101
|
Liu M, Fang X, Wang H, Ji R, Guo Q, Chen Z, Ren Q, Wang Y, Zhou Y. Characterization of lipid droplet metabolism patterns identified prognosis and tumor microenvironment infiltration in gastric cancer. Front Oncol 2023; 12:1038932. [PMID: 36713557 PMCID: PMC9875057 DOI: 10.3389/fonc.2022.1038932] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023] Open
Abstract
Background Gastric cancer is one of the common malignant tumors of the digestive system worldwide, posing a serious threat to human health. A growing number of studies have demonstrated the important role that lipid droplets play in promoting cancer progression. However, few studies have systematically evaluated the role of lipid droplet metabolism-related genes (LDMRGs) in patients with gastric cancer. Methods We identified two distinct molecular subtypes in the TCGA-STAD cohort based on LDMRGs expression. We then constructed risk prediction scoring models in the TCGA-STAD cohort by lasso regression analysis and validated the model with the GSE15459 and GSE66229 cohorts. Moreover, we constructed a nomogram prediction model by cox regression analysis and evaluated the predictive efficacy of the model by various methods in STAD. Finally, we identified the key gene in LDMRGs, ABCA1, and performed a systematic multi-omics analysis in gastric cancer. Results Two molecular subtypes were identified based on LDMRGs expression with different survival prognosis and immune infiltration levels. lasso regression models were effective in predicting overall survival (OS) of gastric cancer patients at 1, 3 and 5 years and were validated in the GEO database with consistent results. The nomogram prediction model incorporated additional clinical factors and prognostic molecules to improve the prognostic predictive value of the current TNM staging system. ABCA1 was identified as a key gene in LDMRGs and multi-omics analysis showed a strong correlation between ABCA1 and the prognosis and immune status of patients with gastric cancer. Conclusion This study reveals the characteristics and possible underlying mechanisms of LDMRGs in gastric cancer, contributing to the identification of new prognostic biomarkers and providing a basis for future research.
Collapse
Affiliation(s)
- Mengxiao Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xidong Fang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Haoying Wang
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xinan, China
| | - Rui Ji
- Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Qinghong Guo
- Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhaofeng Chen
- Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Qian Ren
- Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yuping Wang
- Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yongning Zhou
- Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
102
|
Gastric Cancer: Innovations in Screening, Diagnosis and Treatment. J Pers Med 2023; 13:jpm13010127. [PMID: 36675788 PMCID: PMC9864353 DOI: 10.3390/jpm13010127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 01/06/2023] [Indexed: 01/10/2023] Open
Abstract
Gastric cancer (GC) is an aggressive and heterogeneous malignancy that is one of the leading cancers in the world and an important global health problem due to its overall high prevalence and high mortality rate [...].
Collapse
|
103
|
Zhang Z, Peng L, Yang W, Li B, Hua Y, Luo S. PHF5A facilitates the development and progression of gastric cancer through SKP2-mediated stabilization of FOS. J Transl Med 2023; 21:5. [PMID: 36609277 PMCID: PMC9817416 DOI: 10.1186/s12967-022-03821-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 12/11/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is the fifth most common cancer and the third most common cause of cancer death worldwide. Plant homeodomain (PHD)-finger domain protein PHF5A has been demonstrated to play a promoting role in a variety of cancers. This study aimed to clarify the role of PHF5A in the progression of GC and its potential mechanism of action. METHODS Immunohistochemical staining experiments were performed based on tissues from clinical GC patients to reveal PHF5A expression. A series of functional experiments in vitro and in vivo were used to clarify the role of PHF5A in GC. RESULTS Clinically, PHF5A was abundantly expressed in GC and existed clinical value indicating poor prognosis. In addition, GC cells with knockdown of PHF5A expression showed slowed proliferation, enhanced sensitivity to apoptosis and inhibition of migration. Mechanically, knockdown of PHF5A led to decreased protein stability of FOS, which was mediated ubiquitination of E3 ubiquitin ligase S-phase kinase-associated protein 2 (SKP2). Moreover, downregulation of FOS attenuated the promotion of PHF5A overexpression on GC cells. Consistently, Pladienolide B (PHF5A inhibitor) treatment reversed the induction of PHF5A overexpression on the malignant phenotypes and tumor formation of GC cells. CONCLUSION Knockdown of PHF5A inhibited the progression of GC through SKP2-mediated ubiquitination of FOS, which may be a promising candidate target with potential therapeutic value.
Collapse
Affiliation(s)
- Zhandong Zhang
- grid.414008.90000 0004 1799 4638Department of General Surgery, Henan Tumor Hospital, Affiliated Tumor Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou, Henan China
| | - Liangqun Peng
- grid.414008.90000 0004 1799 4638Department of General Surgery, Henan Tumor Hospital, Affiliated Tumor Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou, Henan China
| | - Wei Yang
- grid.414008.90000 0004 1799 4638Department of General Surgery, Henan Tumor Hospital, Affiliated Tumor Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou, Henan China
| | - Baodong Li
- grid.414008.90000 0004 1799 4638Department of General Surgery, Henan Tumor Hospital, Affiliated Tumor Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou, Henan China
| | - Yawei Hua
- grid.414008.90000 0004 1799 4638Department of General Surgery, Henan Tumor Hospital, Affiliated Tumor Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou, Henan China
| | - Suxia Luo
- grid.414008.90000 0004 1799 4638Department of General Surgery, Henan Tumor Hospital, Affiliated Tumor Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou, Henan China
| |
Collapse
|
104
|
Development and Evaluation of a Novel Cuproptosis-Related lncRNA Signature for Gastric Cancer Prognosis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2023; 2023:6354212. [PMID: 36820319 PMCID: PMC9938768 DOI: 10.1155/2023/6354212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/27/2022] [Accepted: 01/06/2023] [Indexed: 02/13/2023]
Abstract
Background According to a growing body of research, long noncoding RNAs (lncRNAs) participate in the progress of gastric cancer (GC). Cuproptosis is a distinct kind of programmed cell death, separating it from several other forms of programmed cell death that may be caused by genetic programming. Consequently, it is crucial to examine cuproptosis-related lncRNAs (CRLs) prognostic importance for the prognosis and treatment response in GC. Method The Cancer Genome Atlas (TCGA) database was used to retrieve RNA-seq data, pertinent clinical information, and somatic mutation data. A list of cuproptosis-related genes (CRGs) was obtained from prior work. We can distinguish prognostic CRLs using coexpression and univariate Cox analysis. Then, using CRLs, we developed a risk prediction model using multivariate Cox regression analysis and the least absolute shrinkage selection operator (LASSO) technique. To evaluate the diagnostic accuracy of this model, a Kaplan-Meier (K-M) survival analysis and a receiver operating characteristic (ROC) analysis were used. Moreover, the relationships between the risk model and immunological function, somatic mutation, and drug sensitivity were also investigated. Results Using the multivariate Cox analysis technique, we developed a signature based on cuproptosis-related four lncRNAs. We then classified patients into high-risk and low-risk groups based on the likelihood of unfavorable outcomes. The model was subjected to further testing, including K-M survival analysis, ROC analysis, and multivariate Cox regression analysis, all of which proved the model's exceptional robustness and predictive capacity. In addition, a nomogram that has a strong capacity for prediction ability was built. This nomogram included age, gender, clinical grade, pathologic stage, T stage, and risk score. Furthermore, we discovered substantial disparities in immune function and the number of mutations carried by tumors between the high-risk and low-risk groups. Moreover, this research also found that the IC50 values for 27 chemotherapeutic drugs varied widely across patients within high- and low-risk groups. Conclusion The proposed 4-CRLs signature is a promising biomarker to predict clinical outcomes in GC.
Collapse
|
105
|
Usman M, Beilerli A, Sufianov A, Kudryashov V, Ilyasova T, Balaev P, Danilov A, Lu H, Gareev I. Investigations into the impact of non-coding RNA on the sensitivity of gastric cancer to radiotherapy. Front Physiol 2023; 14:1149821. [PMID: 36909247 PMCID: PMC9998927 DOI: 10.3389/fphys.2023.1149821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/16/2023] [Indexed: 02/26/2023] Open
Abstract
Non-coding RNAs (ncRNAs) are a newly discovered functional RNA different from messenger RNA, which can participate in regulating the occurrence and development of tumors. More and more research results show that ncRNAs can participate in the regulation of gastric cancer (GC) radiotherapy response, and its mechanism may be related to its effect on DNA damage repair, gastric cancer cell stemness, cell apoptosis, activation of epidermal growth factor receptor signaling pathway, etc. This article summarizes the relevant mechanisms of ncRNAs regulating the response to radiotherapy in gastric cancer, which will be directly important for the introduction of ncRNAs particularly microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) into clinical medicine as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Muhammad Usman
- Department of Medical Imaging, Central Hospital Affiliated to Chongqing University of Technology, Chongqing, China
| | - Aferin Beilerli
- Department of Obstetrics and Gynecology, Tyumen State Medical University, Tyumen, Russia
| | - Albert Sufianov
- Department of Neurosurgery, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.,Department of Internal Diseases, Bashkir State Medical University, Ufa, Russia
| | - Valentin Kudryashov
- Gastric Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Tatiana Ilyasova
- Department of Internal Diseases, Bashkir State Medical University, Ufa, Russia
| | - Pavel Balaev
- Department of Oncology and Radiology, Ural State Medical University, Yekaterinburg, Russia
| | - Andrei Danilov
- Department of Clinical Pharmacology, Smolensk State Medical University, Smolensk, Russia
| | - Hong Lu
- Department of Medical Imaging, Central Hospital Affiliated to Chongqing University of Technology, Chongqing, China
| | - Ilgiz Gareev
- Educational and Scientific Institute of Neurosurgery, Рeoples' Friendship University of Russia (RUDN University), Moscow, Russia
| |
Collapse
|
106
|
Feng J, Tang X, Song L, Zhou Z, Jiang Y, Huang Y. A telomerase regulation-related lncRNA signature predicts prognosis and immunotherapy response for gastric cancer. J Cancer Res Clin Oncol 2023; 149:135-146. [PMID: 36333566 DOI: 10.1007/s00432-022-04456-6] [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: 09/10/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Telomeres are involved in the development and progression of gastric cancer (GC). However, the association of telomerase regulation-related lncRNAs with prognosis and immunotherapy responsiveness in gastric cancer is unclear. METHODS This study systematically evaluated the relationship between lncRNAs co-expressed with 67 telomerase regulatory genes and gastric cancer prognosis. The risk scores of the samples were calculated based on telomerase regulation-related lncRNAs with prognostic value, and the samples were classified into high-/low-risk groups. The prognostic value of risk groups was then evaluated, a GC prognostic prediction model based on risk groups and clinical characteristics was established, and the prediction accuracy of the model was clarified by receiving operating characteristic (ROC) curves and calibration curves. Finally, the value of risk grouping in GC immunotherapy sensitivity was predicted by comparing MSI status and tumor mutation load between the high- and low-risk groups. RESULTS We identified 13 lncRNAs with prognostic value co-expressed with telomerase regulatory genes and observed that the prognosis of the low-risk group was significantly better than that of the high-risk group. Meanwhile, a GC overall survival (OS) prediction model based on risk grouping and clinical characteristics was developed, and ROC curves and calibration curves confirmed the good predictive ability of the model. In addition, the low-risk group exhibited a higher tumor mutation load and MSI-H, suggesting a possible benefit of immunotherapy. CONCLUSION We found that telomerase regulation-related lncRNAs have prognostic value in GC patients and contribute to the exploration of more effective immunotherapeutic strategies.
Collapse
Affiliation(s)
- Jinggao Feng
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan Province, China.
| | - Xiayu Tang
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan Province, China
| | - Liusong Song
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan Province, China
| | - Zhipeng Zhou
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan Province, China
| | - Yuan Jiang
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan Province, China
| | - Yao Huang
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan Province, China
| |
Collapse
|
107
|
Li M, Tao J, Qian R, Jiang F, Song Y, Zeng Z, Cai C. Development of alternative herbals remedy for gastric cancer based on transcriptomic analysis of immune infiltration and ferroptosis. Front Genet 2023; 14:1086368. [PMID: 36936437 PMCID: PMC10020191 DOI: 10.3389/fgene.2023.1086368] [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: 11/01/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Objective: Screening out potential herbal medicines and herbal ingredients for the treatment of gastric cancer based on transcriptomic analysis of immune infiltration and ferroptosis. Methods: Gene expression profiles of gastric tumour tissues and normal tissue samples were obtained from the GEO database and the samples were analysed for immune cell infiltration condition and differential expressed genes of ferroptosis. Key genes were screened by protein-protein interaction (PPI) and enrichment analysis, and molecular docking was used to predict and preliminary validate potential herbal and traditional Chinese medicine components for gastric cancer based on the key genes. Finally, RT-QPCR was used to validate the prediction results. Results: Immune cell infiltration analysis revealed high levels of infiltration of activated CD4 memory T cells, monocytes, M0 macrophages in gastric tumor tissues, while plasma cells and resting mast cells had higher levels of infiltration in the paraneoplastic tissues. Differential gene expression analysis identified 1,012 upregulated genes and 880 downregulated genes, of which 84 immune related differentially expressed genes such as CTSB, PGF and PLAU and 10 ferroptosis-related differentially expressed genes such as HSF1, NOX4 and NF2 were highly expressed in gastric cancer tissues. The results of enrichment analysis showed that they mainly involve 343 biological processes such as extracellular matrix organization and extracellular structural organization; 37 cellular components such as complexes of collagen trimer and basement membrane; 35 molecular functions such as signal receptor activator activity and receptor ligand activity; 19 regulatory pathways such as cytokine-cytokine receptor interactions and retinol metabolism. Finally, two key genes, TLR4 and KRAS, were selected and 12 herbal medicines such as Radix Salviae liguliobae, Rhizoma Coptidis, Rhizoma Polygoni cuspidati and 27 herbal ingredients such as resveratrol, salvianolic acid b were predicted on the basis of key genes. Molecular docking results showed that KRAS binds tightly to coumarin and magnolol, while TLR4 can bind tightly to resveratrol, curcumin, salvianolic acid b, shikonin. Subsequently, the effect of resveratrol and magnolol was experimentally verified. Conclusion: Herbal medicines such as S. liguliobae, Rhizoma Coptidis, Rhizoma P. cuspidati and herbal ingredients such as resveratrol, curcumin, salvianolic acid b may provide research directions and alternative therapeutic approaches for immunomodulation of TME and ferroptosis of tumour cells in gastric cancer.
