7,042 research outputs found

    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

    AI in drug discovery and its clinical relevance

    Get PDF
    The COVID-19 pandemic has emphasized the need for novel drug discovery process. However, the journey from conceptualizing a drug to its eventual implementation in clinical settings is a long, complex, and expensive process, with many potential points of failure. Over the past decade, a vast growth in medical information has coincided with advances in computational hardware (cloud computing, GPUs, and TPUs) and the rise of deep learning. Medical data generated from large molecular screening profiles, personal health or pathology records, and public health organizations could benefit from analysis by Artificial Intelligence (AI) approaches to speed up and prevent failures in the drug discovery pipeline. We present applications of AI at various stages of drug discovery pipelines, including the inherently computational approaches of de novo design and prediction of a drug's likely properties. Open-source databases and AI-based software tools that facilitate drug design are discussed along with their associated problems of molecule representation, data collection, complexity, labeling, and disparities among labels. How contemporary AI methods, such as graph neural networks, reinforcement learning, and generated models, along with structure-based methods, (i.e., molecular dynamics simulations and molecular docking) can contribute to drug discovery applications and analysis of drug responses is also explored. Finally, recent developments and investments in AI-based start-up companies for biotechnology, drug design and their current progress, hopes and promotions are discussed in this article.  Other InformationPublished in:HeliyonLicense: https://creativecommons.org/licenses/by/4.0/See article on publisher's website: https://doi.org/10.1016/j.heliyon.2023.e17575 </p

    Artificial intelligence in cancer target identification and drug discovery

    Get PDF
    Artificial intelligence is an advanced method to identify novel anticancer targets and discover novel drugs from biology networks because the networks can effectively preserve and quantify the interaction between components of cell systems underlying human diseases such as cancer. Here, we review and discuss how to employ artificial intelligence approaches to identify novel anticancer targets and discover drugs. First, we describe the scope of artificial intelligence biology analysis for novel anticancer target investigations. Second, we review and discuss the basic principles and theory of commonly used network-based and machine learning-based artificial intelligence algorithms. Finally, we showcase the applications of artificial intelligence approaches in cancer target identification and drug discovery. Taken together, the artificial intelligence models have provided us with a quantitative framework to study the relationship between network characteristics and cancer, thereby leading to the identification of potential anticancer targets and the discovery of novel drug candidates

    Predicting potential drugs and drug-drug interactions for drug repositioning

    Get PDF
    The purpose of drug repositioning is to predict novel treatments for existing drugs. It saves time and reduces cost in drug discovery, especially in preclinical procedures. In drug repositioning, the challenging objective is to identify reasonable drugs with strong evidence. Recently, benefiting from various types of data and computational strategies, many methods have been proposed to predict potential drugs. Signature-based methods use signatures to describe a specific disease condition and match it with drug-induced transcriptomic profiles. For a disease signature, a list of potential drugs is produced based on matching scores. In many studies, the top drugs on the list are identified as potential drugs and verified in various ways. However, there are a few limitations in existing methods: (1) For many diseases, especially cancers, the tissue samples are often heterogeneous and multiple subtypes are involved. It is challenging to identify a signature from such a group of profiles. (2) Genes are treated as independent elements in many methods, while they may associate with each other in the given condition. (3) The disease signatures cannot identify potential drugs for personalized treatments. In order to address those limitations, I propose three strategies in this dissertation. (1) I employ clustering methods to identify sub-signatures from the heterogeneous dataset, then use a weighting strategy to concatenate them together. (2) I utilize human protein complex (HPC) information to reflect the dependencies among genes and identify an HPC signature to describe a specific type of cancer. (3) I use an HPC strategy to identify signatures for drugs, then predict a list of potential drugs for each patient. Besides predicting potential drugs directly, more indications are essential to enhance my understanding in drug repositioning studies. The interactions between biological and biomedical entities, such as drug-drug interactions (DDIs) and drug-target interactions (DTIs), help study mechanisms behind the repurposed drugs. Machine learning (ML), especially deep learning (DL), are frontier methods in predicting those interactions. Network strategies, such as constructing a network from interactions and studying topological properties, are commonly used to combine with other methods to make predictions. However, the interactions may have different functions, and merging them in a single network may cause some biases. In order to solve it, I construct two networks for two types of DDIs and employ a graph convolutional network (GCN) model to concatenate them together. In this dissertation, the first chapter introduces background information, objectives of studies, and structure of the dissertation. After that, a comprehensive review is provided in Chapter 2. Biological databases, methods and applications in drug repositioning studies, and evaluation metrics are discussed. I summarize three application scenarios in Chapter 2. The first method proposed in Chapter 3 considers the issue of identifying a cancer gene signature and predicting potential drugs. The k-means clustering method is used to identify highly reliable gene signatures. The identified signature is used to match drug profiles and identify potential drugs for the given disease. The second method proposed in Chapter 4 uses human protein complex (HPC) information to identify a protein complex signature, instead of a gene signature. This strategy improves the prediction accuracy in the experiments of cancers. Chapter 5 introduces the signature-based method in personalized cancer medicine. The profiles of a given drug are used to identify a drug signature, under the HPC strategy. Each patient has a profile, which is matched with the drug signature. Each patient has a different list of potential drugs. Chapter 6 propose a graph convolutional network with multi-kernel to predict DDIs. This method constructs two DDI kernels and concatenates them in the GCN model. It achieves higher performance in predicting DDIs than three state-of-the-art methods. In summary, this dissertation has proposed several computational algorithms for drug repositioning. Experimental results have shown that the proposed methods can achieve very good performance

