation of autophagy. In this review, we summarize how lncRNAs play a behind the scenes role in the pathogenesis of GI cancers through regulation of autophagy.
Collapse
Affiliation(s)
- Rana Shafabakhsh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Farzaneh Arianfar
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Mahjoubin-Tehran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hashem Khanbabaei
- Medical Physics Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hamed Kowsari
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Layla Shojaie
- Research Center for Liver Diseases, Keck School of Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
40
|
Steiniche T, Rha SY, Chung HC, Georgsen JB, Ladekarl M, Nordsmark M, Jespersen ML, Kim HS, Kim H, Fein C, Tang LH, Wu T, Marton MJ, Peter S, Kelsen DP, Ku G. Prognostic significance of T-cell-inflamed gene expression profile and PD-L1 expression in patients with esophageal cancer. Cancer Med 2021; 10:8365-8376. [PMID: 34693652 PMCID: PMC8633232 DOI: 10.1002/cam4.4333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose The ability of the T‐cell–inflamed gene expression profile (GEP) to predict clinical outcome in esophageal cancer (EC) is unknown. This retrospective observational study assessed the prognostic value of GEP and programmed death ligand 1 (PD‐L1) expression in patients with EC treated in routine clinical practice. Methods Tumor samples of 294 patients from three centers in Denmark, South Korea, and the United States, collected between 2005 and 2017, were included. T‐cell–inflamed GEP score was defined as non‐low or low using a cutoff of −1.54. A combined positive score (CPS) ≥10 was defined as PD‐L1 expression positivity. Associations between overall survival (OS) and GEP status and PD‐L1 expression were explored by Cox proportional hazards models adjusting for age, sex, histology, stage, and performance status. Results Median age was 65 years; 63% of patients had adenocarcinoma (AC) and 37% had squamous cell carcinoma (SCC). Thirty‐six percent of tumors were GEP non‐low, with higher prevalence in AC (46%) than SCC (18%). Twenty‐one percent were PD‐L1–positive: 32% in South Korean samples versus 16% in non‐Asian samples and 26% in SCC versus 18% in AC. GEP scores and PD‐L1 CPS were weakly correlated (Spearman’s R = 0.363). OS was not significantly associated with GEP status (non‐low vs low; adjusted hazard ratio, 0.91 [95% CI, 0.69–1.19]) or PD‐L1 expression status. Conclusion Neither GEP nor PD‐L1 expression was a prognostic marker in Asian and non‐Asian patients with EC.
Collapse
Affiliation(s)
- Torben Steiniche
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Sun Young Rha
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Cheol Chung
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Morten Ladekarl
- Department of Oncology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | | | | | - Hyo Song Kim
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyunki Kim
- Department of Pathology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Carly Fein
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Laura H Tang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ting Wu
- BARDS-Epidemiology, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Matthew J Marton
- Department of Translational Medicine, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Senaka Peter
- BARDS-Epidemiology, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - David P Kelsen
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Geoffrey Ku
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
41
|
Bonnot PE, Lintis A, Mercier F, Benzerdjeb N, Passot G, Pocard M, Meunier B, Bereder JM, Abboud K, Marchal F, Quenet F, Goere D, Msika S, Arvieux C, Pirro N, Wernert R, Rat P, Gagnière J, Lefevre JH, Courvoisier T, Kianmanesh R, Vaudoyer D, Rivoire M, Meeus P, Villeneuve L, Piessen G, Glehen O. Prognosis of poorly cohesive gastric cancer after complete cytoreductive surgery with or without hyperthermic intraperitoneal chemotherapy (CYTO-CHIP study). Br J Surg 2021; 108:1225-1235. [PMID: 34498666 DOI: 10.1093/bjs/znab200] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/07/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND The incidence of gastric poorly cohesive carcinoma (PCC) is increasing. The prognosis for patients with peritoneal metastases remains poor and the role of cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) is controversial. The aim was to clarify the impact of gastric PCC with peritoneal metastases treated by CRS with or without HIPEC. METHODS All patients with peritoneal metastases from gastric