Cancer subtypes

癌症亚型
  • 文章类型: Journal Article
    背景:美国心脏协会最近推出了一种新的心血管健康(CVH)指标,生命的本质8(LE8),促进健康。然而,LE8与癌症死亡风险之间的关系仍不确定.
    方法:我们调查了来自美国国家健康与营养调查(USNHANES)的17,076名参与者和来自英国生物银行的272,727名参与者,基线时全部无癌。CVH分数,基于LE8指标,包含四种健康行为(饮食,身体活动,吸烟,和睡眠)和四个健康因素(体重指数,脂质,血糖,和血压)。自我报告问卷评估健康行为。主要结果是总癌症及其亚型的死亡率。使用带调整的Cox模型检查CVH评分(连续和分类变量)与结果之间的关联。构建与癌症亚型相关的多基因风险评分(PRS),以评估其与CVH对癌症死亡风险的相互作用。
    结果:美国NHANES超过141,526人年,发生了424例癌症相关死亡,在英国生物银行,在3,690,893人年期间记录了8,872例癌症死亡。与低CVH相比,高CVH与总体癌症死亡率降低相关(美国NHANES中HR0.58,95%CI0.37-0.91;英国生物银行中HR0.51,0.46-0.57)。在美国NHANES中,CVH评分的每一个标准差增加与癌症死亡率降低19%(HR:0.81;95%CI:0.73-0.91)和英国生物银行降低19%(HR:0.81;95%CI:0.79-0.83)相关。坚持理想的CVH与降低肺部死亡风险呈线性关系,膀胱,肝脏,肾,食道,乳房,结直肠,胰腺,和英国生物银行的胃癌。此外,整合遗传数据显示,与PRS和CVH较高的患者相比,PRS较低和CVH较高的患者在8种癌症中死亡率最低(HRs为0.36~0.57).未观察到因遗传易感性导致的CVH与八种癌症的死亡风险之间的关联的显着改变。亚组分析显示,在年轻参与者和社会经济地位较低的参与者中,总体癌症死亡率具有更明显的保护性关联。
    结论:维持最佳CVH与总体癌症死亡率风险的显著降低相关。对理想CVH的坚持与多种癌症亚型的死亡风险降低呈线性关系。具有理想CVH和高遗传易感性的个体表现出显著的健康益处。这些发现支持采用理想的CVH作为干预策略,以减轻癌症死亡风险并促进健康衰老。
    BACKGROUND: The American Heart Association recently introduced a novel cardiovascular health (CVH) metric, Life\'s Essential 8 (LE8), for health promotion. However, the relationship between LE8 and cancer mortality risk remains uncertain.
    METHODS: We investigated 17,076 participants from US National Health and Nutrition Examination Survey (US NHANES) and 272,727 participants from UK Biobank, all free of cancer at baseline. The CVH score, based on LE8 metrics, incorporates four health behaviors (diet, physical activity, smoking, and sleep) and four health factors (body mass index, lipid, blood glucose, and blood pressure). Self-reported questionnaires assessed health behaviors. Primary outcomes were mortality rates for total cancer and its subtypes. The association between CVH score (continuous and categorical variable) and outcomes was examined using Cox model with adjustments. Cancer subtypes-related polygenic risk score (PRS) was constructed to evaluate its interactions with CVH on cancer death risk.
    RESULTS: Over 141,526 person-years in US NHANES, 424 cancer-related deaths occurred, and in UK Biobank, 8,872 cancer deaths were documented during 3,690,893 person-years. High CVH was associated with reduced overall cancer mortality compared to low CVH (HR 0.58, 95% CI 0.37-0.91 in US NHANES; 0.51, 0.46-0.57 in UK Biobank). Each one-standard deviation increase in CVH score was linked to a 19% decrease in cancer mortality (HR: 0.81; 95% CI: 0.73-0.91) in US NHANES and a 19% decrease (HR: 0.81; 95% CI: 0.79-0.83) in UK Biobank. Adhering to ideal CVH was linearly associated with decreased risks of death from lung, bladder, liver, kidney, esophageal, breast, colorectal, pancreatic, and gastric cancers in UK Biobank. Furthermore, integrating genetic data revealed individuals with low PRS and high CVH exhibited the lowest mortality from eight cancers (HRs ranged from 0.36 to 0.57) compared to those with high PRS and low CVH. No significant modification of the association between CVH and mortality risk for eight cancers by genetic predisposition was observed. Subgroup analyses showed a more pronounced protective association for overall cancer mortality among younger participants and those with lower socio-economic status.
