SwePub
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "L773:1758 2946 srt2:(2020-2023)"

Sökning: L773:1758 2946 > (2020-2023)

  • Resultat 1-10 av 21
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  • Aghdam, Rosa, et al. (författare)
  • Using informative features in machine learning based method for COVID-19 drug repurposing
  • 2021
  • Ingår i: Journal of Cheminformatics. - : Springer Nature. - 1758-2946. ; 13:1
  • Tidskriftsartikel (refereegranskat)abstract
    • Coronavirus disease 2019 (COVID-19) is caused by a novel virus named Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). This virus induced a large number of deaths and millions of confirmed cases worldwide, creating a serious danger to public health. However, there are no specific therapies or drugs available for COVID-19 treatment. While new drug discovery is a long process, repurposing available drugs for COVID-19 can help recognize treatments with known clinical profiles. Computational drug repurposing methods can reduce the cost, time, and risk of drug toxicity. In this work, we build a graph as a COVID-19 related biological network. This network is related to virus targets or their associated biological processes. We select essential proteins in the constructed biological network that lead to a major disruption in the network. Our method from these essential proteins chooses 93 proteins related to COVID-19 pathology. Then, we propose multiple informative features based on drug-target and protein-protein interaction information. Through these informative features, we find five appropriate clusters of drugs that contain some candidates as potential COVID-19 treatments. To evaluate our results, we provide statistical and clinical evidence for our candidate drugs. From our proposed candidate drugs, 80% of them were studied in other studies and clinical trials.
  •  
2.
  • Ahmed, Laeeq, et al. (författare)
  • Predicting target profiles with confidence as a service using docking scores
  • 2020
  • Ingår i: Journal of Cheminformatics. - : Springer Nature. - 1758-2946. ; 12:1
  • Tidskriftsartikel (refereegranskat)abstract
    • Background: Identifying and assessing ligand-target binding is a core component in early drug discovery as one or more unwanted interactions may be associated with safety issues. Contributions: We present an open-source, extendable web service for predicting target profiles with confidence using machine learning for a panel of 7 targets, where models are trained on molecular docking scores from a large virtual library. The method uses conformal prediction to produce valid measures of prediction efficiency for a particular confidence level. The service also offers the possibility to dock chemical structures to the panel of targets with QuickVina on individual compound basis. Results: The docking procedure and resulting models were validated by docking well-known inhibitors for each of the 7 targets using QuickVina. The model predictions showed comparable performance to molecular docking scores against an external validation set. The implementation as publicly available microservices on Kubernetes ensures resilience, scalability, and extensibility.
  •  
3.
  • Bahl, A, et al. (författare)
  • PROTEOMAS: a workflow enabling harmonized proteomic meta-analysis and proteomic signature mapping
  • 2023
  • Ingår i: Journal of cheminformatics. - : Springer Science and Business Media LLC. - 1758-2946. ; 15:1, s. 34-
  • Tidskriftsartikel (refereegranskat)abstract
    • Toxicological evaluation of substances in regulation still often relies on animal experiments. Understanding the substances’ mode-of-action is crucial to develop alternative test strategies. Omics methods are promising tools to achieve this goal. Until now, most attention was focused on transcriptomics, while proteomics is not yet routinely applied in toxicology despite the large number of datasets available in public repositories. Exploiting the full potential of these datasets is hampered by differences in measurement procedures and follow-up data processing. Here we present the tool PROTEOMAS, which allows meta-analysis of proteomic data from public origin. The workflow was designed for analyzing proteomic studies in a harmonized way and to ensure transparency in the analysis of proteomic data for regulatory purposes. It agrees with the Omics Reporting Framework guidelines of the OECD with the intention to integrate proteomics to other omic methods in regulatory toxicology. The overarching aim is to contribute to the development of AOPs and to understand the mode of action of substances. To demonstrate the robustness and reliability of our workflow we compared our results to those of the original studies. As a case study, we performed a meta-analysis of 25 proteomic datasets to investigate the toxicological effects of nanomaterials at the lung level. PROTEOMAS is an important contribution to the development of alternative test strategies enabling robust meta-analysis of proteomic data. This workflow commits to the FAIR principles (Findable, Accessible, Interoperable and Reusable) of computational protocols.
  •  
4.
