2,497 research outputs found

    A bioinformatics approach for precision medicine off-label drug drug selection among triple negative breast cancer patients

    Get PDF
    Cancer has been extensively characterized on the basis of genomics. The integration of genetic information about cancers with data on how the cancers respond to target based therapy to help to optimum cancer treatment. OBJECTIVE: The increasing usage of sequencing technology in cancer research and clinical practice has enormously advanced our understanding of cancer mechanisms. The cancer precision medicine is becoming a reality. Although off-label drug usage is a common practice in treating cancer, it suffers from the lack of knowledge base for proper cancer drug selections. This eminent need has become even more apparent considering the upcoming genomics data. METHODS: In this paper, a personalized medicine knowledge base is constructed by integrating various cancer drugs, drug-target database, and knowledge sources for the proper cancer drugs and their target selections. Based on the knowledge base, a bioinformatics approach for cancer drugs selection in precision medicine is developed. It integrates personal molecular profile data, including copy number variation, mutation, and gene expression. RESULTS: By analyzing the 85 triple negative breast cancer (TNBC) patient data in the Cancer Genome Altar, we have shown that 71.7% of the TNBC patients have FDA approved drug targets, and 51.7% of the patients have more than one drug target. Sixty-five drug targets are identified as TNBC treatment targets and 85 candidate drugs are recommended. Many existing TNBC candidate targets, such as Poly (ADP-Ribose) Polymerase 1 (PARP1), Cell division protein kinase 6 (CDK6), epidermal growth factor receptor, etc., were identified. On the other hand, we found some additional targets that are not yet fully investigated in the TNBC, such as Gamma-Glutamyl Hydrolase (GGH), Thymidylate Synthetase (TYMS), Protein Tyrosine Kinase 6 (PTK6), Topoisomerase (DNA) I, Mitochondrial (TOP1MT), Smoothened, Frizzled Class Receptor (SMO), etc. Our additional analysis of target and drug selection strategy is also fully supported by the drug screening data on TNBC cell lines in the Cancer Cell Line Encyclopedia. CONCLUSIONS: The proposed bioinformatics approach lays a foundation for cancer precision medicine. It supplies much needed knowledge base for the off-label cancer drug usage in clinics

    ClinOmicsTrailbc: a visual analytics tool for breast cancer treatment stratification

    Get PDF
    Motivation: Breast cancer is the second leading cause of cancer death among women. Tumors, even of the same histopathological subtype, exhibit a high genotypic diversity that impedes therapy stratification and that hence must be accounted for in the treatment decision-making process. Results: Here, we present ClinOmicsTrailbc, a comprehensive visual analytics tool for breast cancer decision support that provides a holistic assessment of standard-of-care targeted drugs, candidates for drug repositioning and immunotherapeutic approaches. To this end, our tool analyzes and visualizes clinical markers and (epi-)genomics and transcriptomics datasets to identify and evaluate the tumor’s main driver mutations, the tumor mutational burden, activity patterns of core cancerrelevant pathways, drug-specific biomarkers, the status of molecular drug targets and pharmacogenomic influences. In order to demonstrate ClinOmicsTrailbc’s rich functionality, we present three case studies highlighting various ways in which ClinOmicsTrailbc can support breast cancer precision medicine. ClinOmicsTrailbc is a powerful integrated visual analytics tool for breast cancer research in general and for therapy stratification in particular, assisting oncologists to find the best possible treatment options for their breast cancer patients based on actionable, evidence-based results. Availability and implementation: ClinOmicsTrailbc can be freely accessed at https://clinomicstrail. bioinf.uni-sb.de