    CONCLUSIONS: Maintaining optimal CVH is associated with a substantial reduction in the risk of overall cancer mortality. Adherence to ideal CVH correlates linearly with decreased mortality risk across multiple cancer subtypes. Individuals with both ideal CVH and high genetic predisposition demonstrated significant health benefits. These findings support adopting ideal CVH as an intervention strategy to mitigate cancer mortality risk and promote healthy aging.
    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
    (1)背景:肿瘤亚型的识别是精准医学准确诊断和个性化治疗的基础。癌症的发展通常是由体细胞突变的积累驱动的,这些突变可以导致组织功能和形态的改变。在这项工作中,提出了一种基于深度神经网络的方法,该方法集成到基于网络的分层框架(D3NS)中,用于根据体细胞突变对肿瘤进行分层.(2)方法:该方法利用深度神经网络的力量,通过结合基因相互作用网络中包含的知识来检测数据中的隐藏信息,作为基于网络的分层方法的典型代表。使用来自膀胱癌症基因组图谱的真实世界数据应用D3NS,卵巢,和肾癌。(3)结果:该技术可以识别具有不同生存率和与几种临床结果显着关联的肿瘤亚型(肿瘤分期,分级或对治疗的反应)。(4)结论:D3NS可为肿瘤研究提供基础模型,可作为肿瘤分层的有效工具。在临床环境中提供潜在的支持。
    (1) Background: The identification of tumor subtypes is fundamental in precision medicine for accurate diagnoses and personalized therapies. Cancer development is often driven by the accumulation of somatic mutations that can cause alterations in tissue functions and morphologies. In this work, a method based on a deep neural network integrated into a network-based stratification framework (D3NS) is proposed to stratify tumors according to somatic mutations. (2) Methods: This approach leverages the power of deep neural networks to detect hidden information in the data by combining the knowledge contained in a network of gene interactions, as typical of network-based stratification methods. D3NS was applied using real-world data from The Cancer Genome Atlas for bladder, ovarian, and kidney cancers. (3) Results: This technique allows for the identification of tumor subtypes characterized by different survival rates and significant associations with several clinical outcomes (tumor stage, grade or response to therapy). (4) Conclusion: D3NS can provide a base model in cancer research and could be considered as a useful tool for tumor stratification, offering potential support in clinical settings.
    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
    当不同组的事件或暴露导致不同的疾病亚型时,就会发生病因异质性。从两阶段结果相关的采样数据中推断特定亚型的暴露效果需要对混杂和采样设计进行调整。推断这些影响的常见方法不一定适当地调整这些偏差源,或允许对不同亚型的效果进行正式比较。在这里,使用逆概率加权(IPW)拟合多项式模型显示,这种采样设计可以对特定亚型的暴露效果及其对比产生有效的推断。将IPW方法与使用模拟评估暴露效应异质性的基于回归的常见方法进行比较。在卡罗莱纳州乳腺癌研究中,该方法用于评估各种暴露对乳腺癌风险的亚型特异性影响。
    Etiologic heterogeneity occurs when distinct sets of events or exposures give rise to different subtypes of disease. Inference about subtype-specific exposure effects from two-phase outcome-dependent sampling data requires adjustment for both confounding and the sampling design. Common approaches to inference for these effects do not necessarily appropriately adjust for these sources of bias, or allow for formal comparisons of effects across different subtypes. Herein, using inverse probability weighting (IPW) to fit a multinomial model is shown to yield valid inference with this sampling design for subtype-specific exposure effects and contrasts thereof. The IPW approach is compared to common regression-based methods for assessing exposure effect heterogeneity using simulations. The methods are applied to estimate subtype-specific effects of various exposures on breast cancer risk in the Carolina Breast Cancer Study.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    抗癌肽(ACPs)在选择性靶向和消除癌细胞中起着至关重要的作用。评估和比较各种机器学习(ML)和深度学习(DL)技术的预测是具有挑战性的,但对于抗癌药物研究至关重要。我们对15个ML和10个DL模型进行了全面分析,包括2022年之后发布的模型,发现具有特征组合和选择的支持向量机(SVM)显著提升了整体性能。DL模型,特别是具有基于光梯度增强机(LGBM)的特征选择方法的卷积神经网络(CNN),展示改进的表征。使用新的测试数据集(ACP10)进行评估,确定ACPred,MLACP2.0,AI4ACP,mACPred,和AntiCP2.0_AAC作为连续的最优预测因子,展示强大的性能。我们的评论强调了当前预测工具的局限性,并主张采用全向ACP预测框架来推动正在进行的研究。