Collapse
Affiliation(s)
- Mingyue Li
- Department of Pharmacy, Shenzhen Bao’an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Jie Tao
- Department of Cardiology, Shenzhen Bao’an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Rui Qian
- Department of Gastroenterology, Shenzhen Bao’an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Feng Jiang
- Department of Cardiology, Shenzhen Bao’an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yinzhi Song
- Department of Cardiology, Shenzhen Bao’an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Zhicong Zeng
- Department of Cardiology, Shenzhen Bao’an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- *Correspondence: Zhicong Zeng, ; Changlong Cai,
| | - Changlong Cai
- Department of Surgery, Shenzhen Bao’an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- *Correspondence: Zhicong Zeng, ; Changlong Cai,
| |
Collapse
|
108
|
LncRNA BBOX1-AS1 Contributes to the Progression of Esophageal Carcinoma by Targeting the miR-361-3p/COL5A1 Axis. Biochem Genet 2022:10.1007/s10528-022-10307-3. [PMID: 36586008 DOI: 10.1007/s10528-022-10307-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/22/2022] [Indexed: 01/01/2023]
Abstract
Long noncoding RNAs (lncRNAs) are known to participate in the progression of several cancers, including esophageal carcinoma (EC), a common malignancy of the digestive system. Although the role of the lncRNA-miRNA-mRNA regulatory network is crucial for the growth and progression of EC, the regulation of lncRNA BBOX1-AS1 (BBOX1 antisense RNA1) remains unclear. We performed reverse transcription-quantitative PCR (RT-qPCR) and western blotting to evaluate miR-361-3p, collagen type V alpha 1 chain (COL5A1), and BBOX1-AS1 expression levels in EC cells and tissues. The colony formation assay (CFA) and Cell Counting Kit-8 (CCK-8) were employed to identify EC cell proliferation, while western blotting was used to examine EC cell apoptosis and Bax and Bcl-2 expression levels. The effect of BBOX1-AS1 on EC proliferation was determined using an in vivo carcinogenesis assay. Correlation between COL5A1, BBOX1-AS1, and miR-361-3p was examined using the luciferase reporter system and RNA immunoprecipitation assay (RIP). Herein, we observed that BBOX1-AS1 expression levels were upregulated in EC cells and tissues. BBOX1-AS1 knockdown inhibited EC cell proliferation and conferred a pro-apoptotic effect. These results indicated a positive interaction between BBOX1-AS1 and miR-361-3p in EC and a negative association with miR-361-3p. COL5A1 was recognized as a downstream miR-361-3p target and was inversely related to miR-361-3p in EC. Therefore, BBOX1-AS1 expression suppressed cell apoptosis and promoted cell proliferation via the downregulation of miR-361-3p and upregulation of COL5A1 expression. Overall, BBOX1-AS1 facilitates EC progression via the miR-361-3p or COL5A1 axis, indicating that BBOX1-AS1 might be a potential therapeutic target for EC therapy.
Collapse
|
109
|
Spagnol LW, Polettini J, Silveira DA, Wegner GRM, Paiva DFF. P16 gene promoter methylation is associated with oncogenesis and progression of gastric carcinomas: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2022; 180:103843. [DOI: 10.1016/j.critrevonc.2022.103843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/02/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
|
110
|
Li N, Meng G, Yang C, Li H, Liu L, Wu Y, Liu B. Changes in epigenetic information during the occurrence and development of gastric cancer. Int J Biochem Cell Biol 2022; 153:106315. [DOI: 10.1016/j.biocel.2022.106315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/22/2022] [Accepted: 10/18/2022] [Indexed: 11/24/2022]
|
111
|
Zeng C, He R, Dai Y, Lu X, Deng L, Zhu Q, Liu Y, Liu Q, Lu W, Wang Y, Jin J. Identification of TGF-β signaling-related molecular patterns, construction of a prognostic model, and prediction of immunotherapy response in gastric cancer. Front Pharmacol 2022; 13:1069204. [PMID: 36467074 PMCID: PMC9715605 DOI: 10.3389/fphar.2022.1069204] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/07/2022] [Indexed: 06/22/2024] Open
Abstract
Background: TGF-β signaling pathway plays an essential role in tumor progression and immune responses. However, the link between TGF-β signaling pathway-related genes (TSRGs) and clinical prognosis, tumor microenvironment (TME), and immunotherapy in gastric cancer is unclear. Methods: Transcriptome data and related clinical data of gastric cancer were downloaded from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, and 54 TSRGs were obtained from the Molecular Signatures Database (MSigDB). We systematically analyzed the expression profile characteristics of 54 TSRGs in 804 gastric cancer samples and examined the differences in prognosis, clinicopathological features, and TME among different molecular subtypes. Subsequently, TGF-β-related prognostic models were constructed using univariate and least absolute shrinkage and selection operator (LASSO) Cox regression analysis to quantify the degree of risk in each patient. Patients were divided into two high- and low-risk groups based on the median risk score. Finally, sensitivity to immune checkpoint inhibitors (ICIs) and anti-tumor agents was assessed in patients in high- and low-risk groups. Results: We identified two distinct TGF-β subgroups. Compared to TGF-β cluster B, TGF-β cluster A exhibits an immunosuppressive microenvironment with a shorter overall survival (OS). Then, a novel TGF-β-associated prognostic model, including SRPX2, SGCE, DES, MMP7, and KRT17, was constructed, and the risk score was demonstrated as an independent prognostic factor for gastric cancer patients. Further studies showed that gastric cancer patients in the low-risk group, characterized by higher tumor mutation burden (TMB), the proportion of high microsatellite instability (MSI-H), immunophenoscore (IPS), and lower tumor immune dysfunction and exclusion (TIDE) score, had a better prognosis, and linked to higher response rate to immunotherapy. In addition, the risk score and anti-tumor drug sensitivity were strongly correlated. Conclusion: These findings highlight the importance of TSRGs, deepen the understanding of tumor immune microenvironment, and guide individualized immunotherapy for gastric cancer patients.
Collapse
Affiliation(s)
- Cheng Zeng
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Rong He
- Department of Medical Oncology, Shanghai Tenths People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuyang Dai
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiaohuan Lu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Linghui Deng
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qi Zhu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Yu Liu
- Department of Internal Medicine, School of Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Qian Liu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Wenbin Lu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Yue Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jianhua Jin
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
112
|
Wang J, Liu T, Huang T, Shang M, Wang X. The mechanisms on evasion of anti-tumor immune responses in gastric cancer. Front Oncol 2022; 12:943806. [PMID: 36439472 PMCID: PMC9686275 DOI: 10.3389/fonc.2022.943806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 09/02/2022] [Indexed: 10/22/2023] Open
Abstract
The immune system and the tumor have been at each other's throats for so long that the neoplasm has learned to avoid detection and avoid being attacked, which is called immune evasion. Malignant tumors, such as gastric cancer (GC), share the ability to evade the body's immune system as a defining feature. Immune evasion includes alterations to tumor-associated antigens (TAAs), antigen presentation mechanisms (APMs), and the tumor microenvironment (TME). While TAA and APM are simpler in nature, they both involve mutations or epigenetic regulation of genes. The TME is comprised of numerous cell types, cytokines, chemokines and extracellular matrix, any one of which might be altered to have an effect on the surrounding ecosystem. The NF-kB, MAPK, PI3K/AKT, JAK/STAT, Wnt/β-catenin, Notch, Hippo and TGF-β/Smad signaling pathways are all associated with gastric cancer tumor immune evasion. In this review, we will delineate the functions of these pathways in immune evasion.
Collapse
Affiliation(s)
| | | | | | | | - Xudong Wang
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
113
|
Lei ZN, Teng QX, Tian Q, Chen W, Xie Y, Wu K, Zeng Q, Zeng L, Pan Y, Chen ZS, He Y. Signaling pathways and therapeutic interventions in gastric cancer. Signal Transduct Target Ther 2022; 7:358. [PMID: 36209270 PMCID: PMC9547882 DOI: 10.1038/s41392-022-01190-w] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/14/2022] [Accepted: 09/07/2022] [Indexed: 11/23/2022] Open
Abstract
Gastric cancer (GC) ranks fifth in global cancer diagnosis and fourth in cancer-related death. Despite tremendous progress in diagnosis and therapeutic strategies and significant improvements in patient survival, the low malignancy stage is relatively asymptomatic and many GC cases are diagnosed at advanced stages, which leads to unsatisfactory prognosis and high recurrence rates. With the recent advances in genome analysis, biomarkers have been identified that have clinical importance for GC diagnosis, treatment, and prognosis. Modern molecular classifications have uncovered the vital roles that signaling pathways, including EGFR/HER2, p53, PI3K, immune checkpoint pathways, and cell adhesion signaling molecules, play in GC tumorigenesis, progression, metastasis, and therapeutic responsiveness. These biomarkers and molecular classifications open the way for more precise diagnoses and treatments for GC patients. Nevertheless, the relative significance, temporal activation, interaction with GC risk factors, and crosstalk between these signaling pathways in GC are not well understood. Here, we review the regulatory roles of signaling pathways in GC potential biomarkers, and therapeutic targets with an emphasis on recent discoveries. Current therapies, including signaling-based and immunotherapies exploited in the past decade, and the development of treatment for GC, particularly the challenges in developing precision medications, are discussed. These advances provide a direction for the integration of clinical, molecular, and genomic profiles to improve GC diagnosis and treatments.
Collapse
Affiliation(s)
- Zi-Ning Lei
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Qin Tian
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China
| | - Wei Chen
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China
| | - Yuhao Xie
- Institute for Biotechnology, St. John's University, Queens, NY, 11439, USA
| | - Kaiming Wu
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China
| | - Qianlin Zeng
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China
| | - Leli Zeng
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China.
| | - Yihang Pan
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
- Institute for Biotechnology, St. John's University, Queens, NY, 11439, USA.
| | - Yulong He
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China.
| |
Collapse
|
114
|
Penpulimab, an anti-PD1 IgG1 antibody in the treatment of advanced or metastatic upper gastrointestinal cancers. Cancer Immunol Immunother 2022; 71:2371-2379. [DOI: 10.1007/s00262-022-03160-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 01/19/2022] [Indexed: 12/24/2022]
|
115
|
Peri S, Ruzzolini J, Urciuoli S, Versienti G, Biagioni A, Andreucci E, Peppicelli S, Bianchini F, Bottari A, Calorini L, Nediani C, Magnelli L, Papucci L. An Oleocanthal-Enriched EVO Oil Extract Induces the ROS Production in Gastric Cancer Cells and Potentiates the Effect of Chemotherapy. Antioxidants (Basel) 2022; 11:antiox11091762. [PMID: 36139836 PMCID: PMC9495378 DOI: 10.3390/antiox11091762] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/26/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Oleocanthal, a minor polar compound in extra-virgin olive (EVO) oil, contains anticancer properties, which should be encouraged in its use in oncology. Gastric Cancer (GC), a very aggressive human cancer, is often diagnosed at advanced stages, when surgery is substituted or supported by chemotherapy (CT). However, CT frequently fails due to the patient’s resistance to the treatment. Thus, the aim of this study is to verify whether an OC-enriched EVO oil extract fraction (OCF) may be useful in order to overcome a resistance to GC. We evaluated the OCF effects on an AGS gastric adenocarcinoma cell line wild type (AGS wt) and on its subpopulations resistant to 5-fluorouracil (5FUr), Paclitaxel (TAXr) or cisplatin (CISr). We found that a 60 µM dose of the OCF acts on the AGS wt, 5FUr and TAXr, leading to the cell cycle inhibition and to a ROS production, but not on CISr cells. Resistance of CISr to the OCF seems to be due to higher levels of antioxidant-enzymes that can counteract the OCF-induced ROS production. Moreover, using the OCF plus 5-fluorouracil, Paclitaxel or cisplatin, we found a potentiating effect compared with a mono-treatment in all resistant GC cells, including CISr. In conclusion, the use of the OCF in the management of GC has shown very interesting advantages, opening-up the possibility to evaluate the efficacy of the OCF in vivo, as a valid adjuvant in the treatment of resistant GC.
