1
|
Lin M, Cai J, Wei Y, Peng X, Luo Q, Li B, Chen Y, Wang L. MalariaFlow: A comprehensive deep learning platform for multistage phenotypic antimalarial drug discovery. Eur J Med Chem 2024; 277:116776. [PMID: 39173285 DOI: 10.1016/j.ejmech.2024.116776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/24/2024]
Abstract
Malaria remains a significant global health challenge due to the growing drug resistance of Plasmodium parasites and the failure to block transmission within human host. While machine learning (ML) and deep learning (DL) methods have shown promise in accelerating antimalarial drug discovery, the performance of deep learning models based on molecular graph and other co-representation approaches warrants further exploration. Current research has overlooked mutant strains of the malaria parasite with varying degrees of sensitivity or resistance, and has not covered the prediction of inhibitory activities across the three major life cycle stages (liver, asexual blood, and gametocyte) within the human host, which is crucial for both treatment and transmission blocking. In this study, we manually curated a benchmark antimalarial activity dataset comprising 407,404 unique compounds and 410,654 bioactivity data points across ten Plasmodium phenotypes and three stages. The performance was systematically compared among two fingerprint-based ML models (RF::Morgan and XGBoost:Morgan), four graph-based DL models (GCN, GAT, MPNN, and Attentive FP), and three co-representations DL models (FP-GNN, HiGNN, and FG-BERT), which reveal that: 1) The FP-GNN model achieved the best predictive performance, outperforming the other methods in distinguishing active and inactive compounds across balanced, more positive, and more negative datasets, with an overall AUROC of 0.900; 2) Fingerprint-based ML models outperformed graph-based DL models on large datasets (>1000 compounds), but the three co-representations DL models were able to incorporate domain-specific chemical knowledge to bridge this gap, achieving better predictive performance. These findings provide valuable guidance for selecting appropriate ML and DL methods for antimalarial activity prediction tasks. The interpretability analysis of the FP-GNN model revealed its ability to accurately capture the key structural features responsible for the liver- and blood-stage activities of the known antimalarial drug atovaquone. Finally, we developed a web server, MalariaFlow, incorporating these high-quality models for antimalarial activity prediction, virtual screening, and similarity search, successfully predicting novel triple-stage antimalarial hits validated through experimental testing, demonstrating its effectiveness and value in discovering potential multistage antimalarial drug candidates.
Collapse
Affiliation(s)
- Mujie Lin
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Junxi Cai
- School of Civil Engineering and Transportation, South China University of Technology, Guangzhou, 510006, China
| | - Yuancheng Wei
- School of Software Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Xinru Peng
- School of Software Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Qianhui Luo
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Biaoshun Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Yihao Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Ling Wang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
2
|
Casanola-Martin GM, Karuth A, Pham-The H, González-Díaz H, Webster DC, Rasulev B. Machine learning analysis of a large set of homopolymers to predict glass transition temperatures. Commun Chem 2024; 7:226. [PMID: 39358434 PMCID: PMC11447034 DOI: 10.1038/s42004-024-01305-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024] Open
Abstract
Glass transition temperature of polymers, Tg, is an important thermophysical property, which sometimes can be difficult to measure experimentally. In this regard, data-driven machine learning approaches are important alternatives to assess Tg values, in a high-throughput way. In this study, a large dataset of more than 900 polymers with reported glass transition temperature (Tg) was assembled from various public sources in order to develop a predictive model depicting the structure-property relationships. The collected dataset was curated, explored via cluster analysis, and then split into training and test sets for validation purposes and then polymer structures characterized by molecular descriptors. To find the models, several machine learning techniques, including multiple linear regression (MLR), k-nearest neighbor (k-NN), support vector machine (SVM), random forest (RF), gaussian processes for regression (GPR), and multi-layer perceptron (MLP) were explored. As result, a model with the subset of 15 descriptors accurately predicting the glass transition temperatures was developed. The electronic effect indices were determined to be important properties that positively contribute to the Tg values. The SVM-based model showed the best performance with determination coefficients (R2) of 0.813 and 0.770, for training and test sets, respectively. Also, the SVM model showed the lowest estimation error, RMSE = 0.062. In addition, the developed structure-property model was implemented as a web app to be used as an online computational tool to design and evaluate new homopolymers with desired glass transition profiles.
