51
|
Jiang S, Zhang Y, Zhang X, Lu B, Sun P, Wu Q, Ding X, Huang J. GARP Correlates With Tumor-Infiltrating T-Cells and Predicts the Outcome of Gastric Cancer. Front Immunol 2021; 12:660397. [PMID: 34421887 PMCID: PMC8378229 DOI: 10.3389/fimmu.2021.660397] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022] Open
Abstract
Accepting the crucial role of the immune microenvironment (TME) in tumor progression enables us to identify immunotherapeutic targets and develop new therapies. Glycoprotein A repetitions predominant (GARP) plays a vital part in maintaining regulatory T cell (Treg)-mediated immune tolerance. The impact of GARP in TME of gastric cancer is still worth exploring. We investigated public genomic datasets from The Cancer Genome Atlas and Gene Expression Omnibus to analyze the possible role of GARP and its relationship with TME of gastric cancer. Fluorescence-based multiplex immunohistochemistry and immunohistochemistry for T-cell immune signatures in a series of tissue microarrays were used to validate the value of GARP in the TME. We initially found that GARP expression was upregulated in gastric carcinoma cells, and diverse levels o3f immune cell infiltration and immune checkpoint expression were detected. Gene expression profiling revealed that GARP expression was related to the TME of gastric cancer. GARP upregulation was usually accompanied by increased FOXP3+ Treg and CD4+ T cell infiltration. In addition, GARP expression had positive relationships with CTLA-4 and PD-L1 expression in gastric cancer. Cox regression analysis and a nomogram highlighted that the probability of poor overall survival was predicted well by GARP or GARP+CD4+ T cell. Taken together, this research underlines the potential effect of GARP in regulating survival and tumor-infiltrating T-cells. In addition, the function of CD4+ T cell immune signatures in the prognosis can be clinically meaningful, thereby providing a new idea for the immunotherapeutic approach.
Collapse
Affiliation(s)
- Sutian Jiang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, China.,Department of Pathology and Pathophysiology, School of Medicine, Nantong University, Nantong, China
| | - Yifan Zhang
- Clinical Medicine, Xian Medical University, Xi'an, China
| | - Xiaojing Zhang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, China
| | - Bing Lu
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, China
| | - Pingping Sun
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, China
| | - Qianqian Wu
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, China
| | - Xuzhong Ding
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, China
| | - Jianfei Huang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, China.,Translational Medicine Center, The Affiliated Kezhou People's Hospital of Nanjing Medical University, Kezhou, China
| |
Collapse
|
52
|
Li F, Li J, Yu J, Pan T, Yu B, Sang Q, Dai W, Hou J, Yan C, Zang M, Zhu Z, Su L, Li YY, Liu B. Identification of ARGLU1 as a potential therapeutic target for gastric cancer based on genome-wide functional screening data. EBioMedicine 2021; 69:103436. [PMID: 34157484 PMCID: PMC8220577 DOI: 10.1016/j.ebiom.2021.103436] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/21/2021] [Accepted: 05/27/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Due to the molecular mechanism complexity and heterogeneity of gastric cancer (GC), mechanistically interpretable biomarkers were required for predicting prognosis and discovering therapeutic targets for GC patients. METHODS Based on a total of 824 GC-specific fitness genes from the Project Score database, LASSOCox regression was performed in TCGA-STAD cohort to construct a GC Prognostic (GCP) model which was then evaluated on 7 independent GC datasets. Targets prioritization was performed in GC organoids. ARGLU1 was selected to further explore the biological function and molecular mechanism. We evaluated the potential of ARGLU1 serving as a promising therapeutic target for GC using patients derived xenograft (PDX) model. FINDINGS The 9-gene GCP model showed a statistically significant prognostic performance for GC patients in 7 validation cohorts. Perturbation of SSX4, DDX24, ARGLU1 and TTF2 inhibited GC organoids tumor growth. The results of tissue microarray indicated lower expression of ARGLU1 was correlated with advanced TNM stage and worse overall survival. Over-expression ARGLU1 significantly inhibited GC cells viability in vitro and in vivo. ARGLU1 could enhance the transcriptional level of mismatch repair genes including MLH3, MSH2, MSH3 and MSH6 by potentiating the recruitment of SP1 and YY1 on their promoters. Moreover, inducing ARGLU1 by LNP-formulated saRNA significantly inhibited tumor growth in PDX model. INTERPRETATION Based on genome-wide functional screening data, we constructed a 9-gene GCP model with satisfactory predictive accuracy and mechanistic interpretability. Out of nine prognostic genes, ARGLU1 was verified to be a potential therapeutic target for GC. FUNDING National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Fangyuan Li
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Jianfang Li
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Junxian Yu
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Tao Pan
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Beiqin Yu
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Qingqing Sang
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Wentao Dai
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China; Shanghai Center for Bioinformation Technology, Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai 201203, PR China
| | - Junyi Hou
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Chao Yan
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Mingde Zang
- Department of Gastric Cancer Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, 200032 Shanghai, PR China
| | - Zhenggang Zhu
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Liping Su
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Yuan-Yuan Li
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China; Shanghai Center for Bioinformation Technology, Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai 201203, PR China.
