scholarly journals Targeting UCHL1 Induces Cell Cycle Arrest in High-Risk Multiple Myeloma with t(4;14)

2021 ◽  
Vol 27 ◽  
Author(s):  
Parin Kamseng ◽  
Teerapong Siriboonpiputtana ◽  
Teeraya Puavilai ◽  
Suporn Chuncharunee ◽  
Karan Paisooksantivatana ◽  
...  

Multiple myeloma (MM) patients considered to be at high cytogenetic risk commonly fail to respond to standard treatment. A thorough understanding of the molecular mechanism of MM development is, therefore, needed. We endeavored to explore the transcriptional signature among different subgroups of newly diagnosed MM using gene chip-based expression microarray. Bone marrow samples of 15 newly diagnosed Thai MM patients were included. The chromosomal translocation t(4;14) was the most frequently identified genetic alteration in the high-risk subgroup. Cluster analysis from expression profiling demonstrated that high-risk MM have a distinctly different expression pattern compared to standard-risk patients. The most significant differentially expressed gene was UCHL1. Functional enrichment analysis by Gene Set Enrichment Analysis, FUNRICH, and Gene Ontology Panther pathway revealed the gene sets involved in cell cycle control to be enriched in the t(4;14) high-risk group. Interestingly, among the well-established downstream targets of UCHL1, only CCND2 was significantly expressed in the t(4;14) high-risk group. Suppression of UCHL1 protein level by LDN-5744 inhibitor could arrest the cell cycle in G1 phase in cell lines. These findings shed light on the molecular mechanism of UCHL1 in t(4;14) high-risk MM and support the evidence that alteration of the UCHL1 pathway may play a role in the pathogenesis of high-risk MM.

2021 ◽  
Author(s):  
Yanjia Hu ◽  
Jing Zhang ◽  
Jing Chen

Abstract Background Hypoxia-related long non-coding RNAs (lncRNAs) have been proven to play a role in multiple cancers and can serve as prognostic markers. Lower-grade gliomas (LGGs) are characterized by large heterogeneity. Methods This study aimed to construct a hypoxia-related lncRNA signature for predicting the prognosis of LGG patients. Transcriptome and clinical data of LGG patients were obtained from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA). LGG cohort in TCGA was chosen as training set and LGG cohorts in CGGA served as validation sets. A prognostic signature consisting of fourteen hypoxia-related lncRNAs was constructed using univariate and LASSO Cox regression. A risk score formula involving the fourteen lncRNAs was developed to calculate the risk score and patients were classified into high- and low-risk groups based on cutoff. Kaplan-Meier survival analysis was used to compare the survival between two groups. Cox regression analysis was used to determine whether risk score was an independent prognostic factor. A nomogram was then constructed based on independent prognostic factors and assessed by C-index and calibration plot. Gene set enrichment analysis and immune cell infiltration analysis were performed to uncover further mechanisms of this lncRNA signature. Results LGG patients with high risk had poorer prognosis than those with low risk in both training and validation sets. Recipient operating characteristic curves showed good performance of the prognostic signature. Univariate and multivariate Cox regression confirmed that the established lncRNA signature was an independent prognostic factor. C-index and calibration plots showed good predictive performance of nomogram. Gene set enrichment analysis showed that genes in the high-risk group were enriched in apoptosis, cell adhesion, pathways in cancer, hypoxia etc. Immune cells were higher in high-risk group. Conclusion The present study showed the value of the 14-lncRNA signature in predicting survival of LGGs and these 14 lncRNAs could be further investigated to reveal more mechanisms involved in gliomas.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5593-5593
Author(s):  
Andrey Garifullin ◽  
Sergei Voloshin ◽  
Vasily Shuvaev ◽  
Irina Martynkevich ◽  
Elizaveta Kleina ◽  
...  