Collapse
Affiliation(s)
| | - Anas Karuth
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, USA
| | - Hai Pham-The
- University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Humbert González-Díaz
- Department of Organic and Inorganic Chemistry, University of Basque Country UPV/EHU, Leioa, Spain
- BIOFISIKA, Basque Center for Biophysics CSIC-UPVEH, Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Biscay, Spain
| | - Dean C Webster
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, USA
| | - Bakhtiyor Rasulev
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, USA.
| |
Collapse
|
3
|
Silva GM, Gomes SQ, Lopes CD, de Albuquerque S, de Paula da Silva CHT. Structural analysis and shape-based identification of novel inhibitors targeting the Trypanosoma cruzi proteasome. Int J Biol Macromol 2024; 277:134290. [PMID: 39084432 DOI: 10.1016/j.ijbiomac.2024.134290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/17/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024]
Abstract
There is an urgent need to develop new, safer, and more effective drugs against Chagas disease (CD) as well as related kinetoplastid diseases. Targeting and inhibiting the Trypanosoma cruzi proteasome has emerged as a promising therapeutic approach in this context. To expand the chemical space for this class of inhibitors, we performed virtual screening campaigns with emphasis on shape-based similarity and ADMET prioritization. We describe the ideation and application of robustly validated shape queries for these campaigns, which furnished 44 compounds for biological evaluation. Five hit compounds demonstrated in vitro antitrypanosomal activity by potential inhibition of T. cruzi proteasome and notable chemical diversities, particularly, LCQFTC11. Structural insights were achieved by homology modeling, sequence/structure alignment, proteasome-species comparison, docking, molecular dynamics, and MMGBSA binding affinity estimations. These methods confirmed key interactions as well as the stability of LCQFTC11 at the β4/β5 subunits' binding site of the T. cruzi proteasome, consistent with known inhibitors. Our results warrant future assay confirmation of our hit as a T. cruzi proteasome inhibitor. Importantly, we also shed light into dynamic details for a proteasome inhibition mechanism that shall be further investigated. We expect to contribute to the development of viable CD drug candidates through such a relevant approach.
Collapse
Affiliation(s)
- Guilherme Martins Silva
- Center for Drug Discovery and Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; Computational Laboratory of Pharmaceutical Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil.
| | - Suzane Quintana Gomes
- Computational Laboratory of Pharmaceutical Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil
| | - Carla Duque Lopes
- Laboratório de Parasitologia, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil
| | - Sérgio de Albuquerque
- Laboratório de Parasitologia, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil
| | - Carlos Henrique Tomich de Paula da Silva
- Computational Laboratory of Pharmaceutical Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil
| |
Collapse
|
4
|
Shah P, Siramshetty VB, Mathé E, Xu X. Developing Robust Human Liver Microsomal Stability Prediction Models: Leveraging Inter-Species Correlation with Rat Data. Pharmaceutics 2024; 16:1257. [PMID: 39458588 PMCID: PMC11510424 DOI: 10.3390/pharmaceutics16101257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/03/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
Objectives: Pharmacokinetic issues were the leading cause of drug attrition, accounting for approximately 40% of all cases before the turn of the century. To this end, several high-throughput in vitro assays like microsomal stability have been developed to evaluate the pharmacokinetic profiles of compounds in the early stages of drug discovery. At NCATS, a single-point rat liver microsomal (RLM) stability assay is used as a Tier I assay, while human liver microsomal (HLM) stability is employed as a Tier II assay. We experimentally screened and collected data on over 30,000 compounds for RLM stability and over 7000 compounds for HLM stability. Although HLM stability screening provides valuable insights, the increasing number of hits generated, along with the time- and resource-intensive nature of the assay, highlights the need for alternative strategies. One promising approach is leveraging in silico models trained on these experimental datasets. Methods: We describe the development of an HLM stability prediction model using our in-house HLM stability dataset. Results: Employing both classical machine learning methods and advanced techniques, such as neural networks, we achieved model accuracies exceeding 80%. Moreover, we validated our model using external test sets and found that our models are comparable to some of the best models in literature. Additionally, the strong correlation observed between our RLM and HLM data was further reinforced by the fact that our HLM model performance improved when using RLM stability predictions as an input descriptor. Conclusions: The best model along with a subset of our dataset (PubChem AID: 1963597) has been made publicly accessible on the ADME@NCATS website for the benefit of the greater drug discovery community. To the best of our knowledge, it is the largest open-source model of its kind and the first to leverage cross-species data.
