51
|
Jin X, Liu Z, Yang D, Yin K, Chang X. Recent Progress and Future Perspectives of Immunotherapy in Advanced Gastric Cancer. Front Immunol 2022; 13:948647. [PMID: 35844558 PMCID: PMC9284215 DOI: 10.3389/fimmu.2022.948647] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/06/2022] [Indexed: 12/15/2022] Open
Abstract
As one of the most common forms of solid tumours, gastric carcinoma has been revealed as the third leading cause of death worldwide. The symptom of gastric cancer is usually not obvious and thus difficult to detect at earlier stages. Therefore, gastric cancer is already in the advanced stage once detected in patients, which has a poor prognosis due to ineffective therapies and multiple resistance. Recent advance in understanding the microenvironment of cancer has significantly promoted the development of immunotherapy for advanced gastric cancer. Immunotherapy can induce immune responses in gastric cancer patients thus leads to the destruction of cancer cells. In comparison of traditional therapy, immunotherapy has demonstrated robust efficacy and tolerable toxicity. Therefore, this novel strategy for treatment of advanced gastric cancer has gain increasingly popularity. In this review, we summarize recent progress of immunotherapy in advanced gastric cancer, such as immune check point inhibitors, adoptive cell therapy, VEGF inhibitors, cancer vaccines and CAR-T cell therapy. We highlight immunotherapies involved in clinical applications and discuss the existing challenges of current immunotherapies and promising strategies to overcome these limitations.
Collapse
Affiliation(s)
- Xin Jin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China
| | - Zhaorui Liu
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dongxiao Yang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Dongxiao Yang, ; Kai Yin, ; Xusheng Chang,
| | - Kai Yin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Dongxiao Yang, ; Kai Yin, ; Xusheng Chang,
| | - Xusheng Chang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Dongxiao Yang, ; Kai Yin, ; Xusheng Chang,
| |
Collapse
|
52
|
Jin X, Liu Z, Yang D, Yin K, Chang X. Recent Progress and Future Perspectives of Immunotherapy in Advanced Gastric Cancer. Front Immunol 2022; 13:948647. [PMID: 35844558 DOI: 10.3389/fimmu.2022.948647bsd3bmst] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/06/2022] [Indexed: 01/29/2024] Open
Abstract
As one of the most common forms of solid tumours, gastric carcinoma has been revealed as the third leading cause of death worldwide. The symptom of gastric cancer is usually not obvious and thus difficult to detect at earlier stages. Therefore, gastric cancer is already in the advanced stage once detected in patients, which has a poor prognosis due to ineffective therapies and multiple resistance. Recent advance in understanding the microenvironment of cancer has significantly promoted the development of immunotherapy for advanced gastric cancer. Immunotherapy can induce immune responses in gastric cancer patients thus leads to the destruction of cancer cells. In comparison of traditional therapy, immunotherapy has demonstrated robust efficacy and tolerable toxicity. Therefore, this novel strategy for treatment of advanced gastric cancer has gain increasingly popularity. In this review, we summarize recent progress of immunotherapy in advanced gastric cancer, such as immune check point inhibitors, adoptive cell therapy, VEGF inhibitors, cancer vaccines and CAR-T cell therapy. We highlight immunotherapies involved in clinical applications and discuss the existing challenges of current immunotherapies and promising strategies to overcome these limitations.