  • Bahl, A, et al. (författare)
  • PROTEOMAS: a workflow enabling harmonized proteomic meta-analysis and proteomic signature mapping
  • 2023
  • Ingår i: Journal of cheminformatics. - : Springer Science and Business Media LLC. - 1758-2946. ; 15:1, s. 34-
  • Tidskriftsartikel (refereegranskat)abstract
    • Toxicological evaluation of substances in regulation still often relies on animal experiments. Understanding the substances’ mode-of-action is crucial to develop alternative test strategies. Omics methods are promising tools to achieve this goal. Until now, most attention was focused on transcriptomics, while proteomics is not yet routinely applied in toxicology despite the large number of datasets available in public repositories. Exploiting the full potential of these datasets is hampered by differences in measurement procedures and follow-up data processing. Here we present the tool PROTEOMAS, which allows meta-analysis of proteomic data from public origin. The workflow was designed for analyzing proteomic studies in a harmonized way and to ensure transparency in the analysis of proteomic data for regulatory purposes. It agrees with the Omics Reporting Framework guidelines of the OECD with the intention to integrate proteomics to other omic methods in regulatory toxicology. The overarching aim is to contribute to the development of AOPs and to understand the mode of action of substances. To demonstrate the robustness and reliability of our workflow we compared our results to those of the original studies. As a case study, we performed a meta-analysis of 25 proteomic datasets to investigate the toxicological effects of nanomaterials at the lung level. PROTEOMAS is an important contribution to the development of alternative test strategies enabling robust meta-analysis of proteomic data. This workflow commits to the FAIR principles (Findable, Accessible, Interoperable and Reusable) of computational protocols.
  •  
5.
  • Bajorath, J., et al. (författare)
  • Chemoinformatics and artificial intelligence colloquium: progress and challenges in developing bioactive compounds
  • 2022
  • Ingår i: Journal of Cheminformatics. - : Springer Science and Business Media LLC. - 1758-2946. ; 14:1
  • Tidskriftsartikel (refereegranskat)abstract
    • We report the main conclusions of the first Chemoinformatics and Artificial Intelligence Colloquium, Mexico City, June 15–17, 2022. Fifteen lectures were presented during a virtual public event with speakers from industry, academia, and non-for-profit organizations. Twelve hundred and ninety students and academics from more than 60 countries. During the meeting, applications, challenges, and opportunities in drug discovery, de novo drug design, ADME-Tox (absorption, distribution, metabolism, excretion and toxicity) property predictions, organic chemistry, peptides, and antibiotic resistance were discussed. The program along with the recordings of all sessions are freely available at https://www.difacquim.com/english/events/2022-colloquium/.
  •  
6.
  • Bohman, Björn (författare)
  • Determining the parent and associated fragment formulae in mass spectrometry via the parent subformula graph
  • 2023
  • Ingår i: Journal of Cheminformatics. - 1758-2946. ; 15
  • Tidskriftsartikel (refereegranskat)abstract
    • BackgroundIdentifying the molecular formula and fragmentation reactions of an unknown compound from its mass spectrum is crucial in areas such as natural product chemistry and metabolomics. We propose a method for identifying the correct candidate formula of an unidentified natural product from its mass spectrum. The method involves scoring the plausibility of parent candidate formulae based on a parent subformula graph (PSG), and two possible metrics relating to the number of edges in the PSG. This method is applicable to both electron-impact mass spectrometry (EI-MS) and tandem mass spectrometry (MS/MS) data. Additionally, this work introduces the two-dimensional fragmentation plot (2DFP) for visualizing PSGs.ResultsOur results suggest that incorporating information regarding the edges of the PSG results in enhanced performance in correctly identifying parent formulae, in comparison to the more well-accepted "MS/MS score", on the 2016 Computational Assessment of Small Molecule Identification (CASMI 2016) data set (76.3 vs 58.9% correct formula identification) and the Research Centre for Toxic Compounds in the Environment (RECETOX) data set (66.2% vs 59.4% correct formula identification). In the extension of our method to identify the correct candidate formula from complex EI-MS data of semiochemicals, our method again performed better (correct formula appearing in the top 4 candidates in 20/23 vs 7/23 cases) than the MS/MS score, and enables the rapid identification of both the correct parent ion mass and the correct parent formula with minimal expert intervention.ConclusionOur method reliably identifies the correct parent formula even when the mass information is ambiguous. Furthermore, should parent formula identification be successful, the majority of associated fragment formulae can also be correctly identified. Our method can also identify the parent ion and its associated fragments in EI-MS spectra where the identity of the parent ion is unclear due to low quantities and overlapping compounds. Finally, our method does not inherently require empirical fitting of parameters or statistical learning, meaning it is easy to implement and extend upon.Scientific contributionDeveloped, implemented and tested new metrics for assessing plausibility of candidate molecular formulae obtained from HR-MS data.
  •  
7.