Background The risk-stratification systems are repeatedly updated in accordance with the emergence of new information about the prognostic impact of anomalies and other factors. The most extensive and modern system in this time is mSMART risk stratification involving many parameters such as genetic anomalies, albumin, beta-2-microglobulin, LDH, Plasma Cell S-phase and GEP levels. It is possible to use risk-adapted treatment programs with or without ASCT. Nevertheless, the role of complex karyotype, combination of genetic abnormalities and ASCT remains unclear. Aims To estimate the genetic abnormalities in patients with newly diagnosed multiple myeloma and define the role of risk-stratification and ASCT in prognosis of disease. Methods The study included 159 patients (median age 63 years, range 28 - 83; male: female ratio - 1:1.37) with NDMM. Metaphase cytogenetics on bone marrow samples was done by standard GTG-method. FISH analyses were performed according to the manufacturer's protocol for detection primary IgH translocations, 13q (13q14/13q34) deletion, 1p32/1q21 amplification/deletion, P53/cen 17 deletion (MetaSystems DNA probes). We additional searched the t(4;14), t(6;14), t(11;14), t(14;16) and t(14;20) in patients with IgH translocation. All patient was treated by bortezomib-based programs (VD, CVD, VMP, PAD). ASCT was performed at 42% patients. Results The frequency of genetic abnormalities in NDMM patients was 49% (78/159). IgH translocation was detected in 26.4% (42/159) patients: t(11;14) - 16.3% (26/159), t(4;14) - 5.0% (8/159); TP53/del17p - 5.6% (9/159); 1p32/1q21 amp/del - 12% (19/159); hypodiploidy - 3.1% (5/159); hyperdiploidy - 1.25% (2/159); del5q - 0,6% (1/159); other - not found. Combination two aberrations was discovered in 11.9% (19/159) patients, complex abnormalities (>3 aberrations) - in 4.4% (7/159) patients. The median OS in "two aberration" and "complex abnormalities" groups were lower than in standard-risk mSMART 3.0 (normal, t(11;14), hypodiploidy, hyperdiploidy and other): 49 months, 26 months and was not reached, respectively (p=.00015). The median PFS for these groups was 12 months, 11 months and 30 months, respectively (p=.011). Differences between "two aberration" and "complex abnormalities" groups were not find (p> .05). We modified high-risk (gain 1q, p53 mutation, del 17p deletion, t(4;14), t(14;16), t(14;20), R-ISS stage III, double and triple hit myeloma) mSMART 3.0 by adding "two aberration" and "complex abnormalities" groups on based the OS and PFS results. The final analysis was based on the results of the complex examination of 87 patients: 53 patients in standard-risk group and 34 patients in high-risk group. The median OS in standard-risk mSMART 3.0 was not reached, in high-risk mSMART 3.0mod - 48 months; 5-years OS was 62% and 38%, respectively (p=0.0073). The median PFS was 43 and 29 months, respectively (p=.09). The best results of OS and PFS were reach in both groups of patient who performed ASCT. The median OS in standard-risk mSMART 3.0 with ASCT (n=37) was not reached, in high-risk mSMART 3.0mod with ASCT - 48 months (n=20); standard-risk mSMART 3.0 without ASCT - 40 months (n=16); in high-risk mSMART 3.0mod without ASCT - 22 months (n=14); 5-years OS was 81%, 60%, 33% and 28%, respectively (p=0.0015). The median PFS was not reached, 46, 22 and 19 months, respectively (p=.017). Conclusions The combination of two aberrations and complex abnormalities is unfavorable prognostic markers. The median OS and PFS was higher in standard-risk than high-risk group according mSMART 3.0mod. The ASCT can improve treatment's outcomes and life expectancy especially in patients with high-risk. It can be useful for update risk stratification in a future. Disclosures Shuvaev: Novartis: Consultancy; Pfize: Honoraria; Fusion Pharma: Consultancy; BMS: Consultancy.


2021 ◽  
Vol 11 ◽  
Author(s):  
Qianwen Cheng ◽  
Li Cai ◽  
Yuyang Zhang ◽  
Lei Chen ◽  
Yu Hu ◽  
...  

Background: To investigate the prognostic value of circulating plasma cells (CPC) and establish novel nomograms to predict individual progression-free survival (PFS) as well as overall survival (OS) of patients with newly diagnosed multiple myeloma (NDMM).Methods: One hundred ninetyone NDMM patients in Wuhan Union Hospital from 2017.10 to 2020.8 were included in the study. The entire cohort was randomly divided into a training (n = 130) and a validation cohort (n = 61). Univariate and multivariate analyses were performed on the training cohort to establish nomograms for the prediction of survival outcomes, and the nomograms were validated by calibration curves.Results: When the cut-off value was 0.038%, CPC could well distinguish patients with higher tumor burden and lower response rates (P < 0.05), and could be used as an independent predictor of PFS and OS. Nomograms predicting PFS and OS were developed according to CPC, lactate dehydrogenase (LDH) and creatinine. The C-index and the area under receiver operating characteristic curves (AUC) of the nomograms showed excellent individually predictive effects in training cohort, validation cohort or entire cohort. Patients with total points of the nomograms ≤ 60.7 for PFS and 75.8 for OS could be defined as low-risk group and the remaining as high-risk group. The 2-year PFS and OS rates of patients in low-risk group was significantly higher than those in high-risk group (p < 0.001).Conclusions: CPC is an independent prognostic factor for NDMM patients. The proposed nomograms could provide individualized PFS and OS prediction and risk stratification.