    Protein Structure-Guided Approaches to Identify Functional Mutations in Cancer

    Get PDF
    Distinguishing driver mutations from passenger mutations within tumor cells continues to be a major challenge in cancer genomics. Many computational tools have been developed to address this challenge; however, they rely heavily on primary protein sequence context and frequency/mutation rate. Rare driver mutations not found in many cancer patients may be missed with these traditional approaches. Additionally, the structural context of mutations on tertiary/quaternary protein structures is not taken into account and may play a more prominent role in determining phenotype and function. This dissertation first presents a novel computational tool called HotSpot3D, which identifies regions of protein structures that are enriched in proximal mutations from cancer patients and identifies clusters of mutations within a single protein as well as along the interface of protein-protein complexes. This tool gives insight to potential rare driver mutations that may cluster closely to known hotspot driver mutations as well as critical regions of proteins specific to certain cancer types. A small subset of predictions from this tool are validated using high throughput phosphorylation data and in vitro cell-based assay to support its biological utility. We then shift to studying the druggability of mutations and apply HotSpot3D to identify potential druggable mutations that cluster with known sensitive actionable mutations. We also demonstrate how utilizing integrative omics approaches better enables precision oncology; Combining multiple data types such as genomic mutations or mRNA/protein expression outliers as biomarkers of druggability can expand the druggable cohort, better inform treatment response, and nominate novel combinatorial therapies for clinical trials. Lastly, we improve driver predictions of HotSpot3D by creating a supervised learning approach that integrates additional biological features related to structural context beyond just positional clustering. Overall, this dissertation provides a suite of computational methods to explore mutations in the context of protein structure and their potential implications in oncogenesis

    Recent applications of quantitative systems pharmacology and machine learning models across diseases

    Get PDF
    Quantitative systems pharmacology (QSP) is a quantitative and mechanistic platform describing the phenotypic interaction between drugs, biological networks, and disease conditions to predict optimal therapeutic response. In this meta-analysis study, we review the utility of the QSP platform in drug development and therapeutic strategies based on recent publications (2019–2021). We gathered recent original QSP models and described the diversity of their applications based on therapeutic areas, methodologies, software platforms, and functionalities. The collection and investigation of these publications can assist in providing a repository of recent QSP studies to facilitate the discovery and further reusability of QSP models. Our review shows that the largest number of QSP efforts in recent years is in Immuno-Oncology. We also addressed the benefits of integrative approaches in this field by presenting the applications of Machine Learning methods for drug discovery and QSP models. Based on this meta-analysis, we discuss the advantages and limitations of QSP models and propose fields where the QSP approach constitutes a valuable interface for more investigations to tackle complex diseases and improve drug development
    • …
    corecore