    Precision Medicine in Solid Tumors

    Get PDF
    In the era of precision medicine, the use of molecularly targeted therapies in selected patients has led to a paradigm change in cancer treatment. Multiple studies have demonstrated the benefits of therapies that are chosen based on the molecular profile of the tumor and also from the liquid biopsy. With genomics' increasing ability, a routine transcriptomics analysis of advanced/metastatic cancers is now feasible in most cancer hospitals, including community cancer centers. This is an unprecedented shift in the management of cancers irrespective of their organ types, which not only improved the outcome but also opened several new avenues in research and practice, such as immune-check-point inhibitors, tumor-TME co-evolution in the development of resistance, longitudinal liquid biopsies, biomarkers screening and the management of electronic medical records.This book brings together these crucial areas of investigation. The research presented here attempts to address the current issues to provoke thoughts for the future. The future of precision medicine will have to embrace a shift from in vitro, in vivo/PDX models for the mechanistic study to a more functional test based on the scientific interrogation of genomic data, in the form of functional precision medicine. We will also have to combat the element of noise in the multitudes of data and impart the regulatory structure to make judicious use of the data. The expectations for functional precision medicine are high. We aspire to witness a tremendous improvement in patient outcomes, from better to best, down the road that will match the clinical guidelines

    Explainable artificial intelligence for patient stratification and drug repositioning

    Get PDF
    Enabling precision medicine requires developing robust patient stratification methods as well as drugs tailored to homogeneous subgroups of patients from a heterogeneous population. Developing de novo drugs is expensive and time consuming with an ultimately low FDA approval rate. These limitations make developing new drugs for a small portion of a disease population unfeasible. Therefore, drug repositioning is an essential alternative for developing new drugs for a disease subpopulation. There is a crucial need to develop data-driven approaches that find druggable homogeneous subgroups within the disease population and reposition the drugs for these subgroups. In this study, we developed an explainable AI approach for patient stratification and drug repositioning. Exploratory mining mimicking the trial recruitment process as well as network analysis were used to discover homogeneous subgroups within a disease population. For each subgroup, a biomedical network analysis was done to find the drugs that are most relevant to a given subgroup of patients. The set of candidate drugs for each subgroup was ranked using an aggregated drug score assigned to each drug. The method represents a human-in-the-loop framework, where medical experts use data-driven results to generate hypotheses and obtain insights into potential therapeutic candidates for patients who belong to a subgroup. To examine the validity of our method, we implemented our method on individual cancer types and on pan-cancer data to consider the inter- and intra-heterogeneity within a cancer type and among cancer types. Patients' phenotypic and genotypic data was utilized with a heterogeneous knowledge base because it gives a multi-view perspective for finding new indications for drugs outside of their original use. Our analysis of the top candidate drugs for the subgroups showed that most of these drugs are FDA-approved drugs for cancer, and others are non-cancer related, but have the potential to be repurposed for cancer. We have discovered novel cancer-related mechanisms that these drugs can target in different cancer types to reduce cancer treatment costs and improve patient survival. Further wet lab experiments to validate these findings are required prior to initiating clinical trials using these repurposed therapies.Includes bibliographical references

    Drug Repurposing

    Get PDF
    This book focuses on various aspects and applications of drug repurposing, the understanding of which is important for treating diseases. Due to the high costs and time associated with the new drug discovery process, the inclination toward drug repurposing is increasing for common as well as rare diseases. A major focus of this book is understanding the role of drug repurposing to develop drugs for infectious diseases, including antivirals, antibacterial and anticancer drugs, as well as immunotherapeutics

    Specialized Named Entity Recognition For Breast Cancer Subtyping

    Get PDF
    The amount of data and analysis being published and archived in the biomedical research community is more than can feasibly be sifted through manually, which limits the information an individual or small group can synthesize and integrate into their own research. This presents an opportunity for using automated methods, including Natural Language Processing (NLP), to extract important information from text on various topics. Named Entity Recognition (NER), is one way to automate knowledge extraction of raw text. NER is defined as the task of identifying named entities from text using labels such as people, dates, locations, diseases, and proteins. There are several NLP tools that are designed for entity recognition, but rely on large established corpus for training data. Biomedical research has the potential to guide diagnostic and therapeutic decisions, yet the overwhelming density of publications acts as a barrier to getting these results into a clinical setting. An exceptional example of this is the field of breast cancer biology where over 2 million people are diagnosed worldwide every year and billions of dollars are spent on research. Breast cancer biology literature and research relies on a highly specific domain with unique language and vocabulary, and therefore requires specialized NLP tools which can generate biologically meaningful results. This thesis presents a novel annotation tool, that is optimized for quickly creating training data for spaCy pipelines as well as exploring the viability of said data for analyzing papers with automated processing. Custom pipelines trained on these annotations are shown to be able to recognize custom entities at levels comparable to large corpus based recognition