Collapse
|
5
|
Dong F, Hardy B, Liu J, Mohoric T, Guo W, Exner T, Tong W, Dohler J, Bachler D, Hong H. Development of a comprehensive open access "molecules with androgenic activity resource (MAAR)" to facilitate risk assessment of chemicals. Exp Biol Med (Maywood) 2024; 249:10279. [PMID: 39364092 PMCID: PMC11446862 DOI: 10.3389/ebm.2024.10279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/27/2024] [Indexed: 10/05/2024] Open
Abstract
The increasing prevalence of endocrine-disrupting chemicals (EDCs) and their potential adverse effects on human health underscore the necessity for robust tools to assess and manage associated risks. The androgen receptor (AR) is a critical component of the endocrine system, playing a pivotal role in mediating the biological effects of androgens, which are male sex hormones. Exposure to androgen-disrupting chemicals during critical periods of development, such as fetal development or puberty, may result in adverse effects on reproductive health, including altered sexual differentiation, impaired fertility, and an increased risk of reproductive disorders. Therefore, androgenic activity data is critical for chemical risk assessment. A large amount of androgenic data has been generated using various experimental protocols. Moreover, the data are reported in different formats and in diverse sources. To facilitate utilization of androgenic activity data in chemical risk assessment, the Molecules with Androgenic Activity Resource (MAAR) was developed. MAAR is the first open-access platform designed to streamline and enhance the risk assessment of chemicals with androgenic activity. MAAR's development involved the integration of diverse data sources, including data from public databases and mining literature, to establish a reliable and versatile repository. The platform employs a user-friendly interface, enabling efficient navigation and extraction of pertinent information. MAAR is poised to advance chemical risk assessment by offering unprecedented access to information crucial for evaluating the androgenic potential of a wide array of chemicals. The open-access nature of MAAR promotes transparency and collaboration, fostering a collective effort to address the challenges posed by androgenic EDCs.
Collapse
Affiliation(s)
- Fan Dong
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, United States
| | - Barry Hardy
- Edelweiss Connect Inc., Durham, NC, United States
| | - Jie Liu
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, United States
| | | | - Wenjing Guo
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, United States
| | - Thomas Exner
- Edelweiss Connect Inc., Durham, NC, United States
| | - Weida Tong
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, United States
| | - Joh Dohler
- Edelweiss Connect Inc., Durham, NC, United States
| | | | - Huixiao Hong
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, United States
| |
Collapse
|
6
|
Johnson H, Gusev F, Dull JT, Seo Y, Priestley RD, Isayev O, Rand BP. Discovery of Crystallizable Organic Semiconductors with Machine Learning. J Am Chem Soc 2024; 146:21583-21590. [PMID: 39051486 PMCID: PMC11311223 DOI: 10.1021/jacs.4c05245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024]
Abstract
Crystalline organic semiconductors are known to have improved charge carrier mobility and exciton diffusion length in comparison to their amorphous counterparts. Certain organic molecular thin films can be transitioned from initially prepared amorphous layers to large-scale crystalline films via abrupt thermal annealing. Ideally, these films crystallize as platelets with long-range-ordered domains on the scale of tens to hundreds of microns. However, other organic molecular thin films may instead crystallize as spherulites or resist crystallization entirely. Organic molecules that have the capability of transforming into a platelet morphology feature both high melting point (Tm) and crystallization driving force (ΔGc). In this work, we employed machine learning (ML) to identify candidate organic materials with the potential to crystallize into platelets by estimating the aforementioned thermal properties. Six organic molecules identified by the ML algorithm were experimentally evaluated; three crystallized as platelets, one crystallized as a spherulite, and two resisted thin film crystallization. These results demonstrate a successful application of ML in the scope of predicting thermal properties of organic molecules and reinforce the principles of Tm and ΔGc as metrics that aid in predicting the crystallization behavior of organic thin films.
Collapse
Affiliation(s)
- Holly
M. Johnson
- Department
of Electrical and Computer Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Filipp Gusev
- Computational
Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Chemistry, Mellon College of Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Jordan T. Dull
- Department
of Electrical and Computer Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Yejoon Seo
- Department
of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Rodney D. Priestley
- Department
of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Olexandr Isayev
- Computational
Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Chemistry, Mellon College of Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Barry P. Rand
- Department
of Electrical and Computer Engineering, Princeton University, Princeton, New Jersey 08544, United States
- Andlinger
Center for Energy and the Environment, Princeton
University, Princeton, New Jersey 08544, United States
| |
Collapse
|
7
|
Martin HJ, Hossain MA, Wellnitz J, Kelestemur E, Hochuli JE, Parveen S, Arrowsmith C, Willson TM, Muratov E, Tropsha A. Heli-SMACC: Helicase-targeting SMAll Molecule Compound Collection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.04.602122. [PMID: 39026851 PMCID: PMC11257486 DOI: 10.1101/2024.07.04.602122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Helicases have emerged as promising targets for the development of antiviral drugs; however, the family remains largely undrugged. To support the focused development of viral helicase inhibitors we identified, collected, and integrated all chemogenomics data for all available helicases from the ChEMBL database. After thoroughly curating and enriching the data with relevant annotations we have created a derivative database of helicase inhibitors which we dubbed Heli-SMACC (Helicase-targeting SMAll Molecule Compound Collection). The current version of Heli-SMACC contains 20,432 bioactivity entries for viral, human, and bacterial helicases. We have selected 30 compounds with promising viral helicase activity and tested them in a SARS-CoV-2 NSP13 ATPase assay. Twelve compounds demonstrated ATPase inhibition and a consistent dose-response curve. The Heli-SMACC database may serve as a reference for virologists and medicinal chemists working on the development of novel helicase inhibitors. Heli-SMACC is publicly available at https://smacc.mml.unc.edu.
