74 research outputs found

    Interactive visual analysis of drug-target interaction networks using Drug Target Profiler, with applications to precision medicine and drug repurposing

    Get PDF
    Knowledge of the full target space of drugs (or drug-like compounds) provides important insights into the potential therapeutic use of the agents to modulate or avoid their various on- and off-targets in drug discovery and precision medicine. However, there is a lack of consolidated databases and associated data exploration tools that allow for systematic profiling of drug target-binding potencies of both approved and investigational agents using a network-centric approach. We recently initiated a community-driven platform, Drug Target Commons (DTC), which is an open-data crowdsourcing platform designed to improve the management, reproducibility and extended use of compound-target bioactivity data for drug discovery and repurposing, as well as target identification applications. In this work, we demonstrate an integrated use of the rich bioactivity data from DTC and related drug databases using Drug Target Profiler (DTP), an open-source software and web tool for interactive exploration of drug-target interaction networks. DTP was designed for network-centric modeling of mode-of-action of multi-targeting anticancer compounds, especially for precision oncology applications. DTP enables users to construct an interaction network based on integrated bioactivity data across selected chemical compounds and their protein targets, further customizable using various visualization and filtering options, as well as cross-links to several drug and protein databases to provide comprehensive information of the network nodes and interactions. We demonstrate here the operation of the DTP tool and its unique features by several use cases related to both drug discovery and drug repurposing applications, using examples of anticancer drugs with shared target profiles. DTP is freely accessible at http://drugtargetprofiler.fimm.fi/

    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

    Predicting COVID-19—Comorbidity Pathway Crosstalk-Based Targets and Drugs: Towards Personalized COVID-19 Management

    Get PDF
    It is well established that pre-existing comorbid conditions such as hypertension, diabetes, obesity, cardiovascular diseases (CVDs), chronic kidney diseases (CKDs), cancers, and chronic obstructive pulmonary disease (COPD) are associated with increased severity and fatality of COVID-19. The increased death from COVID-19 is due to the unavailability of a gold standard therapeutic and, more importantly, the lack of understanding of how the comorbid conditions and COVID-19 interact at the molecular level, so that personalized management strategies can be adopted. Here, using multi-omics data sets and bioinformatics strategy, we identified the pathway crosstalk between COVID-19 and diabetes, hypertension, CVDs, CKDs, and cancers. Further, shared pathways and hub gene-based targets for COVID-19 and its associated specific and combination of comorbid conditions are also predicted towards developing personalized management strategies. The approved drugs for most of these identified targets are also provided towards drug repurposing. Literature supports the involvement of our identified shared pathways in pathogenesis of COVID-19 and development of the specific comorbid condition of interest. Similarly, shared pathways- and hub gene-based targets are also found to have potential implementations in managing COVID-19 patients. However, the identified targets and drugs need further careful evaluation for their repurposing towards personalized treatment of COVID-19 cases having pre-existing specific comorbid conditions we have considered in this analysis. The method applied here may also be helpful in identifying common pathway components and targets in other disease-disease interactions too

    Drug repurposing prediction for COVID-19 using probabilistic networks and crowdsourced curation

    Get PDF
    Severe acute respiratory syndrome coronavirus two (SARS-CoV-2), the virus responsible for the coronavirus disease 2019 (COVID-19) pandemic, represents an unprecedented global health challenge. Consequently, a large amount of research into the disease pathogenesis and potential treatments has been carried out in a short time frame. However, developing novel drugs is a costly and lengthy process, and is unlikely to deliver a timely treatment for the pandemic. Drug repurposing, by contrast, provides an attractive alternative, as existing drugs have already undergone many of the regulatory requirements. In this work we used a combination of network algorithms and human curation to search integrated knowledge graphs, identifying drug repurposing opportunities for COVID-19. We demonstrate the value of this approach, reporting on eight potential repurposing opportunities identified, and discuss how this approach could be incorporated into future studies

