2,805 research outputs found

    Translational bioinformatics: linking knowledge across biological and clinical realms

    Get PDF
    Nearly a decade since the completion of the first draft of the human genome, the biomedical community is positioned to usher in a new era of scientific inquiry that links fundamental biological insights with clinical knowledge. Accordingly, holistic approaches are needed to develop and assess hypotheses that incorporate genotypic, phenotypic, and environmental knowledge. This perspective presents translational bioinformatics as a discipline that builds on the successes of bioinformatics and health informatics for the study of complex diseases. The early successes of translational bioinformatics are indicative of the potential to achieve the promise of the Human Genome Project for gaining deeper insights to the genetic underpinnings of disease and progress toward the development of a new generation of therapies

    Doctor of Philosophy

    Get PDF
    dissertationThe widespread use of genomic information to improve clinical care has long been a goal of clinicians, researchers, and policy-makers. With the completion of the Human Genome Project over a decade ago, the feasibility of attaining this goal on a widespread basis is becoming a greater reality. In fact, new genome sequencing technologies are bringing the cost of obtaining a patient's genomic information within reach of the general population. While this is an exciting prospect to health care, many barriers still remain to effectively use genomic information in a clinically meaningful way. These barriers, if not overcome, will limit the ability of genomic information to provide a significant impact on health care. Nevertheless, clinical decision support (CDS), which entails the provision of patient-specific knowledge to clinicians at appropriate times to enhance health care, offers a feasible solution. As such, this body of work represents an effort to develop a functional CDS solution capable of leveraging whole genome sequence information on a widespread basis. Many considerations were made in the design of the CDS solution in order to overcome the complexities of genomic information while aligning with common health information technology approaches and standards. This work represents an important advancement in the capabilities of integrating actionable genomic information within the clinical workflow using health informatics approaches

    A national clinical decision support infrastructure to enable the widespread and consistent practice of genomic and personalized medicine

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>In recent years, the completion of the Human Genome Project and other rapid advances in genomics have led to increasing anticipation of an era of genomic and personalized medicine, in which an individual's health is optimized through the use of all available patient data, including data on the individual's genome and its downstream products. Genomic and personalized medicine could transform healthcare systems and catalyze significant reductions in morbidity, mortality, and overall healthcare costs.</p> <p>Discussion</p> <p>Critical to the achievement of more efficient and effective healthcare enabled by genomics is the establishment of a robust, nationwide clinical decision support infrastructure that assists clinicians in their use of genomic assays to guide disease prevention, diagnosis, and therapy. Requisite components of this infrastructure include the standardized representation of genomic and non-genomic patient data across health information systems; centrally managed repositories of computer-processable medical knowledge; and standardized approaches for applying these knowledge resources against patient data to generate and deliver patient-specific care recommendations. Here, we provide recommendations for establishing a national decision support infrastructure for genomic and personalized medicine that fulfills these needs, leverages existing resources, and is aligned with the <it>Roadmap for National Action on Clinical Decision Support </it>commissioned by the U.S. Office of the National Coordinator for Health Information Technology. Critical to the establishment of this infrastructure will be strong leadership and substantial funding from the federal government.</p> <p>Summary</p> <p>A national clinical decision support infrastructure will be required for reaping the full benefits of genomic and personalized medicine. Essential components of this infrastructure include standards for data representation; centrally managed knowledge repositories; and standardized approaches for leveraging these knowledge repositories to generate patient-specific care recommendations at the point of care.</p

    Collaborative, Multidisciplinary Evaluation of Cancer Variants Through Virtual Molecular Tumor Boards Informs Local Clinical Practices.

    Get PDF
    PURPOSE: The cancer research community is constantly evolving to better understand tumor biology, disease etiology, risk stratification, and pathways to novel treatments. Yet the clinical cancer genomics field has been hindered by redundant efforts to meaningfully collect and interpret disparate data types from multiple high-throughput modalities and integrate into clinical care processes. Bespoke data models, knowledgebases, and one-off customized resources for data analysis often lack adequate governance and quality control needed for these resources to be clinical grade. Many informatics efforts focused on genomic interpretation resources for neoplasms are underway to support data collection, deposition, curation, harmonization, integration, and analytics to support case review and treatment planning. METHODS: In this review, we evaluate and summarize the landscape of available tools, resources, and evidence used in the evaluation of somatic and germline tumor variants within the context of molecular tumor boards. RESULTS: Molecular tumor boards (MTBs) are collaborative efforts of multidisciplinary cancer experts equipped with genomic interpretation resources to aid in the delivery of accurate and timely clinical interpretations of complex genomic results for each patient, within an institution or hospital network. Virtual MTBs (VMTBs) provide an online forum for collaborative governance, provenance, and information sharing between experts outside a given hospital network with the potential to enhance MTB discussions. Knowledge sharing in VMTBs and communication with guideline-developing organizations can lead to progress evidenced by data harmonization across resources, crowd-sourced and expert-curated genomic assertions, and a more informed and explainable usage of artificial intelligence. CONCLUSION: Advances in cancer genomics interpretation aid in better patient and disease classification, more streamlined identification of relevant literature, and a more thorough review of available treatments and predicted patient outcomes

    Practical, ethical and regulatory considerations for the evolving medical and research genomics landscape.