Collapse
Affiliation(s)
- Sara Peri
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Jessica Ruzzolini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Silvia Urciuoli
- PHYTOLAB (Pharmaceutical, Cosmetic, Food Supplement, Technology and Analysis)-DiSIA, University of Florence, Via U. Schiff, 6, 50019 Florence, Italy
| | - Giampaolo Versienti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Andrea Bottari
- Digestive Surgery Unit, AOU Careggi University Hospital, 50134 Florence, Italy
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
- Center of Excellence for Research, Transfer, and High Education (DENOTHE), University of Florence, 50134 Florence, Italy
| | - Chiara Nediani
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
- Correspondence: ; Tel.: +39-0552751203
| | - Lucia Magnelli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Laura Papucci
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| |
Collapse
|
116
|
Xue C, Xu YH. Trastuzumab combined chemotherapy for the treatment of HER2-positive advanced gastric cancer: A systematic review and meta-analysis of randomized controlled trial. Medicine (Baltimore) 2022; 101:e29992. [PMID: 36042610 PMCID: PMC9410626 DOI: 10.1097/md.0000000000029992] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND This systematic review and meta-analysis aimed to assess the efficacy of trastuzumab combined with chemotherapy for the treatment in HER2-positive advanced gastric cancer (HER2-PAGC). METHODS This systematic review and meta-analysis was designed using randomized controlled trials that compared trastuzumab in combination with chemotherapy and chemotherapy alone. A comprehensive search was conducted in the following databases from their inception onwards: PubMed, EMBASE, Cochrane Library, WANGFANG, and CNKI. We also searched other literature sources to avoid missing relevant studies. Two reviewers independently performed all record selection, data collection, and methodological assessments. Any confusion was resolved by discussion or referral to a third reviewer. If there were ample data from eligible studies, we performed a fixed-effects meta-analysis. Whenever this was not possible, we conducted a narrative synthesis. RESULTS Meta-analysis results showed that trastuzumab in combination with chemotherapy achieved better outcomes on response rate (trastuzumab plus CFC vs CFC: odds ratio [OR] = 1.56, 95% confidence interval [CI] [1.17-2.09], I2 = 0%, P < .003; trastuzumab plus OT vs OT: OR = 2.97, 95% CI [1.74-5.09], I2 = 0%, P < .0001; and trastuzumab plus CC vs CC: OR = 2.62, 95% CI [1.84-3.73], I2 = 0%, P < .0001), and disease control rate (trastuzumab plus CFC vs CFC: OR = 1.61, 95% CI [1.17-2.21], I2 = 0%, P = .004; trastuzumab plus OT vs OT: OR = 4.29, 95% CI [2.33-7.90], I2 = 0%, P < .0001; and trastuzumab plus CC vs CC: OR = 2.99, 95% CI [1.99-4.48], I2 = 0%, P < .0001). However, there were no significant differences in the adverse events. CONCLUSIONS The results of this study revealed that the efficacy of trastuzumab combined with chemotherapy was superior to that of chemotherapy alone for the treatment of HER2-PAGC. The 2 modalities showed similar safety profiles.
Collapse
Affiliation(s)
- Chuan Xue
- Department of Gastroenterology, Sunshine Union Hospital, Weifang, China
| | - Yong-Hong Xu
- Department of Gastroenterology, Sunshine Union Hospital, Weifang, China
- *Correspondence: Yong-Hong Xu, MM, Department of Gastroenterology, Sunshine Union Hospital, No. 9000 Yingqian Street, High-tech District, Weifang, Shandong Province 261061, China (e-mail: )
| |
Collapse
|
117
|
Guo L, Qin X, Xue L, Yang JY, Zhang Y, Zhu S, Ye G, Tang R, Yang W. A novel form of docetaxel polymeric micelles demonstrates anti-tumor and ascites-inhibitory activities in animal models as monotherapy or in combination with anti-angiogenic agents. Front Pharmacol 2022; 13:964076. [PMID: 36091776 PMCID: PMC9449419 DOI: 10.3389/fphar.2022.964076] [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: 06/08/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022] Open
Abstract
Malignant ascites (MA) is caused by intraperitoneal spread of solid tumor cells and results in a poor quality of life. Chemotherapy is a common first-line treatment for patients with MA. Taxotere ® (DTX) is widely used in solid tumor therapies. However, the low water solubility and side effects caused by additives in the formulation restrict the clinical application of docetaxel. HT001 is a clinical stage docetaxel micelle developed to overcome the solubility issue with improved safety profiles. To support clinical development and expand clinical application of HT001, this study used in vitro and in vivo approaches to investigate the anti-tumor effects of HT001 when applied as monotherapy or in combination with anti-angiogenic agents. HT001 demonstrated comparable anti-proliferative activities as docetaxel in a broad range of cancer cell lines in vitro. Furthermore, HT001 suppressed tumor growth in a dose-dependent manner in A549, MCF-7, and SKOV-3 xenograft tumor mouse models in vivo. In a hepatocellular carcinoma H22 malignant ascites-bearing mouse model, HT001 presented a dose-dependent inhibition of ascites production, prolonged animal survival, and reduced VEGF levels. When dosed at 20 mg/kg, the HT001-treated group exhibited curative results, with no ascites formation in 80% of mice at the end of the study while all the mice in the vehicle control group succumbed. Similar results were obtained in HT001 treatment of mice bearing malignant ascites produced by human ovarian cancer ES-2 cells. Notably, the combination of HT001 with Endostar not only significantly reduced ascites production but also prolonged survival of H22 ascites-bearing mice. HT001 showed similar PK and tissue distribution profiles as DTX in non-rodent hosts. Collectively, these results demonstrate potent anti-tumor activity of HT001 in multiple solid tumor models or malignant ascites models, and reveal synergistic effects with anti-angiogenic agents, supporting the clinical development and clinical expansion plans for HT001.
Collapse
Affiliation(s)
- Leilei Guo
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Xiaokang Qin
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Liting Xue
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Janine Y. Yang
- Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Yumei Zhang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shunwei Zhu
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Gang Ye
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Renhong Tang
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
- *Correspondence: WenQing Yang, ; Renhong Tang,
| | - WenQing Yang
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
- *Correspondence: WenQing Yang, ; Renhong Tang,
| |
Collapse
|
118
|
Wang Z, He T, Yu D, Qin X, Geng A, Yang H. Neoadjuvant apatinib plus tegafur/gimeracil/oteracil potassium (S‑1)/oxaliplatin chemotherapy vs. chemotherapy alone in patients with locally advanced gastric carcinoma. Exp Ther Med 2022; 24:625. [PMID: 36160880 PMCID: PMC9468841 DOI: 10.3892/etm.2022.11562] [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: 03/23/2022] [Accepted: 05/20/2022] [Indexed: 02/03/2023] Open
Abstract
The current study aimed to evaluate the efficacy and safety of neoadjuvant apatinib plus tegafur/gimeracil/oteracil potassium (S-1) plus oxaliplatin (SOX) chemotherapy in patients with locally advanced gastric carcinoma (LAGC). Therefore, patients with LAGC treated with neoadjuvant apatinib plus SOX chemotherapy (apatinib + SOX group; n=25) or SOX chemotherapy (SOX group; n=35) were enrolled in the present study. Subsequently, the objective response (ORR) and disease control rates (DCR), pathological response, disease-free survival (DFS), overall survival (OS) and adverse events were recorded. The results showed that patients in the apatinib + SOX group exhibited a higher ORR (64.0 vs. 37.1%; P=0.040), but a similar DCR (96.0 vs. 88.6%; P=0.580), compared with those in the SOX group. The pathological response rates in patients with grade 0, 1, 2 and 3 LAGC were 0.0, 20.8, 62.5 and 16.7%, respectively, in the apatinib + SOX group, while in those treated with SOX alone they were 9.1, 39.4, 42.4 and 9.1%, respectively. By contrast, the pathological response was elevated in the apatinib + SOX group compared with the SOX group (P=0.030). During a median follow-up period of 21.0 months (range, 6.4-38.1 months), median DFS and OS were not reached. More specifically, the 1-, 2- and 3-year DFS rates were 91.7, 75.2 and 75.2% in the apatinib + SOX group and 71.8, 59.6 and 44.7% in the SOX group, respectively. In addition, the 1-, 2- and 3-year OS rates were 100.0, 89.6 and 78.4% in the apatinib + SOX group, while those in the SOX group were 90.3, 69.2 and 55.4%, respectively. However, no differences in DFS (P=0.094) or OS (P=0.155) were observed between the two groups. Additionally, the most common adverse events in the SOX group were mild leukopenia (42.9%) and fatigue (34.3%), while those in the apatinib + SOX group were tolerable leukopenia (44.0%) and hypertension (44.0%). In conclusion, the present study suggested that neoadjuvant apatinib plus SOX chemotherapy could be more effective and tolerable compared with SOX chemotherapy alone in patients with LAGC.
Collapse
Affiliation(s)
- Zhenfeng Wang
- Department of General Surgery, The Second People's Hospital of Liaocheng, Linqing, Liaocheng, Shandong 252699, P.R. China
| | - Tingbang He
- Department of General Surgery, The People's Hospital of Xiajin Affiliated to Shandong First Medical University, Xiajin, Dezhou, Shandong 253299, P.R. China
| | - Deguo Yu
- Department of Emergency Surgery, The Second People's Hospital of Liaocheng, Linqing, Liaocheng, Shandong 252699, P.R. China
| | - Xiantao Qin
- Department of General Surgery, The Second People's Hospital of Liaocheng, Linqing, Liaocheng, Shandong 252699, P.R. China
| | - Aizhi Geng
- Department of Gynecology, The Second People's Hospital of Liaocheng, Linqing, Liaocheng, Shandong 252699, P.R. China
| | - Hailei Yang
- Department of Gynecology, The Second People's Hospital of Liaocheng, Linqing, Liaocheng, Shandong 252699, P.R. China
| |
Collapse
|
119
|
Liu ZH, Zhu BW, Shi M, Qu YR, He XJ, Yuan HL, Ma J, Li W, Zhao DD, Liu ZC, Wang BM, Wang CY, Tao HQ, Ma TH. Profiling of gene fusion involving targetable genes in Chinese gastric cancer. World J Gastrointest Oncol 2022; 14:1528-1539. [PMID: 36160735 PMCID: PMC9412921 DOI: 10.4251/wjgo.v14.i8.1528] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/14/2022] [Accepted: 07/19/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Approximately half of all new cases of gastric cancer (GC) and related deaths occur in China. More than 80% of patients with GC are diagnosed at an advanced stage, which results in poor prognosis. Although HER2-directed therapy and immune checkpoint inhibitors have been somewhat successful, new drugs are still needed for the treatment of GC. Notably, several gene fusion-targeted drugs have been approved by the United States Food and Drug Administration for solid tumors, including GC, such as larotrectinib for NTRK fusion-positive cancers and zenocutuzumab for NRG1 fusion-positive cancers. However, gene fusions involving targetable genes have not been well characterized in Chinese patients with GC.
AIM To identify the profile of fusions involving targetable genes in Chinese patients with GC using clinical specimens and determine the distribution of patients with gene fusion variants among the molecular subtypes of GC.
METHODS We retrospectively analyzed gene fusion events in tumor tissue samples from 954 Chinese patients with GC. Clinicopathological characteristics were obtained from their medical records. Genetic alterations, such as single nucleotide variants, indels, amplifications, and gene fusions, were identified using a targeted sequencing panel containing 825 genes. Fusions were validated by fluorescence in situ hybridization (FISH) using break-apart probes. The microsatellite instability (MSI) status was evaluated using MSIsensor from the targeted sequencing panel data. Tumor mutational burden (TMB) was calculated using the total number of nonsynonymous mutations divided by the total genomic targeted region. Chi-square analysis was used to determine the enrichment of gene fusions associated with the molecular subtypes of GC.
RESULTS We found that 1.68% (16/954) of patients harbored 20 fusion events involving targetable genes. RARA fusions (n = 5) were the most common, followed by FGFR2, BRAF, MET, FGFR3, RET, ALK, EGFR, NTRK2, and NRG1 fusions. Two of the RARA fusions, EML4-ALK (E6:E20) and EGFR-SEPTIN14 (E7:E10), have been identified in other tumors but not in GC. Surprisingly, 18 gene fusion events were previously not reported in any cancer types. Twelve of the eighteen novel gene fusions included complete exons encoding functional domains of targetable genes, such as the tyrosine kinase domain of receptor tyrosine kinases and the DNA- and ligand-binding domains of RARA. Consistent with the results of detection using the targeted sequencing fusion panel, the results of FISH (fluorescence in situ hybridization) confirmed the rearrangement of FGFR2 and BRAF in tumors from patients 04 and 09, respectively. Genetic analysis indicated that the fusion genes were significantly enriched in patients with ERBB2 amplification (P = 0.02); however, there were no significant differences between fusion-positive and fusion-negative patients in age, sex, MSI status, and TMB.
CONCLUSION We characterized the landscape of fusions involving targetable genes in a Chinese GC cohort and found that 1.68% of patients with GC harbor potential targetable gene fusions, which were enriched in patients with ERBB2 amplification. Gene fusion detection may provide a potential treatment strategy for patients with GC with disease progression following standard therapy.