Collapse
Affiliation(s)
- Holli-Joi Martin
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Mohammad A. Hossain
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - James Wellnitz
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Enes Kelestemur
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Joshua E. Hochuli
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Sumera Parveen
- The Structural Genomics Consortium, University of Toronto, Canada
| | | | - Timothy M. Willson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Eugene Muratov
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Alexander Tropsha
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
8
|
Boczar D, Michalska K. A Review of Machine Learning and QSAR/QSPR Predictions for Complexes of Organic Molecules with Cyclodextrins. Molecules 2024; 29:3159. [PMID: 38999108 PMCID: PMC11243237 DOI: 10.3390/molecules29133159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
Cyclodextrins are macrocyclic rings composed of glucose residues. Due to their remarkable structural properties, they can form host-guest inclusion complexes, which is why they are frequently used in the pharmaceutical, cosmetic, and food industries, as well as in environmental and analytical chemistry. This review presents the reports from 2011 to 2023 on the quantitative structure-activity/property relationship (QSAR/QSPR) approach, which is primarily employed to predict the thermodynamic stability of inclusion complexes. This article extensively discusses the significant developments related to the size of available experimental data, the available sets of descriptors, and the machine learning (ML) algorithms used, such as support vector machines, random forests, artificial neural networks, and gradient boosting. As QSAR/QPR analysis only requires molecular structures of guests and experimental values of stability constants, this approach may be particularly useful for predicting these values for complexes with randomly substituted cyclodextrins, as well as for estimating their dependence on pH. This work proposes solutions on how to effectively use this knowledge, which is especially important for researchers who will deal with this topic in the future. This review also presents other applications of ML in relation to CD complexes, including the prediction of physicochemical properties of CD complexes, the development of analytical methods based on complexation with CDs, and the optimisation of experimental conditions for the preparation of the complexes.
Collapse
Affiliation(s)
- Dariusz Boczar
- Department of Synthetic Drugs, National Medicines Institute, Chełmska 30/34, 00-725 Warsaw, Poland
| | - Katarzyna Michalska
- Department of Synthetic Drugs, National Medicines Institute, Chełmska 30/34, 00-725 Warsaw, Poland
| |
Collapse
|
9
|
Wossnig L, Furtmann N, Buchanan A, Kumar S, Greiff V. Best practices for machine learning in antibody discovery and development. Drug Discov Today 2024; 29:104025. [PMID: 38762089 DOI: 10.1016/j.drudis.2024.104025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/25/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
In the past 40 years, therapeutic antibody discovery and development have advanced considerably, with machine learning (ML) offering a promising way to speed up the process by reducing costs and the number of experiments required. Recent progress in ML-guided antibody design and development (D&D) has been hindered by the diversity of data sets and evaluation methods, which makes it difficult to conduct comparisons and assess utility. Establishing standards and guidelines will be crucial for the wider adoption of ML and the advancement of the field. This perspective critically reviews current practices, highlights common pitfalls and proposes method development and evaluation guidelines for various ML-based techniques in therapeutic antibody D&D. Addressing challenges across the ML process, best practices are recommended for each stage to enhance reproducibility and progress.
Collapse
Affiliation(s)
- Leonard Wossnig
- LabGenius Ltd, The Biscuit Factory, 100 Drummond Road, London SE16 4DG, UK; Department of Computer Science, University College London, 66-72 Gower St, London WC1E 6EA, UK.