| | - Bingya Liu
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China.
| |
Collapse
|
53
|
Liu F, Yang Z, Zheng L, Shao W, Cui X, Wang Y, Jia J, Fu Y. A Tumor Progression Related 7-Gene Signature Indicates Prognosis and Tumor Immune Characteristics of Gastric Cancer. Front Oncol 2021; 11:690129. [PMID: 34195091 PMCID: PMC8238374 DOI: 10.3389/fonc.2021.690129] [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: 04/02/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Gastric cancer is a common gastrointestinal malignancy. Since it is often diagnosed in the advanced stage, its mortality rate is high. Traditional therapies (such as continuous chemotherapy) are not satisfactory for advanced gastric cancer, but immunotherapy has shown great therapeutic potential. Gastric cancer has high molecular and phenotypic heterogeneity. New strategies for accurate prognostic evaluation and patient selection for immunotherapy are urgently needed. METHODS Weighted gene coexpression network analysis (WGCNA) was used to identify hub genes related to gastric cancer progression. Based on the hub genes, the samples were divided into two subtypes by consensus clustering analysis. After obtaining the differentially expressed genes between the subtypes, a gastric cancer risk model was constructed through univariate Cox regression, least absolute shrinkage and selection operator (LASSO) regression and multivariate Cox regression analysis. The differences in prognosis, clinical features, tumor microenvironment (TME) components and immune characteristics were compared between subtypes and risk groups, and the connectivity map (CMap) database was applied to identify potential treatments for high-risk patients. RESULTS WGCNA and screening revealed nine hub genes closely related to gastric cancer progression. Unsupervised clustering according to hub gene expression grouped gastric cancer patients into two subtypes related to disease progression, and these patients showed significant differences in prognoses, TME immune and stromal scores, and suppressive immune checkpoint expression. Based on the different expression patterns between the subtypes, we constructed a gastric cancer risk model and divided patients into a high-risk group and a low-risk group based on the risk score. High-risk patients had a poorer prognosis, higher TME immune/stromal scores, higher inhibitory immune checkpoint expression, and more immune characteristics suitable for immunotherapy. Multivariate Cox regression analysis including the age, stage and risk score indicated that the risk score can be used as an independent prognostic factor for gastric cancer. On the basis of the risk score, we constructed a nomogram that relatively accurately predicts gastric cancer patient prognoses and screened potential drugs for high-risk patients. CONCLUSIONS Our results suggest that the 7-gene signature related to tumor progression could predict the clinical prognosis and tumor immune characteristics of gastric cancer.