cancer treated with CRS with or without HIPEC, in 19 French centres, between 1989 and 2014, were identified from institutional databases. Clinicopathological characteristics and outcomes were compared between PCC and non-PCC subtypes, and the possible benefit of HIPEC was assessed. RESULTS In total, 277 patients were included (188 PCC, 89 non-PCC). HIPEC was performed in 180 of 277 patients (65 per cent), including 124 of 188 with PCC (66 per cent). Median overall survival (OS) was 14.7 (95 per cent c.i. 12.7 to 17.3) months in the PCC group versus 21.2 (14.7 to 36.4) months in the non-PCC group (P < 0.001). In multivariable analyses, PCC (hazard ratio (HR) 1.51, 95 per cent c.i. 1.01 to 2.25; P = 0.044) was associated with poorer OS, as were pN3, Peritoneal Cancer Index (PCI), and resection with a completeness of cytoreduction score of 1, whereas HIPEC was associated with improved OS (HR 0.52; P < 0.001). The benefit of CRS-HIPEC over CRS alone was consistent, irrespective of histology, with a median OS of 16.7 versus 11.3 months (HR 0.60, 0.39 to 0.92; P = 0.018) in the PCC group, and 34.5 versus 14.3 months (HR 0.43, 0.25 to 0.75; P = 0.003) in the non-PCC group. Non-PCC and HIPEC were independently associated with improved recurrence-free survival and fewer peritoneal recurrences. In patients who underwent HIPEC, PCI values of below 7 and less than 13 were predictive of OS in PCC and non-PCC populations respectively. CONCLUSION In selected patients, CRS-HIPEC offers acceptable outcomes among those with gastric PCC and long survival for patients without PCC.
Collapse
Affiliation(s)
- P E Bonnot
- Department of Surgical Oncology, Centre Georges Francois Leclerc, Dijon, France.,Department of Surgical Oncology, CHU Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - A Lintis
- Department of Surgical Oncology, CHU Lyon Sud, Hospices Civils de Lyon, Lyon, France.,Department of General Surgery, CHU Lille, Lille, France
| | - F Mercier
- Department of Surgical Oncology, CHU Lyon Sud, Hospices Civils de Lyon, Lyon, France.,Department of Surgical Oncology, Centre Hospitalier Universitaire de Montreal, Montreal, Quebec, Canada
| | - N Benzerdjeb
- Pathology Department, CHU Lyon Sud, Hospices Civils de Lyon, University of Lyon, Lyon, France
| | - G Passot
- Department of Surgical Oncology, CHU Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - M Pocard
- Department of Surgical Oncology, Hôpital Lariboisière, Paris, France
| | - B Meunier
- Department of Surgical Oncology, CHU Pontchaillou, Rennes, France
| | - J M Bereder
- Department of Surgical Oncology, CHU L'Archet, Nice, France
| | - K Abboud
- Department of Surgical Oncology, CHU St Etienne, St Etienne, France
| | - F Marchal
- Department of Surgical Oncology, Institut de Cancérologie de Lorraine, Université de Lorraine, Nancy, France
| | - F Quenet
- Department of Surgical Oncology, Centre Val D'Aurelle, Montpellier, France
| | - D Goere
- Department of Surgical Oncology, Institut Gustave Roussy, Villejuif, France
| | - S Msika
- Department of Surgical Oncology, CHU Louis Mourier, Paris, France
| | - C Arvieux
- Department of Surgical Oncology, CHU La Tronche, Grenoble, France
| | - N Pirro
- Department of Surgical Oncology, CHU La Timone, Marseille, France
| | - R Wernert
- Department of Surgical Oncology, Institut Paul Papin, Angers, France
| | - P Rat
- Department of Surgical Oncology, CHU Le Bocage, Dijon, France
| | - J Gagnière
- Department of Surgical Oncology, CHU Clermont-Ferrand, Clermont Ferrand, France
| | - J H Lefevre
- Department of Surgical Oncology, Hôpital Saint-Antoine, AP-HP, Paris, Sorbonne Université, Paris, France
| | - T Courvoisier
- Department of Surgical Oncology, CHU Poitiers, Poitiers, France
| | - R Kianmanesh
- Department of Surgical Oncology, CHU Reims, Reims, France
| | - D Vaudoyer
- Department of Surgical Oncology, CHU Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - M Rivoire
- Department of Surgical Oncology, Centre Léon Bérard, Lyon, France
| | - P Meeus
- Department of Surgical Oncology, Centre Léon Bérard, Lyon, France
| | - L Villeneuve
- Department of Surgical Oncology, CHU Lyon Sud, Hospices Civils de Lyon, Lyon, France.,Unité de Recherche Clinique, Pôle Information Médicale Evaluation Recherche, Hospices Civils de Lyon, Lyon, France
| | - G Piessen
- Department of General Surgery, CHU Lille, Lille, France
| | - O Glehen
- Department of Surgical Oncology, CHU Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | | |
Collapse
|
42
|