| | - Norbert Furtmann
- R&D Large Molecules Research Platform, Sanofi Deutschland GmbH, Industriepark Höchst, Frankfurt Am Main, Germany
| | - Andrew Buchanan
- Biologics Engineering, R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Sandeep Kumar
- Computational Protein Design and Modeling Group, Computational Science, Moderna Therapeutics, 200 Technology Square, Cambridge, MA 02139, USA
| | - Victor Greiff
- Department of Immunology and Oslo University Hospital, University of Oslo, Oslo, Norway
| |
Collapse
|
10
|
Sanches IH, Braga RC, Alves VM, Andrade CH. Enhancing hERG Risk Assessment with Interpretable Classificatory and Regression Models. Chem Res Toxicol 2024; 37:910-922. [PMID: 38781421 PMCID: PMC11187631 DOI: 10.1021/acs.chemrestox.3c00400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/22/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
The human Ether-à-go-go-Related Gene (hERG) is a transmembrane protein that regulates cardiac action potential, and its inhibition can induce a potentially deadly cardiac syndrome. In vitro tests help identify hERG blockers at early stages; however, the high cost motivates searching for alternative, cost-effective methods. The primary goal of this study was to enhance the Pred-hERG tool for predicting hERG blockage. To achieve this, we developed new QSAR models that incorporated additional data, updated existing classificatory and multiclassificatory models, and introduced new regression models. Notably, we integrated SHAP (SHapley Additive exPlanations) values to offer a visual interpretation of these models. Utilizing the latest data from ChEMBL v30, encompassing over 14,364 compounds with hERG data, our binary and multiclassification models outperformed both the previous iteration of Pred-hERG and all publicly available models. Notably, the new version of our tool introduces a regression model for predicting hERG activity (pIC50). The optimal model demonstrated an R2 of 0.61 and an RMSE of 0.48, surpassing the only available regression model in the literature. Pred-hERG 5.0 now offers users a swift, reliable, and user-friendly platform for the early assessment of chemically induced cardiotoxicity through hERG blockage. The tool provides versatile outcomes, including (i) classificatory predictions of hERG blockage with prediction reliability, (ii) multiclassificatory predictions of hERG blockage with reliability, (iii) regression predictions with estimated pIC50 values, and (iv) probability maps illustrating the contribution of chemical fragments for each prediction. Furthermore, we implemented explainable AI analysis (XAI) to visualize SHAP values, providing insights into the contribution of each feature to binary classification predictions. A consensus prediction calculated based on the predictions of the three developed models is also present to assist the user's decision-making process. Pred-hERG 5.0 has been designed to be user-friendly, making it accessible to users without computational or programming expertise. The tool is freely available at http://predherg.labmol.com.br.
Collapse
Affiliation(s)
- Igor H. Sanches
- Laboratory
for Molecular Modeling and Drug Design (LabMol), Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO 74690-900, Brazil
- Center
for Excellence in Artificial Intelligence (CEIA), Institute of Informatics, Universidade Federal de Goiás, Goiânia, GO 74690-900, Brazil
- Center
for the Research and Advancement in Fragments and Molecular Targets
(CRAFT), School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto, SP 05508-220, Brazil
| | | | - Vinicius M. Alves
- University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Carolina Horta Andrade
- Laboratory
for Molecular Modeling and Drug Design (LabMol), Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO 74690-900, Brazil
- Center
for Excellence in Artificial Intelligence (CEIA), Institute of Informatics, Universidade Federal de Goiás, Goiânia, GO 74690-900, Brazil
- Center
for the Research and Advancement in Fragments and Molecular Targets
(CRAFT), School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto, SP 05508-220, Brazil
| |
Collapse
|
11
|
Cogo RM, Pavani TFA, Mengarda ACA, Cajas RA, Teixeira TR, Fukui-Silva L, Sun YU, Liu LJ, Amarasinghe DK, Yoon MC, Santos-Filho OA, de Moraes J, Caffrey CR, G G Rando D. Pharmacophore Virtual Screening Identifies Riboflavin as an Inhibitor of the Schistosome Cathepsin B1 Protease with Antiparasitic Activity. ACS OMEGA 2024; 9:25356-25369. [PMID: 38882094 PMCID: PMC11170711 DOI: 10.1021/acsomega.4c03376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/18/2024]
Abstract
Schistosomiasis is a neglected disease of poverty that affects over 200 million people worldwide and relies on a single drug for therapy. The cathepsin B1 cysteine protease (SmCB1) of Schistosoma mansoni has been investigated as a potential target. Here, a structure-based pharmacophore virtual screening (VS) approach was used on a data set of approved drugs to identify potential antischistosomal agents targeting SmCB1. Pharmacophore (PHP) models underwent validation through receiver operating characteristics curves achieving values >0.8. The data highlighted riboflavin (RBF) as a compound of particular interest. A 1 μs molecular dynamics simulation demonstrated that RBF altered the conformation of SmCB1, causing the protease's binding site to close around RBF while maintaining the protease's overall integrity. RBF inhibited the activity of SmCB1 at low micromolar values and killed the parasite in vitro. Finally, in a murine model of S. mansoni infection, oral administration of 100 mg/kg RBF for 7 days significantly decreased worm burdens by ∼20% and had a major impact on intestinal and fecal egg burdens, which were decreased by ∼80%.