2021 ◽  
Author(s):  
Yong Lv ◽  
ShuGuang Jin ◽  
Bo Xiang

Abstract BackgroundTreatment of neuroblastoma is evolving toward precision medicine. LncRNAs can be used as prognostic biomarkers in many types of cancer.MethodsBased on the RNA-seq data from GSE49710, we built a lncRNAs-based risk score using the least absolute shrinkage and selection operation (LASSO) regression. Cox regression, receiver operating characteristic curves were used to evaluate the association of the LASSO risk score with overall survival. Nomograms were created and then validated in an external cohort from TARGET database. Gene set enrichment analysis was performed to identify the significantly changed biological pathways. ResultsThe 16-lncRNAs-based LASSO risk score was used to separate patients into high-risk and low-risk groups. In GSE49710 cohort, the high-risk group exhibited a poorer OS than those in the low-risk group (P<0.001). Moreover, multivariate Cox regression analysis demonstrated that LASSO risk score was an independent risk factor (HR=6.201;95%CI:2.536-15.16). The similar prognostic powers of the 16-lncRNAs were also achieved in the external cohort and in stratified analysis. In addition, a nomogram was established and worked well both in the internal validation cohort (C-index=0.831) and external validation cohort (C-index=0.773). The calibration plot indicated the good clinical utility of the nomogram. Gene set enrichment analysis (GSEA) indicated that high-risk group was related with cancer recurrence, metastasis and inflammatory associated pathways.ConclusionThe lncRNA-based LASSO risk score is a promising and potential prognostic tool in predicting the survival of patients with neuroblastoma. The nomogram combined the lncRNAs and clinical parameters allows for accurate risk assessment in guiding clinical management.


2021 ◽  
Author(s):  
Jianxin Li ◽  
Ting Han ◽  
Xin Wang ◽  
Yinchun Wang ◽  
Qingqiang Yang

Abstract Background Long non-coding RNA (lncRNA) is an important regulator of gene expression and serves fundamental role in immune regulation. The present study aimed to develop a novel immune-related lncRNA signature to accurately assess the prognosis of patients with colorectal cancer (CRC). Methods Transcriptome data and clinical information of patients with CRC were downloaded from The Cancer Genome Atlas (TCGA), and the immune-related mRNAs were extracted from immunomodulatory gene datasets IMMUNE RESPONSE and IMMUNE SYSTEM PROCESS based on the Molecular Signatures Database (MSigDB). Then, the immune-related lncRNAs were identified by a correlation analysis between immune-related mRNAs and lncRNAs. Subsequently, univariate, lasso and multivariate Cox regression were used to identify an immune-related lncRNA signature in training cohort, and the predict ability of the signature was further confirmed in the testing cohort and the entire TCGA cohort. Finally, the lncRNA-mRNA co-expression network was established to explore the biological role of the immune-related lncRNA signature. Results In total, 272 Immune-related lncRNAs were identified, five of which were applied to construct an immune-related lncRNA signature based on univariate, lasso and multivariate Cox regression analyses. The signature divided patients with CRC into low- and high-risk groups, and patients with CRC in high-risk group had poorer overall survival than those in low-risk group. Univariate and multivariate Cox regression analyses confirmed that the signature could be an independent prognostic factor in human CRC. Furthermore, functional enrichment analysis revealed that the immune-related lncRNA signature was significantly enriched in immune process and tumor classical pathways. Conclusions The present study revealed that the novel immune-related lncRNA signature could be exploited as underlying molecular biomarkers and therapeutic targets for the patients with CRC.


Author(s):  
Mei Chen ◽  
Zhenyu Nie ◽  
Yan Li ◽  
Yuanhui Gao ◽  
Xiaohong Wen ◽  
...  