Thakkar PV, Kita K, Castillo UD, Galletti G, Madhukar N, Navarro EV, Barasoain I, Goodson HV, Sackett D, Díaz JF, Lu Y, RoyChoudhury A, Molina H, Elemento O, Shah MA, Giannakakou P. CLIP-170S is a microtubule +TIP variant that confers resistance to taxanes by impairing drug-target engagement. Dev Cell 2021; 56:3264-3275.e7. [PMID: 34672971 DOI: 10.1016/j.devcel.2021.09.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 07/14/2021] [Accepted: 09/24/2021] [Indexed: 01/22/2023]
Abstract
Taxanes are widely used cancer chemotherapeutics. However, intrinsic resistance limits their efficacy without any actionable resistance mechanism. We have discovered a microtubule (MT) plus-end-binding CLIP-170 protein variant, hereafter CLIP-170S, which we found enriched in taxane-resistant cell lines and patient samples. CLIP-170S lacks the first Cap-Gly motif, forms longer comets, and impairs taxane access to its MT luminal binding site. CLIP-170S knockdown reversed taxane resistance in cells and xenografts, whereas its re-expression led to resistance, suggesting causation. Using a computational approach in conjunction with the connectivity map, we unexpectedly discovered that Imatinib was predicted to reverse CLIP-170S-mediated taxane resistance. Indeed, Imatinib treatment selectively depleted CLIP-170S, thus completely reversing taxane resistance. Other RTK inhibitors also depleted CLIP-170S, suggesting a class effect. Herein, we identify CLIP-170S as a clinically prevalent variant that confers taxane resistance, whereas the discovery of Imatinib as a CLIP-170S inhibitor provides novel therapeutic opportunities for future trials.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Isabel Barasoain
- Centro de Investigaciones Biológicas Margarita Salas, Madrid, Spain
| | | | - Dan Sackett
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD 20892, USA
| | | | - Yao Lu
- Division of Biostatistics and Epidemiology, Department of Healthcare Policy & Research, Weill Cornell Medicine, New York, NY 10065, USA
| | - Arindam RoyChoudhury
- Division of Biostatistics and Epidemiology, Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, USA
| | - Henrik Molina
- Proteomics Resource Center, the Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | | | | | | |
Collapse
|
43
|
Huynh J, Patel K, Gong J, Cho M, Malla M, Parikh A, Klempner S. Immunotherapy in Gastroesophageal Cancers: Current Evidence and Ongoing Trials. Curr Treat Options Oncol 2021; 22:100. [PMID: 34524553 DOI: 10.1007/s11864-021-00893-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2021] [Indexed: 12/13/2022]
Abstract
OPINION STATEMENT Data supporting the use of immunotherapy in the treatment of gastroesophageal cancer continues to evolve. The promising results from adjuvant immunotherapy and trials combining immunotherapy plus chemotherapy in the 1L setting have led to broad US FDA approvals. Among the PD-L1 negative subgroups, the magnitude of benefit is diminished; effective therapy for this population remains an unmet need. A detailed biologic understanding of the PD-L1 negative (and low) population represents a barrier to developing effective combination therapies, although combination angiogenesis inhibitors and immunotherapy look encouraging. Early phase clinical trials, particularly with pembrolizumab plus lenvatinib (EPOC 1706), demonstrated a clear signal independent of PD-L1, and a confirmatory phase III trial of pembrolizumab plus lenvatinib is planned. Conceptually, it is important to think of immune checkpoint inhibitor therapy as targeted therapy, most active in clearly defined biomarker-selected populations. Pre-planned analyses have reliably shown a clear trend toward a greater magnitude of benefit in patients with higher PD-L1 expression, particularly CPS ≥ 5 and ≥ 10. Whether there is a linear relationship at higher cutoffs is not well known, though it likely represents smaller and smaller populations. Although beyond the scope of this clinically oriented review, recognition of the spatial and temporal heterogeneity in PD-L1 expression is important and repeat testing from progression samples across lines of therapy should be considered. Questions about additional predictive biomarkers, particularly plasma-derived, remain. Responses by tumor histology and location also differ, and special attention to these factors as well as MSI-H, HER2, and EBV subgroups in future trials is warranted. Questions regarding the incorporation of immunotherapy after progression on 1L immunotherapy plus chemotherapy combinations will arise as these combinations are used more frequently, and this represents a key area of future investigation. Overall, the role of immunotherapy continues to expand in GEA, and we welcome any additional tools for this difficult-to-treat group of cancers.