Collapse
Affiliation(s)
- Ramon M Cogo
- Universidade Federal de São Paulo-Campus Diadema, Curso de Pós-Graduação em Biologia Química da Unifesp, Rua São Nicolau 210, 2o andar, Centro, Diadema, São Paulo 09972-270, Brazil
| | - Thaís F A Pavani
- Universidade Federal de São Paulo-Campus Diadema, Curso de Pós-Graduação em Biologia Química da Unifesp, Rua São Nicolau 210, 2o andar, Centro, Diadema, São Paulo 09972-270, Brazil
| | - Ana C A Mengarda
- Universidade Guarulhos, Núcleo de Pesquisa em Doenças Negligenciadas-NPDN, Praça Tereza Cristina 88, Guarulhos 09972-270, Brazil
| | - Rayssa A Cajas
- Universidade Guarulhos, Núcleo de Pesquisa em Doenças Negligenciadas-NPDN, Praça Tereza Cristina 88, Guarulhos 09972-270, Brazil
| | - Thainá R Teixeira
- Universidade Guarulhos, Núcleo de Pesquisa em Doenças Negligenciadas-NPDN, Praça Tereza Cristina 88, Guarulhos 09972-270, Brazil
| | - Lucas Fukui-Silva
- Universidade Guarulhos, Núcleo de Pesquisa em Doenças Negligenciadas-NPDN, Praça Tereza Cristina 88, Guarulhos 09972-270, Brazil
| | - Yujie Uli Sun
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093-0021, United States
| | - Lawrence J Liu
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093-0021, United States
| | - Dilini K Amarasinghe
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093-0021, United States
| | - Michael C Yoon
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093-0021, United States
| | - Osvaldo A Santos-Filho
- Instituto de Pesquisas de Produtos Naturais Walter Mors, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco H, Rio de Janeiro 21941-853, Brazil
| | - Josué de Moraes
- Universidade Guarulhos, Núcleo de Pesquisa em Doenças Negligenciadas-NPDN, Praça Tereza Cristina 88, Guarulhos 09972-270, Brazil
| | - Conor R Caffrey
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093-0021, United States
| | - Daniela G G Rando
- Grupo de Pesquisas Químico-Farmacêuticas da Unifesp, Department of Pharmaceutical Sciences Rua São Nicolau, Universidade Federal de São Paulo-Campus Diadema, 210, 2o andar, Centro, Diadema, São Paulo 09972-270, Brazil
| |
Collapse
|
12
|
Daghighi A, Casanola-Martin GM, Iduoku K, Kusic H, González-Díaz H, Rasulev B. Multi-Endpoint Acute Toxicity Assessment of Organic Compounds Using Large-Scale Machine Learning Modeling. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:10116-10127. [PMID: 38797941 DOI: 10.1021/acs.est.4c01017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
In recent years, alternative animal testing methods such as computational and machine learning approaches have become increasingly crucial for toxicity testing. However, the complexity and scarcity of available biomedical data challenge the development of predictive models. Combining nonlinear machine learning together with multicondition descriptors offers a solution for using data from various assays to create a robust model. This work applies multicondition descriptors (MCDs) to develop a QSTR (Quantitative Structure-Toxicity Relationship) model based on a large toxicity data set comprising more than 80,000 compounds and 59 different end points (122,572 data points). The prediction capabilities of developed single-task multi-end point machine learning models as well as a novel data analysis approach with the use of Convolutional Neural Networks (CNN) are discussed. The results show that using MCDs significantly improves the model and using them with CNN-1D yields the best result (R2train = 0.93, R2ext = 0.70). Several structural features showed a high level of contribution to the toxicity, including van der Waals surface area (VSA), number of nitrogen-containing fragments (nN+), presence of S-P fragments, ionization potential, and presence of C-N fragments. The developed models can be very useful tools to predict the toxicity of various compounds under different conditions, enabling quick toxicity assessment of new compounds.