    Anticancer peptides (ACPs) play a vital role in selectively targeting and eliminating cancer cells. Evaluating and comparing predictions from various machine learning (ML) and deep learning (DL) techniques is challenging but crucial for anticancer drug research. We conducted a comprehensive analysis of 15 ML and 10 DL models, including the models released after 2022, and found that support vector machines (SVMs) with feature combination and selection significantly enhance overall performance. DL models, especially convolutional neural networks (CNNs) with light gradient boosting machine (LGBM) based feature selection approaches, demonstrate improved characterization. Assessment using a new test data set (ACP10) identifies ACPred, MLACP 2.0, AI4ACP, mACPred, and AntiCP2.0_AAC as successive optimal predictors, showcasing robust performance. Our review underscores current prediction tool limitations and advocates for an omnidirectional ACP prediction framework to propel ongoing research.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    下一代测序(NGS)领域的进步已经为相同的一组受试者产生了大量的数据。出现的挑战是如何结合和调和来自不同组学研究的结果,如表观基因组和转录组,以提高疾病亚型的分类。在这项研究中,我们介绍sCClust(稀疏典型相关分析与聚类),一种使用稀疏典型相关分析(SCCA)组合高维组学数据的技术,使得数据集之间的相关性最大化。此阶段之后是在较低维空间中对集成数据进行聚类。我们将sCClust应用于来自癌症基因组图谱(TCGA)的三个癌症基因组学数据集的基因表达和DNA甲基化数据,以区分潜在的亚型。我们使用Kaplan-Meier图和风险比分析对三种类型的癌症-GBM(多形性胶质母细胞瘤)评估已识别的亚型,肺癌和结肠癌。与通过单组和多组研究鉴定的亚型的比较意味着改善的临床关联。我们还进行了通路过度表达分析,以鉴定上调和下调的基因作为暂定的药物靶标。本文的主要目标是双重的:整合表观基因组和转录组数据集,然后阐明潜在空间中的亚型。这项研究的意义在于增强了癌症数据的分类,这对精准医学至关重要。
    Advancements in the field of next generation sequencing (NGS) have generated vast amounts of data for the same set of subjects. The challenge that arises is how to combine and reconcile results from different omics studies, such as epigenome and transcriptome, to improve the classification of disease subtypes. In this study, we introduce sCClust (sparse canonical correlation analysis with clustering), a technique to combine high-dimensional omics data using sparse canonical correlation analysis (sCCA), such that the correlation between datasets is maximized. This stage is followed by clustering the integrated data in a lower-dimensional space. We apply sCClust to gene expression and DNA methylation data for three cancer genomics datasets from the Cancer Genome Atlas (TCGA) to distinguish between underlying subtypes. We evaluate the identified subtypes using Kaplan-Meier plots and hazard ratio analysis on the three types of cancer-GBM (glioblastoma multiform), lung cancer and colon cancer. Comparison with subtypes identified by both single- and multi-omics studies implies improved clinical association. We also perform pathway over-representation analysis in order to identify up-regulated and down-regulated genes as tentative drug targets. The main goal of the paper is twofold: the integration of epigenomic and transcriptomic datasets followed by elucidating subtypes in the latent space. The significance of this study lies in the enhanced categorization of cancer data, which is crucial to precision medicine.