Collapse
Affiliation(s)
- Xin Jin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China
| | - Zhaorui Liu
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dongxiao Yang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Kai Yin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xusheng Chang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
53
|
Jin X, Liu Z, Yang D, Yin K, Chang X. Recent Progress and Future Perspectives of Immunotherapy in Advanced Gastric Cancer. Front Immunol 2022; 13:948647. [PMID: 35844558 DOI: 10.3389/fimmu.2022.948647' and 2*3*8=6*8 and 'fifm'='fifm] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/06/2022] [Indexed: 01/29/2024] Open
Abstract
As one of the most common forms of solid tumours, gastric carcinoma has been revealed as the third leading cause of death worldwide. The symptom of gastric cancer is usually not obvious and thus difficult to detect at earlier stages. Therefore, gastric cancer is already in the advanced stage once detected in patients, which has a poor prognosis due to ineffective therapies and multiple resistance. Recent advance in understanding the microenvironment of cancer has significantly promoted the development of immunotherapy for advanced gastric cancer. Immunotherapy can induce immune responses in gastric cancer patients thus leads to the destruction of cancer cells. In comparison of traditional therapy, immunotherapy has demonstrated robust efficacy and tolerable toxicity. Therefore, this novel strategy for treatment of advanced gastric cancer has gain increasingly popularity. In this review, we summarize recent progress of immunotherapy in advanced gastric cancer, such as immune check point inhibitors, adoptive cell therapy, VEGF inhibitors, cancer vaccines and CAR-T cell therapy. We highlight immunotherapies involved in clinical applications and discuss the existing challenges of current immunotherapies and promising strategies to overcome these limitations.
Collapse
Affiliation(s)
- Xin Jin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China
| | - Zhaorui Liu
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dongxiao Yang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Kai Yin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xusheng Chang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
54
|
Jin X, Liu Z, Yang D, Yin K, Chang X. Recent Progress and Future Perspectives of Immunotherapy in Advanced Gastric Cancer. Front Immunol 2022; 13:948647. [PMID: 35844558 DOI: 10.3389/fimmu.2022.948647����%2527%2522\'\"] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/06/2022] [Indexed: 01/29/2024] Open
Abstract
As one of the most common forms of solid tumours, gastric carcinoma has been revealed as the third leading cause of death worldwide. The symptom of gastric cancer is usually not obvious and thus difficult to detect at earlier stages. Therefore, gastric cancer is already in the advanced stage once detected in patients, which has a poor prognosis due to ineffective therapies and multiple resistance. Recent advance in understanding the microenvironment of cancer has significantly promoted the development of immunotherapy for advanced gastric cancer. Immunotherapy can induce immune responses in gastric cancer patients thus leads to the destruction of cancer cells. In comparison of traditional therapy, immunotherapy has demonstrated robust efficacy and tolerable toxicity. Therefore, this novel strategy for treatment of advanced gastric cancer has gain increasingly popularity. In this review, we summarize recent progress of immunotherapy in advanced gastric cancer, such as immune check point inhibitors, adoptive cell therapy, VEGF inhibitors, cancer vaccines and CAR-T cell therapy. We highlight immunotherapies involved in clinical applications and discuss the existing challenges of current immunotherapies and promising strategies to overcome these limitations.
Collapse
Affiliation(s)
- Xin Jin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China
| | - Zhaorui Liu
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dongxiao Yang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Kai Yin
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xusheng Chang
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
55
|
Fan Z, He H, Chen L. The Combined Clinical Efficacy and Safety Analysis of Adoptive Immunotherapy with Radiotherapy and Chemotherapy in Non-Small-Cell Lung Cancer: Systematic Review and Meta-Analysis. Appl Bionics Biomech 2022; 2022:2731744. [PMID: 35706510 PMCID: PMC9192301 DOI: 10.1155/2022/2731744] [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: 04/30/2022] [Revised: 05/19/2022] [Accepted: 05/21/2022] [Indexed: 12/03/2022] Open
Abstract
Objective To explore the differential efficacy of chemoradiotherapy combined with adoptive immunotherapy and radiochemotherapy alone in patients with non-small-cell lung cancer (NSCLC). Methods Qualified randomized controlled trial (randomized controlled trial, RCT), or nonrandomized concurrent controlled trial (NRCCT), published in various databases, including PubMed, EMBASE, Chinese journal full-text database, Medline, Cochrane database, and VIP Chinese database, and the Revman5. 0 software performed the data analysis. Results We found the significantly different curative effect between the experimental and control groups (OR = 1.94, 95% CI (1.46, 2.58), P < 0.001, I 2 = 0%, Z = 4.59), effect of adoptive immunotherapy on the progression of disease (OR = 1.80, 95% CI (1.38, 2.35), P < 0.001, I 2 = 0%, Z = 4.33), adoptive immunotherapy on overall survival (OR = 2.19, 95% CI (1.60, 2.99), P < 0.001, I 2 = 0%, Z = 4.91), and adverse effects of adoptive immunotherapy (OR = 1.76, 95% CI (1.25, 2.48), P = 0.001, I 2 = 0%, Z = 3.26). Conclusion Adoptive immunotherapy combined with microradiotherapy can decrease the recurrence of NSCLC and improve patient survival, as well as early patients can be benefited more significantly from immunotherapy.
