lung adenocarcinoma (LUAD)

肺腺癌 (LUAD)
  • 文章类型: Journal Article
    肺腺癌(LUAD)是一种以高度肿瘤异质性为特征的肿瘤。尽管LUAD有许多预后和免疫治疗选择,缺乏精确的,个体化治疗方案。我们整合了mRNA,lncRNA,microRNA,来自LUADTCGA数据库的甲基化和突变数据。利用十种聚类算法,我们确定了稳定的多组学共识簇(MOCs)。然后将这些数据与十种机器学习方法合并,以开发能够可靠地识别患者预后和预测免疫治疗结果的强大模型。通过十种聚类算法,确定了两个预后相关的MOC,MOC2显示出更有利的结果。随后,我们基于8个MOCs特异性hub基因构建了MOCs相关机器学习模型(MOCM)。以MOCM评分较低为特征的患者表现出更好的总体生存率和对免疫疗法的反应。这些发现在多个数据集中是一致的,与许多以前发表的LUAD生物标志物相比,我们的MOCM评分显示出优异的预测性能。值得注意的是,低MOCM组更倾向于“热”肿瘤,以高水平的免疫细胞浸润为特征。有趣的是,发现GJB3与MOCM评分之间存在显着正相关(R=0.77,p<0.01)。进一步实验证实GJB3显著增强LUAD增殖,入侵和迁移,表明其作为LUAD治疗的关键靶标的潜力。我们开发的MOCM评分可以准确预测LUAD患者的预后,并确定免疫治疗的潜在受益者。提供广泛的临床适用性。
    Lung adenocarcinoma (LUAD) is a tumour characterized by high tumour heterogeneity. Although there are numerous prognostic and immunotherapeutic options available for LUAD, there is a dearth of precise, individualized treatment plans. We integrated mRNA, lncRNA, microRNA, methylation and mutation data from the TCGA database for LUAD. Utilizing ten clustering algorithms, we identified stable multi-omics consensus clusters (MOCs). These data were then amalgamated with ten machine learning approaches to develop a robust model capable of reliably identifying patient prognosis and predicting immunotherapy outcomes. Through ten clustering algorithms, two prognostically relevant MOCs were identified, with MOC2 showing more favourable outcomes. We subsequently constructed a MOCs-associated machine learning model (MOCM) based on eight MOCs-specific hub genes. Patients characterized by a lower MOCM score exhibited better overall survival and responses to immunotherapy. These findings were consistent across multiple datasets, and compared to many previously published LUAD biomarkers, our MOCM score demonstrated superior predictive performance. Notably, the low MOCM group was more inclined towards \'hot\' tumours, characterized by higher levels of immune cell infiltration. Intriguingly, a significant positive correlation between GJB3 and the MOCM score (R = 0.77, p < 0.01) was discovered. Further experiments confirmed that GJB3 significantly enhances LUAD proliferation, invasion and migration, indicating its potential as a key target for LUAD treatment. Our developed MOCM score accurately predicts the prognosis of LUAD patients and identifies potential beneficiaries of immunotherapy, offering broad clinical applicability.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    在肿瘤免疫微环境中,先天和适应性免疫细胞对肿瘤进展的贡献已得到一致证明.然而,肺腺癌(LUAD)的可靠预后生物标志物尚未确定.因此,我们开发并验证了免疫学长非编码RNA(lncRNA)签名(ILLS),以促进高风险和低风险患者的分类,并提供潜在的“定制”治疗选择。
    从癌症基因组图谱(TCGA)和基因表达综合(GEO)的公共数据库获得并处理了LUAD数据集。通过共识聚类计算免疫浸润的丰度及其相关途径,加权基因共表达网络分析(WGCNA),和整合的ImmLnc以鉴定免疫相关的lncRNAs并提取免疫相关的预后lncRNAs。基于整合程序,最佳的算法组成是最小绝对收缩和选择算子(LASSO)和逐步Cox回归在两个方向上开发的ILLSTCGA-LUAD数据集和验证4个独立数据集的预测能力,通过生存分析GSE31210、GSE37745、GSE30219和GSE50081,接收机工作特性(ROC)分析,和多元Cox回归。将一致性指数(C-index)分析与上述5个数据集中49个已发表的签名进行横向比较,以进一步证实其稳定性和优越性。最后,进行药物敏感性分析以探索潜在的治疗药物.
    与低风险组相比,来自高风险组的患者的总生存期(OS)始终较差。ILLS被证明是独立的预后因素,具有良好的敏感性和特异性。在4个GEO数据集中,与其他文献报道的相比,ILLS保持稳定的预测能力,更适合作为共识风险分层工具。然而,癌症免疫图谱和IMsporch210数据集证明了在识别具有有效免疫疗法的目标人群方面的实际实用性。虽然高危人群表现出某些化疗药物的潜在靶点,比如卡莫司汀,依托泊苷,三氧化二砷,和阿列替尼。
    ILLS表现出优越且稳定的预后预测能力,因此有可能作为辅助LUAD患者进行风险分类和临床决策的工具。
    UNASSIGNED: In the tumor immune microenvironment, the contribution of innate and adaptive immune cells to tumor progression has been consistently demonstrated. However, reliable prognostic biomarkers for lung adenocarcinoma (LUAD) have not yet been identified. We thus developed and validated an immunologic long noncoding RNA (lncRNA) signature (ILLS) to facilitate the classification of patients with high and low risk and provide potential \"made-to-measure\" treatment choices.
    UNASSIGNED: The LUAD data sets were obtained and processed from public databases of The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). The abundance of immune infiltration and its related pathways were calculated through consensus clustering, weighted gene coexpression network analysis (WGCNA), and an integrated ImmLnc to identify immune-related lncRNAs and extract immune-related prognostic lncRNAs. Based on the integrative procedure, the best algorithm composition was least absolute shrinkage and selection operator (LASSO) and stepwise Cox regression in both directions to develop the ILLS in the TCGA-LUAD data set and validate the predictive power of 4 independent data sets, GSE31210, GSE37745, GSE30219, and GSE50081 through survival analysis, receiver operating characteristic (ROC) analysis, and multivariate Cox regression. The concordance index (C-index) analysis was transversely compared with 49 published signatures in the above 5 data sets to further confirm its stability and superiority. Finally, drug sensitivity analysis was conducted to explore potential therapeutic agents.
    UNASSIGNED: Patients from the high-risk groups consistently had worse overall survival (OS) compared to the low-risk groups. ILLS proved to be an independent prognostic factor with favorable sensitivity and specificity. Among the 4 GEO data sets, compared to those reported in the other literature, ILLS maintained stable prediction ability and was more suitable as a consensus risk-stratification tool. However, The Cancer Immunome Atlas and IMvigor210 data sets demonstrated practical utility in recognizing target populations with effective immunotherapy, while the high-risk group exhibited potential targets for certain chemotherapy drugs, such as carmustine, etoposide, arsenic trioxide, and alectinib.
    UNASSIGNED: ILLS demonstrated superior and stable prognostic prediction ability and thus has potential as a tool for assisting in risk classification and clinical decision-making in patients with LUAD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号