    In Silico Strategies for Prospective Drug Repositionings

    Get PDF
    The discovery of new drugs is one of pharmaceutical research's most exciting and challenging tasks. Unfortunately, the conventional drug discovery procedure is chronophagous and seldom successful; furthermore, new drugs are needed to address our clinical challenges (e.g., new antibiotics, new anticancer drugs, new antivirals).Within this framework, drug repositioning—finding new pharmacodynamic properties for already approved drugs—becomes a worthy drug discovery strategy.Recent drug discovery techniques combine traditional tools with in silico strategies to identify previously unaccounted properties for drugs already in use. Indeed, big data exploration techniques capitalize on the ever-growing knowledge of drugs' structural and physicochemical properties, drug–target and drug–drug interactions, advances in human biochemistry, and the latest molecular and cellular biology discoveries.Following this new and exciting trend, this book is a collection of papers introducing innovative computational methods to identify potential candidates for drug repositioning. Thus, the papers in the Special Issue In Silico Strategies for Prospective Drug Repositionings introduce a wide array of in silico strategies such as complex network analysis, big data, machine learning, molecular docking, molecular dynamics simulation, and QSAR; these strategies target diverse diseases and medical conditions: COVID-19 and post-COVID-19 pulmonary fibrosis, non-small lung cancer, multiple sclerosis, toxoplasmosis, psychiatric disorders, or skin conditions

    Shu-Xie decoction alleviates oxidative stress and colon injury in acute sleep-deprived mice by suppressing p62/KEAP1/NRF2/HO1/NQO1 signaling

    Get PDF
    Introduction: Sleep disorders are common clinical psychosomatic disorders that can co-exist with a variety of conditions. In humans and animal models, sleep deprivation (SD) is closely related with gastrointestinal diseases. Shu-Xie Decoction (SX) is a traditional Chinese medicine (TCM) with anti-nociceptive, anti-inflammatory, and antidepressant properties. SX is effective in the clinic for treating patients with abnormal sleep and/or gastrointestinal disorders, but the underlying mechanisms are not known. This study investigated the mechanisms by which SX alleviates SD-induced colon injury in vivo.Methods: C57BL/6 mice were placed on an automated sleep deprivation system for 72 h to generate an acute sleep deprivation (ASD) model, and low-dose SX (SXL), high-dose SX (SXH), or S-zopiclone (S-z) as a positive control using the oral gavage were given during the whole ASD-induced period for one time each day. The colon length was measured and the colon morphology was visualized using hematoxylin and eosin (H&E) staining. ROS and the redox biomarkers include reduced glutathione (GSH), malondialdehyde (MDA), and superoxide dismutase (SOD) were detected. Quantitative real-time PCR (qRT-PCR), molecular docking, immunofluorescence and western blotting assays were performed to detect the antioxidant signaling pathways.Results: ASD significantly increased FBG levels, decreased colon length, moderately increased the infiltration of inflammatory cells in the colon mucosa, altered the colon mucosal structure, increased the levels of ROS, GSH, MDA, and SOD activity compared with the controls. These adverse effects were significantly alleviated by SX treatment. ASD induced nuclear translocation of NRF2 in the colon mucosal cells and increased the expression levels of p62, NQO1, and HO1 transcripts and proteins, but these effects were reversed by SX treatment.Conclusion: SX decoction ameliorated ASD-induced oxidative stress and colon injury by suppressing the p62/KEAP1/NRF2/HO1/NQO1 signaling pathway. In conclusion, combined clinical experience, SX may be a promising drug for sleep disorder combined with colitis