Collapse
Affiliation(s)
- Zhiming Fan
- Departments of Oncology, The First Naval Hospital of Southern Theater of People's Liberation Army, Zhanjiang, Guangdong, China
| | - Honggui He
- Departments of Oncology, The First Naval Hospital of Southern Theater of People's Liberation Army, Zhanjiang, Guangdong, China
| | - Liqun Chen
- Departments of Oncology, The First Naval Hospital of Southern Theater of People's Liberation Army, Zhanjiang, Guangdong, China
| |
Collapse
|
56
|
The Effects of CD73 on Gastrointestinal Cancer Progression and Treatment. JOURNAL OF ONCOLOGY 2022; 2022:4330329. [PMID: 35620732 PMCID: PMC9130010 DOI: 10.1155/2022/4330329] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 05/04/2022] [Indexed: 11/18/2022]
Abstract
Gastrointestinal (GI) cancer is a common and deadly malignant tumor. CD73, a cell-surface protein, acts as a switch of the adenosine-related signaling pathway that can cause significant immunosuppression. Recent evidence has emerged that CD73 is a promising immunotherapy target for regaining immune cell function and restraining tumorigenesis, and a growing stream of research indicates that combining immunotherapy with other therapies can effectively improve the prognosis and survival of GI cancer patients. Several immune checkpoint inhibitors have been approved for use in GI cancer recently; however, they have demonstrated limited efficacy. Solving the problem of immunosuppression in GI cancer is the key to developing an effective therapeutic option and the modulation of CD73 expression may provide an answer. In this review, we discuss current research on CD73 in gastric, liver, pancreatic, and colorectal cancer to evaluate its therapeutic potential as an immunotherapy target in GI cancers.
Collapse
|
57
|
Feasibility and Tolerance of Apatinib plus PD-1 Inhibitors for Previously Treated Advanced Gastric Cancer: A Real-World Exploratory Study. DISEASE MARKERS 2022; 2022:4322404. [PMID: 35531474 PMCID: PMC9076296 DOI: 10.1155/2022/4322404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/10/2022] [Accepted: 04/15/2022] [Indexed: 12/24/2022]
Abstract
Background Apatinib is established to be the standard of care as third-line therapy for patients with previously treated advanced gastric cancer (GC). Programmed cell death protein 1 (PD-1) blockades also exhibited promising efficacy and safety for patients with treatment-refractory advanced GC. Objective This study was to explore the feasibility and tolerance of apatinib plus PD-1 inhibitors for patients with previously treated advanced GC. Methods This study was performed as a real-world study; patients with advanced GC who were treated with previous systemic chemotherapy were screened retrospectively. Eligible patients were administered with apatinib combined with PD-1 blockade treatment. Efficacy of the patients was assessed with the change of target lesion using radiological evidence according to RECIST 1.1 criteria, and follow-up was carried out regularly. A safety profile was collected and documented during the combination treatment. Univariate analysis based on baseline characteristic subgroup was implemented in univariate analysis to identify the potential factor that might contribute to progression-free survival (PFS). Results Between August 2018 and October 2021, a total of 39 patients with advanced GC or gastroesophageal junction adenocarcinoma participated in this study consecutively and all the patients were available for efficacy and safety assessment. The best overall response during apatinib plus PD-1 blockade administration exhibited that PR was observed in 8 patients, SD was noted in 19 patients, and PD was found in 12 patients, which yielded an ORR of 20.5% (95% CI: 9.3%-36.5%), and DCR was 69.2% (95% CI: 52.4%-83.0%). Furthermore, the relatively enough follow-up had resulted in the mature PFS and overall survival (OS) data, suggesting that the median PFS of the 39 patients with advanced GC was 3.9 months (95% CI: 2.74-5.06). Additionally, the median OS of the 39 patients with advanced GC was 7.8 months (95% CI: 4.82-10.78). Furthermore, the most common adverse reactions of the 39 patients who received apatinib plus PD-1 blockades treatment were fatigue (61.5%), nausea and vomiting (56.4%), diarrhea (48.7%), hypertension (46.2%), hand-foot syndrome (38.5%), and rash (28.2%). Furthermore, performance status was independently associated with PFS of apatinib plus PD-1 inhibitor combination administration in baseline characteristic subgroup analysis. Conclusion Apatinib plus PD-1 inhibitors exhibited promising effectiveness and acceptable tolerance for previously treated advanced GC preliminarily. And this conclusion should be confirmed in clinical trials in the future.