    Get PDF
    We are entering a fascinating and uncertain period of medical history, as today’s DNA sequencing technology has the potential to help each of us direct our care and predict our future based on knowledge of our own individual inherited and acquired genetics. However, from a global and local economic perspective, these are lean years, and this adds a significant degree of uncertainty to the immediate future of this enterprise. It is therefore incumbent upon us as a community to show that personalized genomic medicine will not just be a luxury or a burdensome cost center, but that it truly has the potential to save both lives and health care expenses via data-driven management, early disease detection/screening and more efficacious pharmaceutical delivery. To do this, we need to determine how to move forward towards expanded clinical use of this technology in a manner both rapid and economical, while ensuring the integrity of the process and the safety and well-being of patients and research participants. Here, we discuss some of the ethical, regulatory and practical considerations that are emerging in the field of genomic medicine. We also propose that many of the cost and safety issues we are facing can be mitigated through expanded reliance on existing clinical regulatory frameworks and the implementation of worksharing strategies designed to leverage the strengths of our genomics centers and clinical interpretive teams

    Research Directions in the Clinical Implementation of Pharmacogenomics: An Overview of US Programs and Projects

    Get PDF
    Response to a drug often differs widely among individual patients. This variability is frequently observed not only with respect to effective responses but also with adverse drug reactions. Matching patients to the drugs that are most likely to be effective and least likely to cause harm is the goal of effective therapeutics. Pharmacogenomics (PGx) holds the promise of precision medicine through elucidating the genetic determinants responsible for pharmacological outcomes and using them to guide drug selection and dosing. Here we survey the US landscape of research programs in PGx implementation, review current advances and clinical applications of PGx, summarize the obstacles that have hindered PGx implementation, and identify the critical knowledge gaps and possible studies needed to help to address them

    Big Data Transforms Discovery-Utilization Therapeutics Continuum.

    Get PDF
    Enabling omic technologies adopt a holistic view to produce unprecedented insights into the molecular underpinnings of health and disease, in part, by generating massive high-dimensional biological data. Leveraging these systems-level insights as an engine driving the healthcare evolution is maximized through integration with medical, demographic, and environmental datasets from individuals to populations. Big data analytics has accordingly emerged to add value to the technical aspects of storage, transfer, and analysis required for merging vast arrays of omic-, clinical-, and eco-datasets. In turn, this new field at the interface of biology, medicine, and information science is systematically transforming modern therapeutics across discovery, development, regulation, and utilization

    Doctor of Philosophy

    Get PDF
    dissertationGenetic testing is becoming increasingly important to medical practice since the completion of the Human Genome project. To realize the full promise of personalized medicine, we need to first integrate genetic and genomic information into Electronic Health Records (EHRs) as coded and structured data using standards. However, EHRs are not ready for genomic medicine; lack of standardized information models and termi-nologies for genetic and genomic data representation is recognized as one of the major barriers. In this study, we have focused on constitutional cytogenetic tests. We first evaluat-ed the Logical Observation Identifiers Names and Codes (LOINC), the de facto vocabu-lary standard for representing laboratory test names and results, and identified that a gap exists in LOINC to support the integration of cytogenetic test results into EHRs. We ana-lyzed sample clinical reports from several large cytogenetics laboratories, and developed LOINC panels and codes for representing constitutional cytogenetic test findings through the LOINC panel approach. The LOINC committee approved the cytogenetic LOINC panels and officially released them as part of the LOINC database in December 2010. We then followed the well vetted standard development process of Health Level Seven (HL7), developed and balloted a HL7 version 2 implementation guide that details how these LOINC panels are coupled with the messaging standard to transfer cytogenetic test iv results over the wire. We also described the advantages of coupling the LOINC panel content to HL7 version 2 messages, and why we think this approach could be a practical and efficient way for implementers to develop interfaces that utilize standard information models bound to standard terminologies. We have filled the gap that there were no standard information models and no standard terminologies for representing constitutional cytogenetic test results, and have developed the foundation to allow incremental enhancement in the future
    corecore