skip to main content


Title: Pseudotime Based Discovery of Breast Cancer Heterogeneity
Breast cancer is highly sporadic and heterogeneous in nature. Even the patients with same clinical stage do not cluster together in terms of genomic profiles such as mRNA expression. In order to prevent and cure breast cancer completely, it is essential to decipher the detailed heterogeneity of breast cancer at genomic level. Putting the cancer patients on a time scale, which represents the trajectory of cancer development, may help discover the detailed heterogeneity. This in turn would help establish the mechanisms for prevention and complete cure of breast cancer. The goal of this study is to discover the heterogeneity of breast cancer by ordering the cancer patients using pseudotime. This is achieved through two objectives: First, a computational framework is developed to place the cancer patients on a time scale, meaning construct a trajectory of cancer development, by inferring pseudotime from static mRNA expression data; Second, discovering breast cancer heterogeneity at different time periods of the trajectory using statistical and machine learning techniques. In this study, the trajectory of breast cancer progression was constructed using static mRNA expression profiles of 1072 breast cancer patients by inferring pseudotime. Three sets of key genes discovered using supervised machine learning techniques are used to develop the trajectories. The first set of genes are PAM50 genes which is available in literature. The second and third sets of genes were discovered in the present study using the clinical stages of breast cancer (Stage-I, Stage-II, Stage-III, and Stage-IV). The proposed computational framework has the capability of deciphering heterogeneity in breast cancer at a granular level. The results also show the existence of multiple parallel trajectories at different time periods of cancer development or progression.  more » « less
Award ID(s):
1901628 1651917
NSF-PAR ID:
10141530
Author(s) / Creator(s):
; ;
Date Published:
Journal Name:
2019 IEEE International Conference on Bioinformatics and Biomedicine (IEEE BIBM)
Page Range / eLocation ID:
2049 to 2054
Format(s):
Medium: X
Sponsoring Org:
National Science Foundation
More Like this
  1. Roy, Sushmita (Ed.)
    Unraveling molecular regulatory networks underlying disease progression is critically important for understanding disease mechanisms and identifying drug targets. The existing methods for inferring gene regulatory networks (GRNs) rely mainly on time-course gene expression data. However, most available omics data from cross-sectional studies of cancer patients often lack sufficient temporal information, leading to a key challenge for GRN inference. Through quantifying the latent progression using random walks-based manifold distance, we propose a latent-temporal progression-based Bayesian method, PROB, for inferring GRNs from the cross-sectional transcriptomic data of tumor samples. The robustness of PROB to the measurement variabilities in the data is mathematically proved and numerically verified. Performance evaluation on real data indicates that PROB outperforms other methods in both pseudotime inference and GRN inference. Applications to bladder cancer and breast cancer demonstrate that our method is effective to identify key regulators of cancer progression or drug targets. The identified ACSS1 is experimentally validated to promote epithelial-to-mesenchymal transition of bladder cancer cells, and the predicted FOXM1-targets interactions are verified and are predictive of relapse in breast cancer. Our study suggests new effective ways to clinical transcriptomic data modeling for characterizing cancer progression and facilitates the translation of regulatory network-based approaches into precision medicine. 
    more » « less
  2. Advanced genomic and molecular profiling technologies accelerated the enlightenment of the regulatory mechanisms behind cancer development and progression, and the targeted therapies in patients. Along this line, intense studies with immense amounts of biological information have boosted the discovery of molecular biomarkers. Cancer is one of the leading causes of death around the world in recent years. Elucidation of genomic and epigenetic factors in Breast Cancer (BRCA) can provide a roadmap to uncover the disease mechanisms. Accordingly, unraveling the possible systematic connections between-omics data types and their contribution to BRCA tumor progression is crucial. In this study, we have developed a novel machine learning (ML) based integrative approach for multi-omics data analysis. This integrative approach combines information from gene expression (mRNA), microRNA (miRNA) and methylation data. Due to the complexity of cancer, this integrated data is expected to improve the prediction, diagnosis and treatment of disease through patterns only available from the 3-way interactions between these 3-omics datasets. In addition, the proposed method bridges the interpretation gap between the disease mechanisms that drive onset and progression. Our fundamental contribution is the 3 Multi-omics integrative tool (3Mint). This tool aims to perform grouping and scoring of groups using biological knowledge. Another major goal is improved gene selection via detection of novel groups of cross-omics biomarkers. Performance of 3Mint is assessed using different metrics. Our computational performance evaluations showed that the 3Mint classifies the BRCA molecular subtypes with lower number of genes when compared to the miRcorrNet tool which uses miRNA and mRNA gene expression profiles in terms of similar performance metrics (95% Accuracy). The incorporation of methylation data in 3Mint yields a much more focused analysis. The 3Mint tool and all other supplementary files are available at https://github.com/malikyousef/3Mint/ . 
