2,850 research outputs found

    Identifying Patterns of Breast Cancer Genetic Signatures using Unsupervised Machine Learning

    Get PDF
    Deploying machine learning to improve medical diagnosis is a promising area. The purpose of this study is to identify and analyze unique genetic signatures for breast cancer grades using publicly available gene expression microarray data. The classification of cancer types is based on unsupervised feature learning. Unsupervised clustering use matrix algebra based on similarity measures which made it suitable for analyzing gene expression. The main advantage of the proposed approach is the ability to use gene expression data from different grades of breast cancer to generate features that automatically identify and enhance the cancer diagnosis. In this paper, we tested different similarity measures in order to find the best way that identifies the sets of genes with a common function using expression microarray data

    Machine Learning and Integrative Analysis of Biomedical Big Data.

    Get PDF
    Recent developments in high-throughput technologies have accelerated the accumulation of massive amounts of omics data from multiple sources: genome, epigenome, transcriptome, proteome, metabolome, etc. Traditionally, data from each source (e.g., genome) is analyzed in isolation using statistical and machine learning (ML) methods. Integrative analysis of multi-omics and clinical data is key to new biomedical discoveries and advancements in precision medicine. However, data integration poses new computational challenges as well as exacerbates the ones associated with single-omics studies. Specialized computational approaches are required to effectively and efficiently perform integrative analysis of biomedical data acquired from diverse modalities. In this review, we discuss state-of-the-art ML-based approaches for tackling five specific computational challenges associated with integrative analysis: curse of dimensionality, data heterogeneity, missing data, class imbalance and scalability issues

    Development of a simple artificial intelligence method to accurately subtype breast cancers based on gene expression barcodes

    Get PDF
    >Magister Scientiae - MScINTRODUCTION: Breast cancer is a highly heterogeneous disease. The complexity of achieving an accurate diagnosis and an effective treatment regimen lies within this heterogeneity. Subtypes of the disease are not simply molecular, i.e. hormone receptor over-expression or absence, but the tumour itself is heterogeneous in terms of tissue of origin, metastases, and histopathological variability. Accurate tumour classification vastly improves treatment decisions, patient outcomes and 5-year survival rates. Gene expression studies aided by transcriptomic technologies such as microarrays and next-generation sequencing (e.g. RNA-Sequencing) have aided oncology researcher and clinician understanding of the complex molecular portraits of malignant breast tumours. Mechanisms governing cancers, which include tumorigenesis, gene fusions, gene over-expression and suppression, cellular process and pathway involvementinvolvement, have been elucidated through comprehensive analyses of the cancer transcriptome. Over the past 20 years, gene expression signatures, discovered with both microarray and RNA-Seq have reached clinical and commercial application through the development of tests such as Mammaprint®, OncotypeDX®, and FoundationOne® CDx, all which focus on chemotherapy sensitivity, prediction of cancer recurrence, and tumour mutational level. The Gene Expression Barcode (GExB) algorithm was developed to allow for easy interpretation and integration of microarray data through data normalization with frozen RMA (fRMA) preprocessing and conversion of relative gene expression to a sequence of 1's and 0's. Unfortunately, the algorithm has not yet been developed for RNA-Seq data. However, implementation of the GExB with feature-selection would contribute to a machine-learning based robust breast cancer and subtype classifier. METHODOLOGY: For microarray data, we applied the GExB algorithm to generate barcodes for normal breast and breast tumour samples. A two-class classifier for malignancy was developed through feature-selection on barcoded samples by selecting for genes with 85% stable absence or presence within a tissue type, and differentially stable between tissues. A multi-class feature-selection method was employed to identify genes with variable expression in one subtype, but 80% stable absence or presence in all other subtypes, i.e. 80% in n-1 subtypes. For RNA-Seq data, a barcoding method needed to be developed which could mimic the GExB algorithm for microarray data. A z-score-to-barcode method was implemented and differential gene expression analysis with selection of the top 100 genes as informative features for classification purposes. The accuracy and discriminatory capability of both microarray-based gene signatures and the RNA-Seq-based gene signatures was assessed through unsupervised and supervised machine-learning algorithms, i.e., K-means and Hierarchical clustering, as well as binary and multi-class Support Vector Machine (SVM) implementations. RESULTS: The GExB-FS method for microarray data yielded an 85-probe and 346-probe informative set for two-class and multi-class classifiers, respectively. The two-class classifier predicted samples as either normal or malignant with 100% accuracy and the multi-class classifier predicted molecular subtype with 96.5% accuracy with SVM. Combining RNA-Seq DE analysis for feature-selection with the z-score-to-barcode method, resulted in a two-class classifier for malignancy, and a multi-class classifier for normal-from-healthy, normal-adjacent-tumour (from cancer patients), and breast tumour samples with 100% accuracy. Most notably, a normal-adjacent-tumour gene expression signature emerged, which differentiated it from normal breast tissues in healthy individuals. CONCLUSION: A potentially novel method for microarray and RNA-Seq data transformation, feature selection and classifier development was established. The universal application of the microarray signatures and validity of the z-score-to-barcode method was proven with 95% accurate classification of RNA-Seq barcoded samples with a microarray discovered gene expression signature. The results from this comprehensive study into the discovery of robust gene expression signatures holds immense potential for further R&F towards implementation at the clinical endpoint, and translation to simpler and cost-effective laboratory methods such as qtPCR-based tests