Collapse
|
58
|
An Immunity-Associated lncRNA Signature for Predicting Prognosis in Gastric Adenocarcinoma. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:3035073. [PMID: 35509706 PMCID: PMC9061059 DOI: 10.1155/2022/3035073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/05/2022] [Accepted: 03/31/2022] [Indexed: 11/18/2022]
Abstract
Background Gastric adenocarcinoma (GAD) is one of the most common tumors in the world and the prognosis is still very poor. Objective We sought to identify reliable prognostic biomarkers for the progression of GAD and the sensitivity to drug therapy. Method The RNA sequencing data of GAD was downloaded from the Cancer Genome Atlas (TCGA) database and used for analysis. Differentially expressed, immune-related lncRNA (DEIRlncRNA) was characterized by differential analysis and correlation analysis. Univariate Cox regression analysis was used to identify DEIRlncRNA associated with prognosis. Least absolute shrinkage and selection operator (LASSO) regression analysis allowed us to determine a signature composed of eight IRlncRNAs. Based on this signature, we further performed gene set enrichment analysis (GSEA) and somatic mutation analysis to evaluate the ability of this signature to predict prognosis. Results In total, 72 immune-related lncRNAs (DEIRlncRNAs) with prognostic value were identified. These lncRNAs were used to construct a model containing eight immune-related lncRNAs (8-IRlncRNAs). Based on this risk model, we divided GAD patients into high-risk and low-risk groups. The analysis showed that the prognosis of the two groups was different and that the high-risk group had worse overall survival (OS). Immune cell infiltration analysis showed that the proportion of memory B cells increased in the high-risk group while the proportion of macrophages M1, T cells, CD4 memory-activated cells, and T cell follicular helpers decreased. GSEA results showed that 8-IRlncRNA was significantly enriched in tumorigenesis pathways such as myc. The results of somatic mutation analysis showed that the CDH1 gene was significantly mutated in the high-risk group. Conclusion A prognostic signature of 8-IRlncRNAs in GAD was established and this signature was able to predict the prognosis of GAD patients.
Collapse
|
59
|
Pan Y, Si H, Deng G, Chen S, Zhang N, Zhou Q, Wang Z, Dai G. A Composite Biomarker of Derived Neutrophil–Lymphocyte Ratio and Platelet–Lymphocyte Ratio Correlates With Outcomes in Advanced Gastric Cancer Patients Treated With Anti-PD-1 Antibodies. Front Oncol 2022; 11:798415. [PMID: 35251952 PMCID: PMC8895371 DOI: 10.3389/fonc.2021.798415] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/29/2021] [Indexed: 12/25/2022] Open
Abstract
BackgroundThe highly heterogeneous characteristics of GC may limit the accuracy of a single biomarker for screening populations benefiting from immunotherapy. However, the combination of multiple indicators can provide more directed information for the detection of potential immune benefit subgroups. At present, there are no recognized complex indexes to identify advanced GC (AGC) in patients who likely benefited from immunotherapy. The objective of this research is to explore whether the composite biomarker of derived neutrophil–lymphocyte ratio (dNLR) and platelet–lymphocyte ratio (PLR) can be used as a reliable prognostic factor for the survival of AGC patients receiving immunotherapy.MethodsFrom December 2014 to May 2021, a total 238 AGC patients at a single Center were included in this retrospective cohort research study. The cutoff value of dNLR was obtained by the ROC curves to predict the disease progression rate at the 8th month and the cutoff value of PLR was estimated by the median value. The cutoff values of dNLR and PLR were 1.95 and 163.63, respectively. The high levels of dNLR (≥1.95) and PLR (≥163.63) were considered to be risk factors. Based on these two risk factors, patients were categorized into 3 groups: the risk factor number for the “good” group was 0, that for the “intermediate” group was 1, and that for the “poor” group was 2. The subjects were divided into two groups: dNLR/PLR-good and dNLR/PLR-intermediate/poor.ResultsOf the 238 patients, the median overall survival (mOS) and progression-free survival (mPFS) were 12.5 and 4.7 months, respectively. Multivariate analysis revealed that the good dNLR/PLR group was independently associated with better prognosis. The intermediate/poor dNLR/PLR group was independently correlated with an over 1.4 times greater risk of disease