Collapse
Affiliation(s)
- Zhen-Hua Liu
- Department of Medical Oncology, Fujian Medical University, Fuzhou 350001, Fujian Province, China
| | - Bo-Wen Zhu
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Min Shi
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Yu-Rong Qu
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Xun-Jun He
- Department of Genetics and Genomic Medicine, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Hong-Ling Yuan
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Jie Ma
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Wei Li
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Dan-Dan Zhao
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Zheng-Chuang Liu
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
- Department of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Bao-Ming Wang
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Chun-Yang Wang
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Hou-Quan Tao
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
- Department of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Tong-Hui Ma
- Department of Translational Medicine, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| |
Collapse
|
120
|
Wang M, Xie C. DNA Damage Repair and Current Therapeutic Approaches in Gastric Cancer: A Comprehensive Review. Front Genet 2022; 13:931866. [PMID: 36035159 PMCID: PMC9412963 DOI: 10.3389/fgene.2022.931866] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/15/2022] [Indexed: 11/23/2022] Open
Abstract
DNA in cells is frequently damaged by endogenous and exogenous agents. However, comprehensive mechanisms to combat and repair DNA damage have evolved to ensure genomic stability and integrity. Improper DNA damage repair may result in various diseases, including some types of tumors and autoimmune diseases. Therefore, DNA damage repair mechanisms have been proposed as novel antitumor drug targets. To date, numerous drugs targeting DNA damage mechanisms have been developed. For example, PARP inhibitors that elicit synthetic lethality are widely used in individualized cancer therapies. In this review, we describe the latent DNA damage repair mechanisms in gastric cancer, the types of DNA damage that can contribute to the development of gastric cancer, and new therapeutic approaches for gastric cancer that target DNA damage repair pathways.
Collapse
Affiliation(s)
| | - Chuan Xie
- Department of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
121
|
HIF in Gastric Cancer: Regulation and Therapeutic Target. Molecules 2022; 27:molecules27154893. [PMID: 35956843 PMCID: PMC9370240 DOI: 10.3390/molecules27154893] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
HIF means hypoxia-inducible factor gene family, and it could regulate various biological processes, including tumor development. In 2021, the FDA approved the new drug Welireg for targeting HIF-2a, and it is mainly used to treat von Hippel-Lindau syndrome, which demonstrated its good prospects in tumor therapy. As the fourth deadliest cancer worldwide, gastric cancer endangers the health of people all across the world. Currently, there are various treatment methods for patients with gastric cancer, but the five-year survival rate of patients with advanced gastric cancer is still not high. Therefore, here we reviewed the regulatory role and target role of HIF in gastric cancer, and provided some references for the treatment of gastric cancer.
Collapse
|
122
|
Abdominal Computed Tomography Enhanced Image Features under an Automatic Segmentation Algorithm in Identification of Gastric Cancer and Gastric Lymphoma. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:2259373. [PMID: 35928973 PMCID: PMC9345719 DOI: 10.1155/2022/2259373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 11/17/2022]
Abstract
To analyze the application value of CT-enhanced scanning based on artificial intelligence algorithm in the diagnosis of gastric cancer and gastric lymphoma, the CT images of 80 patients with Borrmann type IV gastric cancer or primary gastric lymphoma diagnosed by endoscopic pathology were retrospectively collected. Meanwhile, a lymph node recognition algorithm based on OTSU threshold segmentation was proposed for CT image processing. The results showed that the missed diagnosis rate of suspected lymph nodes and the missed lymph node detection rate of this algorithm were substantially lower than those of other algorithms (P < 0.05). The probability of gastric wall motility disappearance, perigastric fat infiltration, and type A enhancement pattern in the Borrmann type IV gastric cancer group was higher than that in the gastric lymphoma group, with remarkable differences (P < 0.05). There was no remarkable difference between the Borrmann type IV gastric cancer group and the gastric lymphoma group in the probability of swollen lymph nodes under the renal hilum (P > 0.05). In addition, 5the sensitivity (83.17%), specificity (95.52%), and accuracy (93.08%) of the combined detection of the three CT signs (stomach wall motility, perigastric fat infiltration, and enhancement mode) were substantially improved compared with those of a single sign (P < 0.05). To sum up, the lymph node recognition algorithm based on OTSU threshold segmentation had better performance in detecting gastric lymph nodes than traditional algorithms. The CT image characteristics of gastric wall motility, perigastric fat infiltration, and enhancement pattern based on artificial intelligence algorithms were effective indicators for distinguishing gastric cancer and gastric lymphoma.
Collapse
|
123
|
Genomically Silent Refractory Gastric Cancer in a Young Patient Exhibits Overexpression of CXCL5. Curr Oncol 2022; 29:4725-4733. [PMID: 35877235 PMCID: PMC9320515 DOI: 10.3390/curroncol29070375] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 12/16/2022] Open
Abstract
Gastric cancer is the third leading cause of cancer-related deaths, with more than one million new cases and approximately 841,000 deaths annually worldwide. We report a case of a young patient (25 years old) with an aggressive form of gastric cancer. The patient had previously been treated for Helicobacter pylori (H. pylori), which is a main risk factor for developing gastric cancer. Genetic testing showed an E-cadherin (CDH1) germline mutation of unknown significance. After eight cycles of chemotherapy, a positron emission tomography (PET) scan showed disease progression with an enlarging hypermetabolic right adnexal mass suspicious for metastatic disease. Tumor pathology demonstrated invasive and poorly differentiated gastric carcinoma. The analysis of the tumor biopsy indicated the very high expression of a chemokine, C-X-C motif chemokine 5 (CXCL5). The combination of H. pylori infection with an existence of a rare CDH1 mutation could have contributed to this aggressive gastric cancer.
Collapse
|
124
|
Zhang J, Gao M, Niu Y, Sun J. Identification of a Novel Ferroptosis Inducer for Gastric Cancer Treatment Using Drug Repurposing Strategy. Front Mol Biosci 2022; 9:860525. [PMID: 35860356 PMCID: PMC9289365 DOI: 10.3389/fmolb.2022.860525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 06/07/2022] [Indexed: 01/10/2023] Open
Abstract
Gastric cancer remains one of the major contributors to global cancer mortality, although there is no promising target drug in clinics. Hence, the identification of novel targeted drugs for gastric cancer is urgent. As a promising strategy for inducing ferroptosis for gastric cancer treatment, the ferroptosis inducer is a potential drug. Nevertheless, no ferroptosis inducer has entered clinics. So, our purpose was to identify a novel ferroptosis inducer for gastric cancer treatment using a drug repurposing strategy. Firstly, using a drug repurposing strategy with the aid of a commercialized compound library, HC-056456, a small molecule bioactive CatSper channel blocker, was characterized to inhibit the growth of gastric cancer line MGC-803. At the same time, this anti-proliferation effect can be blocked by ferrostatin-1, a ferroptosis inhibitor, indicating that HC-056456 is a ferroptosis inducer. Then, HC-056456 was identified to decrease GSH content via p53/SLC7A11 signaling pathway. Then Fe2+ and lipid peroxide were accumulated when cells were exposed to HC-056456. Finally, HC-056456 was found to suppress the growth of gastric cancer cells by increasing p53 and repressing SLC7A11 in vivo but not in the presence of ferrostatin-1. In sum, we systematically elucidate that HC-056456 exerts anti-gastric cancer effect by provoking ferroptosis in vitro and in vivo, suggesting its potential role in gastric cancer treatment.
Collapse
Affiliation(s)
- Jinping Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meimei Gao
- Henan Key Laboratory of Precision Clinical Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying Niu
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiangang Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
125
|
Sun W, Wang X, Wang D, Lu L, Lin H, Zhang Z, Jia Y, Nie X, Liu T, Fu W. CD40×HER2 bispecific antibody overcomes the CCL2-induced trastuzumab resistance in HER2-positive gastric cancer. J Immunother Cancer 2022; 10:jitc-2022-005063. [PMID: 35851310 PMCID: PMC9295658 DOI: 10.1136/jitc-2022-005063] [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] [Accepted: 06/21/2022] [Indexed: 02/06/2023] Open
Abstract
Background There was much hard work to study the trastuzumab resistance in HER2-positive gastric cancer (GC), but the information which would reveal this abstruse mechanism is little. In this study, we aimed to investigate the roles of tumor cell-derived CCL2 on trastuzumab resistance and overcome the resistance by treatment with the anti-CD40-scFv-linked anti-HER2 (CD40 ×HER2) bispecific antibody (bsAb). Methods We measured the levels of CCL2 expression in HER2-positive GC tissues, and revealed biological functions of tumor cell-derived CCL2 on tumor-associated macrophages (TAMs) and the trastuzumab resistance. Then, we developed CD40 ×HER2 bsAb, and examined the targeting roles on HER2 and CD40, to overcome the trastuzumab resistance without systemic toxicity. Results We found the level of CCL2 expression in HER2-postive GC was correlated with infiltration of TAMs, polarization status of infiltrated TAMs, trastuzumab resistance and survival outcomes of GC patients. On exposure to CCL2, TAMs decreased the M1-like phenotype, thereby eliciting the trastuzumab resistance. CCL2 activated the transcription of ZC3H12A, which increased K63-linked deubiquitination and K48-linked auto-ubiquitination of TRAF6/3 to inactivate NF-κB signaling in TAMs. CD40 ×HER2 bsAb, which targeted the CD40 to restore the ubiquitination level of TRAF6/3, increased the M1-like phenotypic transformation of TAMs, and overcame trastuzumab resistance without immune-related adversary effects (irAEs). Conclusions We revealed a novel mechanism of trastuzumab resistance in HER2-positive GC via the CCL2-ZC3H12A-TRAF6/3 signaling axis, and presented a CD40 ×HER2 bsAb which showed great antitumor efficacy with few irAEs.
Collapse
Affiliation(s)
- Weilin Sun
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xi Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Daohan Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Lu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hai Lin
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhaoxiong Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yangpu Jia
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinyang Nie
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Tong Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Weihua Fu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
126
|
Liu G, Xu X, Geng H, Li J, Zou S, Li X. FGA inhibits metastases and induces autophagic cell death in gastric cancer via inhibiting ITGA5 to regulate the FAK/ERK pathway. Tissue Cell 2022; 76:101767. [DOI: 10.1016/j.tice.2022.101767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/23/2022] [Accepted: 02/22/2022] [Indexed: 11/28/2022]
|
127
|
Li TC, Chen NJ, Chen YY, He BJ, Zhou ZF. Solasonine induces apoptosis of the SGC-7901 human gastric cancer cell line in vitro via the mitochondria-mediated pathway. J Cell Mol Med 2022; 26:3387-3395. [PMID: 35524577 PMCID: PMC9189336 DOI: 10.1111/jcmm.17343] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 12/24/2022] Open
Abstract
Solasonine, a steroidal glycoalkaloid isolated from the herbal plant Solanum nigrum Linn., has shown active against multiple human cancers; however, there is little knowledge on the activity of solasonine against gastric cancer until now. This study aimed to examine the effect of solasonine on the biological behaviours of human gastric cancer SGC‐7901 cells. The results showed that solasonine suppressed SGC‐7901 cell proliferation in a dose‐dependent manner. Solasonine treatment mainly induced the cell cycle arrest at G2 phase in SGC‐7901 cells. Treatment with solasonine resulted in significant down‐regulation of Bcl‐2 and Caspase‐3 protein expression and reduced Bax and Bcl‐xL protein expression in SGC‐7901 cells. Solasonine shows a comparable inhibitory effect on the proliferation of human gastric cancer SGC‐7901 cells with cisplatin, and solasonine induces of SGC‐7901 cell apoptosis through triggering the endoplasmic reticulum stress pathway and the mitochondrial pathway. Our data indicate that solasonine may be a promising agent for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Tian-Chuan Li
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Department of Integrated Traditional and Western Medicine, Fujian Medical University Cancer Hospital & Fujian Provincial Cancer Hospital, Fuzhou, China
| | - Nai-Jie Chen
- Department of Integrated Traditional and Western Medicine, Fujian Medical University Cancer Hospital & Fujian Provincial Cancer Hospital, Fuzhou, China
| | - Yun-Ying Chen
- Department of Integrated Traditional and Western Medicine, Fujian Medical University Cancer Hospital & Fujian Provincial Cancer Hospital, Fuzhou, China
| | - Bing-Jing He
- Department of Integrated Traditional and Western Medicine, Fujian Medical University Cancer Hospital & Fujian Provincial Cancer Hospital, Fuzhou, China
| | - Zhi-Feng Zhou
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, China.,Laboratory of Immuno-Oncology, Fujian Medical University Cancer Hospital & Fujian Provincial Cancer Hospital, Fuzhou, China
| |
Collapse
|
128
|
Wu H, Miao X, Liu Y, Zhang S, Li C, Hao J. Clinical Efficacy of Modified Yiwei Shengyang Decoction Combined with FOLFOX4 Chemotherapy Regimen in the Treatment of Advanced Gastric Cancer and Its Effect on Tumor Marker Levels. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:6234032. [PMID: 35571732 PMCID: PMC9098293 DOI: 10.1155/2022/6234032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/28/2022] [Accepted: 04/08/2022] [Indexed: 11/18/2022]
Abstract
Objective To study the clinical efficacy of modified Yiwei Shengyang decoction combined with FOLFOX4 chemotherapy regimen in the treatment of advanced gastric cancer and its effect on tumor marker levels. Methods A total of 106 patients with advanced gastric cancer who were treated in our hospital from September 2019 to September 2021 were recruited and assigned via random number allocation to receive either FOLFOX4 chemotherapy (control group) or modified Yiwei Shengyang decoction plus FOLFOX4 chemotherapy (observation group). Outcome measures included clinical efficacy and tumor marker levels. Results Modified Yiwei Shengyang decoction plus FOLFOX4 chemotherapy was associated with a significantly higher efficacy (86.79%, including 22 (41.51%) cases of complete response (CR), 24 (45.28%) cases of partial response (PR), 6 (11.32%) cases of stable disease (SD), and 1 (1.89%) case of progressive disease (PD)) compared to FOLFOX4 chemotherapy alone (47.16%, including 10 (18.87%) cases of CR, 15 (28.30%) cases of PR, 21 (39.62%) cases of SD, and 7 (13.21%) cases of PD) (P < 0.05). There was no significant difference in the levels of CEA and CA19-9 between the two groups before treatment (P > 0.05). Modified Yiwei Shengyang decoction plus FOLFOX4 chemotherapy resulted in significantly lower levels of carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9) (2.08 ± 0.47, 15.12 ± 6.74) compared to FOLFOX4 chemotherapy alone (5.46 ± 1.84, 31.82 ± 7.48) (P < 0.05). Conclusion Modified Yiwei Shengyang decoction plus FOLFOX4 chemotherapy regimen is effective in the treatment of advanced gastric cancer. It regulates the levels of various serum tumor markers in patients and controls the disease, so it is worthy of clinical application and promotion.
