778 research outputs found

    INSIdE NANO : a systems biology framework to contextualize the mechanism-of-action of engineered nanomaterials

    Get PDF
    Engineered nanomaterials (ENMs) are widely present in our daily lives. Despite the efforts to characterize their mechanism of action in multiple species, their possible implications in human pathologies are still not fully understood. Here we performed an integrated analysis of the effects of ENMs on human health by contextualizing their transcriptional mechanism-of-action with respect to drugs, chemicals and diseases. We built a network of interactions of over 3,000 biological entities and developed a novel computational tool, INSIdE NANO, to infer new knowledge about ENM behavior. We highlight striking association of metal and metal-oxide nanoparticles and major neurodegenerative disorders. Our novel strategy opens possibilities to achieve fast and accurate read-across evaluation of ENMs and other chemicals based on their biosignatures.Peer reviewe

    The landscape of the methodology in drug repurposing using human genomic data:a systematic review

    Get PDF
    The process of drug development is expensive and time-consuming. In contrast, drug repurposing can be introduced to clinical practice more quickly and at a reduced cost. Over the last decade, there has been a significant expansion of large biobanks that link genomic data to electronic health record (EHR) data, public availability of various databases containing biological and clinical information, and rapid development of novel methodologies and algorithms in integrating different sources of data. This review aims to provide a thorough summary of different strategies that utilize genomic data to seek drug-repositioning opportunities. We searched MEDLINE and EMBASE databases to identify eligible studies up until 1st May 2023, with a total of 102 studies finally included after two-step parallel screening. We summarized commonly used strategies for drug repurposing, including Mendelian randomization, multi-omic-based and network-based studies, and illustrated each strategy with examples, as well as the data sources implemented. By leveraging existing knowledge and infrastructure to expedite the drug discovery process and reduce costs, drug repurposing potentially identifies new therapeutic uses for approved drugs in a more efficient and targeted manner. However, technical challenges when integrating different types of data and biased or incomplete understanding of drug interactions are important hindrances that cannot be disregarded in the pursuit of identifying novel therapeutic applications. This review offers an overview of drug repurposing methodologies, providing valuable insights and guiding future directions for advancing drug repurposing studies

    Systems biology approaches to a rational drug discovery paradigm

    Full text link
    The published manuscript is available at EurekaSelect via http://www.eurekaselect.com/openurl/content.php?genre=article&doi=10.2174/1568026615666150826114524.Prathipati P., Mizuguchi K.. Systems biology approaches to a rational drug discovery paradigm. Current Topics in Medicinal Chemistry, 16, 9, 1009. https://doi.org/10.2174/1568026615666150826114524

    Identification of drug candidates and repurposing opportunities through compound-target interaction networks

    Get PDF
    Introduction: System-wide identification of both on- and off-targets of chemical probes provides improved understanding of their therapeutic potential and possible adverse effects, thereby accelerating and de-risking drug discovery process. Given the high costs of experimental profiling of the complete target space of drug-like compounds, computational models offer systematic means for guiding these mapping efforts. These models suggest the most potent interactions for further experimental or pre-clinical evaluation both in cell line models and in patient-derived material.Areas covered: The authors focus here on network-based machine learning models and their use in the prediction of novel compound-target interactions both in target-based and phenotype-based drug discovery applications. While currently being used mainly in complementing the experimentally mapped compound-target networks for drug repurposing applications, such as extending the target space of already approved drugs, these network pharmacology approaches may also suggest completely unexpected and novel investigational probes for drug development.Expert opinion: Although the studies reviewed here have already demonstrated that network-centric modeling approaches have the potential to identify candidate compounds and selective targets in disease networks, many challenges still remain. In particular, these challenges include how to incorporate the cellular context and genetic background into the disease networks to enable more stratified and selective target predictions, as well as how to make the prediction models more realistic for the practical drug discovery and therapeutic applications.Peer reviewe

    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

    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

    Mouse model phenotypes provide information about human drug targets

    Get PDF
    Motivation: Methods for computational drug target identification use information from diverse information sources to predict or prioritize drug targets for known drugs. One set of resources that has been relatively neglected for drug repurposing is animal model phenotype. Results: We investigate the use of mouse model phenotypes for drug target identification. To achieve this goal, we first integrate mouse model phenotypes and drug effects, and then systematically compare the phenotypic similarity between mouse models and drug effect profiles. We find a high similarity between phenotypes resulting from loss-of-function mutations and drug effects resulting from the inhibition of a protein through a drug action, and demonstrate how this approach can be used to suggest candidate drug targets. Availability and implementation: Analysis code and supplementary data files are available on the project Web site at https://drugeffects.googlecode.com. Contact: [email protected] or [email protected] Supplementary information: Supplementary data are available at Bioinformatics online