Background: Ferroptosis is closely related to the occurrence and development of cancer. An increasing number of studies have induced ferroptosis as a treatment strategy for cancer. However, the predictive value of ferroptosis-related lncRNAs in bladder cancer (BC) still need to be further elucidated. The purpose of this study was to construct a predictive signature based on ferroptosis-related long noncoding RNAs (lncRNAs) to predict the prognosis of BC patients.Methods: We downloaded RNA-seq data and the corresponding clinical and prognostic data from The Cancer Genome Atlas (TCGA) database and performed univariate and multivariate Cox regression analyses to obtain ferroptosis-related lncRNAs to construct a predictive signature. The Kaplan-Meier method was used to analyze the overall survival (OS) rate of the high-risk and low-risk groups. Gene set enrichment analysis (GSEA) was performed to explore the functional differences between the high- and low-risk groups. Single-sample gene set enrichment analysis (ssGSEA) was used to explore the relationship between the predictive signature and immune status. Finally, the correlation between the predictive signature and the treatment response of BC patients was analyzed.Results: We constructed a signature composed of nine ferroptosis-related lncRNAs (AL031775.1, AL162586.1, AC034236.2, LINC01004, OCIAD1-AS1, AL136084.3, AP003352.1, Z84484.1, AC022150.2). Compared with the low-risk group, the high-risk group had a worse prognosis. The ferroptosis-related lncRNA signature could independently predict the prognosis of patients with BC. Compared with clinicopathological variables, the ferroptosis-related lncRNA signature has a higher diagnostic efficiency, and the area under the receiver operating characteristic curve was 0.707. When patients were stratified according to different clinicopathological variables, the OS of patients in the high-risk group was shorter than that of those in the low-risk group. GSEA showed that tumor- and immune-related pathways were mainly enriched in the high-risk group. ssGSEA showed that the predictive signature was significantly related to the immune status of BC patients. High-risk patients were more sensitive to anti-PD-1/L1 immunotherapy and the conventional chemotherapy drugs sunitinib, paclitaxel, cisplatin, and docetaxel.Conclusion: The predictive signature can independently predict the prognosis of BC patients, provides a basis for the mechanism of ferroptosis-related lncRNAs in BC and provides clinical treatment guidance for patients with BC.


2022 ◽  
Author(s):  
Xingyun Wang ◽  
Jinli Ji ◽  
Ying Jiang ◽  
Yiyang Zhao ◽  
Zheyao Song ◽  
...  

Abstract Venous thromboembolism (VTE) is one of the major complications of digestive system cancer, and coagulation-fibrinolysis genes play an important role in VTE. We used univariate Cox analysis, least absolute shrinkage and selection operator (LASSO), and multivariate Cox analysis to construct 3-PCFGs (prognostic coagulation-fibrinolysis genes) model based on six prognostic coagulation-fibrinolysis genes. Gene set enrichment analysis (GSEA) was used to analyze the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of the high- and low-risk groups. In addition, we classified digestive system pancancer patients into three clusters A, B, and C based on 3-PCFGs by K means. High-risk group and cluster C were associated with poor prognosis in digestive system pancancer. The m6A-related genes ALKBH5, FTO, RBM15, YTHDC1, and YTHDC2 (P<0.001) were highly expressed in the high-risk group and cluster C. The risk score was positively correlated with cancer-associated fibroblasts and endothelial cells. Cluster C had the highest immune score and stromal score. The poor prognosis in the high-risk group and cluster C may be affected by m6A epigenetic modification and immune microenvironment components in the digestive system pancancer.


BMC Cancer ◽  
2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Yi Fu ◽  
Xindong Wei ◽  
Qiuqin Han ◽  
Jiamei Le ◽  
Yujie Ma ◽  
...  

Abstract Background Early recurrence is the major cause of poor prognosis in hepatocellular carcinoma (HCC). Long non-coding RNAs (lncRNAs) are deeply involved in HCC prognosis. In this study, we aimed to establish a prognostic lncRNA signature for HCC early recurrence. Methods The lncRNA expression profile and corresponding clinical data were retrieved from total 299 HCC patients in TCGA database. LncRNA candidates correlated to early recurrence were selected by differentially expressed gene (DEG), univariate Cox regression and least absolute shrinkage and selection operator (LASSO) regression analyses. A 25-lncRNA prognostic signature was constructed according to receiver operating characteristic curve (ROC). Kaplan-Meier and multivariate Cox regression analyses were used to evaluate the performance of this signature. ROC and nomogram were used to evaluate the integrated models based on this signature with other independent clinical risk factors. Gene set enrichment analysis (GSEA) was used to reveal enriched gene sets in the high-risk group. Tumor infiltrating lymphocytes (TILs) levels were analyzed with single sample Gene Set Enrichment Analysis (ssGSEA). Immune therapy response prediction was performed with TIDE and SubMap. Chemotherapeutic response prediction was conducted by using Genomics of Drug Sensitivity in Cancer (GDSC) pharmacogenomics database. Results Compared to low-risk group, patients in high-risk group showed reduced disease-free survival (DFS) in the training (p < 0.0001) and validation cohort (p = 0.0132). The 25-lncRNA signature, AFP, TNM and vascular invasion could serve as independent risk factors for HCC early recurrence. Among them, the 25-lncRNA signature had the best predictive performance, and combination of those four risk factors further improves the prognostic potential. Moreover, GSEA showed significant enrichment of “E2F TARGETS”, “G2M CHECKPOINT”, “MYC TARGETS V1” and “DNA REPAIR” pathways in the high-risk group. In addition, increased TILs were observed in the low-risk group compared to the high-risk group. The 25-lncRNA signature negatively associates with the levels of some types of antitumor immune cells. Immunotherapies and chemotherapies prediction revealed differential responses to PD-1 inhibitor and several chemotherapeutic drugs in the low- and high-risk group. Conclusions Our study proposed a 25-lncRNA prognostic signature for predicting HCC early recurrence, which may guide postoperative treatment and recurrence surveillance in HCC patients.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 781-781 ◽  
Author(s):  
Michele Cavo ◽  
Sara Bringhen ◽  
Carolina Terragna ◽  
Paola Omedè ◽  
Giulia Marzocchi ◽  
...  

Abstract Abstract 781 Aim of the present study was to evaluate the impact of bortezomib-based induction treatments on clinical outcomes of newly diagnosed multiple myeloma (MM) patients with unfavorable cytogenetic abnormalities. For this purpose, we analyzed 590 bortezomib-treated patients who were screened at diagnosis for the presence of del(13q), t(4;14) and del(17p) by fluorescence in situ hybridization (FISH) on highly purified bone marrow plasma cells. Patients were stratified into the following 3 groups based on 1) the absence of any cytogenetic abnormality (n=261, or 44%) or 2) the presence of del(13q) alone (n=175, or 30%) or 3) positivity for t(4;14) and/or del(17p) (n=154, or 26%). In the great majority of the patients, loss of 17p was detected in more than 70% of bone marrow plasma cells, a finding which precluded a comparison with patients carrying del(17p) in a lower percentage of plasma cells. After diagnosis, 218 patients received induction therapy with bortezomib-thalidomide-dexamethasone (VTD), while the remaining 372 patients were treated with bortezomib-melphalan-prednisone (VMP) (n=181) or VMP plus thalidomide (VMPT) (n=191). The median number of bortezomib infusions (1.3 mg/m2) actually received was 24. Baseline characteristics of the 3 groups of patients were comparable, with the exception of a higher frequency of ISS stage 3 among patients with t(4;14) and/or del(17p) as compared with the cytogenetic-negative group (29% vs 17%, respectively; p=0.003). The rates of absence or presence of del (13q), t(4;14) and/or del(17p) were comparable among patients receiving VTD or VMP or VMPT treatments. Best CR to overall treatment protocols was 39% for the cytogenetic-negative group and 44% for high-risk patients carrying t(4;14) and/or del(17p). With a median follow-up of 27.5 months, median PFS was 40.5 months for patients without cytogenetic abnormalities as compared with 34 months for the high-risk group (p=0.7), while it was not reached after 38 months in the group with del(13q) alone (p not statistically significant for comparison with the other two groups). Overall, the frequency of events was 31% for patients without cytogenetic abnormalities or with del(13q) alone in comparison with 38% for those with high-risk cytogenetic profiles (p=0.15). Median OS was not reached in any of the 3 groups. Forty-month projected OS rates were 89% for the cytogenetic-negative group, 81% for the group with del(13q) alone (p=0.6) and 77% for the high-risk group (p=0.003 for comparison between this latter and the cytogenetic-negative group). Patients with t(4;14) and/or del(17p) had a shorter OS after relapse in comparison with the cytogenetic-negative group (20-month projected rates: 60% vs 76%, respectively; p=0.01). To more carefully evaluate the prognostic relevance of high-risk cytogenetic abnormalities, we stratified patients in the high-risk group into the following 3 subgroups: 1) t(4;14)-positive but del(17p)-negative (84 patients); 2) del(17p)-positive in the absence of t(4;14) (54 patients); t(4;14)-positive and del(17p)-positive (16 patients). Median PFS was not reached after 40 months for patients with t(4;14) alone, while it was 33 months for patients with del(17p) alone (p=0.1) and was 18.5 months for those who carried both these abnormalities (p=0.0008 for comparison between these latter patients and t(4;14)-positive patients). Overall, the frequency of events was 30% and 41% for patients carrying either t(4;14) or del(17p), respectively (p=0.13), while it was as high as 69% for patients with both these abnormalities. The 40-month projected OS rates for these 3 subgroups were 79%, 82% and 64%, respectively (p not significant). In conclusion, the present analysis of a large series of newly diagnosed MM patients receiving bortezomib-based induction treatments showed that: 1) del(13q) alone had no adverse effect on both PFS and OS; 2) the presence of t(4;14) and/or del(17p) did not adversely influence PFS, but was associated with a shorter OS, due at least in part to a worse outcome after relapse; 3) in comparison with t(4;14), del(17p) alone did not predicted for shorter PFS and OS, possibly as a result of the relatively long-term exposure to bortezomib); 4) the presence of both del(17p) and t(4;14) was likely to confer a particularly dismal clinical outlook, a finding which needs to be confirmed in larger series of patients. Disclosures: Cavo: Janssen-Cilag: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Millennium Pharmaceuticals: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees. Off Label Use: Use of bortezomib-based treatment for newly diagnosed multiple myeloma. Petrucci:CELGENE: Honoraria; JANSSEN-CILAG: Honoraria. Boccadoro:NOVARTIS: Honoraria; CELGENE: Honoraria; JANSSEN-CILAG: Honoraria. Palumbo:Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau, no; Janssen-Cilag: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, no.


2021 ◽  
Vol 12 ◽  
Author(s):  
Lei Zhu ◽  
Fugui Yang ◽  
Xinrui Li ◽  
Qinchuan Li ◽  
Chunlong Zhong

Background: Esophageal cancer is one of the most leading and lethal malignancies. Glycolysis and the tumor microenvironment (TME) are responsible for cancer progressions. We aimed to study the relationships between glycolysis, TME, and therapeutic response in esophageal adenocarcinoma (EAC).Materials and Methods: We used the ESTIMATE algorithm to divide EAC patients into ESTIMATE high and ESTIMATE low groups based on the gene expression data downloaded from TCGA. Weighted gene co-expression network analysis (WGCNA) and Gene Set Enrichment Analysis (GSEA) were performed to identify different glycolytic genes in the TME between the two groups. The prognostic gene signature for overall survival (OS) was established through Cox regression analysis. Impacts of glycolytic genes on immune cells were assessed and validated. Next, we conducted the glycolytic gene mutation analysis and drug therapeutic response analysis between the two groups. Finally, the GEO database was employed to validate the impact of glycolysis on TME in patients with EAC.Results: A total of 78 EAC patients with gene expression profiles and clinical information were included for analysis. Functional enrichment results showed that the genes between ESTIMATE high and ESTIMATE low groups (N = 39, respectively) were strongly related with glycolytic and ATP/ADP metabolic pathways. Patients in the low-risk group had probabilities to survive longer than those in the high-risk group (p &lt; 0.001). Glycolytic genes had significant impacts on the components of immune cells in TME, especially on the T-cells and dendritic cells. In the high-risk group, the most common mutant genes were TP53 and TTN, and the most frequent mutation type was missense mutation. Glycolysis significantly influenced drug sensitivity, and high tumor mutation burden (TMB) was associated with better immunotherapeutic response. GEO results confirmed that glycolysis had significant impacts on immune cell contents in TME.Conclusion: We performed a comprehensive study of glycolysis and TME and demonstrated that glycolysis could influence the microenvironment and drug therapeutic response in EAC. Evaluation of the glycolysis pattern could help identify the individualized therapeutic regime.


Sign in / Sign up

Export Citation Format

Share Document