Collapse
Affiliation(s)
- Fen Liu
- Department of Microbiology/Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zongcheng Yang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Lixin Zheng
- Department of Microbiology/Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Shao
- Department of Microbiology/Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiujie Cui
- Department of Microbiology/Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue Wang
- Department of Microbiology/Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jihui Jia
- Department of Microbiology/Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue Fu
- School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
54
|
Shao W, Yang Z, Fu Y, Zheng L, Liu F, Chai L, Jia J. The Pyroptosis-Related Signature Predicts Prognosis and Indicates Immune Microenvironment Infiltration in Gastric Cancer. Front Cell Dev Biol 2021; 9:676485. [PMID: 34179006 PMCID: PMC8226259 DOI: 10.3389/fcell.2021.676485] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/19/2021] [Indexed: 01/20/2023] Open
Abstract
Gastric cancer (GC) is one of the leading causes of cancer-related deaths and shows high levels of heterogeneity. The development of a specific prognostic model is important if we are to improve treatment strategies. Pyroptosis can arise in response to H. pylori, a primary carcinogen, and also in response to chemotherapy drugs. However, the prognostic evaluation of GC to pyroptosis is insufficient. Consensus clustering by pyroptosis-related regulators was used to classify 618 patients with GC from four GEO cohorts. Following Cox regression with differentially expressed genes, our prognosis model (PS-score) was built by LASSO-Cox analysis. The TCGA-STAD cohort was used as the validation set. ESTIMATE, CIBERSORTx, and EPIC were used to investigate the tumor microenvironment (TME). Immunotherapy cohorts by blocking PD1/PD-L1 were used to investigate the treatment response. The subtyping of GC based on pyroptosis-related regulators was able to classify patients according to different clinical traits and TME. The difference between the two subtypes identified in this study was used to develop a prognosis model which we named “PS-score.” The PS-score could predict the prognosis of patients with GC and his/her overall survival time. A low PS-score implies greater inflammatory cell infiltration and better response of immunotherapy by PD1/PD-L1 blockers. Our findings provide a foundation for future research targeting pyroptosis and its immune microenvironment to improve prognosis and responses to immunotherapy.
Collapse
Affiliation(s)
- Wei Shao
- Key Laboratory for Experimental Teratology of The Chinese Ministry of Education, Department of Microbiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zongcheng Yang
- School of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue Fu
- Key Laboratory for Experimental Teratology of The Chinese Ministry of Education, Department of Microbiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China.,School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lixin Zheng
- Key Laboratory for Experimental Teratology of The Chinese Ministry of Education, Department of Microbiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fen Liu
- Key Laboratory for Experimental Teratology of The Chinese Ministry of Education, Department of Microbiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Li Chai
- Key Laboratory for Experimental Teratology of The Chinese Ministry of Education, Department of Microbiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jihui Jia
- Key Laboratory for Experimental Teratology of The Chinese Ministry of Education, Department of Microbiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong University-Karolinska Institutet Collaborative Laboratory for Cancer Research, Jinan, China
| |
Collapse
|
55
|
Hu J, Yu H, Sun L, Yan Y, Zhang L, Jiang G, Zhang P. Identification of an Individualized Metabolism Prognostic Signature and Related Therapy Regimens in Early Stage Lung Adenocarcinoma. Front Oncol 2021; 11:650853. [PMID: 33996569 PMCID: PMC8113858 DOI: 10.3389/fonc.2021.650853] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/30/2021] [Indexed: 12/25/2022] Open
Abstract
Objective The choice of adjuvant therapy for early stage lung adenocarcinoma (LUAD) remains controversial. Identifying the metabolism characteristics leading to worse prognosis may have clinical utility in offering adjuvant therapy. Methods The gene expression profiles of LUAD were collected from 22 public datasets. The patients were divided into a meta-training cohort (n = 790), meta-testing cohort (n = 716), and three independent validation cohorts (n = 345, 358, and 321). A metabolism-related gene pair index (MRGPI) was trained and validated in the cohorts. Subgroup analyses regarding tumor stage and adjuvant chemotherapy (ACT) were performed. To explore potential therapeutic targets, we performed in silico analysis of the MRGPI. Results Through machine learning, MRGPI consisting of 12 metabolism-related gene pairs was constructed. MRGPI robustly stratified patients into high- vs low-risk groups in terms of overall survival across and within subpopulations with stage I or II disease in all cohorts. Multivariable analysis confirmed that MRGPI was an independent prognostic factor. ACT could not improve prognosis in high-risk patients with stage I disease, but could improve prognosis in the high-risk patients with stage II disease. In silico analysis indicated that B3GNT3 (overexpressed in high-risk patients) and HSD17B6 (down-expressed in high-risk patients) may make synergic reaction in immune evasion by the PD-1/PD-L1 pathway. When integrated with clinical characteristics, the composite clinical and metabolism signature showed improved prognostic accuracy. Conclusions MRGPI could effectively predict prognosis of the patients with early stage LUAD. The patients at high risk may get survival benefit from PD-1/PD-L1 blockade (stage I) or combined with chemotherapy (stage II).