progression (4.1 months vs. 5.5 months; p = 0.016) and an over 1.54 times greater risk of death (11.1 months vs. 26.3 months; p = 0.033) than the good dNLR/PLR group. However, no clear differences in the disease control rate (DCR) and overall response rate (ORR) were observed between the intermediate/poor dNLR/PLR group and the good dNLR/PLR group (51.5% vs. 56.3%, 26.3% vs. 29.6%; p = 0.494, p = 0.609).ConclusionOur study firstly verifies that the composite biomarker of dNLR and PLR is an independent prognostic factor affecting survival of advanced AGC patients receiving immunotherapy. It may be difficult for patients with the intermediate/poor dNLR/PLR group to benefit from immunotherapy.
Collapse
Affiliation(s)
- Yuting Pan
- Chinese People’s Liberation Army Medical School, Beijing, China
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Haiyan Si
- Chinese People’s Liberation Army Medical School, Beijing, China
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Guochao Deng
- Chinese People’s Liberation Army Medical School, Beijing, China
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Shiyun Chen
- Chinese People’s Liberation Army Medical School, Beijing, China
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Nan Zhang
- Chinese People’s Liberation Army Medical School, Beijing, China
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Qian Zhou
- Chinese People’s Liberation Army Medical School, Beijing, China
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - ZhiKuan Wang
- Chinese People’s Liberation Army Medical School, Beijing, China
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
- *Correspondence: ZhiKuan Wang, ; Guanghai Dai,
| | - Guanghai Dai
- Chinese People’s Liberation Army Medical School, Beijing, China
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
- *Correspondence: ZhiKuan Wang, ; Guanghai Dai,
| |
Collapse
|
60
|
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
|
61
|
Jiang H, Yu D, Yang P, Guo R, Kong M, Gao Y, Yu X, Lu X, Fan X. Revealing the transcriptional heterogeneity of organ-specific metastasis in human gastric cancer using single-cell RNA Sequencing. Clin Transl Med 2022; 12:e730. [PMID: 35184420 PMCID: PMC8858624 DOI: 10.1002/ctm2.730] [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: 07/06/2021] [Revised: 01/22/2022] [Accepted: 01/25/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Deciphering intra- and inter-tumoural heterogeneity is essential for understanding the biology of gastric cancer (GC) and its metastasis and identifying effective therapeutic targets. However, the characteristics of different organ-tropism metastases of GC are largely unknown. METHODS Ten fresh human tissue samples from six patients, including primary tumour and adjacent non-tumoural samples and six metastases from different organs or tissues (liver, peritoneum, ovary, lymph node) were evaluated using single-cell RNA sequencing. Validation experiments were performed using histological assays and bulk transcriptomic datasets. RESULTS Malignant epithelial subclusters associated with invasion features, intraperitoneal metastasis propensity, epithelial-mesenchymal transition-induced tumour stem cell phenotypes, or dormancy-like characteristics were discovered. High expression of the first three subcluster-associated genes displayed worse overall survival than those with low expression in a GC cohort containing 407 samples. Immune and stromal cells exhibited cellular heterogeneity and created a pro-tumoural and immunosuppressive microenvironment. Furthermore, a 20-gene signature of lymph node-derived exhausted CD8+ T cells was acquired to forecast lymph node metastasis and validated in GC cohorts. Additionally, although anti-NKG2A (KLRC1) antibody have not been used to treat GC patients even in clinical trials, we uncovered not only malignant tumour cells but one endothelial subcluster, mucosal-associated invariant T cells, T cell-like B cells, plasmacytoid dendritic cells, macrophages, monocytes, and neutrophils may contribute to HLA-E-KLRC1/KLRC2 interaction with cytotoxic/exhausted CD8+ T cells and/or natural killer (NK) cells, suggesting novel clinical therapeutic opportunities in GC. Additionally, our findings suggested that PD-1 expression in CD8+ T cells might predict clinical responses to PD-1 blockade therapy in GC. CONCLUSIONS This study provided insights into heterogeneous microenvironment of GC primary tumours and organ-specific metastases and provide support for precise diagnosis and treatment.