  • Guo, Jeff, et al. (författare)
  • DockStream: a docking wrapper to enhance de novo molecular design
  • 2021
  • Ingår i: Journal of Cheminformatics. - : Springer Science and Business Media LLC. - 1758-2946 .- 1758-2946. ; 13:1
  • Tidskriftsartikel (refereegranskat)abstract
    • Recently, we have released the de novo design platform REINVENT in version 2.0. This improved and extended iteration supports far more features and scoring function components, which allows bespoke and tailor-made protocols to maximize impact in small molecule drug discovery projects. A major obstacle of generative models is producing active compounds, in which predictive (QSAR) models have been applied to enrich target activity. However, QSAR models are inherently limited by their applicability domains. To overcome these limitations, we introduce a structure-based scoring component for REINVENT. DockStream is a flexible, stand-alone molecular docking wrapper that provides access to a collection of ligand embedders and docking backends. Using the benchmarking and analysis workflow provided in DockStream, execution and subsequent analysis of a variety of docking configurations can be automated. Docking algorithms vary greatly in performance depending on the target and the benchmarking and analysis workflow provides a streamlined solution to identifying productive docking configurations. We show that an informative docking configuration can inform the REINVENT agent to optimize towards improving docking scores using public data. With docking activated, REINVENT is able to retain key interactions in the binding site, discard molecules which do not fit the binding cavity, harness unused (sub-)pockets, and improve overall performance in the scaffold-hopping scenario. The code is freely available at https://github.com/MolecularAI/DockStream.
  •  
8.
  • He, Jiazhen, et al. (författare)
  • Molecular optimization by capturing chemist’s intuition using deep neural networks
  • 2021
  • Ingår i: Journal of Cheminformatics. - : BioMed Central. - 1758-2946. ; 13:1
  • Tidskriftsartikel (refereegranskat)abstract
    • A main challenge in drug discovery is finding molecules with a desirable balance of multiple properties. Here, we focus on the task of molecular optimization, where the goal is to optimize a given starting molecule towards desirable properties. This task can be framed as a machine translation problem in natural language processing, where in our case, a molecule is translated into a molecule with optimized properties based on the SMILES representation. Typically, chemists would use their intuition to suggest chemical transformations for the starting molecule being optimized. A widely used strategy is the concept of matched molecular pairs where two molecules differ by a single transformation. We seek to capture the chemist’s intuition from matched molecular pairs using machine translation models. Specifically, the sequence-to-sequence model with attention mechanism, and the Transformer model are employed to generate molecules with desirable properties. As a proof of concept, three ADMET properties are optimized simultaneously: logD, solubility, and clearance, which are important properties of a drug. Since desirable properties often vary from project to project, the user-specified desirable property changes are incorporated into the input as an additional condition together with the starting molecules being optimized. Thus, the models can be guided to generate molecules satisfying the desirable properties. Additionally, we compare the two machine translation models based on the SMILES representation, with a graph-to-graph translation model HierG2G, which has shown the state-of-the-art performance in molecular optimization. Our results show that the Transformer can generate more molecules with desirable properties by making small modifications to the given starting molecules, which can be intuitive to chemists. A further enrichment of diverse molecules can be achieved by using an ensemble of models.
  •  
9.
  • He, Jiazhen, et al. (författare)
  • Transformer-based molecular optimization beyond matched molecular pairs
  • 2022
  • Ingår i: Journal of Cheminformatics. - : Springer Science and Business Media LLC. - 1758-2946 .- 1758-2946. ; 14:1
  • Tidskriftsartikel (refereegranskat)abstract
    • Molecular optimization aims to improve the drug profile of a starting molecule. It is a fundamental problem in drug discovery but challenging due to (i) the requirement of simultaneous optimization of multiple properties and (ii) the large chemical space to explore. Recently, deep learning methods have been proposed to solve this task by mimicking the chemist's intuition in terms of matched molecular pairs (MMPs). Although MMPs is a widely used strategy by medicinal chemists, it offers limited capability in terms of exploring the space of structural modifications, therefore does not cover the complete space of solutions. Often more general transformations beyond the nature of MMPs are feasible and/or necessary, e.g. simultaneous modifications of the starting molecule at different places including the core scaffold. This study aims to provide a general methodology that offers more general structural modifications beyond MMPs. In particular, the same Transformer architecture is trained on different datasets. These datasets consist of a set of molecular pairs which reflect different types of transformations. Beyond MMP transformation, datasets reflecting general structural changes are constructed from ChEMBL based on two approaches: Tanimoto similarity (allows for multiple modifications) and scaffold matching (allows for multiple modifications but keep the scaffold constant) respectively. We investigate how the model behavior can be altered by tailoring the dataset while using the same model architecture. Our results show that the models trained on differently prepared datasets transform a given starting molecule in a way that it reflects the nature of the dataset used for training the model. These models could complement each other and unlock the capability for the chemists to pursue different options for improving a starting molecule.
