Predicting the Prognosis and Immunotherapeutic Response of Triple-Negative Breast Cancer by Constructing a Prognostic Model Based on CD8+ T Cell-Related Immune Genes.

Na Ni Li, Xiao Ting Qiu, Jing Song Xue, Li Mu Yi, Mu Lan Chen, Zhi Jian Huang
{"title":"Predicting the Prognosis and Immunotherapeutic Response of Triple-Negative Breast Cancer by Constructing a Prognostic Model Based on CD8+ T Cell-Related Immune Genes.","authors":"Na Ni Li, Xiao Ting Qiu, Jing Song Xue, Li Mu Yi, Mu Lan Chen, Zhi Jian Huang","doi":"10.3967/bes2024.065","DOIUrl":null,"url":null,"abstract":"<p><strong>Objective: </strong>Triple-negative breast cancer (TNBC) poses a significant challenge for treatment efficacy. CD8+ T cells, which are pivotal immune cells, can be effectively analyzed for differential gene expression across diverse cell populations owing to rapid advancements in sequencing technology. By leveraging these genes, our objective was to develop a prognostic model that accurately predicts the prognosis of patients with TNBC and their responsiveness to immunotherapy.</p><p><strong>Methods: </strong>Sample information and clinical data of TNBC were sourced from The Cancer Genome Atlas and METABRIC databases. In the initial stage, we identified 67 differentially expressed genes associated with immune response in CD8+ T cells. Subsequently, we narrowed our focus to three key genes, namely <i>CXCL13</i>, <i>GBP2</i>, and <i>GZMB</i>, which were used to construct a prognostic model. The accuracy of the model was assessed using the validation set data and receiver operating characteristic (ROC) curves. Furthermore, we employed various methods, including Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, immune infiltration, and correlation analyses with CD274 (PD-L1) to explore the model's predictive efficacy in immunotherapeutic responses. Additionally, we investigated the potential underlying biological pathways that contribute to divergent treatment responses.</p><p><strong>Results: </strong>We successfully developed a model capable of predicting the prognosis of patients with TNBC. The areas under the curve (AUC) values for the 1-, 3-, and 5-year survival predictions were 0.618, 0.652, and 0.826, respectively. Employing this risk model, we stratified the samples into high- and low-risk groups. Through KEGG enrichment analysis, we observed that the high-risk group predominantly exhibited enrichment in metabolism-related pathways such as drug and chlorophyll metabolism, whereas the low-risk group demonstrated significant enrichment in cytokine pathways. Furthermore, immune landscape analysis revealed noteworthy variations between (PD-L1) expression and risk scores, indicating that our model effectively predicted the response of patients to immune-based treatments.</p><p><strong>Conclusion: </strong>Our study demonstrates the potential of <i>CXCL13, GBP2,</i> and <i>GZMB</i> as prognostic indicators of clinical outcomes and immunotherapy responses in patients with TNBC. These findings provide valuable insights and novel avenues for developing immunotherapeutic approaches targeting TNBC.</p>","PeriodicalId":93903,"journal":{"name":"Biomedical and environmental sciences : BES","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Biomedical and environmental sciences : BES","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.3967/bes2024.065","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Objective: Triple-negative breast cancer (TNBC) poses a significant challenge for treatment efficacy. CD8+ T cells, which are pivotal immune cells, can be effectively analyzed for differential gene expression across diverse cell populations owing to rapid advancements in sequencing technology. By leveraging these genes, our objective was to develop a prognostic model that accurately predicts the prognosis of patients with TNBC and their responsiveness to immunotherapy.

Methods: Sample information and clinical data of TNBC were sourced from The Cancer Genome Atlas and METABRIC databases. In the initial stage, we identified 67 differentially expressed genes associated with immune response in CD8+ T cells. Subsequently, we narrowed our focus to three key genes, namely CXCL13, GBP2, and GZMB, which were used to construct a prognostic model. The accuracy of the model was assessed using the validation set data and receiver operating characteristic (ROC) curves. Furthermore, we employed various methods, including Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, immune infiltration, and correlation analyses with CD274 (PD-L1) to explore the model's predictive efficacy in immunotherapeutic responses. Additionally, we investigated the potential underlying biological pathways that contribute to divergent treatment responses.

Results: We successfully developed a model capable of predicting the prognosis of patients with TNBC. The areas under the curve (AUC) values for the 1-, 3-, and 5-year survival predictions were 0.618, 0.652, and 0.826, respectively. Employing this risk model, we stratified the samples into high- and low-risk groups. Through KEGG enrichment analysis, we observed that the high-risk group predominantly exhibited enrichment in metabolism-related pathways such as drug and chlorophyll metabolism, whereas the low-risk group demonstrated significant enrichment in cytokine pathways. Furthermore, immune landscape analysis revealed noteworthy variations between (PD-L1) expression and risk scores, indicating that our model effectively predicted the response of patients to immune-based treatments.

Conclusion: Our study demonstrates the potential of CXCL13, GBP2, and GZMB as prognostic indicators of clinical outcomes and immunotherapy responses in patients with TNBC. These findings provide valuable insights and novel avenues for developing immunotherapeutic approaches targeting TNBC.

通过构建基于 CD8+ T 细胞相关免疫基因的预后模型预测三阴性乳腺癌的预后和免疫治疗反应
目的:三阴性乳腺癌(TNBC)对治疗效果提出了巨大挑战。CD8+ T 细胞是关键的免疫细胞,由于测序技术的快速发展,可以有效地分析不同细胞群的不同基因表达。通过利用这些基因,我们的目标是建立一个预后模型,准确预测 TNBC 患者的预后及其对免疫疗法的反应性:TNBC的样本信息和临床数据来自癌症基因组图谱(The Cancer Genome Atlas)和METABRIC数据库。在最初阶段,我们发现了 67 个与 CD8+ T 细胞免疫反应相关的差异表达基因。随后,我们将重点缩小到三个关键基因,即 CXCL13、GBP2 和 GZMB,并用它们构建了一个预后模型。我们利用验证集数据和接收者操作特征曲线(ROC)评估了模型的准确性。此外,我们还采用了多种方法,包括京都基因组百科全书(KEGG)通路、免疫浸润以及与 CD274(PD-L1)的相关性分析,以探讨该模型在免疫治疗反应中的预测功效。此外,我们还研究了导致不同治疗反应的潜在潜在生物通路:我们成功建立了一个能够预测 TNBC 患者预后的模型。1年、3年和5年生存预测的曲线下面积(AUC)值分别为0.618、0.652和0.826。利用该风险模型,我们将样本分为高风险组和低风险组。通过 KEGG 富集分析,我们发现高风险组主要富集于药物和叶绿素代谢等代谢相关通路,而低风险组则显著富集于细胞因子通路。此外,免疫景观分析显示,(PD-L1)表达与风险评分之间存在值得注意的差异,这表明我们的模型能有效预测患者对基于免疫的治疗的反应:我们的研究证明了CXCL13、GBP2和GZMB作为TNBC患者临床结局和免疫治疗反应预后指标的潜力。这些发现为开发针对 TNBC 的免疫治疗方法提供了宝贵的见解和新途径。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
自引率
0.00%
发文量
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信