Collapse
Affiliation(s)
- Haiping Jiang
- Department of Medical OncologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Dingyi Yu
- Pharmaceutical Informatics InstituteCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Penghui Yang
- Pharmaceutical Informatics InstituteCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Rongfang Guo
- Pharmaceutical Informatics InstituteCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Mei Kong
- Department of PathologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Yuan Gao
- Department of Gastro‐Intestinal SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Xiongfei Yu
- Department of Surgical OncologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Xiaoyan Lu
- Pharmaceutical Informatics InstituteCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- State Key Laboratory of Component‐Based Chinese MedicineInnovation Center in Zhejiang UniversityHangzhouChina
| | - Xiaohui Fan
- Pharmaceutical Informatics InstituteCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- State Key Laboratory of Component‐Based Chinese MedicineInnovation Center in Zhejiang UniversityHangzhouChina
- Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
| |
Collapse
|
62
|
The LINC00152/miR-138 Axis Facilitates Gastric Cancer Progression by Mediating SIRT2. JOURNAL OF ONCOLOGY 2021; 2021:1173869. [PMID: 34697541 PMCID: PMC8541877 DOI: 10.1155/2021/1173869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/15/2021] [Accepted: 10/06/2021] [Indexed: 02/06/2023]
Abstract
Gastric cancer (GC) is the most common gastrointestinal cancer and the main cause of tumor-related death. Exploring markers for early diagnosis and new therapeutic targets is always on the way. In the last 10 years, long noncoding RNAs (lncRNAs) have been widely proved to be involved in the progress of many tumors and are regarded as potential targets for tumor therapy. We found that LINC00152, a newly identified lncRNA, was significantly upregulated in GC tissues and affected clinicopathological characteristics in GC patients. Furthermore, we observed that LINC00152 knockdown can significantly reduce cell proliferation and promote apoptosis in human gastric cancer cells. Further bioinformatic analysis indicated that LINC00152 competitively bound with miR-138 and regulated the expression of miR-138. Moreover, SIRT2 was further proved to be a downstream target of miR-138. Overall, this study elucidates the molecular mechanism of LINC00152 underlying the malignant phenotype of GC cells by mediating miR-138/SIRT2 axis, which provides a new understanding of the role and molecular mechanism of lncRNA in GC and also provides a new way for the treatment of gastric cancer.