Collapse
Affiliation(s)
- Hongying Wu
- National Physician Hall, Cangzhou Central Hospital, Cangzhou, China
| | - Xiaomei Miao
- National Physician Hall, Cangzhou Central Hospital, Cangzhou, China
| | - Yan Liu
- Department of Hepatopancreatobiliary Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Shu Zhang
- National Physician Hall, Cangzhou Central Hospital, Cangzhou, China
| | - Chaohui Li
- National Physician Hall, Cangzhou Central Hospital, Cangzhou, China
| | - Jie Hao
- Department of Oncology, Cangzhou Central Hospital, Cangzhou, China
| |
Collapse
|
129
|
Qing X, Xu W, Liu S, Chen Z, Ye C, Zhang Y. Molecular Characteristics, Clinical Significance, and Cancer Immune Interactions of Angiogenesis-Associated Genes in Gastric Cancer. Front Immunol 2022; 13:843077. [PMID: 35273618 PMCID: PMC8901990 DOI: 10.3389/fimmu.2022.843077] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/03/2022] [Indexed: 12/21/2022] Open
Abstract
Background Immunotherapy has evolved as a critical option to treat diverse cancers. The active response to immunotherapy relies on the unique interaction between cancer and the tumor microenvironment (TME). Angiogenesis is one of the hallmarks of cancer. However, the association between angiogenesis and clinical outcome, immune cell infiltration, and immunotherapy remains unknown in gastric cancer (GC). Methods We systematically assessed 36 angiogenesis-associated genes (AAGs) and comprehensively identified the correlation between angiogenesis and transcriptional patterns, prognosis, and immune cell infiltration. The AAG_score was applied to quantify the angiogenesis subtypes of each patient. We then evaluated their values in prognostic prediction and therapeutic responses in GC. Results We discussed the mutations of AAGs in GC specimens from genetic levels and identified their expression patterns from TCGA and GEO cohorts. We determined two different molecular subtypes and observed that AAG mutations were related to patients’ clinicopathological characteristics, prognosis, and infiltrating TME. Next, an AAG_score for predicting overall survival (OS) was established and its reliable predictive ability in GC patients was confirmed. Furthermore, we created a highly reliable nomogram to facilitate the clinical viability of the AAG_score. A low AAG_score, characterized by elevated microsatellite instability-high, mutation burden, and immune activation, demonstrated a superior OS. Additionally, the AAG_score was remarkedly correlated with the cancer stem cell index and drug susceptibility. Conclusion Collectively, we identified a prognostic AAG signature for GC patients. This signature may contribute to clarifying the characteristics of TME and enable the exploration of more potent immunotherapy strategies.
Collapse
Affiliation(s)
- Xin Qing
- School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Wenjing Xu
- School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Shengli Liu
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhencheng Chen
- School of Life and Environmental Sciences, Guilin University of Electronic Technology, Guilin, China
| | - Chunping Ye
- Department of Obstetrics and Gynecology, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Yewei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
130
|
Chang C, Zheng A, Wang P, Teng X. Circular RNA mitochondrial translation optimization 1 correlates with less lymph node metastasis, longer disease-free survival, and higher chemotherapy sensitivity in gastric cancer. J Clin Lab Anal 2022; 36:e23918. [PMID: 35478417 PMCID: PMC9169224 DOI: 10.1002/jcla.23918] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
Objective Circular‐mitochondrial translation optimization 1 (circ‐MTO1) inhibits the progression of gastric cancer by regulating the growth, apoptosis, and invasion of tumor cells. However, its clinical potential as a biomarker for gastric cancer remains to be further evaluated. This study aimed to assess circ‐MTO1 expression and its correlation with clinical features and prognosis in gastric cancer patients, as well as the effect of circ‐MTO1 on the sensitivity to chemotherapy in gastric cancer cells. Methods Circ‐MTO1 in tumor and adjacent tissues of 97 gastric cancer patients undergoing resection was examined by reverse transcription‐quantitative polymerase chain reaction. HGC‐27 and NCI‐N87 cells transfected by circ‐MOT1 overexpression plasmid (OE‐circ‐MOT1) and negative control (OE‐NC) were treated with 0‒6.4 μM oxaliplatin. Relative cell viability was detected using Cell Counting Kit‐8. Results Circ‐MTO1 was insufficiently expressed in gastric tumor tissue (median (interquartile range): 0.403 (0.288‒0.518)) compared with adjacent tissue (median (interquartile range): 1.000 (0.715‒1.524)) (p < 0.001). Besides, tumor circ‐MTO1 was correlated with less lymph node metastasis (p = 0.014) and low TNM stage (p = 0.039), while was not correlated with demographic features or other clinical characteristics (all p > 0.05). Furthermore, tumor circ‐MTO1 high expression was independently correlated with prolonged disease‐free survival (DFS) (p = 0.013, adjusted hazard ratio (95% confidential interval): 0.314 (0.126‒0.782)), but was not correlated with overall survival (p > 0.05). Lastly, in gastric cancer cells, OE‐circ‐MTO1 apparently decreased relative cell viabilities at oxaliplatin concentrations of 0.4, 0.8, 1.6, and 3.2 μM (all p < 0.05). Conclusion Circ‐MTO1 correlates with less lymph node metastasis, prolonged DFS, and improved chemotherapy sensitivity in gastric cancer.
Collapse
Affiliation(s)
- Cheng Chang
- Department of Gastroenterology, Wuhan Hospital of Traditional Chinese medicine, Wuhan, China.,Department of Gastroenterology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University,Edong Healthcare Group, Huangshi, China
| | - Anrui Zheng
- Department of Gastroenterology, Wuhan Hospital of Traditional Chinese medicine, Wuhan, China
| | - Pinfa Wang
- Department of Gastroenterology, Wuhan Hospital of Traditional Chinese medicine, Wuhan, China
| | - Xiaojun Teng
- Department of Gastroenterology, Wuhan Hospital of Traditional Chinese medicine, Wuhan, China
| |
Collapse
|
131
|
Zeng Y, Zhang X, Li F, Wang Y, Wei M. AFF3 is a novel prognostic biomarker and a potential target for immunotherapy in gastric cancer. J Clin Lab Anal 2022; 36:e24437. [PMID: 35478418 PMCID: PMC9169183 DOI: 10.1002/jcla.24437] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/25/2022] [Accepted: 04/04/2022] [Indexed: 12/26/2022] Open
Abstract
Background Gastric cancer (GC) is one of the most common cancers worldwide with a poor prognosis. The tumor microenvironment (TME) serves a pivotal role in affecting the prognosis and efficacy of immunotherapy. Given the poor prognosis of GC patients and the limitation of immunotherapy, we urged to identify new prognostic and immunotherapeutic biomarkers. Methods The transcriptome data were downloaded from the TCGA, GEO, and GEPIA databases, and performed differential analysis of AFF3 in tumor samples and normal samples. The UALCAN, Kaplan–Meier plotter and GEPIA databases were employed to assess the correlation of AFF3 with clinicopathological characteristics and prognosis. The potential mechanism of AFF3 was explored by the GO and KEGG enrichment. The potential role of AFF3 on tumor‐infiltrating immune cells (TIICs) was explored by TIMER2.0 and TISIDB. TIMER2.0 and SangerBox3.0 databases were, respectively, used to determine the correlation of AFF3 with immune checkpoint (ICs), tumor mutational burden (TMB), and microsatellite instability (MSI) in GC. Results We found significant downregulation of AFF3 in GC tissues as compared with normal tissues. However, GC patients having a higher expression of AFF3 were found to have worse clinicopathological characteristics and prognosis. Moreover, the GO enrichment analysis illustrated that AFF3 might regulate the immune cells in the TME. In addition, the AFF3 was positively correlated with TIICs, ICs, TMB, and MSI. Conclusion Here, we conclude that AFF3 may be a promising potential marker for the diagnosis and prognosis of GC patients, and may influence response to ICIs by affecting TIICs and ICs expression in the TME.
Collapse
Affiliation(s)
- Yuling Zeng
- Department of Blood Transfusion, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou City, China
| | - Xueping Zhang
- Department of Hepatobiliary Surgery, Zhengzhou Central Hospital Affiliated of Zhengzhou University, Zhengzhou City, China
| | - Fazhan Li
- Marshall Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou City, China
| | - Ying Wang
- Department of Blood Transfusion, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou City, China
| | - Ming Wei
- Department of Blood Transfusion, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou City, China
| |
Collapse
|
132
|
Liu H, Wang H, Dong A, Huo X, Wang H, Wang J, Si J. The Inhibition of Gastric Cancer Cells’ Progression by 23,24-Dihydrocucurbitacin E through Disruption of the Ras/Raf/ERK/MMP9 Signaling Pathway. Molecules 2022; 27:molecules27092697. [PMID: 35566048 PMCID: PMC9100127 DOI: 10.3390/molecules27092697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/11/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is considered to be one of the most common causes of cancer death worldwide due to its high recurrence and metastasis rates. The molecule 23,24-Dihydrocucurbitacin E (DHCE) is a cucurbitacin-derived tetracyclic triterpenoid compound that has anti-tumor activity, but the exact mechanism remains to be elucidated. This research aimed to explore the effects of DHCE on human gastric cancer cells and the possible mechanisms. The results showed that DHCE suppressed proliferation, migration, and invasion of gastric cancer cells, as well as induced apoptosis and G2/M phase arrest. Mechanistically, the potential targets and pathways of DHCE were predicted using database screening and verified using a molecular docking study, fluorescence staining, and Western blot. The results indicated that DHCE obviously inhibited the kinase activity of ERK2 via targeting its ATP-binding domain, destroyed F-actin microfilament, and reduced the expression levels of Ras, p-c-Raf, ERK, p-ERK, and MMP9 proteins. Collectively, our study demonstrated that DHCE suppressed gastric cancer cells’ proliferation, migration, and invasion through targeting ERK2 and disrupting the Ras/Raf/ERK/MMP9 signaling pathway. These properties make DHCE a promising candidate drug for the further design and development of novel and effective Ras/Raf/ERK/MMP9 pathway inhibitors for treating gastric cancer.
Collapse
|
133
|
Su PF, Yu JC. Progress in neoadjuvant therapy for gastric cancer (Review). Oncol Lett 2022; 23:172. [PMID: 35497934 PMCID: PMC9019865 DOI: 10.3892/ol.2022.13292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/28/2022] [Indexed: 02/03/2023] Open
Affiliation(s)
- Peng-Fei Su
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Jian-Chun Yu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
134
|
Effect of Apatinib Combined with Seggio on the Expression of Serum AFP and CA724 and Long-Term Survival Rate in Patients with Advanced Gastric Cancer Undergoing Comfortable Nursing Intervention. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:2004973. [PMID: 35432838 PMCID: PMC9010191 DOI: 10.1155/2022/2004973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/04/2022] [Accepted: 03/11/2022] [Indexed: 11/17/2022]
Abstract
Objective To study the effect of apatinib combined with seggio on the expression of serum AFP and CA724 and the long-term survival rate in advanced gastric cancer patients undergoing comfort nursing intervention. Methods 98 advanced gastric cancer patients were divided into single-drug group and joint group. Both groups of patients were given comfort nursing intervention, the single-drug group was treated with seggio, and the joint group was treated with apatinib and seggio. The clinical efficacy, survival rate, relationship between the tumor markers and the survival time, serum tumor markers levels (CA724 and AFP), inflammatory factors (IL-4, IL-10) levels, quality-of-life scores, and immunity function were measured after treatment. Results The clinical efficacy in the joint group was better than that in the single-drug group. The three-year survival time in the joint group was upregulated relative to the single-drug group. The patients with high expression of CA724 or AFP had a lower survival time than the patients with low expression of CA724 or AFP. After treatment, IL-10 and IL-4 levels were obviously decreased, and the joint group showed a more obvious decrease compared with the single-drug group. The quality-of-life scores were significantly upregulated after treatment, and compared with the joint group, the scores in the single drug-group were obviously higher. The CD4+/CD8+, CD4+, and CD3+ levels were increased, while CD8+ levels were decreased after treatment, and the changes of each index in the joint group were more significant than those in the single-drug group. The content of CA724 and AFP were significantly decreased after treatment, and the joint group showed a more significant decrease than the single-drug group. Conclusion Apatinib combined with seggio for advanced gastric cancer patients' treatment based on comfort nursing intervention can improve the clinical efficacy and survival time, reduce inflammatory factors and serum tumor markers levels, enhance patients' immune function, and quality of life.