  •  
10.
  • Joeres, R., et al. (författare)
  • GlyLES: Grammar-based Parsing of Glycans from IUPAC-condensed to SMILES
  • 2023
  • Ingår i: Journal of Cheminformatics. - : Springer Science and Business Media LLC. - 1758-2946. ; 15:1
  • Tidskriftsartikel (refereegranskat)abstract
    • Glycans are important polysaccharides on cellular surfaces that are bound to glycoproteins and glycolipids. These are one of the most common post-translational modifications of proteins in eukaryotic cells. They play important roles in protein folding, cell-cell interactions, and other extracellular processes. Changes in glycan structures may influence the course of different diseases, such as infections or cancer. Glycans are commonly represented using the IUPAC-condensed notation. IUPAC-condensed is a textual representation of glycans operating on the same topological level as the Symbol Nomenclature for Glycans (SNFG) that assigns colored, geometrical shapes to the main monomers. These symbols are then connected in tree-like structures, visualizing the glycan structure on a topological level. Yet for a representation on the atomic level, notations such as SMILES should be used. To our knowledge, there is no easy-to-use, general, open-source, and offline tool to convert the IUPAC-condensed notation to SMILES. Here, we present the open-access Python package GlyLES for the generalizable generation of SMILES representations out of IUPAC-condensed representations. GlyLES uses a grammar to read in the monomer tree from the IUPAC-condensed notation. From this tree, the tool can compute the atomic structures of each monomer based on their IUPAC-condensed descriptions. In the last step, it merges all monomers into the atomic structure of a glycan in the SMILES notation. GlyLES is the first package that allows conversion from the IUPAC-condensed notation of glycans to SMILES strings. This may have multiple applications, including straightforward visualization, substructure search, molecular modeling and docking, and a new featurization strategy for machine-learning algorithms. GlyLES is available at https://github. com/kalininalab/GlyLES.
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-10 av 21
Typ av publikation
tidskriftsartikel (20)
forskningsöversikt (1)
Typ av innehåll
refereegranskat (21)
Författare/redaktör
Spjuth, Ola, Profess ... (5)
Engkvist, Ola, 1967 (4)
Tyrchan, Christian (3)
Nymark, P (2)
Norinder, Ulf, 1956- (2)
Schaal, Wesley, PhD (2)
visa fler...
Spjuth, Ola, Docent, ... (2)
Voronov, Alexey (2)
Svensson, Fredrik (2)
Oprea, Tudor I (2)
Bahl, A (2)
Ibrahim, C (2)
Plate, K (2)
Haase, A (2)
Dengjel, J (2)
Dumit, VI (2)
aut (1)
Larsson, Anders (1)
Kultima, Kim (1)
Aghdam, Rosa (1)
Habibi, Mahnaz (1)
Taheri, Golnaz (1)
Laure, Erwin (1)
Engkvist, Ola (1)
Ahmed, Laeeq (1)
Alogheli, Hiba (1)
Arvidsson Mc Shane, ... (1)
Alvarsson, Jonathan, ... (1)
Berg, Arvid (1)
Lampa, Samuel (1)
Arvidsson McShane, S ... (1)
Mestres, J (1)
Sandström, Emil (1)
Bajorath, J. (1)
Chávez-Hernández, A. ... (1)
Duran-Frigola, M. (1)
Fernández-de Gortari ... (1)
Gasteiger, J. (1)
López-López, E. (1)
Maggiora, G. M. (1)
Medina-Franco, J. L. (1)
Méndez-Lucio, O. (1)
Miranda-Quintana, R. ... (1)
Plisson, F. (1)
Prieto-Martínez, F. ... (1)
Rodríguez-Pérez, R. (1)
Rondón-Villarreal, P ... (1)
Saldívar-Gonzalez, F ... (1)
Sánchez-Cruz, N. (1)
Valli, M. (1)
visa färre...
Lärosäte
Uppsala universitet (9)
Chalmers tekniska högskola (4)
Göteborgs universitet (3)
Kungliga Tekniska Högskolan (3)
Karolinska Institutet (3)
Stockholms universitet (2)
visa fler...
Örebro universitet (2)
Umeå universitet (1)
Sveriges Lantbruksuniversitet (1)
visa färre...
Språk
Engelska (21)
Forskningsämne (UKÄ/SCB)
Naturvetenskap (15)
Medicin och hälsovetenskap (5)
Teknik (2)

År

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy