Radiotranscriptomics

  • 文章类型: Journal Article
    本研究旨在确定动态对比增强磁共振成像(DCE-MRI)衍生的放射组学模型在胆管癌的肿瘤免疫谱分析和免疫治疗中的预测作用。要执行放射学分析,免疫相关亚组聚类首先通过单样本基因集富集分析(ssGSEA)进行。第二,使用Python软件包Pyradiomics共提取了DCE-MRI每个阶段的806个影像组学特征.然后,经过三步特征缩减和选择,构建了一个预测影像组学特征模型,并采用受试者工作特性(ROC)曲线评价模型的性能。最后,我们使用一个独立的检测队列,包括胆管癌患者术后接受抗PD-1Sindilimab治疗,以验证建立的影像组学模型在胆管癌免疫治疗中的潜在应用.使用基于转录组测序的ssGSEA对两个不同的免疫相关亚组进行分类。对于放射学分析,最终确定了总共10个预测放射学特征,以建立用于免疫景观分类的放射学特征模型。关于预测性能,在训练/验证队列中,ROC曲线的平均AUC为0.80.对于独立测试队列,影像组学模型的个体预测概率与来自ssGSEA的相应免疫评分显著相关.总之,基于DCE-MRI的影像学特征模型能够预测淋巴结癌的免疫格局。因此,本研究提出了这种已开发的放射学模型在指导胆管癌免疫治疗方面的潜在临床应用.
    This study aims to determine the predictive role of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) derived radiomic model in tumor immune profiling and immunotherapy for cholangiocarcinoma. To perform radiomic analysis, immune related subgroup clustering was first performed by single sample gene set enrichment analysis (ssGSEA). Second, a total of 806 radiomic features for each phase of DCE-MRI were extracted by utilizing the Python package Pyradiomics. Then, a predictive radiomic signature model was constructed after a three-step features reduction and selection, and receiver operating characteristic (ROC) curve was employed to evaluate the performance of this model. In the end, an independent testing cohort involving cholangiocarcinoma patients with anti-PD-1 Sintilimab treatment after surgery was used to verify the potential application of the established radiomic model in immunotherapy for cholangiocarcinoma. Two distinct immune related subgroups were classified using ssGSEA based on transcriptome sequencing. For radiomic analysis, a total of 10 predictive radiomic features were finally identified to establish a radiomic signature model for immune landscape classification. Regarding to the predictive performance, the mean AUC of ROC curves was 0.80 in the training/validation cohort. For the independent testing cohort, the individual predictive probability by radiomic model and the corresponding immune score derived from ssGSEA was significantly correlated. In conclusion, radiomic signature model based on DCE-MRI was capable of predicting the immune landscape of chalangiocarcinoma. Consequently, a potentially clinical application of this developed radiomic model to guide immunotherapy for cholangiocarcinoma was suggested.
    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
    随着近年来心血管成像领域的巨大进步,计算机断层扫描(CT)已成为动脉粥样硬化性冠状动脉疾病的表型。使用人工智能(AI)的新分析方法可以分析动脉粥样硬化斑块的复杂表型信息。特别是,使用卷积神经网络(CNN)的基于深度学习的方法促进了病变检测等任务,分割,和分类。新的放射转录组学技术甚至通过对CT图像上的体素进行高阶结构分析来捕获潜在的生物组织化学过程。在不久的将来,国际大规模牛津危险因素和非侵入性成像(ORFAN)研究将为测试和验证基于AI的预后模型提供强大的平台。目标是将这些新方法从研究环境转变为临床工作流程。在这次审查中,我们概述了现有的基于AI的技术,重点是成像生物标志物以确定冠状动脉炎症的程度,冠状动脉斑块,以及相关风险。Further,将讨论使用基于AI的方法的当前限制以及解决这些挑战的优先事项。这将为AI启用的风险评估工具铺平道路,以检测易损的动脉粥样硬化斑块并指导患者的治疗策略。
    With the enormous progress in the field of cardiovascular imaging in recent years, computed tomography (CT) has become readily available to phenotype atherosclerotic coronary artery disease. New analytical methods using artificial intelligence (AI) enable the analysis of complex phenotypic information of atherosclerotic plaques. In particular, deep learning-based approaches using convolutional neural networks (CNNs) facilitate tasks such as lesion detection, segmentation, and classification. New radiotranscriptomic techniques even capture underlying bio-histochemical processes through higher-order structural analysis of voxels on CT images. In the near future, the international large-scale Oxford Risk Factors And Non-invasive Imaging (ORFAN) study will provide a powerful platform for testing and validating prognostic AI-based models. The goal is the transition of these new approaches from research settings into a clinical workflow. In this review, we present an overview of existing AI-based techniques with focus on imaging biomarkers to determine the degree of coronary inflammation, coronary plaques, and the associated risk. Further, current limitations using AI-based approaches as well as the priorities to address these challenges will be discussed. This will pave the way for an AI-enabled risk assessment tool to detect vulnerable atherosclerotic plaques and to guide treatment strategies for patients.