    more » « less
  3. There is currently no gene expression assay that can assess if premalignant lesions will develop into invasive breast cancer. This study sought to identify biomarkers for selecting patients with a high potential for developing invasive carcinoma in the breast with normal histology, benign lesions, or premalignant lesions. A set of 26-gene mRNA expression profiles were used to identify invasive ductal carcinomas from histologically normal tissue and benign lesions and to select those with a higher potential for future cancer development (ADHC) in the breast associated with atypical ductal hyperplasia (ADH). The expression-defined model achieved an overall accuracy of 94.05% (AUC = 0.96) in classifying invasive ductal carcinomas from histologically normal tissue and benign lesions (n = 185). This gene signature classified cancer development in ADH tissues with an overall accuracy of 100% (n = 8). The mRNA expression patterns of these 26 genes were validated using RT-PCR analyses of independent tissue samples (n = 77) and blood samples (n = 48). The protein expression of PBX2 and RAD52 assessed with immunohistochemistry were prognostic of breast cancer survival outcomes. This signature provided significant prognostic stratification in The Cancer Genome Atlas breast cancer patients (n = 1100), as well as basal-like and luminal A subtypes, and was associated with distinct immune infiltration and activities. The mRNA and protein expression of the 26 genes was associated with sensitivity or resistance to 18 NCCN-recommended drugs for treating breast cancer. Eleven genes had significant proliferative potential in CRISPR-Cas9/RNAi screening. Based on this gene expression signature, the VEGFR inhibitor ZM-306416 was discovered as a new drug for treating breast cancer.

     
    more » « less
  4. Abstract Motivation Detecting cancer gene expression and transcriptome changes with mRNA-sequencing (RNA-Seq) or array-based data are important for understanding the molecular mechanisms underlying carcinogenesis and cellular events during cancer progression. In previous studies, the differentially expressed genes were detected across patients in one cancer type. These studies ignored the role of mRNA expression changes in driving tumorigenic mechanisms that are either universal or specific in different tumor types. To address the problem, we introduce two network-based multi-task learning frameworks, NetML and NetSML, to discover common differentially expressed genes shared across different cancer types as well as differentially expressed genes specific to each cancer type. The proposed frameworks consider the common latent gene co-expression modules and gene-sample biclusters underlying the multiple cancer datasets to learn the knowledge crossing different tumor types. Results Large-scale experiments on simulations and real cancer high-throughput datasets validate that the proposed network-based multi-task learning frameworks perform better sample classification compared with the models without the knowledge sharing across different cancer types. The common and cancer specific molecular signatures detected by multi-task learning frameworks on TCGA ovarian cancer, breast cancer, and prostate cancer datasets are correlated with the known marker genes and enriched in cancer relevant KEGG pathways and Gene Ontology terms. Availability and Implementation Source code is available at: https://github.com/compbiolabucf/NetML Supplementary information Supplementary data are available at Bioinformatics 
    more » « less
  5. Finding the network biomarkers of cancers and the analysis of cancer driving genes that are involved in these biomarkers are essential for understanding the dynamics of cancer. Clusters of genes in co-expression networks are commonly known as functional units. This work is based on the hypothesis that the dense clusters or communities in the gene co-expression networks of cancer patients may represent functional units regarding cancer initiation and progression. In this study, RNA-seq gene expression data of three cancers - Breast Invasive Carcinoma (BRCA), Colorectal Adenocarcinoma (COAD) and Glioblastoma Multiforme (GBM) - from The Cancer Genome Atlas (TCGA) are used to construct gene co-expression networks using Pearson Correlation. Six well-known community detection algorithms are applied on these networks to identify communities with five or more genes. A permutation test is performed to further mine the communities that are conserved in other cancers, thus calling them conserved communities. Then survival analysis is performed on clinical data of three cancers using the conserved community genes as prognostic co-variates. The communities that could distinguish the cancer patients between high- and low-risk groups are considered as cancer biomarkers. In the present study, 16 such network biomarkers are discovered. 
    more » « less