Collapse
|
135
|
Zhou Y, Zhang Q, Liao B, Qiu X, Hu S, Xu Q. Circ_0006089 promotes gastric cancer growth, metastasis, glycolysis and angiogenesis by regulating miR‐361‐3p/TGFB1. Cancer Sci 2022; 113:2044-2055. [PMID: 35347818 PMCID: PMC9207367 DOI: 10.1111/cas.15351] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/24/2022] [Accepted: 03/26/2022] [Indexed: 11/30/2022] Open
Abstract
Circular RNA (circRNA) participates in a variety of pathophysiological processes, including the development of gastric cancer (GC). However, the role of circ_0006089 in GC progression and its underlying molecular mechanism need to be further revealed. Quantitative real‐time PCR was utilized for detecting circ_0006089, microRNA (miR)‐361‐3p and transforming growth factor‐β1 (TGFB1) expression. The interaction between miR‐361‐3p and circ_0006089 or TGFB1 was confirmed using a dual‐luciferase reporter assay and an RNA immunoprecipitation (RIP) assay. Cell proliferation, metastasis, apoptosis, and angiogenesis were determined using colony formation assay, EdU assay, transwell assay, flow cytometry, and tube formation assay. Cell glycolysis was evaluated by detecting glucose consumption, lactate production, and ATP levels. In addition, western blot (WB) analysis was used to measure protein expression. Xenograft tumor models were used to assess the effect of circ_0006089 knockdown on GC tumorigenesis. circ_0006089 had been found to be upregulated in GC tissues and cells, and it could act as an miR‐361‐3p sponge. circ_0006089 knockdown suppressed GC proliferation, metastasis, glycolysis, angiogenesis, and increased apoptosis, while this effect could be revoked by miR‐361‐3p inhibitor. TGFB1 was targeted by miR‐361‐3p, and its overexpression reversed the effects of miR‐361‐3p on GC cell function. Also, circ_0006089 promoted TGFB1 expression via sponging miR‐361‐3p. Animal experiments showed that silenced circ_0006089 inhibited GC tumorigenesis through the miR‐361‐3p/TGFB1 pathway. Our results revealed that the circ_0006089/miR‐361‐3p/TGFB1 axis contributed to GC progression, confirming that circ_0006089 might be a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Gastroenterology Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine Shanghai 200120 China
| | - Qilin Zhang
- Department of General Surgery Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine Shanghai 200120 China
| | - Bingling Liao
- Department of Gastroenterology Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine Shanghai 200120 China
| | - Xiaofeng Qiu
- Department of General Surgery Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine Shanghai 200120 China
| | - Sheng Hu
- Department of General Surgery Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine Shanghai 200120 China
| | - Qihua Xu
- Department of Gastroenterology Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine Shanghai 200120 China
| |
Collapse
|
136
|
Chen P, Fan W, Hou Y, Wang F, Luo N. Role of kinesin family member 14 in disease monitoring and prognosis in patients with gastrointestinal cancer. Oncol Lett 2022; 23:156. [PMID: 35836481 PMCID: PMC9258591 DOI: 10.3892/ol.2022.13276] [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: 09/02/2021] [Accepted: 01/13/2022] [Indexed: 11/06/2022] Open
Abstract
Kinesin family member 14 (KIF14) is not only involved in numerous essential biological activities, such as cytokinesis and myelination, but also regulates several malignant behaviors and progression of cancer. However, its role in gastrointestinal cancer is rarely reported. Therefore, the present study aimed to investigate the association of KIF14 expression with disease-free survival (DFS) and overall survival (OS) times in patients with gastrointestinal cancer. A total of 101 patients with gastrointestinal cancer (36 patients with gastric cancer and 65 patients with colorectal cancer) were retrospectively reviewed, and their cancer samples were collected to detect the protein and mRNA expression levels of KIF14 using immunohistochemistry and reverse transcription-quantitative PCR, respectively. KIF14 protein expression was increased in cancer tissues compared with adjacent tissues (all P<0.001). The protein expression levels of KIF14 were positively associated with T stage (P<0.001), distant metastases (P=0.007) and TNM stage (P<0.001), while KIF14 mRNA expression was positively associated with T stage (P<0.001), lymph node metastasis (P=0.004), distant metastases (P=0.001) and TNM stage (P<0.001). High protein and mRNA expression levels of KIF14 were associated with worse DFS (P<0.001) and OS (P=0.016) times. In addition, high KIF14 protein expression independently predicted unfavorable DFS times (P=0.007). Subgroup analysis revealed that in patients with gastric cancer, KIF14 expression was associated with DFS and OS times, while in patients with colorectal cancer, KIF14 expression was only associated with DFS time, but not with OS time. In conclusion, KIF14 expression was not only associated with advanced pathological differentiation and TNM stage but was also associated with poor survival time in patients with gastrointestinal cancer. These results indicate the potential of KIF14 as a biomarker for gastrointestinal cancer prognosis.
Collapse
Affiliation(s)
- Ping Chen
- Department of Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Weining Fan
- Department of Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yujin Hou
- Department of Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Fang Wang
- Department of Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Na Luo
- Department of Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| |
Collapse
|
137
|
Hu Y, Zhang P, Shi Y, Dong X, Wu Y, Dong D, Li E, Fan Y. Inhibition of Ras protein activator like 2 produces antitumor effects in gastric cancer via the suppression of YAP1 activation. ENVIRONMENTAL TOXICOLOGY 2022; 37:527-538. [PMID: 34826200 DOI: 10.1002/tox.23418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/28/2021] [Accepted: 11/14/2021] [Indexed: 06/13/2023]
Abstract
Ras protein activator like 2 (RASAL2) has a cancer-related function, but plays inconsistent roles in different malignancies. This project was designed to determine the role of RASAL2 in carcinogenesis in gastric cancer. The Cancer Genome Atlas data revealed high levels of RASAL2 in gastric cancer tissue, which was confirmed in clinical specimens of gastric cancer via real-time quantitative PCR and western blotting assays. High RASAL2 was correlated with a reduced survival rate in gastric cancer patients. In gastric cancer cell lines, the silencing of RASAL2 restrained cellular proliferation, invasion and epithelial-to-mesenchymal transition, while enhancing chemosensitivity to cisplatin. Mechanistically, the silencing of RASAL2 was found to inhibit the activation of Yes-associated protein 1 (YAP1), a pro-oncogenic protein in gastric cancer, and decrease the expression of YAP1 target genes. The re-expression of constitutively active YAP1 substantially reversed RASAL2-silencing-produced antitumor effects. Moreover, treatment with YAP1 inhibitors could diminish RASAL2-overexpression-evoked oncogenic effects in gastric cancer cells. Additionally, gastric cancer cells with RASAL2 silencing exhibited a reduced ability to form xenograft tumors in nude mice. Collectively, our data demonstrate that the silencing of RASAL2 has noteworthy antitumor effects in gastric cancer cells via the suppression of YAP1 activation. This project underscores a vital role of the RASAL2/YAP1 axis in gastric progression and indicates that targeting this oncogenic axis may be applied as a potential therapeutic option for gastric cancer.
Collapse
Affiliation(s)
- Yuan Hu
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Pengchuang Zhang
- Department of Gynecologic Cancer, Shaanxi Provincial Tumor Hospital, China
| | - Yu Shi
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Xuyuan Dong
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Yinying Wu
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Danfeng Dong
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Enxiao Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Yangwei Fan
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| |
Collapse
|
138
|
Clinical Efficacy and Safety of Bevacizumab, Apatinib, and Recombinant Human Endothelial Inhibitor in the Treatment of Advanced Gastric Cancer. JOURNAL OF ONCOLOGY 2022; 2022:6189833. [PMID: 35251174 PMCID: PMC8894022 DOI: 10.1155/2022/6189833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/14/2022] [Accepted: 01/25/2022] [Indexed: 11/17/2022]
Abstract
Objective To investigate the clinical efficacy and safety of bevacizumab, apatinib, and recombinant human endothelial inhibitor in the treatment of advanced gastric cancer. Methods The medical data of 204 patients with a medium to advanced gastric cancer assessed for eligibility treated in our hospital from February 2019 to April 2020 were retrospectively analyzed. The eligible patients were assigned at a ratio of 1 : 1:1 : 1 to either the control group (chemotherapy), study group I (bevacizumab combined with chemotherapy), study group II (apatinib combined with chemotherapy), or study group III (recombinant human endothelial inhibitor combined with chemotherapy) according to different treatment methods. The treatment efficacy, drug toxicity, quality of life, and serum tumor marker levels before and after treatment were compared among the four groups. Results Regarding the treatment effects, the effective rate of study group II (68.63%) was significantly higher than that of the control group (33.33%), study group I (58.82%), and study group III (49.02%) (P < 0.05). The four groups showed similar safety and tolerability profiles (P > 0.05). The treatment in study group II led to a significantly higher physiological function score vs. the other three groups, but the scores of other items were not significantly different. Significant reduction was observed in the serum tumor markers after treatment in the four groups (P < 0.05), but treatment in study group II led to a significantly greater reduction than the other three groups (P < 0.05). Conclusion The addition of apatinib, bevacizumab, and recombinant human endothelial inhibitor injection to chemotherapy for the treatment of medium to advanced gastric cancer can significantly improve the clinical treatment efficacy, among which the use of apatinib combined with chemotherapy achieves the best results, which is worthy of clinical promotion.
Collapse
|
139
|
Liu Z, Hu K, Wang X, Zhang Y, Wang W, Wu Y. lncRNA ACTA2-AS1 inhibits malignant phenotypes of gastric cancer cells. Open Med (Wars) 2022; 17:266-279. [PMID: 35274046 PMCID: PMC8854910 DOI: 10.1515/med-2021-0406] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 09/02/2021] [Accepted: 11/12/2021] [Indexed: 01/19/2023] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies in digestive system. Accumulating evidence reveals the critical role of long noncoding RNAs (lncRNAs) in GC development. The study aimed to explore the functions and mechanism of lncRNA actin alpha 2, smooth muscle antisense RNA 1 (ACTA2-AS1) in GC. Reverse transcription-quantitative polymerase chain reaction analyses and subcellular fractionation assays showed that ACTA2-AS1 was lowly expressed in GC cells and was mainly distributed in the cytoplasm. Overexpressed ACTA2-AS1 inhibited GC cell viability, proliferation, migration, invasion, and epithelial-mesenchymal transition process, as suggested by cell counting kit-8 assays, colony formation assays, wound healing assays, Transwell assays and Western blot analyses. Mechanistically, ACTA2-AS1 served as a competing endogenous RNA (ceRNA) to bind with miR-378a-3p and thereby, antagonized the inhibitory effect of miR-378a-3p on the expression of messenger RNA phosphatidylinositol specific phospholipase C X domain containing 2 (PLCXD2). The binding capacity between miR-378a-3p and ACTA2-AS1 (or PLCXD2) was detected by RNA pulldown assays, luciferase reporter assays and RNA immunoprecipitation assays. Moreover, PLCXD2 knockdown rescued the inhibitory effect of ACTA2-AS1 overexpression on malignant behaviors of GC cells. Overall, ACTA2-AS1 inhibits malignant phenotypes of GC cells by acting as a ceRNA to target miR-378a-3p/PLCXD2 axis.