    Systems approaches to drug repositioning

    Get PDF
    PhD ThesisDrug discovery has overall become less fruitful and more costly, despite vastly increased biomedical knowledge and evolving approaches to Research and Development (R&D). One complementary approach to drug discovery is that of drug repositioning which focusses on identifying novel uses for existing drugs. By focussing on existing drugs that have already reached the market, drug repositioning has the potential to both reduce the timeframe and cost of getting a disease treatment to those that need it. Many marketed examples of repositioned drugs have been found via serendipitous or rational observations, highlighting the need for more systematic methodologies. Systems approaches have the potential to enable the development of novel methods to understand the action of therapeutic compounds, but require an integrative approach to biological data. Integrated networks can facilitate systems-level analyses by combining multiple sources of evidence to provide a rich description of drugs, their targets and their interactions. Classically, such networks can be mined manually where a skilled person can identify portions of the graph that are indicative of relationships between drugs and highlight possible repositioning opportunities. However, this approach is not scalable. Automated procedures are required to mine integrated networks systematically for these subgraphs and bring them to the attention of the user. The aim of this project was the development of novel computational methods to identify new therapeutic uses for existing drugs (with particular focus on active small molecules) using data integration. A framework for integrating disparate data relevant to drug repositioning, Drug Repositioning Network Integration Framework (DReNInF) was developed as part of this work. This framework includes a high-level ontology, Drug Repositioning Network Integration Ontology (DReNInO), to aid integration and subsequent mining; a suite of parsers; and a generic semantic graph integration platform. This framework enables the production of integrated networks maintaining strict semantics that are important in, but not exclusive to, drug repositioning. The DReNInF is then used to create Drug Repositioning Network Integration (DReNIn), a semantically-rich Resource Description Framework (RDF) dataset. A Web-based front end was developed, which includes a SPARQL Protocol and RDF Query Language (SPARQL) endpoint for querying this dataset. To automate the mining of drug repositioning datasets, a formal framework for the definition of semantic subgraphs was established and a method for Drug Repositioning Semantic Mining (DReSMin) was developed. DReSMin is an algorithm for mining semantically-rich networks for occurrences of a given semantic subgraph. This algorithm allows instances of complex semantic subgraphs that contain data about putative drug repositioning opportunities to be identified in a computationally tractable fashion, scaling close to linearly with network data. The ability of DReSMin to identify novel Drug-Target (D-T) associations was investigated. 9,643,061 putative D-T interactions were identified and ranked, with a strong correlation between highly scored associations and those supported by literature observed. The 20 top ranked associations were analysed in more detail with 14 found to be novel and six found to be supported by the literature. It was also shown that this approach better prioritises known D-T interactions, than other state-of-the-art methodologies. The ability of DReSMin to identify novel Drug-Disease (Dr-D) indications was also investigated. As target-based approaches are utilised heavily in the field of drug discovery, it is necessary to have a systematic method to rank Gene-Disease (G-D) associations. Although methods already exist to collect, integrate and score these associations, these scores are often not a reliable re flection of expert knowledge. Therefore, an integrated data-driven approach to drug repositioning was developed using a Bayesian statistics approach and applied to rank 309,885 G-D associations using existing knowledge. Ranked associations were then integrated with other biological data to produce a semantically-rich drug discovery network. Using this network it was shown that diseases of the central nervous system (CNS) provide an area of interest. The network was then systematically mined for semantic subgraphs that capture novel Dr-D relations. 275,934 Dr-D associations were identified and ranked, with those more likely to be side-effects filtered. Work presented here includes novel tools and algorithms to enable research within the field of drug repositioning. DReNIn, for example, includes data that previous comparable datasets relevant to drug repositioning have neglected, such as clinical trial data and drug indications. Furthermore, the dataset may be easily extended using DReNInF to include future data as and when it becomes available, such as G-D association directionality (i.e. is the mutation a loss-of-function or gain-of-function). Unlike other algorithms and approaches developed for drug repositioning, DReSMin can be used to infer any types of associations captured in the target semantic network. Moreover, the approaches presented here should be more generically applicable to other fields that require algorithms for the integration and mining of semantically rich networks.European and Physical Sciences Research Council (EPSRC) and GS
    • …
    corecore