    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
    人乳头瘤病毒相关(HPV+)头颈部鳞状细胞癌(HNSCC)是美国最常见的HPV相关癌症,在过去的二十年中,发病率迅速增加。HPV+HNSCC的负担可能会继续上升,考虑到感染和HPV+HNSCC发展之间的潜伏期长,据估计,HPV疫苗的效果要到2060年才能反映在HNSCC的流行中.已经开始努力降低这种疾病的标准疗法的发病率,其改进的表征正被用来识别和瞄准分子漏洞。用于新疗法的伴侣生物标志物将识别反应性肿瘤。对头颈部HPV癌变的两种机制的更基本的理解已经确定了HPV+HNSCC的亚型,其与不同的致癌程序相关并且确定具有良好或不良预后的肿瘤。目前可靠地识别这两种亚型的生物标志物的发展,以及可以在更早的时间检测复发性疾病的生物标志物,将有立即的临床应用。
    Human papillomavirus-associated (HPV+) head and neck squamous cell carcinoma (HNSCC) is the most common HPV-associated cancer in the United States, with a rapid increase in incidence over the last two decades. The burden of HPV+ HNSCC is likely to continue to rise, and given the long latency between infection and the development of HPV+ HNSCC, it is estimated that the effect of the HPV vaccine will not be reflected in HNSCC prevalence until 2060. Efforts have begun to decrease morbidity of standard therapies for this disease, and its improved characterization is being leveraged to identify and target molecular vulnerabilities. Companion biomarkers for new therapies will identify responsive tumors. A more basic understanding of two mechanisms of HPV carcinogenesis in the head and neck has identified subtypes of HPV+ HNSCC that correlate with different carcinogenic programs and that identify tumors with good or poor prognosis. Current development of biomarkers that reliably identify these two subtypes, as well as biomarkers that can detect recurrent disease at an earlier time, will have immediate clinical application.
    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
    简介:作为评价指标,癌症分级和亚型有不同的临床,病态,以及具有预后和治疗意义的分子特征。尽管研究人员已经开始研究癌症分化和亚型预测,大多数相关方法都基于传统的机器学习,依赖于单一的组学数据。有必要探索一种集成多组学数据的深度学习算法,以实现癌症分化和亚型的分类预测。方法:本文提出了一种基于多视图图神经网络(MVGNN)的多组数据融合算法,用于预测癌症分化和亚型分类。该模型框架由用于从不同组学数据中学习特征的图卷积网络(GCN)模块和用于整合多组学数据的注意力模块组成。使用三种不同类型的组学数据。对于每种类型的组学数据,使用卡方检验和最小冗余最大相关性(mRMR)等方法进行特征选择。基于选定的组学特征构建加权患者相似度网络,GCN使用组学特征和相应的相似性网络进行训练。最后,注意模块集成了不同类型的组学特征,并执行最终的癌症分类预测。结果:为了验证MVGNN模型的癌症分类预测性能,我们与传统机器学习模型和当前流行的方法进行了实验比较,这些方法基于5倍交叉验证整合了多组学数据.此外,我们基于单个组学数据对癌症分化及其亚型进行了比较实验,两个组学数据,和三个组学数据。讨论:本文提出了MVGNN模型,该模型在基于多个组学数据的癌症分类预测中表现良好。
    Introduction: As the evaluation indices, cancer grading and subtyping have diverse clinical, pathological, and molecular characteristics with prognostic and therapeutic implications. Although researchers have begun to study cancer differentiation and subtype prediction, most of relevant methods are based on traditional machine learning and rely on single omics data. It is necessary to explore a deep learning algorithm that integrates multi-omics data to achieve classification prediction of cancer differentiation and subtypes. Methods: This paper proposes a multi-omics data fusion algorithm based on a multi-view graph neural network (MVGNN) for predicting cancer differentiation and subtype classification. The model framework consists of a graph convolutional network (GCN) module for learning features from different omics data and an attention module for integrating multi-omics data. Three different types of omics data are used. For each type of omics data, feature selection is performed using methods such as the chi-square test and minimum redundancy maximum relevance (mRMR). Weighted patient similarity networks are constructed based on the selected omics features, and GCN is trained using omics features and corresponding similarity networks. Finally, an attention module integrates different types of omics features and performs the final cancer classification prediction. Results: To validate the cancer classification predictive performance of the MVGNN model, we conducted experimental comparisons with traditional machine learning models and currently popular methods based on integrating multi-omics data using 5-fold cross-validation. Additionally, we performed comparative experiments on cancer differentiation and its subtypes based on single omics data, two omics data, and three omics data. Discussion: This paper proposed the MVGNN model and it performed well in cancer classification prediction based on multiple omics data.