Collapse
Affiliation(s)
- Jasmine Huynh
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Kanishka Patel
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Jun Gong
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Health System, Los Angeles, CA, USA
| | - May Cho
- University of California Irvine Chao Family Comprehensive Cancer Center, Orange, CA, USA
| | - Midhun Malla
- West Virginia Cancer Institute, Morgantown, WV, USA
| | - Aparna Parikh
- Division of Hematology-Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Samuel Klempner
- Division of Hematology-Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
| |
Collapse
|
44
|
Huang Y, Yang Q, Zheng Y, Lin L, Xu X, Xu XE, Silva TC, Hazawa M, Peng L, Cao H, Ding Y, Lu D, Berman BP, Xu LY, Li EM, Yin D. Activation of bivalent factor DLX5 cooperates with master regulator TP63 to promote squamous cell carcinoma. Nucleic Acids Res 2021; 49:9246-9263. [PMID: 34370013 PMCID: PMC8450110 DOI: 10.1093/nar/gkab679] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 07/21/2021] [Accepted: 07/27/2021] [Indexed: 12/31/2022] Open
Abstract
To reconstruct systematically hyperactive transcription factor (TF)-dependent transcription networks in squamous cell carcinomas (SCCs), a computational method (ELMER) was applied to 1293 pan-SCC patient samples, and 44 hyperactive SCC TFs were identified. As a top candidate, DLX5 exhibits a notable bifurcate re-configuration of its bivalent promoter in cancer. Specifically, DLX5 maintains a bivalent state in normal tissues; its promoter is hypermethylation, leading to DLX5 transcriptional silencing in esophageal adenocarcinoma (EAC). In stark contrast, DLX5 promoter gains active histone marks and becomes transcriptionally activated in ESCC, which is directly mediated by SOX2. Functionally, silencing of DLX5 substantially inhibits SCC viability both in vitro and in vivo. Mechanistically, DLX5 cooperates with TP63 in regulating ∼2000 enhancers and promoters, which converge on activating cancer-promoting pathways. Together, our data establish a novel and strong SCC-promoting factor and elucidate a new epigenomic mechanism - bifurcate chromatin re-configuration - during cancer development.
Collapse
Affiliation(s)
- Yongsheng Huang
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Qian Yang
- Institute of Oncologic Pathology, Medical College of Shantou University, Shantou, China
| | - Yueyuan Zheng
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lehang Lin
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xin Xu
- Institute of Oncologic Pathology, Medical College of Shantou University, Shantou, China
| | - Xiu-E Xu
- Institute of Oncologic Pathology, Medical College of Shantou University, Shantou, China
| | - Tiago C Silva
- Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Masaharu Hazawa
- Cell-Bionomics Research Unit, Innovative Integrated Bio-Research Core, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, 920-1192 Ishikawa, Japan
| | - Li Peng
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Haotian Cao
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yanbing Ding
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Jiangsu, China
| | - Daning Lu
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Benjamin P Berman
- Center for Bioinformatics and Functional Genomics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Li-Yan Xu
- Institute of Oncologic Pathology, Medical College of Shantou University, Shantou, China
| | - En-Min Li
- Institute of Oncologic Pathology, Medical College of Shantou University, Shantou, China
| | - Dong Yin
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| |
Collapse
|
45
|
Du S, Yang Z, Lu X, Yousuf S, Zhao M, Li W, Miao J, Wang X, Yu H, Zhu X, Chen H, Shi L, Xu E, Xia X, Guan W. Anoikis resistant gastric cancer cells promote angiogenesis and peritoneal metastasis through C/EBPβ-mediated PDGFB autocrine and paracrine signaling. Oncogene 2021; 40:5764-5779. [PMID: 34341514 DOI: 10.1038/s41388-021-01988-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 11/09/2022]
Abstract
Anoikis is a type of programmed cell death induced by loss of anchorage to the extracellular matrix (ECM). Anoikis resistance (AR) is crucial for the survival of metastatic cancer cells in blood, lymphatic circulation and distant organs. Compared to ordinary cancer cells, anoikis resistant cancer cells undergo various cellular and molecular alterations, probably characterizing the cells with unique features not limited to anoikis resistance. However, the molecular mechanisms connecting anoikis resistance to other metastatic properties are still poorly understood. Here, the biological interaction between anoikis resistance and angiogenesis as well as their involvement into peritoneal metastasis of gastric cancer (GC) were investigated in vitro and in vivo. The prognostic value of key