Collapse
Affiliation(s)
- Amirreza Daghighi
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58102, United States
- Biomedical Engineering Program, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Gerardo M Casanola-Martin
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Kweeni Iduoku
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58102, United States
- Biomedical Engineering Program, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Hrvoje Kusic
- Faculty of Chemical Engineering and Technology, University of Zagreb, Marulicev Trg 19, Zagreb 10000, Croatia
| | - Humberto González-Díaz
- Department of Organic and Inorganic Chemistry, University of Basque Country UPV/EHU, Leioa 48940, Spain
- BIOFISIKA, Basque Center for Biophysics CSIC-UPVEH, Leioa 48940, Spain
- IKERBASQUE, Basque Foundation for Science,Bilbao, Biscay 48011, Spain
| | - Bakhtiyor Rasulev
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58102, United States
- Biomedical Engineering Program, North Dakota State University, Fargo, North Dakota 58102, United States
| |
Collapse
|
13
|
Bessa CDPB, Feu AE, de Menezes RPB, Scotti MT, Lima JMG, Lima ML, Tempone AG, de Andrade JP, Bastida J, Borges WDS. Multitarget anti-parasitic activities of isoquinoline alkaloids isolated from Hippeastrum aulicum (Amaryllidaceae). PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155414. [PMID: 38503155 DOI: 10.1016/j.phymed.2024.155414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/02/2024] [Accepted: 02/03/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND Chagas disease and leishmaniasis affect a significant portion of the Latin American population and still lack efficient treatments. In this context, natural products emerge as promising compounds for developing more effective therapies, aiming to mitigate side effects and drug resistance. Notably, species from the Amaryllidaceae family emerge as potential reservoirs of antiparasitic agents due to the presence of diverse biologically active alkaloids. PURPOSE To assess the anti-Trypanosoma cruzi and anti-Leishmania infantum activity of five isolated alkaloids from Hippeastrum aulicum Herb. (Amaryllidaceae) against different life stages of the parasites using in silico and in vitro assays. Furthermore, molecular docking was employed to evaluate the interaction of the most active alkaloids. METHODS Five natural isoquinoline alkaloids isolated in suitable quantities for in vitro testing underwent preliminary in silico analysis to predict their potential efficacy against Trypanosoma cruzi (amastigote and trypomastigote forms) and Leishmania infantum (amastigote and promastigote forms). The in vitro antiparasitic activity and mammalian cytotoxicity were investigated with a subsequent comparison of both analysis (in silico and in vitro) findings. Additionally, this study employed the molecular docking technique, utilizing cruzain (T. cruzi) and sterol 14α-demethylase (CYP51, L. infantum) as crucial biological targets for parasite survival, specifically focusing on compounds that exhibited promising activities against both parasites. RESULTS Through computational techniques, it was identified that the alkaloids haemanthamine (1) and lycorine (8) were the most active against T. cruzi (amastigote and trypomastigote) and L. infantum (amastigote and promastigote), while also revealing unprecedented activity of alkaloid 7‑methoxy-O-methyllycorenine (6). The in vitro analysis confirmed the in silico tests, in which compound 1 presented the best activities against the promastigote and amastigote forms of L. infantum with half-maximal inhibitory concentration (IC50) 0.6 µM and 1.78 µM, respectively. Compound 8 exhibited significant activity against the amastigote form of T. cruzi (IC50 7.70 µM), and compound 6 demonstrated activity against the trypomastigote forms of T. cruzi and amastigote of L. infantum, with IC50 values of 89.55 and 86.12 µM, respectively. Molecular docking analyses indicated that alkaloids 1 and 8 exhibited superior interaction energies compared to the inhibitors. CONCLUSION The hitherto unreported potential of compound 6 against T. cruzi trypomastigotes and L. infantum amastigotes is now brought to the forefront. Furthermore, the acquired dataset signifies that the isolated alkaloids 1 and 8 from H. aulicum might serve as prototypes for subsequent structural refinements aimed at the exploration of novel leads against both T. cruzi and L. infantum parasites.
Collapse
Affiliation(s)
- Carliani Dal Piero Betzel Bessa
- Programa de Pós-Graduação em Química, Departamento de Química, Universidade Federal do Espírito Santo, Vitória-ES 29075-910, Brazil
| | - Amanda Eiriz Feu
- Programa de Pós-Graduação em Química, Departamento de Química, Universidade Federal do Espírito Santo, Vitória-ES 29075-910, Brazil
| | - Renata Priscila Barros de Menezes
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos (PgPNSB), Universidade Federal da Paraíba, Campus I, Cidade Universitária, João Pessoa 58051-900, Brazil
| | - Marcus Tullius Scotti
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos (PgPNSB), Universidade Federal da Paraíba, Campus I, Cidade Universitária, João Pessoa 58051-900, Brazil
| | | | - Marta Lopes Lima
- School of Life Sciences, University of Dundee, Scotland DD1 4HN, United Kingdom
| | | | - Jean Paulo de Andrade
- Departamento de Medicina Traslacional, Facultad de Medicina, Escuela de Química y Farmacia, Universidad Católica del Maule, Talca 3480112, Chile
| | - Jaume Bastida
- Departament de Biologia, Sanitat i Medi Ambient, Facultat de Farmàcia i Ciències de l´Alimentació, Universitat de Barcelona, Barcelona 08028, Spain
| | - Warley de Souza Borges
- Programa de Pós-Graduação em Química, Departamento de Química, Universidade Federal do Espírito Santo, Vitória-ES 29075-910, Brazil.