Collapse
|
63
|
Song Q, Zheng Y, Wu J, Wang S, Meng L, Yao Q, Li Z, Lian S. PTP4A3 Is a Prognostic Biomarker Correlated With Immune Infiltrates in Papillary Renal Cell Carcinoma. Front Immunol 2021; 12:717688. [PMID: 34630392 PMCID: PMC8495008 DOI: 10.3389/fimmu.2021.717688] [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: 05/31/2021] [Accepted: 08/31/2021] [Indexed: 12/24/2022] Open
Abstract
PTP4A3 plays an important role in the tumorigenesis and metastasis of multiple tumors, but its prognostic role in renal cancer is not well understood. We utilized the Oncomine and Tumor Immunoassay Resource databases to examine the differential expression of PTP4A3 in tumor tissues and normal tissues in breast, urinary tract, gastrointestinal tract and skin. Using the GEPIA and PrognoScan databases, the independent prognostic role of PTP4A3 was confirmed in clear cell renal cell cancer and papillary renal cell cancer. Expression of PTP4A3 were obviously higher in tumor tissue compare with normal tissues (P=0.028). We haven’t found the associations of PTP4A3 and clinicopathological features in our IHC cohort. Ectopic expression of PTP4A3 promotes proliferation, migration and invasion and increased the mRNA level of TGFB1 in RCC cell lines. Immunohistochemical staining indicated that the expression of PTP4A3 associates with CD3+ (P =0.037)/CD8+ (P =0.037) intratumor TILs, not with invasive margins in renal cancer. Comprehensive analysis of immune infiltration in the TIMER database correlated PTP4A3 expression with the infiltration of B cells, CD8+ T cells, CD4+ T cells and neutrophils in both clear cell renal cell carcinoma and papillary renal cell carcinoma. PTP4A3 expression was associated with the infiltration of dendritic cells in papillary renal cell carcinoma. We further confirmed that the infiltration of B cells and CD8+ T cells was associated with poor prognosis in papillary renal cell carcinoma patients, consistent with the prognostic role of PTP4A3 in papillary renal cell carcinoma. PTP4A3 expression correlated genes involved in B cells, monocytes, M1 macrophages, Th2 and Treg cells in papillary renal cell carcinoma. These results suggest PTP4A3 as a prognostic factor with a role in regulating immune cell infiltration in papillary renal cell carcinoma.
Collapse
Affiliation(s)
- Qian Song
- Department of Clinical Laboratory, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital); Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yutian Zheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Junzhou Wu
- Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital); Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Sheng Wang
- Department of Clinical Laboratory, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital); Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Lin Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qian Yao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhongwu Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Shenyi Lian
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
64
|
Wei XL, Luo TQ, Li JN, Xue ZC, Wang Y, Zhang Y, Chen YB, Peng C. Development and Validation of a Prognostic Classifier Based on Lipid Metabolism-Related Genes in Gastric Cancer. Front Mol Biosci 2021; 8:691143. [PMID: 34277706 PMCID: PMC8277939 DOI: 10.3389/fmolb.2021.691143] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/07/2021] [Indexed: 01/23/2023] Open
Abstract
Background: Dysregulation of lipid metabolism plays important roles in the tumorigenesis and progression of gastric cancer (GC). The present study aimed to establish a prognostic model based on the lipid metabolism–related genes in GC patients. Materials and Methods: Two GC datasets from the Gene Expression Atlas, GSE62254 (n = 300) and GSE26942 (n = 217), were used as training and validation cohorts to establish a risk predictive scoring model. The efficacy of this model was assessed by ROC analysis. The association of the risk predictive scores with patient characteristics and immune cell subtypes was evaluated. A nomogram was constructed based on the risk predictive score model and other prognostic factors. Results: A risk predictive score model was established based on the expression of 19 lipid metabolism–related genes (LPL, IPMK, PLCB3, CDIPT, PIK3CA, DPM2, PIGZ, GPD2, GPX3, LTC4S, CYP1A2, GALC, SGMS1, SMPD2, SMPD3, FUT6, ST3GAL1, B4GALNT1, and ACADS). The time-dependent ROC analysis revealed that the risk predictive score model was stable and robust. Patients with high risk scores had significantly unfavorable overall survival compared with those with low risk scores in both the training and validation cohorts. A higher risk score was associated with more aggressive features, including a higher tumor grade, a more advanced TNM stage, and diffuse type of Lauren classification of GC. Moreover, distinct immune cell subtypes and signaling pathways were found between the high–risk and low–risk score groups. A nomogram containing patients’ age, tumor stage, adjuvant chemotherapy, and the risk predictive score could accurately predict the survival probability of patients at 1, 3, and 5 years. Conclusion: A novel 19-gene risk predictive score model was developed based on the lipid metabolism–related genes, which could be a potential prognostic indicator and therapeutic target of GC.
Collapse
Affiliation(s)
- Xiao-Li Wei
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Tian-Qi Luo
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jia-Ning Li
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zhi-Cheng Xue
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yun Wang
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - You Zhang
- Zhongshan School of Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ying-Bo Chen
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Chuan Peng
- Department of Ultrasound, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|