    Spatial Organization and Molecular Correlation of Tumor-Infiltrating Lymphocytes Using Deep Learning on Pathology Images

    Get PDF
    Beyond sample curation and basic pathologic characterization, the digitized H&E-stained images of TCGA samples remain underutilized. To highlight this resource, we present mappings of tumorinfiltrating lymphocytes (TILs) based on H&E images from 13 TCGA tumor types. These TIL maps are derived through computational staining using a convolutional neural network trained to classify patches of images. Affinity propagation revealed local spatial structure in TIL patterns and correlation with overall survival. TIL map structural patterns were grouped using standard histopathological parameters. These patterns are enriched in particular T cell subpopulations derived from molecular measures. TIL densities and spatial structure were differentially enriched among tumor types, immune subtypes, and tumor molecular subtypes, implying that spatial infiltrate state could reflect particular tumor cell aberration states. Obtaining spatial lymphocytic patterns linked to the rich genomic characterization of TCGA samples demonstrates one use for the TCGA image archives with insights into the tumor-immune microenvironment

    Pathway-Based Multi-Omics Data Integration for Breast Cancer Diagnosis and Prognosis.

    Get PDF
    Ph.D. Thesis. University of Hawaiʻi at Mānoa 2017

    Molecular Signature as Optima of Multi-Objective Function with Applications to Prediction in Oncogenomics

    Get PDF
    Náplní této práce je teoretický úvod a následné praktické zpracování tématu Molekulární signatura jako optimální multi-objektivní funkce s aplikací v predikci v onkogenomice. Úvodní kapitoly jsou zaměřeny na téma rakovina, zejména pak rakovina prsu a její podtyp triple negativní rakovinu prsu. Následuje literární přehled z oblasti optimalizačních metod, zejména se zaměřením na metaheuristické metody a problematiku strojového učení. Část se odkazuje na onkogenomiku a principy microarray a také na statistiku a s důrazem na výpočet p-hodnoty a bimodálního indexu. Praktická část je pak zaměřena na konkrétní průběh výzkumu a nalezené závěry, vedoucí k dalším krokům výzkumu. Implementace vybraných metod byla provedena v programech Matlab a R, s využitím dalších programovacích jazyků a to konkrétně programů Java a Python.Content of this work is theoretical introduction and follow-up practical processing of topic Molecular signature as optima of multi-objective function with applications to prediction in oncogenomics. Opening chapters are targeted on topic of cancer, mainly on breast cancer and its subtype Triple Negative Breast Cancer. Succeeds the literature review of optimization methods, mainly on meta-heuristic methods for multi-objective optimization and problematic of machine learning. Part is focused on the oncogenomics and on the principal of microarray and also to statistics methods with emphasis on the calculation of p-value and Bimodality Index. Practical part of work consists from concrete research and conclusions lead to next steps of research. Implementation of selected methods was realised in Matlab and R, with use of other programming languages Java and Python.

    Deep Domain Adaptation Learning Framework for Associating Image Features to Tumour Gene Profile

    Get PDF
    While medical imaging and general pathology are routine in cancer diagnosis, genetic sequencing is not always assessable due to the strong phenotypic and genetic heterogeneity of human cancers. Image-genomics integrates medical imaging and genetics to provide a complementary approach to optimise cancer diagnosis by associating tumour imaging traits with clinical data and has demonstrated its potential in identifying imaging surrogates for tumour biomarkers. However, existing image-genomics research has focused on quantifying tumour visual traits according to human understanding, which may not be optimal across different cancer types. The challenge hence lies in the extraction of optimised imaging representations in an objective data-driven manner. Such an approach requires large volumes of annotated image data that are difficult to acquire. We propose a deep domain adaptation learning framework for associating image features to tumour genetic information, exploiting the ability of domain adaptation technique to learn relevant image features from close knowledge domains. Our proposed framework leverages the current state-of-the-art in image object recognition to provide image features to encode subtle variations of tumour phenotypic characteristics with domain adaptation techniques. The proposed framework was evaluated with current state-of-the-art in: (i) tumour histopathology image classification and; (ii) image-genomics associations. The proposed framework demonstrated improved accuracy of tumour classification, as well as providing additional data-derived representations of tumour phenotypic characteristics that exhibit strong image-genomics association. This thesis advances and indicates the potential of image-genomics research to reveal additional imaging surrogates to genetic biomarkers, which has the potential to facilitate cancer diagnosis
    corecore