    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
    本研究调查了反映前列腺癌(PCa)相关基因突变的超声(US)表型。在这里,整合放射性转录组学数据,US和对比增强超声(CEUS)影像,RNA测序旨在显著提高PCa预后的准确性.我们进行了临床的放射性转录组学分析,成像,和来自48和22名PCa和良性前列腺增生(BPH)男性的两个基因组(mRNA和microRNA表达)数据集,分别。23个US纹理特征和4个微血管灌注特征与52个与PCa相关的差异表达基因的不同模式相关(p<0.05);17个过表达基因与两个关键纹理特征相关。使用微血管灌注特征鉴定了十二个过表达的基因。此外,mRNA和miRNA生物标志物可用于区分PCa和BPH。与RNA测序相比,B型和CEUS特征反映了与激素受体状态相关的基因组改变,血管生成,PCa患者的预后。这些发现表明US评估PCa患者生物标志物水平的潜力。
    The present study investigated ultrasound (US) phenotypes reflecting prostate cancer (PCa)-related genetic mutations. Herein, integration of radiotranscriptomic data, US and contrast-enhanced ultrasound (CEUS) radiomic images, and RNA sequencing was performed with the aim of significantly improving the accuracy of PCa prognosis. We performed radiotranscriptomic analysis of clinical, imaging, and two genomic (mRNA and microRNA expression) datasets from 48 and 22 men with PCa and benign prostatic hyperplasia (BPH), respectively. Twenty-three US texture features and four microvascular perfusion features were associated with various patterns of 52 differentially expressed genes related to PCa (p < 0.05); 17 overexpressed genes were associated with two key texture features. Twelve overexpressed genes were identified using microvascular perfusion features. Furthermore, mRNA and miRNA biomarkers could be used to distinguish between PCa and BPH. Compared with RNA sequencing, B-mode and CEUS features reflected genomic alterations associated with hormone receptor status, angiogenesis, and prognosis in patients with PCa. These findings indicate the potential of US to assess biomarker levels in patients with PCa.