components involved in this interaction was evaluated in the GC cohort. Compared to ordinary GC cells, GCAR cells exhibited stronger metastatic and pro-angiogenic traits corresponding to elevated PDGFB secretion. Mechanistically, transcription factor C/EBPβ facilitated PDGFB transcription by directly binding to and interacting with PDGFB promoter elements, subsequently increasing PDGFB secretion. Secreted PDGFB promoted the survival of detached GC cells through a C/EBPβ-dependent self-feedback loop. Moreover, secreted PDGFB promoted angiogenesis in metastases via activation of the MAPK/ERK signaling pathway in vascular endothelial cells. Both C/EBPβ activation level and PDGFB expression were significantly elevated in GC and correlated with metastatic progression and poor prognosis of patients with GC. Overall, interaction between GCAR cells and vascular endothelial cells promotes angiogenesis and peritoneal metastasis of GC based on C/EBPβ-mediated PDGFB autocrine and paracrine signaling. C/EBPβ-PDGFB-PDGFRβ-MAPK axis promises to be potential prognostic biomarkers and therapeutic targets for peritoneal metastasis of GC.
Collapse
Affiliation(s)
- Shangce Du
- Department of Gastrointestinal Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, P.R. China.,Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, P.R. China
| | - Zhi Yang
- Department of Gastrointestinal Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, P.R. China.,Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, P.R. China
| | - Xiaofeng Lu
- Department of Gastrointestinal Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, P.R. China.,Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, P.R. China
| | - Suhail Yousuf
- Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Min Zhao
- Department of Gastrointestinal Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, P.R. China
| | - Wenxi Li
- Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| | - Ji Miao
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, P.R. China
| | - Xingzhou Wang
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, P.R. China
| | - Heng Yu
- Department of Gastrointestinal Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, P.R. China
| | - Xinya Zhu
- Department of Gastrointestinal Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, P.R. China
| | - Hong Chen
- Department of Gastrointestinal Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, P.R. China
| | - Linseng Shi
- Department of Gastrointestinal Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, P.R. China
| | - En Xu
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, P.R. China.
| | - Xuefeng Xia
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, P.R. China.
| | - Wenxian Guan
- Department of Gastrointestinal Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, P.R. China. .,Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, P.R. China.
| |
Collapse
|
46
|
Tian S, Liu Z, Zhou Q, Wu R, Huang X, Liang Z, Zhang Z, Tian X. Upregulation of MiR-340-5p Reverses Cisplatin Sensitivity by Inhibiting the Expression of CDK6 in HepG2 Cells. Folia Biol (Praha) 2021. [DOI: 10.3409/fb_69-2.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Cisplatin (CDDP) has been successfully used in chemotherapy for liver cancer. However, the development of CDDP resistance in HepG2 cells usually leads to relapse and a worsening prognosis. MiR-340-5p has attracted much attention because of its ability to affect cell resistance. This
project is intended to explore the role of miR-340-5p and CDK6 in CDDP-R HepG2 cells and provide new ideas for the treatment of liver cancer. A dual-luciferase reporter assay was used to confirm the targeting relationship between miR-340-5p and CDK6. We constructed a CDDP-resistant model of
HepG2 cells to examine the effect of miR-340-5p on the drug sensitivity of HepG2 cells. CDDP-R HepG2 cells were transfected with miR-340-5p overexpression plasmid and CDK6 silencing plasmid. QRT-PCR was used to detect the expression of miR-340-5p and CDK6. A western blot was performed to determine
the expression of CDK6, CyclinD1, and CyclinD2. CCK-8, flow cytometry, TUNEL and Clonogenic assays were also carried out to detect CDDP-R HepG2 cells. There is a targeting relationship between miR-340-5p and CDK6. The drug resistance of CDDP-R HepG2 cells was significantly higher than that
of CDDP-S HepG2 cells. CDDP-R HepG2 cells transfected with both miR-340-5p overexpressing plasmid and CDK6 silencing plasmid showed a lower proliferation ability, cell cycle arrest in the G0/G1 phase, and lower drug resistance compared with single CDDP-R HepG2 cells. Overexpression of miR-340-5p
aggravated CDDP-R HepG2 cells' apoptosis and inhibited cell viability. Overexpression of miR-340-5p could reverse the sensitivity of HepG2 cells to CDDP by inhibiting the expression of CDK6 in HepG2 cells.