    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
    背景:二硫化物下垂,一种由二硫化物的异常细胞内积累诱导的细胞死亡形式,是一种新认识到的细胞死亡。透明细胞肾细胞癌(ccRCC)是一种常见的泌尿系统肿瘤,具有严重的健康风险。到目前为止,对ccRCC中的二硫化物凋亡相关基因(DRGs)的研究很少。
    方法:表达式,转录变异,并评估了DRGs的预后作用。基于DRG,采用共识无监督聚类分析将ccRCC患者分为不同亚型,并构建DRG风险评分模型.通过该模型将患者分为高危组或低危组。我们专注于评估预后的差异,TME,化疗易感性,和两个风险组之间的免疫景观。最后,我们通过体外实验验证了风险评分基因FLRT3的表达并探讨了其生物学功能。
    结果:不同亚型的基因表达差异显著,免疫,和预后景观。在两个风险组中,高危人群的TME评分较高,更显著的免疫细胞浸润,从免疫疗法中获益的可能性更高,但预后较差.两个风险组之间的化疗敏感性也存在显着差异。在ccRCC细胞中,显示FLRT3的表达较低,其过表达导致细胞增殖和转移能力降低。
    结论:从二硫键下降开始,我们建立了一个新的风险评分模型,可以为医生预测患者生存和确定临床治疗方案提供新的思路。
    Disulfidptosis, a form of cell death induced by abnormal intracellular accumulation of disulfides, is a newly recognized variety of cell death. Clear cell renal cell carcinoma (ccRCC) is a usual urological tumor that poses serious health risks. There are few studies of disulfidptosis-related genes (DRGs) in ccRCC so far.
    The expression, transcriptional variants, and prognostic role of DRGs were assessed. Based on DRGs, consensus unsupervised clustering analysis was performed to stratify ccRCC patients into various subtypes and constructed a DRG risk scoring model. Patients were stratified into high or low-risk groups by this model. We focused on assessing the discrepancy in prognosis, TME, chemotherapeutic susceptibility, and landscape of immune between the two risk groups. Finally, we validated the expression and explored the biological function of the risk scoring gene FLRT3 through in vitro experiments.
    The different subtypes had significantly different gene expression, immune, and prognostic landscapes. In the two risk groups, the high-risk group had higher TME scores, more significant immune cell infiltration, and a higher probability of benefiting from immunotherapy, but had a worse prognosis. There were also remarkable differences in chemotherapeutic susceptibility between the two risk groups. In ccRCC cells, the expression of FLRT3 was shown to be lower and its overexpression caused a decrease in cell proliferation and metastatic capacity.
    Starting from disulfidptosis, we established a new risk scoring model which can provide new ideas for doctors to forecast patient survival and determine clinical treatment plans.