    COMPUTATIONAL PHENOTYPING AND DRUG REPURPOSING FROM ELECTRONIC MEDICAL RECORDS

    Get PDF
    Using electronic medical records (EMR) for research involves selecting cohorts and manipulating data for tasks like predictive analysis. Computational phenotyping for cohort characterization and stratification is becoming increasingly important for researchers to produce clinically relevant findings. There are significant amounts of time and effort devoted to manual chart abstraction by subject matter experts and researchers, which creates a large bottleneck for progress in clinical research. I focus on developing computational phenotyping pipelines, and I also focus on using EMR for drug repurposing in breast cancer. Drug repurposing is defined as the process of applying known drugs that are already on the market to new disease indications. Using EMR data for drug repurposing has the unique advantage of being able to observe a patient cohort over time and see drug effects on outcomes. In this dissertation, I present work on computational phenotyping and EMR-based drug repurposing. First, I use embedding models and foundational natural language processing methods to predict oral cancer risk with pathology notes. Second, I use natural language processing methods and transfer learning for breast cancer cohort selection and information extraction. Third, I present a pipeline for producing drug repurposing candidates from EMR and provide supporting evidence for predictions with biomedical literature and existing clinical trials.Doctor of Philosoph

    Computational methods for personalized cancer genomics

    Get PDF
    In recent years, cancer treatment strategies have moved towards personalized approaches, specifically tailoring cancer treatments on a single-patient basis using molecular profiles from the patients’ tumor genomes. Knowledge of a patient’s molecular profile can be used to 1) identify the disease mechanisms and underlying cause of a single patient’s cancer, 2) assign patients into treatment groups based on the molecular prognosis, and 3) recommend potential treatments for individual patients based on the patient’s molecular signature data. However, the bottleneck of the personalized medicine approach lies in the challenge of translating the vast amount of sequencing data to meaningful clinical insights. This dissertation explores several computational methods that utilize molecular signature data to understand disease mechanisms of cancer, categorize patients into biologically relevant subtypes, and recommend drug treatments to patients. In the dissertation, we present a method, DawnRank, a patient-specific method that determines the potential driving genomic alterations (the drivers) of cancer. We expand on DawnRank’s capabilities by using the DawnRank scores in key driver mutations and copy number variants (CNVs) to identify breast cancer subtypes. We found 5 alternative subtypes based on potentially clinically relevant driver genes, each with unique defining target features and pathways. These subtypes correspond to and build upon our previous knowledge of breast cancer subtypes. We also identify disease mechanisms in identifying key novel cancer pathways in which driver genes interact. We developed a method, C3, which pinpoints patterns of cancer mutations in a pathway context from a patient population to detect novel cancer pathways that consist of significant driver genes. C3 improves on current methods in driver pathway detection both on a technical aspect and a results-oriented aspect. C3 can detect larger and more consistent pathways than previous methods as well as discovering more biologically relevant drivers. Finally, we address the issue of drug recommendation in the wake of molecular signature data. We develop a method, Scattershot, which combines genomic information along with biological insights on cancer disease mechanisms to predict drug response and prioritize drug treatments. Scattershot outperforms previous methods in predicting drug response and produces recommendations that largely comply with known medical treatment protocols.Scattershot recommends drugs to cancer patients that are in line with the actual drugs prescribed by the physician
    • …
    corecore