Collapse
|
47
|
Huang W, Zhan D, Li Y, Zheng N, Wei X, Bai B, Zhang K, Liu M, Zhao X, Ni X, Xia X, Shi J, Zhang C, Lu Z, Ji J, Wang J, Wang S, Ji G, Li J, Nie Y, Liang W, Wu X, Cui J, Meng Y, Cao F, Shi T, Zhu W, Wang Y, Chen L, Zhao Q, Wang H, Shen L, Qin J. Proteomics provides individualized options of precision medicine for patients with gastric cancer. SCIENCE CHINA-LIFE SCIENCES 2021; 64:1199-1211. [PMID: 34258712 DOI: 10.1007/s11427-021-1966-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/17/2021] [Indexed: 12/19/2022]
Abstract
While precision medicine driven by genome sequencing has revolutionized cancer care, such as lung cancer, its impact on gastric cancer (GC) has been minimal. GC patients are routinely treated with chemotherapy, but only a fraction of them receive the clinical benefit. There is an urgent need to develop biomarkers or algorithms to select chemo-sensitive patients or apply targeted therapy. Here, we carried out retrospective analyses of 1,020 formalin-fixed, paraffin-embedded GC surgical resection samples from 5 hospitals and developed a mass spectrometry-based workflow for proteomic subtyping of GC. We identified two proteomic subtypes: the chemo-sensitive group (CSG) and the chemo-insensitive group (CIG) in the discovery set. The 5-year overall survival of CSG was significantly improved in patients who had received adjuvant chemotherapy after surgery compared with those who received surgery only (64.2% vs. 49.6%; Cox P-value=0.002), whereas no such improvement was observed in CIG (50.0% vs. 58.6%; Cox P-value=0.495). We validated these results in an independent validation set. Further, differential proteome analysis uncovered 9 FDA-approved drugs that may be applicable for targeted therapy of GC. A prospective study is warranted to test these findings for future GC patient care.
Collapse
Affiliation(s)
- Wenwen Huang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Dongdong Zhan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China.,Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yazhuo Li
- Department of Pathology, The Fourth Medical Center of PLA General Hospital, Beijing, 100048, China
| | - Nairen Zheng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Xin Wei
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China.,Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Bin Bai
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Kecheng Zhang
- Department of General Surgery & Institute of General Surgery, Chinese PLA General Hospital First Medical Center, Beijing, 100853, China
| | - Mingwei Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Xuefei Zhao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Xiaotian Ni
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Xia Xia
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Jinwen Shi
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Cheng Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Zhihao Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Jiafu Ji
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Juan Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Shiqi Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Gang Ji
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Jipeng Li
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Wenquan Liang
- Department of General Surgery & Institute of General Surgery, Chinese PLA General Hospital First Medical Center, Beijing, 100853, China
| | - Xiaosong Wu
- Department of General Surgery & Institute of General Surgery, Chinese PLA General Hospital First Medical Center, Beijing, 100853, China
| | - Jianxin Cui
- Department of General Surgery & Institute of General Surgery, Chinese PLA General Hospital First Medical Center, Beijing, 100853, China
| | - Yongsheng Meng
- Department of tumor biobank, Shanxi Cancer Hospital, Taiyuan, 030013, China
| | - Feilin Cao
- Department of tumor biobank, Shanxi Cancer Hospital, Taiyuan, 030013, China
| | - Tieliu Shi
- Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Weimin Zhu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Lin Chen
- Department of General Surgery & Institute of General Surgery, Chinese PLA General Hospital First Medical Center, Beijing, 100853, China.
| | - Qingchuan Zhao
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China.
| | - Hongwei Wang
- Department of Pathology, The Fourth Medical Center of PLA General Hospital, Beijing, 100048, China.
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China. .,State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
48
|
Shirakawa Y, Tazawa H, Tanabe S, Kanaya N, Noma K, Koujima T, Kashima H, Kato T, Kuroda S, Kikuchi S, Kagawa S, Katsui K, Kanazawa S, Urata Y, Fujiwara T. Phase I dose-escalation study of endoscopic intratumoral injection of OBP-301 (Telomelysin) with radiotherapy in oesophageal cancer patients unfit for standard treatments. Eur J Cancer 2021; 153:98-108. [PMID: 34153720 DOI: 10.1016/j.ejca.2021.04.043] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/19/2021] [Accepted: 04/30/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE OBP-301 (Telomelysin) is an attenuated type-5 adenovirus that contains the human telomerase reverse transcriptase promoter to regulate viral replication. OBP-301 sensitises human cancer cells to ionising radiation by inhibiting DNA repair, and radiation enhances coxsackievirus and adenovirus receptor-mediated OBP-301 infection on the contrary. We assessed OBP-301 with radiotherapy in oesophageal cancer patients unfit for standard chemoradiation treatments. METHODS A phase I dose-escalation study of OBP-301 with radiotherapy was conducted in 13 histologically confirmed oesophageal cancer patients deemed unfit to undergo surgery or chemotherapy. Study treatment consisted of OBP-301 administration by intratumoural needle injection using a flexible endoscope on days 1, 18 and 32. Radiotherapy was administered concurrently over 6 weeks, beginning on day 4, to a total of 60 Gy. RESULTS Of the 13 patients, 7, 3 and 3 patients were treated with 1010, 1011 and 1012 virus particles, respectively. Study group comprised 10 males and 3 females, with a median age of 82 years (range, 53-91 years). All patients developed a transient, self-limited lymphopenia. Distribution studies revealed transient virus shedding in the plasma. Eight patients had local complete response (CR); all of them exhibited no pathologically viable malignant cells in biopsy specimens, and 3 patients had a partial response. The objective response rate was 91.7%. The clinical CR rate was 83.3% in stage I and 60.0% in stage II/III. Histopathological examination revealed massive infiltration of CD8+ cells and increased PD-L1 expression. CONCLUSION Multiple courses of endoscopic intratumoural OBP-301 injection with radiotherapy are feasible and provide clinical benefits in patients with oesophageal cancer unfit for standard treatments.
Collapse
Affiliation(s)
- Yasuhiro Shirakawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hiroshi Tazawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama 700-8558, Japan
| | - Shunsuke Tanabe
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Nobuhiko Kanaya
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Kazuhiro Noma
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Takeshi Koujima
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hajime Kashima
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Takuya Kato
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shinji Kuroda
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Satoru Kikuchi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shunsuke Kagawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Kuniaki Katsui
- Department of Radiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Susumu Kanazawa
- Department of Radiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Yasuo Urata
- Oncolys BioPharma, Inc., Tokyo 106-0032, Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| |
Collapse
|
49
|
Gou M, Zhang Y, Liu T, Qu T, Si H, Wang Z, Yan H, Qian N, Dai G. The Prognostic Value of Pre-treatment Hemoglobin (Hb) in Patients With Advanced or Metastatic Gastric Cancer Treated With Immunotherapy. Front Oncol 2021; 11:655716. [PMID: 34211839 PMCID: PMC8239234 DOI: 10.3389/fonc.2021.655716] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/11/2021] [Indexed: 11/13/2022] Open
Abstract
Background Biomarkers such as prevailing PD-L1 expression and TMB have been proposed as a way of predicting the outcome of immunotherapy in patients with advanced gastric cancer (AGC) and metastatic gastric cancer (MGC). Our study aims to investigate whether there is a link between pretreatment hemoglobin (Hb) levels and survival to immunotherapy in patients with AGC and MGC. Methods We retrospectively reviewed patients with AGC or MGC treated at the oncology department of the Chinese PLA general hospital receiving PD-1 inhibitor. The Propensity Score Matching (PSM) (1:1) was performed to balance potential baseline confounding factors. Progression-free survival (PFS) and overall survival (OS) was analyzed among different Hb level (normal Hb group and decreased Hb group). Objective response rate (ORR), disease control rate (DCR) were also analyzed. Univariate analysis and multivariate analysis were performed further to validate the