    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
    高级别浆液性肿瘤卵巢癌(HGSTOC)是女性生殖道最致命的肿瘤。上述治疗包括细胞减少,然后是标准的铂/紫杉烷化疗;或者,对于原发性无法切除的肿瘤,新辅助铂/紫杉烷化疗,然后延迟间隔细胞减少。在手术欠佳或晚期疾病的患者中,不同形式的靶向治疗已在临床试验中被接受或测试.对HGSTOC的研究发现了其遗传和蛋白质组异质性,表观遗传调控,以及肿瘤微环境的作用。这些发现将注意力转移到HGSTOC有几种不同的原发性肿瘤亚型和原发性肿瘤的独特生物学上。转移性,和复发性肿瘤可能导致不同的药物反应。这导致一些原发性肿瘤的化学难治性,更频繁和更具破坏性的东西,转移性和复发性HGSTOC肿瘤的继发性化学耐药性。铂抗性疾病的治疗可能性包括几种具有中等活性的化学疗法和具有难以耐受的作用的不同靶向药物。因此,问题似乎是为什么不同的卵巢癌亚型主要基于相同的治疗方案而不是以个性化的方式进行治疗,适应特定肿瘤亚型的生物学特性和疾病的时间矩。这篇论文回顾了基因组,突变,HGSTOC亚型和肿瘤微环境的表观遗传特征。关于个性化治疗的临床试验和新的总体结果,已经提出并讨论了卵巢癌个性化治疗的综合方法。
    High-grade serous tubo-ovarian cancer (HGSTOC) is the most lethal tumor of the female genital tract. The foregoing therapy consists of cytoreduction followed by standard platinum/taxane chemotherapy; alternatively, for primary unresectable tumors, neo-adjuvant platinum/taxane chemotherapy followed by delayed interval cytoreduction. In patients with suboptimal surgery or advanced disease, different forms of targeted therapy have been accepted or tested in clinical trials. Studies on HGSTOC discovered its genetic and proteomic heterogeneity, epigenetic regulation, and the role of the tumor microenvironment. These findings turned attention to the fact that there are several distinct primary tumor subtypes of HGSTOC and the unique biology of primary, metastatic, and recurrent tumors may result in a differential drug response. This results in both chemo-refractoriness of some primary tumors and, what is significantly more frequent and destructive, secondary chemo-resistance of metastatic and recurrent HGSTOC tumors. Treatment possibilities for platinum-resistant disease include several chemotherapeutics with moderate activity and different targeted drugs with difficult tolerable effects. Therefore, the question appears as to why different subtypes of ovarian cancer are predominantly treated based on the same therapeutic schemes and not in an individualized way, adjusted to the biology of a specific tumor subtype and temporal moment of the disease. The paper reviews the genomic, mutational, and epigenetic signatures of HGSTOC subtypes and the tumor microenvironment. The clinical trials on personalized therapy and the overall results of a new, comprehensive approach to personalized therapy for ovarian cancer have been presented and discussed.
    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
    整合多个组学数据集以区分癌症亚型是一项强大的技术,可利用多组学数据中的一致和互补信息。矩阵分解是集成聚类中用于识别跨多组数据的潜在亚型结构的常用技术。组学数据的高维度和长计算时间一直是聚类方法的共同挑战。为了应对挑战,我们提出了使用非负矩阵分解的随机奇异值分解(RSVD)用于集成聚类:intNMF-rsvd。该方法利用RSVD通过将数据投影到用户指定的较低秩的特征向量空间来降低维数。然后,聚类分析是通过估计预测的多组数据集的公共基础矩阵来进行的。使用模拟数据集评估了所提出方法的性能,并使用来自癌症基因组图谱研究的真实数据集与六种最先进的综合聚类方法进行了比较。与标准的intNMF和其他多组学聚类方法相比,发现intNMF-rsvd有效且具有竞争力。最重要的是,intNMF-rsvd可以处理大量的功能,并显著减少计算时间。鉴定的亚型可用于进一步的临床关联研究以了解疾病的病因。
    Integration of multiple \'omics datasets for differentiating cancer subtypes is a powerful technic that leverages the consistent and complementary information across multi-omics data. Matrix factorization is a common technique used in integrative clustering for identifying latent subtype structure across multi-omics data. High dimensionality of the omics data and long computation time have been common challenges of clustering methods. In order to address the challenges, we propose randomized singular value decomposition (RSVD) for integrative clustering using Non-negative Matrix Factorization: intNMF-rsvd. The method utilizes RSVD to reduce the dimensionality by projecting the data into eigen vector space with user specified lower rank. Then, clustering analysis is carried out by estimating common basis matrix across the projected multi-omics datasets. The performance of the proposed method was assessed using the simulated datasets and compared with six state-of-the-art integrative clustering methods using real-life datasets from The Cancer Genome Atlas Study. intNMF-rsvd was found working efficiently and competitively as compared to standard intNMF and other multi-omics clustering methods. Most importantly, intNMF-rsvd can handle large number of features and significantly reduce the computation time. The identified subtypes can be utilized for further clinical association studies to understand the etiology of the disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号