3,647 research outputs found

    Computational models and approaches for lung cancer diagnosis

    Full text link
    The success of treatment of patients with cancer depends on establishing an accurate diagnosis. To this end, the aim of this study is to developed novel lung cancer diagnostic models. New algorithms are proposed to analyse the biological data and extract knowledge that assists in achieving accurate diagnosis results

    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

    Artificial Neural Network Inference (ANNI): A Study on Gene-Gene Interaction for Biomarkers in Childhood Sarcomas

    Get PDF
    Objective: To model the potential interaction between previously identified biomarkers in children sarcomas using artificial neural network inference (ANNI). Method: To concisely demonstrate the biological interactions between correlated genes in an interaction network map, only 2 types of sarcomas in the children small round blue cell tumors (SRBCTs) dataset are discussed in this paper. A backpropagation neural network was used to model the potential interaction between genes. The prediction weights and signal directions were used to model the strengths of the interaction signals and the direction of the interaction link between genes. The ANN model was validated using Monte Carlo cross-validation to minimize the risk of over-fitting and to optimize generalization ability of the model. Results: Strong connection links on certain genes (TNNT1 and FNDC5 in rhabdomyosarcoma (RMS); FCGRT and OLFM1 in Ewing’s sarcoma (EWS)) suggested their potency as central hubs in the interconnection of genes with different functionalities. The results showed that the RMS patients in this dataset are likely to be congenital and at low risk of cardiomyopathy development. The EWS patients are likely to be complicated by EWS-FLI fusion and deficiency in various signaling pathways, including Wnt, Fas/Rho and intracellular oxygen. Conclusions: The ANN network inference approach and the examination of identified genes in the published literature within the context of the disease highlights the substantial influence of certain genes in sarcomas

    The importance of data classification using machine learning methods in microarray data

    Get PDF
    The detection of genetic mutations has attracted global attention. several methods have proposed to detect diseases such as cancers and tumours. One of them is microarrays, which is a type of representation for gene expression that is helpful in diagnosis. To unleash the full potential of microarrays, machine-learning algorithms and gene selection methods can be implemented to facilitate processing on microarrays and to overcome other potential challenges. One of these challenges involves high dimensional data that are redundant, irrelevant, and noisy. To alleviate this problem, this representation should be simplified. For example, the feature selection process can be implemented by reducing the number of features adopted in clustering and classification. A subset of genes can be selected from a pool of gene expression data recorded on DNA micro-arrays. This paper reviews existing classification techniques and gene selection methods. The effectiveness of emerging techniques, such as the swarm intelligence technique in feature selection and classification in microarrays, are reported as well. These emerging techniques can be used in detecting cancer. The swarm intelligence technique can be combined with other statistical methods for attaining better results

    Significant Gene Array Analysis and Cluster-Based Machine Learning for Disease Class Prediction

    Get PDF
    Gene expression analysis has been of major interest to biostatisticians for many decades. Such studies are necessary for the understanding of disease risk assessment and prediction, so that medical professionals and scientists alike may learn how to better create treatment plans to lessen symptoms and perhaps even find cures. In this study, we will investigate various gene expression analyses and machine learning techniques for disease class prediction, as well as assess predictive validity of these models and uncover differentially expressed (DE) genes for their relevant pathology datasets. Multiple gene expression datasets will be used to test model accuracies and will be obtained using the Affymetrix U133A platform (GPL96). Significant Analysis of Microarrays (SAM) had been used to identify potential disease biomarkers, followed by these predictive models: (a) random forest, (b) random forest with Gene eXpression Network Analysis (GXNA), (c) RF++, (d) LASSO, and (e) Bayesian Neural Networks. One of the intended goals for this study is to find clusters of co-expressed genes and identify the effect of clustering classification based on knowledge in gene expression data/microarray data. The other goal is to determine the usefulness of Automatic Relevancy Determination in Bayesian neural networks

    A voting-based machine learning approach for classifying biological and clinical datasets.

    Get PDF
    BACKGROUND: Different machine learning techniques have been proposed to classify a wide range of biological/clinical data. Given the practicability of these approaches accordingly, various software packages have been also designed and developed. However, the existing methods suffer from several limitations such as overfitting on a specific dataset, ignoring the feature selection concept in the preprocessing step, and losing their performance on large-size datasets. To tackle the mentioned restrictions, in this study, we introduced a machine learning framework consisting of two main steps. First, our previously suggested optimization algorithm (Trader) was extended to select a near-optimal subset of features/genes. Second, a voting-based framework was proposed to classify the biological/clinical data with high accuracy. To evaluate the efficiency of the proposed method, it was applied to 13 biological/clinical datasets, and the outcomes were comprehensively compared with the prior methods. RESULTS: The results demonstrated that the Trader algorithm could select a near-optimal subset of features with a significant level of p-value \u3c 0.01 relative to the compared algorithms. Additionally, on the large-sie datasets, the proposed machine learning framework improved prior studies by ~ 10% in terms of the mean values associated with fivefold cross-validation of accuracy, precision, recall, specificity, and F-measure. CONCLUSION: Based on the obtained results, it can be concluded that a proper configuration of efficient algorithms and methods can increase the prediction power of machine learning approaches and help researchers in designing practical diagnosis health care systems and offering effective treatment plans

    Machine Learning Models for Deciphering Regulatory Mechanisms and Morphological Variations in Cancer