Collapse
Affiliation(s)
- Junjie Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huansha Yu
- Experimental Animal Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liangdong Sun
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yilv Yan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lele Zhang
- Central Laboratory of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Gening Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
56
|
Guan X, Xu ZY, Chen R, Qin JJ, Cheng XD. Identification of an Immune Gene-Associated Prognostic Signature and Its Association With a Poor Prognosis in Gastric Cancer Patients. Front Oncol 2021; 10:629909. [PMID: 33628738 PMCID: PMC7898907 DOI: 10.3389/fonc.2020.629909] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/21/2020] [Indexed: 12/13/2022] Open
Abstract
The immune response plays a critical role in gastric cancer (GC) development, metastasis, and treatment. A better understanding of the tumor-immune system interactions in gastric cancer may provide promising diagnostic, prognostic, and therapeutic biomarkers for patients with this disease. In the present study, we aimed to identify a prognostic signature of GC through a comprehensive bioinformatics analysis on the tumor-immune interactions as well as the molecular characteristics. We firstly identified two immunophenotypes and immunological characteristics by employing multiple algorithms, such as the single sample Gene Sets Enrichment Analysis and Cell type Identification By Estimating Relative Subsets of RNA Transcripts. Next, we developed a six-immune-gene signature as a promising independent prognostic biomarker for GC using Lasso Cox regression and verified it via the external validation set and systematically correlated the immune signature with GC clinicopathologic features and genomic characteristics. Finally, a nomogram was successfully constructed based on the immune signature and clinical characteristics and showed a high potential for GC prognosis prediction. This study may shed light on the treatment strategies for GC patients from the perspective of immunology.
Collapse
Affiliation(s)
- Xiaoqing Guan
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhi-Yuan Xu
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Runzhe Chen
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jiang-Jiang Qin
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xiang-Dong Cheng
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
57
|
Wen C, Wang H, Wang H, Mo H, Zhong W, Tang J, Lu Y, Zhou W, Tan A, Liu Y, Xie W. A three-gene signature based on tumour microenvironment predicts overall survival of osteosarcoma in adolescents and young adults. Aging (Albany NY) 2020; 13:619-645. [PMID: 33281116 PMCID: PMC7835013 DOI: 10.18632/aging.202170] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023]
Abstract
Evidences shows that immune and stroma related genes in the tumour microenvironment (TME) play a key regulator in the prognosis of Osteosarcomas (OSs). The purpose of this study was to develop a TME-related risk model for assessing the prognosis of OSs. 82 OSs cases aged ≤25 years from TARGET were divided into two groups according to the immune/stromal scores that were analyzed by the Estimate algorithm. The differentially expressed genes (DEGs) between the two groups were analyzed and 122 DEGs were revealed. Finally, three genes (COCH, MYOM2 and PDE1B) with the minimum AIC value were derived from 122 DEGs by multivariate cox analysis. The three-gene risk model (3-GRM) could distinguish patients with high risk from the training (TARGET) and validation (GSE21257) cohort. Furthermore, a nomogram model included 3-GRM score and clinical features were developed, with the AUC values in predicting 1, 3 and 5-year survival were 0.971, 0.853 and 0.818, respectively. In addition, in the high 3-GRM score group, the enrichment degrees of infiltrating immune cells were significantly lower and immune-related pathways were markedly suppressed. In summary, this model may be used as a marker to predict survival for OSs patients in adolescent and young adults.
Collapse
Affiliation(s)
- Chunkai Wen
- Department of Breast, Bone and Soft Tissue Oncology, the Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China.,Graduate School of Guangxi Medical University, Nanning 530021, China
| | - Hongxue Wang
- Department of Breast, Bone and Soft Tissue Oncology, the Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Han Wang
- Department of Breast, Bone and Soft Tissue Oncology, the Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Hao Mo
- Department of Bone and Soft Tissue Tumor Surgery, the Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Wuning Zhong
- Department of Breast, Bone and Soft Tissue Oncology, the Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jing Tang
- Department of Breast, Bone and Soft Tissue Oncology, the Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yongkui Lu
- Department of Breast, Bone and Soft Tissue Oncology, the Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Wenxian Zhou
- Department of Breast, Bone and Soft Tissue Oncology, the Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Aihua Tan
- Department of Breast, Bone and Soft Tissue Oncology, the Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yan Liu
- Department of Breast, Bone and Soft Tissue Oncology, the Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Weimin Xie
- Department of Breast, Bone and Soft Tissue Oncology, the Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| |
Collapse
|