prognostic value of Hb level. Results We included 137 patients with AGC and MGC who received PD-1 inhibitors (including Pembrolizumab, Nivolumab, Sintilimab, Toripalimab) in this study. After PSM matching, there were no significant differences between the two groups for baseline characteristics. Within the matched cohort, the median PFS was 7.8 months in the normal Hb level group and 4.3 months in the decreased Hb group (HR 95% CI 0.5(0.31, 0.81), P=0.004). The OS was 14.4 months with normal Hb level as compared with 8.2 months with decreased Hb level(HR 95% CI 0.59(0.37, 0.94), P=0.024). The ORR was 40.7% and DCR was 83.0% in the normal Hb group, while the ORR was 25.5% and DCR was 85.1% in the decreased Hb group. No significant differences were found in the ORR and DCR between the two groups (P=0.127, P=0.779). Univariate analysis and multivariate analysis showed that Hb level was only independent predictor for PFS and baseline Hb level was significant prognostic factor influencing the OS. Only when patients had normal Hb level, anti-pd-1 monotherapy or combined with chemotherapy was superior to anti-pd-1 plus anti-angiogenic therapy with respect to PFS (10.3 m vs 2.8 m, HR 95% CI 0.37(0.15, 0.95), P=0.031) and OS(15 m vs 5.7 m, HR 95% CI 0.21 (0.08, 0.58), P=0.001). Conclusions Our study have demonstrated that pretreatment Hb level was an independent prognostic biomarker in term of PFS and OS with immunotherapy for AGC and MGC patients. Correction of anemia for GC patients as immunotherapy would be a strategy to improve the survival. More data was warranted to further influence this finding.
Collapse
Affiliation(s)
- Miaomiao Gou
- Medical Oncology Department, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yong Zhang
- Medical Oncology Department, The Second Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Tiee Liu
- Medical Oncology Department, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Tongtong Qu
- Medical Oncology Department, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Haiyan Si
- Medical Oncology Department, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhikuan Wang
- Medical Oncology Department, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Huan Yan
- Medical Oncology Department, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Niansong Qian
- Medical Oncology Department, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.,The Hainan Medical Center, Chinese People's Liberation Army General Hospital, Sanya, China
| | - Guanghai Dai
- Medical Oncology Department, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
50
|
Lu Y, Guan L, Xu M, Wang F. The efficacy and safety of antibodies targeting PD-1 for treatment in advanced esophageal cancer: A systematic review and meta-analysis. Transl Oncol 2021; 14:101083. [PMID: 33784583 PMCID: PMC8042180 DOI: 10.1016/j.tranon.2021.101083] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND A novel therapy based on programmed death 1 (PD-1) inhibitors has been proved to be effective in advanced esophageal cancer. This article is a meta-analysis that aims to compare the efficacy and safety of anti-PD-1 therapy with chemotherapy in esophageal cancer. PATIENTS AND METHODS Data were collected from eligible studies searched from PubMed, Web of Science, Cochrane Library, and Embase. Pooled hazard ratio (HR) for overall survival (OS), progression-free survival (PFS), and objective response rate (ORR) was estimated to assess the efficacy of PD-1 inhibitors versus chemotherapy. The subgroup analysis was also performed to evaluate the OS benefits. The OR for the occurrence of treatment-related adverse effects was calculated to assess the safety of anti-PD-1 therapy. RESULTS A total of 4 studies were analyzed. Compared with patients with chemotherapy, patients with anti-PD-1 therapy had a significant improvement in OS (HR = 0.79, 95% CI: 0.71-0.88, and P<0.001), but no significant relationship was observed in PFS (HR = 0.96, 95% CI: 0.76-1.20, and P = 0.69) and ORR (OR = 1.92, 95% CI: 0.98-3.72, and P = 0.06). A similar result was observed in esophageal squamous cell carcinoma. The significant predictor for treatment benefit alone was histology (P = 0.009). The incidence of grade 3 - 5 treatment-related adverse effects in anti-PD-1 therapy was distinctly lower than that in chemotherapy, but there is no statistical difference in all treatment-related adverse effects. CONCLUSION Anti-PD-1 therapy significantly prolonged the OS, simultaneously lowered grade 3 - 5 treatment-related adverse effects versus chemotherapy.
Collapse
Affiliation(s)
- Yao Lu
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P R China
| | - Lulu Guan
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P R China
| | - Mengli Xu
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P R China
| | - Feng Wang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P R China.
| |
Collapse
|