    Get PDF
    The exponential growth of multi-omics biological datasets is resulting in an emerging paradigm shift in fundamental biological research. In recent years, imaging and transcriptomics datasets are increasingly incorporated into biological studies, pushing biology further into the domain of data-intensive-sciences. New approaches and tools from statistics, computer science, and data engineering are profoundly influencing biological research. Harnessing this ever-growing deluge of multi-omics biological data requires the development of novel and creative computational approaches. In parallel, fundamental research in data sciences and Artificial Intelligence (AI) has advanced tremendously, allowing the scientific community to generate a massive amount of knowledge from data. Advances in Deep Learning (DL), in particular, are transforming many branches of engineering, science, and technology. Several of these methodologies have already been adapted for harnessing biological datasets; however, there is still a need to further adapt and tailor these techniques to new and emerging technologies. In this dissertation, we present computational algorithms and tools that we have developed to study gene-regulation and cellular morphology in cancer. The models and platforms that we have developed are general and widely applicable to several problems relating to dysregulation of gene expression in diseases. Our pipelines and software packages are disseminated in public repositories for larger scientific community use. This dissertation is organized in three main projects. In the first project, we present Causal Inference Engine (CIE), an integrated platform for the identification and interpretation of active regulators of transcriptional response. The platform offers visualization tools and pathway enrichment analysis to map predicted regulators to Reactome pathways. We provide a parallelized R-package for fast and flexible directional enrichment analysis to run the inference on custom regulatory networks. Next, we designed and developed MODEX, a fully automated text-mining system to extract and annotate causal regulatory interaction between Transcription Factors (TFs) and genes from the biomedical literature. MODEX uses putative TF-gene interactions derived from high-throughput ChIP-Seq or other experiments and seeks to collect evidence and meta-data in the biomedical literature to validate and annotate the interactions. MODEX is a complementary platform to CIE that provides auxiliary information on CIE inferred interactions by mining the literature. In the second project, we present a Convolutional Neural Network (CNN) classifier to perform a pan-cancer analysis of tumor morphology, and predict mutations in key genes. The main challenges were to determine morphological features underlying a genetic status and assess whether these features were common in other cancer types. We trained an Inception-v3 based model to predict TP53 mutation in five cancer types with the highest rate of TP53 mutations. We also performed a cross-classification analysis to assess shared morphological features across multiple cancer types. Further, we applied a similar methodology to classify HER2 status in breast cancer and predict response to treatment in HER2 positive samples. For this study, our training slides were manually annotated by expert pathologists to highlight Regions of Interest (ROIs) associated with HER2+/- tumor microenvironment. Our results indicated that there are strong morphological features associated with each tumor type. Moreover, our predictions highly agree with manual annotations in the test set, indicating the feasibility of our approach in devising an image-based diagnostic tool for HER2 status and treatment response prediction. We have validated our model using samples from an independent cohort, which demonstrates the generalizability of our approach. Finally, in the third project, we present an approach to use spatial transcriptomics data to predict spatially-resolved active gene regulatory mechanisms in tissues. Using spatial transcriptomics, we identified tissue regions with differentially expressed genes and applied our CIE methodology to predict active TFs that can potentially regulate the marker genes in the region. This project bridged the gap between inference of active regulators using molecular data and morphological studies using images. The results demonstrate a significant local pattern in TF activity across the tissue, indicating differential spatial-regulation in tissues. The results suggest that the integrative analysis of spatial transcriptomics data with CIE can capture discriminant features and identify localized TF-target links in the tissue

    Passively mode-locked laser using an entirely centred erbium-doped fiber

    Get PDF
    This paper describes the setup and experimental results for an entirely centred erbium-doped fiber laser with passively mode-locked output. The gain medium of the ring laser cavity configuration comprises a 3 m length of two-core optical fiber, wherein an undoped outer core region of 9.38 μm diameter surrounds a 4.00 μm diameter central core region doped with erbium ions at 400 ppm concentration. The generated stable soliton mode-locking output has a central wavelength of 1533 nm and pulses that yield an average output power of 0.33 mW with a pulse energy of 31.8 pJ. The pulse duration is 0.7 ps and the measured output repetition rate of 10.37 MHz corresponds to a 96.4 ns pulse spacing in the pulse train

    Biological Factor Regulatory Neural Network

    Full text link
    Genes are fundamental for analyzing biological systems and many recent works proposed to utilize gene expression for various biological tasks by deep learning models. Despite their promising performance, it is hard for deep neural networks to provide biological insights for humans due to their black-box nature. Recently, some works integrated biological knowledge with neural networks to improve the transparency and performance of their models. However, these methods can only incorporate partial biological knowledge, leading to suboptimal performance. In this paper, we propose the Biological Factor Regulatory Neural Network (BFReg-NN), a generic framework to model relations among biological factors in cell systems. BFReg-NN starts from gene expression data and is capable of merging most existing biological knowledge into the model, including the regulatory relations among genes or proteins (e.g., gene regulatory networks (GRN), protein-protein interaction networks (PPI)) and the hierarchical relations among genes, proteins and pathways (e.g., several genes/proteins are contained in a pathway). Moreover, BFReg-NN also has the ability to provide new biologically meaningful insights because of its white-box characteristics. Experimental results on different gene expression-based tasks verify the superiority of BFReg-NN compared with baselines. Our case studies also show that the key insights found by BFReg-NN are consistent with the biological literature
    corecore