8,140 research outputs found

    11th German Conference on Chemoinformatics (GCC 2015) : Fulda, Germany. 8-10 November 2015.

    Get PDF

    Systems Analytics and Integration of Big Omics Data

    Get PDF
    A “genotype"" is essentially an organism's full hereditary information which is obtained from its parents. A ""phenotype"" is an organism's actual observed physical and behavioral properties. These may include traits such as morphology, size, height, eye color, metabolism, etc. One of the pressing challenges in computational and systems biology is genotype-to-phenotype prediction. This is challenging given the amount of data generated by modern Omics technologies. This “Big Data” is so large and complex that traditional data processing applications are not up to the task. Challenges arise in collection, analysis, mining, sharing, transfer, visualization, archiving, and integration of these data. In this Special Issue, there is a focus on the systems-level analysis of Omics data, recent developments in gene ontology annotation, and advances in biological pathways and network biology. The integration of Omics data with clinical and biomedical data using machine learning is explored. This Special Issue covers new methodologies in the context of gene–environment interactions, tissue-specific gene expression, and how external factors or host genetics impact the microbiome

    Artificial intelligence, machine learning, and drug repurposing in cancer

    Get PDF
    Introduction: Drug repurposing provides a cost-effective strategy to re-use approved drugs for new medical indications. Several machine learning (ML) and artificial intelligence (AI) approaches have been developed for systematic identification of drug repurposing leads based on big data resources, hence further accelerating and de-risking the drug development process by computational means. Areas covered: The authors focus on supervised ML and AI methods that make use of publicly available databases and information resources. While most of the example applications are in the field of anticancer drug therapies, the methods and resources reviewed are widely applicable also to other indications including COVID-19 treatment. A particular emphasis is placed on the use of comprehensive target activity profiles that enable a systematic repurposing process by extending the target profile of drugs to include potent off-targets with therapeutic potential for a new indication. Expert opinion: The scarcity of clinical patient data and the current focus on genetic aberrations as primary drug targets may limit the performance of anticancer drug repurposing approaches that rely solely on genomics-based information. Functional testing of cancer patient cells exposed to a large number of targeted therapies and their combinations provides an additional source of repurposing information for tissue-aware AI approaches.Peer reviewe

    Network-based methods for biological data integration in precision medicine

    Full text link
    [eng] The vast and continuously increasing volume of available biomedical data produced during the last decades opens new opportunities for large-scale modeling of disease biology, facilitating a more comprehensive and integrative understanding of its processes. Nevertheless, this type of modelling requires highly efficient computational systems capable of dealing with such levels of data volumes. Computational approximations commonly used in machine learning and data analysis, namely dimensionality reduction and network-based approaches, have been developed with the goal of effectively integrating biomedical data. Among these methods, network-based machine learning stands out due to its major advantage in terms of biomedical interpretability. These methodologies provide a highly intuitive framework for the integration and modelling of biological processes. This PhD thesis aims to explore the potential of integration of complementary available biomedical knowledge with patient-specific data to provide novel computational approaches to solve biomedical scenarios characterized by data scarcity. The primary focus is on studying how high-order graph analysis (i.e., community detection in multiplex and multilayer networks) may help elucidate the interplay of different types of data in contexts where statistical power is heavily impacted by small sample sizes, such as rare diseases and precision oncology. The central focus of this thesis is to illustrate how network biology, among the several data integration approaches with the potential to achieve this task, can play a pivotal role in addressing this challenge provided its advantages in molecular interpretability. Through its insights and methodologies, it introduces how network biology, and in particular, models based on multilayer networks, facilitates bringing the vision of precision medicine to these complex scenarios, providing a natural approach for the discovery of new biomedical relationships that overcomes the difficulties for the study of cohorts presenting limited sample sizes (data-scarce scenarios). Delving into the potential of current artificial intelligence (AI) and network biology applications to address data granularity issues in the precision medicine field, this PhD thesis presents pivotal research works, based on multilayer networks, for the analysis of two rare disease scenarios with specific data granularities, effectively overcoming the classical constraints hindering rare disease and precision oncology research. The first research article presents a personalized medicine study of the molecular determinants of severity in congenital myasthenic syndromes (CMS), a group of rare disorders of the neuromuscular junction (NMJ). The analysis of severity in rare diseases, despite its importance, is typically neglected due to data availability. In this study, modelling of biomedical knowledge via multilayer networks allowed understanding the functional implications of individual mutations in the cohort under study, as well as their relationships with the causal mutations of the disease and the different levels of severity observed. Moreover, the study presents experimental evidence of the role of a previously unsuspected gene in NMJ activity, validating the hypothetical role predicted using the newly introduced methodologies. The second research article focuses on the applicability of multilayer networks for gene priorization. Enhancing concepts for the analysis of different data granularities firstly introduced in the previous article, the presented research provides a methodology based on the persistency of network community structures in a range of modularity resolution, effectively providing a new framework for gene priorization for patient stratification. In summary, this PhD thesis presents major advances on the use of multilayer network-based approaches for the application of precision medicine to data-scarce scenarios, exploring the potential of integrating extensive available biomedical knowledge with patient-specific data

    Knowledge-based Biomedical Data Science 2019

    Full text link
    Knowledge-based biomedical data science (KBDS) involves the design and implementation of computer systems that act as if they knew about biomedicine. Such systems depend on formally represented knowledge in computer systems, often in the form of knowledge graphs. Here we survey the progress in the last year in systems that use formally represented knowledge to address data science problems in both clinical and biological domains, as well as on approaches for creating knowledge graphs. Major themes include the relationships between knowledge graphs and machine learning, the use of natural language processing, and the expansion of knowledge-based approaches to novel domains, such as Chinese Traditional Medicine and biodiversity.Comment: Manuscript 43 pages with 3 tables; Supplemental material 43 pages with 3 table
    • 

    corecore