    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
    放射转录组学是一个新兴领域,旨在研究从医学图像中提取的放射组学特征与有助于诊断的基因表达谱之间的关系。治疗计划,和癌症的预后。这项研究提出了一个方法学框架,用于研究这些关联与非小细胞肺癌(NSCLC)的应用。使用具有转录组学数据的六个公开可用的NSCLC数据集来导出和验证转录组特征,以用于区分癌症和非恶性肺组织的能力。24名NSCLC诊断患者的公开数据集,转录组和成像数据,用于联合放射转录组学分析。对于每个病人来说,提取了749个计算机断层扫描(CT)的影像学特征,并通过DNA微阵列提供了相应的转录组学数据。使用迭代K均值算法对放射学特征进行聚类,得到77个同质聚类,以元放射学特征为代表。通过进行微阵列的显著性分析(SAM)和2倍变化来选择最显著的差异表达基因(DEGs)。使用SAM和Spearman等级相关检验研究了CT成像特征与所选DEG之间的相互作用,错误发现率(FDR)为5%,导致73个DEGs的提取与放射学特征显着相关。这些基因被用来产生meta-radiomics特征的预测模型,定义为p-元组学特征,通过执行Lasso回归。在77个元放射学特征中,图51可以根据转录组特征进行建模。这些重要的放射转录组学关系形成了可靠的基础,可以在生物学上证明从解剖成像方式中提取的放射组学特征。因此,通过对基于转录组学的回归模型进行富集分析,证明了这些放射学特征的生物学价值,揭示密切相关的生物过程和途径。总的来说,拟议的方法学框架提供了联合放射转录组学标记和模型,以支持转录组和癌症表型之间的联系和互补性,如在NSCLC的情况下所证明的。
    Radiotranscriptomics is an emerging field that aims to investigate the relationships between the radiomic features extracted from medical images and gene expression profiles that contribute in the diagnosis, treatment planning, and prognosis of cancer. This study proposes a methodological framework for the investigation of these associations with application on non-small-cell lung cancer (NSCLC). Six publicly available NSCLC datasets with transcriptomics data were used to derive and validate a transcriptomic signature for its ability to differentiate between cancer and non-malignant lung tissue. A publicly available dataset of 24 NSCLC-diagnosed patients, with both transcriptomic and imaging data, was used for the joint radiotranscriptomic analysis. For each patient, 749 Computed Tomography (CT) radiomic features were extracted and the corresponding transcriptomics data were provided through DNA microarrays. The radiomic features were clustered using the iterative K-means algorithm resulting in 77 homogeneous clusters, represented by meta-radiomic features. The most significant differentially expressed genes (DEGs) were selected by performing Significance Analysis of Microarrays (SAM) and 2-fold change. The interactions among the CT imaging features and the selected DEGs were investigated using SAM and a Spearman rank correlation test with a False Discovery Rate (FDR) of 5%, leading to the extraction of 73 DEGs significantly correlated with radiomic features. These genes were used to produce predictive models of the meta-radiomics features, defined as p-metaomics features, by performing Lasso regression. Of the 77 meta-radiomic features, 51 can be modeled in terms of the transcriptomic signature. These significant radiotranscriptomics relationships form a reliable basis to biologically justify the radiomics features extracted from anatomic imaging modalities. Thus, the biological value of these radiomic features was justified via enrichment analysis on their transcriptomics-based regression models, revealing closely associated biological processes and pathways. Overall, the proposed methodological framework provides joint radiotranscriptomics markers and models to support the connection and complementarities between the transcriptome and the phenotype in cancer, as demonstrated in the case of NSCLC.
    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
    Radiogenomic and radiotranscriptomic studies have the potential to pave the way for a holistic decision support system built on genomics, transcriptomics, radiomics, deep features and clinical parameters to assess treatment evaluation and care planning. The integration of invasive and routine imaging data into a common feature space has the potential to yield robust models for inferring the drivers of underlying biological mechanisms. In this non-small cell lung carcinoma study, a multi-omics representation comprised deep features and transcriptomics was evaluated to further explore the synergetic and complementary properties of these diverse multi-view data sources by utilizing data-driven machine learning models. The proposed deep radiotranscriptomic analysis is a feature-based fusion that significantly enhances sensitivity by up to 0.174 and AUC by up to 0.22, compared to the baseline single source models, across all experiments on the unseen testing set. Additionally, a radiomics-based fusion was also explored as an alternative methodology yielding radiomic signatures that are comparable to several previous publications in the field of radiogenomics. Furthermore, the machine learning multi-omics analysis based on deep features and transcriptomics achieved an AUC performance of up to 0.831 ± 0.09/0.925 ± 0.04 for the examined molecular and histology subtypes analysis, respectively. The clinical impact of such high-performing models can add prognostic value and lead to optimal treatment assessment by targeting specific oncogenes, namely the response of tyrosine kinase inhibitors of EGFR mutated or predicting the chemotherapy resistance of KRAS mutated tumors.
    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