Collapse
Affiliation(s)
- Zhiping Liu
- Department of General Surgery, Hefei Hospital Affiliated to Medical University of Anhui, Hefei 230011, Anhui, China
| | - Kaibing Hu
- Department of General Surgery, Hefei Hospital Affiliated to Medical University of Anhui, Hefei 230011, Anhui, China
| | - Xiang Wang
- Department of General Surgery, Hefei Hospital Affiliated to Medical University of Anhui, Hefei 230011, Anhui, China
| | - Youqian Zhang
- Department of General Surgery, Hefei Hospital Affiliated to Medical University of Anhui, Hefei 230011, Anhui, China
| | - Weiping Wang
- Department of General Surgery, Hefei Hospital Affiliated to Medical University of Anhui, Hefei 230011, Anhui, China
| | - Yindi Wu
- Department of Pediatrics, Hefei First People’s Group Hospital, 390 Huaihe Road, Luyang District, Hefei 230000, Anhui, China
| |
Collapse
|
140
|
Pang J, Li G, Qian H, Wu Y, Chen Y. Secretory PKG II blocks activation of PDGFRβ via Ser254 in gastric cancer cells. Cell Biol Int 2022; 46:747-754. [PMID: 35066967 PMCID: PMC9305209 DOI: 10.1002/cbin.11766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/28/2021] [Accepted: 01/08/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Ji Pang
- Department of Physiology, School of Medicine, Jiangsu UniversityZhenjiang CityJiangsu Province212013People's Republic of China
| | - Guorui Li
- Department of Physiology, School of Medicine, Jiangsu UniversityZhenjiang CityJiangsu Province212013People's Republic of China
| | - Hai Qian
- Department of Physiology, School of Medicine, Jiangsu UniversityZhenjiang CityJiangsu Province212013People's Republic of China
| | - Yan Wu
- Department of Physiology, School of Medicine, Jiangsu UniversityZhenjiang CityJiangsu Province212013People's Republic of China
| | - Yongchang Chen
- Department of Physiology, School of Medicine, Jiangsu UniversityZhenjiang CityJiangsu Province212013People's Republic of China
| |
Collapse
|
141
|
Huo J, Guan G, Cai J, Wu L. Integrated analysis of 1804 samples of six centers to construct and validate a robust immune-related prognostic signature associated with stromal cell abundance in tumor microenvironment for gastric cancer. World J Surg Oncol 2022; 20:4. [PMID: 34983559 PMCID: PMC8728957 DOI: 10.1186/s12957-021-02485-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/22/2021] [Indexed: 12/24/2022] Open
Abstract
Background Stromal cells in tumor microenvironment could promote immune escape through a variety of mechanisms, but there are lacking research in the field of gastric cancer (GC). Methods We identified differential expressed immune-related genes (DEIRGs) between the high- and low-stromal cell abundance GC samples in The Cancer Genome Atlas and GSE84437 datasets. A risk score was constructed basing on univariate cox regression analysis, LASSO regression analysis, and multivariate cox regression analysis in the training cohort (n=772). The median value of the risk score was used to classify patients into groups with high and low risk. We conducted external validation of the prognostic signature in four independent cohorts (GSE26253, n=432; GSE62254, n=300; GSE15459, n=191; GSE26901, n=109) from the Gene Expression Omnibus (GEO) database. The immune cell infiltration was quantified by the CIBERSORT method. Results The risk score contained 6 genes (AKT3, APOD, FAM19A5, LTBP3, NOV, and NOX4) showed good performance in predicting 5-year overall survival (OS) rate and 5-year recurrence-free survival (RFS) rate of GC patients. The risk death and recurrence of GC patients growing with the increasing risk score. The patients were clustered into three subtypes according to the infiltration of 22 kinds of immune cells quantified by the CIBERSORT method. The proportion of cluster A with the worst prognosis in the high-risk group was significantly higher than that in the low-risk group; the risk score of cluster C subtype with the best prognosis was significantly lower than that of the other two subtypes. Conclusion This study established and validated a robust prognostic model for gastric cancer by integrated analysis 1804 samples of six centers, and its mechanism was explored in combination with immune cell infiltration characterization.
Collapse
Affiliation(s)
- Junyu Huo
- Liver Disease Center, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266003, China
| | - Ge Guan
- Liver Disease Center, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266003, China
| | - Jinzhen Cai
- Liver Disease Center, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266003, China
| | - Liqun Wu
- Liver Disease Center, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266003, China.
| |
Collapse
|
142
|
Wang X, Wang H, Cao L, Wu J, Lu T, Li S, Li J. Efficacy and Safety of Brucea javanica Oil Emulsion Injection in the Treatment of Gastric Cancer: A Systematic Review and Meta-Analysis. Front Nutr 2021; 8:784164. [PMID: 34957186 PMCID: PMC8696120 DOI: 10.3389/fnut.2021.784164] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/05/2021] [Indexed: 12/25/2022] Open
Abstract
Background: Gastric cancer (GC) is one of the most common digestive tract cancers and ranks fifth in the incidence of malignant tumors worldwide. Brucea javanica oil emulsion injection (BJOEI), a Chinese patent medicine extracted from Brucea javanica (Yadanzi in Chinese Pinyin), is widely used as an adjuvant treatment for GC in China. This systematic review and meta-analysis aimed to evaluate the available data on the efficacy and safety of BJOEI in the treatment of GC and assess the quality of the synthesized evidence. Methods: A comprehensive search was performed on PubMed, EMBASE, CENTRAL, Web of Science, Chinese Biomedical Literature Database (CBM), China National Knowledge Infrastructure (CNKI), Wanfang database and Chinese Scientific Journals Database (VIP database), and other potential resources, such as the Chinese Clinical Trial Registry (ChiCTR) and ClinicalTrials.gov from their inception to July 31, 2021. Randomized controlled trials (RCTs) comparing the therapeutic effects of BJOEI combined with conventional therapy to those of conventional therapy alone were included. We used RevMan 5.3 for data analysis and quality evaluation of the included studies and assessed the evidence quality based on the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) criteria. Results: Eighteen RCTs involving 1,210 patients were included, and the meta-analysis results demonstrated that compared with the control group (conventional therapy), the experimental group (BJOEI combined with conventional therapy) showed a significantly improved overall response rate (ORR) (risk ratio [RR] = 1.52, 95% CI: 1.36–1.69, P < 0.00001), clinical benefit rate (CBR) (RR = 1.17, 95% CI: 1.11–1.23, P < 0.00001), performance status (RR = 1.72, 95% CI: 1.46–2.01, P < 0.00001), and reduced incidence of the following adverse drug reactions (ADRs): neutropenia, leukopenia, nausea and vomiting, diarrhea, liver damage, hand-foot syndrome, and peripheral sensory nerve toxicity. Subgroup analysis showed that the BJOEI intervention could significantly improve the ORR and CBR in patients with GC when combined with FOLFOX4, XELOX, and other chemotherapeutics. Conclusion: The evidence presented in this study supports the fact that BJOEI combined with conventional chemotherapy provides a statistically significant and clinically important effect in the improvement of ORR, CBR, performance status, and ADR reduction in patients with GC. To further support this conclusion, more rigorously designed, large-scale, and multicenter RCTs are needed in the future.
Collapse
Affiliation(s)
- Xinmiao Wang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Heping Wang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Luchang Cao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingyuan Wu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Graduate College, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Taicheng Lu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Graduate College, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Shixin Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Graduate College, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Jie Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
143
|
Identification of MATN3 as a Novel Prognostic Biomarker for Gastric Cancer through Comprehensive TCGA and GEO Data Mining. DISEASE MARKERS 2021; 2021:1769635. [PMID: 34900024 PMCID: PMC8660198 DOI: 10.1155/2021/1769635] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022]
Abstract
Gastric cancer (GC) is still a vital malignant cancer across the world with unsatisfactory prognostic results. Matrilin-3 (MATN3) is a member of the extracellular matrix (ECM) protein family. The present research intends to explore the expression level of MATN3 in patients with GC and to explore the prognosis significance of MATN3. In this study, we observed that the MATN3 expression was remarkably upregulated in GC samples in contrast to noncancer samples. Clinical analyses unveiled that high MATN3 expression was related to age, tumor status, and clinical stages. Survival analyses unveiled that patients with high MATN3 expression displayed a poorer overall survival and progression-free survival than those with low MATN3 expression. The AUC of the relevant ROC curve for 1 year, 3 years, and 5 years of survival is 0.571, 0.596, and 0.720, separately. Multivariate assays revealed that MATN3 expression and stage were independent predictors of poor prognosis of GC patients. A meta-analysis unveiled that high MATN3 expression was tightly associated with better overall survival. Overall, our data indicated that MATN3 may have a diagnostic and prognostic value for patients with advanced gastric cancer and assist to improve clinical outcomes for GC patients.
Collapse
|
144
|
Wen L, Xia L, Guo X, Huang HF, Wang F, Yang XT, Yang Z, Zhu H. Multimodal Imaging Technology Effectively Monitors HER2 Expression in Tumors Using Trastuzumab-Coupled Organic Nanoparticles in Patient-Derived Xenograft Mice Models. Front Oncol 2021; 11:778728. [PMID: 34869025 PMCID: PMC8637767 DOI: 10.3389/fonc.2021.778728] [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: 09/17/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
Trastuzumab is a monoclonal antibody targeting human epidermal growth factor 2 (HER2), which has been successfully used in the treatment of patients with breast cancer and gastric cancer; however, problems concerning its cardiotoxicity, drug resistance, and unpredictable efficacy still remain. Herein, we constructed novel organic dopamine–melanin nanoparticles (dMNs) as a carrier and then surface-loaded them with trastuzumab to construct a multifunctional nanoprobe named Her-PEG-dMNPs. We used micro-PET/CT and PET/MRI multimodality imaging to evaluate the retention effect of the nanoprobe in HER2 expression in gastric cancer patient-derived xenograft (PDX) mice models after labeling of the radionuclides 64Cu or 124I and MRI contrast agent Mn2+. The nanoprobes can specifically target the HER2-expressing SKOV-3 cells in vitro (3.61 ± 0.74 vs. 1.24 ± 0.43 for 2 h, P = 0.002). In vivo, micro-PET/CT and PET/MRI showed that the 124I-labeled nanoprobe had greater contrast and retention effect in PDX models than unloaded dMNPs as carrier (1.63 ± 0.07 vs. 0.90 ± 0.04 at 24 h, P = 0.002), a similarity found in 64Cu-labeled Her-PEG-dMNPs. Because 124I has a longer half-life and matches the pharmacokinetics of the nanoparticles, we focused on the further evaluation of 124I-Her-PEG-dMNPs. Furthermore, immunohistochemistry staining confirmed the overexpression of HER2 in the animal model. This study developed and validated novel HER2-specific multimodality imaging nanoprobes for quantifying HER2 expression in mice. Through the strong retention effect of the tumor site, it can be used for the promotion of monoclonal antibody treatment effect and process monitoring.
Collapse
Affiliation(s)
- Li Wen
- Medical College, Guizhou University, Guiyang, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lei Xia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaoyi Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hai-Feng Huang
- Department of Orthopaedics, Guizhou Provincial People's Hospital, Guiyang, China
| | - Feng Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xian-Teng Yang
- Medical College, Guizhou University, Guiyang, China.,Department of Orthopaedics, Guizhou Provincial People's Hospital, Guiyang, China
| | - Zhi Yang
- Medical College, Guizhou University, Guiyang, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hua Zhu
- Medical College, Guizhou University, Guiyang, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
145
|
Yue F, Peng K, Zhang L, Zhang J. Circ_0004104 Accelerates the Progression of Gastric Cancer by Regulating the miR-539-3p/RNF2 Axis. Dig Dis Sci 2021; 66:4290-4301. [PMID: 33449226 DOI: 10.1007/s10620-020-06802-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Circular RNA (circRNA) has been shown to be closely associated with cancer progression, including gastric cancer (GC). However, the function of circ_0004104 in GC progression has not been clarified. AIMS The purpose of this study was to explore the role of circ_0004104 in GC progression. METHODS The expression levels of circ_0004104, miR-539-3p, and ring finger protein 2 (RNF2) were assessed using quantitative real-time polymerase chain reaction. Cell proliferation was measured by 3-(4,5-dimethyl-2 thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay, and cell migration and invasion were detected using transwell assay. The levels of glutamine, glutamate, and α-ketoglutarate were determined to evaluate the glutaminolysis of cells, and the protein levels of glutaminolysis-related markers and RNF2 were detected using western blot analysis. Furthermore, Dual-Luciferase Reporter Assay was employed to assess the interaction between miR-539-3p and circ_0004104 or RNF2. Animal experiments were carried out to evaluate the effect of circ_0004104 silencing on GC tumor growth in vivo. RESULTS Circ_0004104 was upregulated in GC, and its knockdown repressed the proliferation, metastasis, and glutaminolysis of GC cells in vitro and reduced GC tumor growth in vivo. Furthermore, we discovered that circ_0004104 could sponge miR-539-3p and miR-539-3p could target RNF2. The rescue experiments suggested that miR-539-3p inhibitor could reverse the suppressive effect of circ_0004104 silencing on GC progression, and RNF2 overexpression also reversed the inhibition effect of miR-539-3p mimic on GC progression. CONCLUSION Circ_0004104 accelerated GC progression via regulating the miR-539-3p/RNF2 axis, indicating that circ_0004104 might be a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Furong Yue
- Department of Oncology, Chongqing Three Gorges Central Hospital and Chongqing University Three Gorges Hospital, No. 165, Xin Cheng Road, Wanzhou District, Chongqing, 404000, China
| | - Keyu Peng
- Department of Oncology, Chongqing Three Gorges Central Hospital and Chongqing University Three Gorges Hospital, No. 165, Xin Cheng Road, Wanzhou District, Chongqing, 404000, China
| | - Li Zhang
- Department of Oncology, Chongqing Three Gorges Central Hospital and Chongqing University Three Gorges Hospital, No. 165, Xin Cheng Road, Wanzhou District, Chongqing, 404000, China
| | - Jun Zhang
- Department of Oncology, Chongqing Three Gorges Central Hospital and Chongqing University Three Gorges Hospital, No. 165, Xin Cheng Road, Wanzhou District, Chongqing, 404000, China.
| |
Collapse
|
146
|
Ma J, Chen X, Zhu X, Pan Z, Hao W, Li D, Zheng Q, Tang X. Luteolin potentiates low-dose oxaliplatin-induced inhibitory effects on cell proliferation in gastric cancer by inducing G 2/M cell cycle arrest and apoptosis. Oncol Lett 2021; 23:16. [PMID: 34820015 PMCID: PMC8607327 DOI: 10.3892/ol.2021.13134] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/13/2021] [Indexed: 01/02/2023] Open
Abstract
Although the reduction of oxaliplatin doses may alleviate deleterious side effects of gastrointestinal and gynecological cancer treatment, it also limits the anticancer therapeutic effects. As a high-efficient and low-priced herbal medicine ingredient, luteolin is an agent with a broad spectrum of anticancer activities and acts as a potential enhancer of therapeutic effects of chemotherapy agents in cancer treatment. This study focused on the antitumor effects and mechanism of combined treatment with luteolin and oxaliplatin on a mouse forestomach carcinoma (MFC) cell line. The study used CCK-8 assay, flow cytometry, Annexin V-FITC/PI double staining assay, reactive oxygen species testing assay, mitochondrial membrane potential testing assay, and western blot assay. The results showed that luteolin and oxaliplatin exerted synergistic effects on inhibiting MFC cell proliferation by inducing G2/M cell cycle arrest and apoptosis. Inhibiting the tumor necrosis factor receptor-associated protein 1/phosphorylated-extracellular-regulated protein kinases1/2/cell division cycle 25 homolog C/cyclin-dependent kinase-1/cyclin B1 pathway was indispensable to the combined treatment with luteolin and oxaliplatin to induce G2/M cell cycle arrest. In addition, luteolin increased oxidative stress in MFC cells treated with a low dose of oxaliplatin. The combined therapy damaged mitochondrial membrane potential and regulated BCL-2-associated X protein and B-cell lymphoma 2 protein expression, leading to apoptosis. Findings of the present study suggest that luteolin may be a qualified chemotherapy enhancer to potentiate the anticancer effects of low-dose oxaliplatin in MFC cells. This work provides a theoretical foundation for future research on applications of luteolin in clinical chemotherapy.