    Structural Cheminformatics for Kinase-Centric Drug Design

    Get PDF
    Drug development is a long, expensive, and iterative process with a high failure rate, while patients wait impatiently for treatment. Kinases are one of the main drug targets studied for the last decades to combat cancer, the second leading cause of death worldwide. These efforts resulted in a plethora of structural, chemical, and pharmacological kinase data, which are collected in the KLIFS database. In this thesis, we apply ideas from structural cheminformatics to the rich KLIFS dataset, aiming to provide computational tools that speed up the complex drug discovery process. We focus on methods for target prediction and fragment-based drug design that study characteristics of kinase binding sites (also called pockets). First, we introduce the concept of computational target prediction, which is vital in the early stages of drug discovery. This approach identifies biological entities such as proteins that may (i) modulate a disease of interest (targets or on-targets) or (ii) cause unwanted side effects due to their similarity to on-targets (off-targets). We focus on the research field of binding site comparison, which lacked a freely available and efficient tool to determine similarities between the highly conserved kinase pockets. We fill this gap with the novel method KiSSim, which encodes and compares spatial and physicochemical pocket properties for all kinases (kinome) that are structurally resolved. We study kinase similarities in the form of kinome-wide phylogenetic trees and detect expected and unexpected off-targets. To allow multiple perspectives on kinase similarity, we propose an automated and production-ready pipeline; user-defined kinases can be inspected complementarily based on their pocket sequence and structure (KiSSim), pocket-ligand interactions, and ligand profiles. Second, we introduce the concept of fragment-based drug design, which is useful to identify and optimize active and promising molecules (hits and leads). This approach identifies low-molecular-weight molecules (fragments) that bind weakly to a target and are then grown into larger high-affinity drug-like molecules. With the novel method KinFragLib, we provide a fragment dataset for kinases (fragment library) by viewing kinase inhibitors as combinations of fragments. Kinases have a highly conserved pocket with well-defined regions (subpockets); based on the subpockets that they occupy, we fragment kinase inhibitors in experimentally resolved protein-ligand complexes. The resulting dataset is used to generate novel kinase-focused molecules that are recombinations of the previously fragmented kinase inhibitors while considering their subpockets. The KinFragLib and KiSSim methods are published as freely available Python tools. Third, we advocate for open and reproducible research that applies FAIR principles ---data and software shall be findable, accessible, interoperable, and reusable--- and software best practices. In this context, we present the TeachOpenCADD platform that contains pipelines for computer-aided drug design. We use open source software and data to demonstrate ligand-based applications from cheminformatics and structure-based applications from structural bioinformatics. To emphasize the importance of FAIR data, we dedicate several topics to accessing life science databases such as ChEMBL, PubChem, PDB, and KLIFS. These pipelines are not only useful to novices in the field to gain domain-specific skills but can also serve as a starting point to study research questions. Furthermore, we show an example of how to build a stand-alone tool that formalizes reoccurring project-overarching tasks: OpenCADD-KLIFS offers a clean and user-friendly Python API to interact with the KLIFS database and fetch different kinase data types. This tool has been used in this thesis and beyond to support kinase-focused projects. We believe that the FAIR-based methods, tools, and pipelines presented in this thesis (i) are valuable additions to the toolbox for kinase research, (ii) provide relevant material for scientists who seek to learn, teach, or answer questions in the realm of computer-aided drug design, and (iii) contribute to making drug discovery more efficient, reproducible, and reusable

    Integrative omics approaches for new target identification and therapeutics development

    Get PDF
    The growing research and commercial pressures for novel therapeutics development accentuate why better strategies are needed for drug discovery. The costly nature of developing a pharmaceutical compound as well as the shrinking pool of ‘easy’ targets are some of the key reasons why there is a research paradigm shift towards integrative and systems biology driven approaches. Moreover, multifactorial aspects of many diseases require more innovative clinical strategies rather than just focusing on a single target. Cardiovascular diseases as well as associated immune components exemplify this complexity well. This thesis aimed to introduce a gradual and highly integrative analytical framework by incorporating a full range of studies from disease target selection to high-throughput virtual screening so that a cost-effective and efficient stratification of targets and associated compounds could be achieved. Heart failure served as a case study for complex diseases where the first in-depth omics study on cardiomyopathies helped to elucidate new therapeutic avenues. This research tied in with a development of a novel scoring function and integrated machine learning approach for multiple therapeutic target classification and exploration. Finally, all pieces of the introduced research were used to create a highly integrative in silico screening workflow. Some of the key results included the first reported molecular dynamics analyses for a complex immunotherapeutic target, c-Rel, as well as 15 new therapeutic compounds that could potentially modulate this transcription factor subunit. Thus, this dissertation provided several important improvements for target identification, validation, and drug discovery that could significantly advance current development strategies and accelerate new therapeutics production
    • 

    corecore