Collapse
Affiliation(s)
- Jun Ma
- College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong 266003, P.R. China.,School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Xiaojie Chen
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Xuejie Zhu
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Zhaohai Pan
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Wenjin Hao
- School of Life Sciences, Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Defang Li
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Qiusheng Zheng
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China.,School of Pharmacy, Shihezi University, Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Ministry of Education, School of Pharmacy, Shihezi, Xinjiang 832002, P.R. China
| | - Xuexi Tang
- College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
147
|
Pellino A, Brignola S, Riello E, Niero M, Murgioni S, Guido M, Nappo F, Businello G, Sbaraglia M, Bergamo F, Spolverato G, Pucciarelli S, Merigliano S, Pilati P, Cavallin F, Realdon S, Farinati F, Dei Tos AP, Zagonel V, Lonardi S, Loupakis F, Fassan M. Association of CLDN18 Protein Expression with Clinicopathological Features and Prognosis in Advanced Gastric and Gastroesophageal Junction Adenocarcinomas. J Pers Med 2021; 11:1095. [PMID: 34834447 PMCID: PMC8624955 DOI: 10.3390/jpm11111095] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/11/2021] [Accepted: 10/22/2021] [Indexed: 12/16/2022] Open
Abstract
The tight junction protein claudin-18 (CLDN18), is often expressed in various cancer types including gastric (GC) and gastroesophageal adenocarcinomas (GECs). In the last years, the isoform CLDN18.2 emerged as a potential drug target in metastatic GCs, leading to the development of monoclonal antibodies against this protein. CLDN18.2 is the dominant isoform of CLDN18 in normal gastric and gastric cancer tissues. In this work, we evaluated the immunohistochemical (IHC) profile of CLDN18 and its correlation with clinical and histopathological features including p53, E-cadherin, MSH2, MSH6, MLH1, PMS2, HER2, EBER and PD-L1 combined positive score, in a large real-world and mono-institutional series of advanced GCs (n = 280) and GECs (n = 70). The association of IHC results with survival outcomes was also investigated. High membranous CLDN18 expression (2+ and 3+ intensity ≥75%) was found in 117/350 (33.4%) samples analyzed. CLDN18 expression correlated with age <70 (p = 0.0035), positive EBV status (p = 0.002), high stage (III, IV) at diagnosis (p = 0.003), peritoneal involvement (p < 0.001) and lower incidence of liver metastases (p = 0.013). CLDN18 did not correlate with overall survival. The predictive value of response of CLDN18 to targeted agents is under investigation in several clinical trials and further studies will be needed to select patients who could benefit from these therapies.
Collapse
Affiliation(s)
- Antonio Pellino
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 35128 Padua, Italy; (A.P.); (S.M.); (F.N.); (F.B.); (V.Z.); (F.L.)
| | - Stefano Brignola
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35122 Padua, Italy; (S.B.); (E.R.); (M.G.); (G.B.); (M.S.); (A.P.D.T.)
- Department of Pathology, Azienda ULSS 2 Marca Trevigiana, 31100 Treviso, Italy;
| | - Erika Riello
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35122 Padua, Italy; (S.B.); (E.R.); (M.G.); (G.B.); (M.S.); (A.P.D.T.)
| | - Monia Niero
- Department of Pathology, Azienda ULSS 2 Marca Trevigiana, 31100 Treviso, Italy;
| | - Sabina Murgioni
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 35128 Padua, Italy; (A.P.); (S.M.); (F.N.); (F.B.); (V.Z.); (F.L.)
| | - Maria Guido
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35122 Padua, Italy; (S.B.); (E.R.); (M.G.); (G.B.); (M.S.); (A.P.D.T.)
- Department of Pathology, Azienda ULSS 2 Marca Trevigiana, 31100 Treviso, Italy;
| | - Floriana Nappo
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 35128 Padua, Italy; (A.P.); (S.M.); (F.N.); (F.B.); (V.Z.); (F.L.)
| | - Gianluca Businello
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35122 Padua, Italy; (S.B.); (E.R.); (M.G.); (G.B.); (M.S.); (A.P.D.T.)
| | - Marta Sbaraglia
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35122 Padua, Italy; (S.B.); (E.R.); (M.G.); (G.B.); (M.S.); (A.P.D.T.)
| | - Francesca Bergamo
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 35128 Padua, Italy; (A.P.); (S.M.); (F.N.); (F.B.); (V.Z.); (F.L.)
| | - Gaya Spolverato
- 1st Surgery Unit, Department of Surgical, Oncological, and Gastroenterological Sciences (DISCOG), University of Padua, 35122 Padua, Italy; (G.S.); (S.P.)
| | - Salvatore Pucciarelli
- 1st Surgery Unit, Department of Surgical, Oncological, and Gastroenterological Sciences (DISCOG), University of Padua, 35122 Padua, Italy; (G.S.); (S.P.)
| | - Stefano Merigliano
- 3rd Surgery Unit, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, 35122 Padua, Italy;
| | - Pierluigi Pilati
- Surgery Unit, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 31033 Castelfranco Veneto, Italy;
| | | | - Stefano Realdon
- Gastroenterology Unit, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 35128 Padua, Italy;
| | - Fabio Farinati
- Gastroenterology Unit, Department of Surgical, Oncological, and Gastroenterological Sciences (DISCOG), University of Padua, 35122 Padua, Italy;
| | - Angelo Paolo Dei Tos
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35122 Padua, Italy; (S.B.); (E.R.); (M.G.); (G.B.); (M.S.); (A.P.D.T.)
| | - Vittorina Zagonel
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 35128 Padua, Italy; (A.P.); (S.M.); (F.N.); (F.B.); (V.Z.); (F.L.)
| | - Sara Lonardi
- Oncology Unit 3, Department of Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 35128 Padua, Italy;
| | - Fotios Loupakis
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 35128 Padua, Italy; (A.P.); (S.M.); (F.N.); (F.B.); (V.Z.); (F.L.)
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35122 Padua, Italy; (S.B.); (E.R.); (M.G.); (G.B.); (M.S.); (A.P.D.T.)
- Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 35128 Padua, Italy
| |
Collapse
|
148
|
Salati M, Venetis K, Fassan M, Malapelle U, Pagni F, Sajjadi E, Fusco N, Ghidini M. ctDNA analysis in the personalized clinical management of gastroesophageal adenocarcinoma: turning hope into reality. Future Oncol 2021; 17:4607-4618. [PMID: 34406032 DOI: 10.2217/fon-2021-0228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Gastroesophageal adenocarcinoma (GEA) is a global health issue with a high fatality-to-case ratio and a 5-year overall survival that has only slightly improved. High-throughput molecular profiling has uncovered a profound complexity and heterogeneity in GEA biology, which limits considerably the treatment advances. Liquid biopsy with circulating tumor (ct)DNA analysis could elucidate GEA molecular heterogeneity and provide diagnostic, prognostic and predictive information to guide clinical decision-making. However, only a handful of studies have shown positive results for the application of ctDNA analysis in GEA clinical management. As a result, no comprehensive information is available to date on this continuously evolving topic. Here, we discuss the current state of knowledge, along with promises and challenges related to ctDNA analysis in GEA.
Collapse
Affiliation(s)
- Massimiliano Salati
- Division of Oncology, Oncology and Hematology Department, University of Modena and Reggio Emilia, Modena, Italy.,Ph.D. Program Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Konstantinos Venetis
- Department of Oncology and Hemato-Oncology, University of Milan, Milan 20122, Italy.,Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan 20141, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology and Cytopathology Unit, University of Padua, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, Pathology, University Milan Bicocca, Milan 20126, Italy
| | - Elham Sajjadi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan 20122, Italy.,Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan 20141, Italy
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, University of Milan, Milan 20122, Italy.,Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan 20141, Italy
| | - Michele Ghidini
- Division of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| |
Collapse
|
149
|
He C, Qi W, Wang Z. Effect and mechanism of downregulating the long-chain noncoding RNA TM4SF1-AS1 on the proliferation, apoptosis and invasion of gastric cancer cells. World J Surg Oncol 2021; 19:226. [PMID: 34330293 PMCID: PMC8325262 DOI: 10.1186/s12957-021-02334-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 07/16/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND To investigate long-chain noncoding TM4SF1-AS1 in gastric cancer (GC) tissues and cells. METHODS TM4SF1-AS1 in 40 GC tissues and adjacent tissues was detected and compared using real-time fluorescence quantitative PCR (qRT-PCR). TM4SF1-AS1 in MKN28 and SGC7901 GC cells was downregulated using small interfering RNA (shRNA). The cells were grouped into an interference group (shTM4SF1-AS1 group) and a control group (shControl group). MTT and Transwell tests were applied to determine the proliferation and invasion of the cells in both groups, and flow cytometry was performed to assess the apoptosis rate in the two groups. Western blotting was performed to determine changes in key proteins in cells during the epithelial-to-mesenchymal transition (EMT) and in the TM4SF1 and PI3K-AKT signalling pathways in response to the downregulation of TM4SF1-AS1. RESULTS The proliferation of MKN28 and SGC7901 in the shTM4SF1-AS1 group was significantly inhibited at 48 h and 72 h compared to that in the shControl group (all P < 0.05). In the shTM4SF1-AS1 group, the number of invaded MKN28 and SGC7901 cells was significantly lower than that in the shControl group (all P < 0.05). Apoptosis in the MKN28 and SGC7901 shTM4SF1-AS1 groups was significantly higher than that in the shControl group (all P < 0.05). Compared to those in the shControl group, levels of E-cadherin in EMT-related proteins were significantly elevated (P < 0.01), while levels of N-cadherin, Snail and Twist1 were significantly decreased (all P < 0.01). After silencing the expression of LncTM4SF1-AS1, the expression levels of TM4SF1 in the shTM4SF1-AS1 group were downregulated compared to those in the shControl group, and the p-PI3K and p-AKT proteins in the PI3K-AKT signalling pathway in the shTM4SF1-AS1 group were downregulated compared to those of the shControl group. CONCLUSIONS TM4SF1-AS1 is upregulated in gastric cancer tissues and cells. Interfering with and downregulating its expression inhibit cancer cell proliferation, invasion and the EMT and promote apoptosis. The underlying mechanism for these effects is related to silencing the TM4SF1 and PI3K-AKT signalling pathways. TM4SF1-AS1 may be a potential therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Chengzhi He
- Department of Gastrointestinal Surgery, Wuhan Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 26 Shengli Street, Jiang'an District, Wuhan, 430000, Hubei Province, China
| | - Wenjing Qi
- Wuhan College of Arts and Sciences, Wuhan, 430345, Hubei Province, China
| | - Zhihui Wang
- Department of Gastrointestinal Surgery, Wuhan Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 26 Shengli Street, Jiang'an District, Wuhan, 430000, Hubei Province, China.
| |
Collapse
|
150
|
Huo J, Wu L, Zang Y. Eight-gene prognostic signature associated with hypoxia and ferroptosis for gastric cancer with general applicability. Epigenomics 2021; 13:875-890. [PMID: 33942671 DOI: 10.2217/epi-2020-0411] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Aims: To investigate the prognostic significance of hypoxia- and ferroptosis-related genes for gastric cancer (GC). Materials & methods: We extracted data on 259 hypoxia- and ferroptosis-related genes from The Cancer Genome Atlas and identified the differentially expressed genes between normal (n = 32) and tumor (n = 375) tissues. A risk score was established by univariate Cox regression analysis and LASSO penalized Cox regression analysis. Results: The risk score contained eight genes showed good performance in predicting overall survival and relapse-free survival in GC patients in both the training cohort (The Cancer Genome Atlas, n = 350) and the testing cohorts (GSE84437, n = 431; GSE62254, n = 300; GSE15459, n = 191; GSE26253, n = 432). Conclusion: The eight-gene signature may help to the improve the prognostic risk classification of GC.
Collapse
Affiliation(s)
- Junyu Huo
- Liver Disease Center, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao 266003, China.,Qingdao University, No. 308 Ningxia Road, Qingdao 266071, China
| | - Liqun Wu
- Liver Disease Center, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao 266003, China
| | - Yunjin Zang
- Liver Disease Center, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao 266003, China
| |
Collapse
|