| |
Collapse
|
14
|
Melo-Filho CC, Su G, Liu K, Muratov EN, Tropsha A, Liu J. Modeling interactions between Heparan sulfate and proteins based on the Heparan sulfate microarray analysis. Glycobiology 2024; 34:cwae039. [PMID: 38836441 PMCID: PMC11180703 DOI: 10.1093/glycob/cwae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/30/2024] [Accepted: 05/29/2024] [Indexed: 06/06/2024] Open
Abstract
Heparan sulfate (HS), a sulfated polysaccharide abundant in the extracellular matrix, plays pivotal roles in various physiological and pathological processes by interacting with proteins. Investigating the binding selectivity of HS oligosaccharides to target proteins is essential, but the exhaustive inclusion of all possible oligosaccharides in microarray experiments is impractical. To address this challenge, we present a hybrid pipeline that integrates microarray and in silico techniques to design oligosaccharides with desired protein affinity. Using fibroblast growth factor 2 (FGF2) as a model protein, we assembled an in-house dataset of HS oligosaccharides on microarrays and developed two structural representations: a standard representation with all atoms explicit and a simplified representation with disaccharide units as "quasi-atoms." Predictive Quantitative Structure-Activity Relationship (QSAR) models for FGF2 affinity were developed using the Random Forest (RF) algorithm. The resulting models, considering the applicability domain, demonstrated high predictivity, with a correct classification rate of 0.81-0.80 and improved positive predictive values (PPV) up to 0.95. Virtual screening of 40 new oligosaccharides using the simplified model identified 15 computational hits, 11 of which were experimentally validated for high FGF2 affinity. This hybrid approach marks a significant step toward the targeted design of oligosaccharides with desired protein interactions, providing a foundation for broader applications in glycobiology.
Collapse
Affiliation(s)
- Cleber C Melo-Filho
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, 301 Beard Hall, University of North Carolina, Chapel Hill, NC 27599, United States
| | - Guowei Su
- Glycan Therapeutics, 617 Hutton Street, Raleigh, NC 27606, United States
| | - Kevin Liu
- Glycan Therapeutics, 617 Hutton Street, Raleigh, NC 27606, United States
| | - Eugene N Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, 301 Beard Hall, University of North Carolina, Chapel Hill, NC 27599, United States
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, 301 Beard Hall, University of North Carolina, Chapel Hill, NC 27599, United States
| | - Jian Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, 1044 Genetic Medicine Bldg., University of North Carolina, Chapel Hill, NC 27599, United States
| |
Collapse
|
15
|
Rath M, Wellnitz J, Martin HJ, Melo-Filho C, Hochuli JE, Silva GM, Beasley JM, Travis M, Sessions ZL, Popov KI, Zakharov AV, Cherkasov A, Alves V, Muratov EN, Tropsha A. Pharmacokinetics Profiler (PhaKinPro): Model Development, Validation, and Implementation as a Web Tool for Triaging Compounds with Undesired Pharmacokinetics Profiles. J Med Chem 2024; 67:6508-6518. [PMID: 38568752 DOI: 10.1021/acs.jmedchem.3c02446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Computational models that predict pharmacokinetic properties are critical to deprioritize drug candidates that emerge as hits in high-throughput screening campaigns. We collected, curated, and integrated a database of compounds tested in 12 major end points comprising over 10,000 unique molecules. We then employed these data to build and validate binary quantitative structure-activity relationship (QSAR) models. All trained models achieved a correct classification rate above 0.60 and a positive predictive value above 0.50. To illustrate their utility in drug discovery, we used these models to predict the pharmacokinetic properties for drugs in the NCATS Inxight Drugs database. In addition, we employed the developed models to predict the pharmacokinetic properties of all compounds in the DrugBank. All models described in this paper have been integrated and made publicly available via the PhaKinPro Web-portal that can be accessed at https://phakinpro.mml.unc.edu/.
Collapse
Affiliation(s)
- Marielle Rath
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - James Wellnitz
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Holli-Joi Martin
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Cleber Melo-Filho
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Joshua E Hochuli
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Guilherme Martins Silva
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Jon-Michael Beasley
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Maxfield Travis
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Zoe L Sessions
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Konstantin I Popov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Alexey V Zakharov
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia V6H3Z6, Canada
| | - Vinicius Alves
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Eugene N Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
16
|
Barbosa H, Espinoza GZ, Amaral M, de Castro Levatti EV, Abiuzi MB, Veríssimo GC, Fernandes PDO, Maltarollo VG, Tempone AG, Honorio KM, Lago JHG. Andrographolide: A Diterpenoid from Cymbopogon schoenanthus Identified as a New Hit Compound against Trypanosoma cruzi Using Machine Learning and Experimental Approaches. J Chem Inf Model 2024; 64:2565-2576. [PMID: 38148604 DOI: 10.1021/acs.jcim.3c01410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
American Trypanosomiasis, also known as Chagas disease, is caused by the protozoan Trypanosoma cruzi and exhibits limited options for treatment. Natural products offer various structurally complex metabolites with biological activities, including those with anti-T. cruzi potential. The discovery and development of prototypes based on natural products frequently display multiple phases that could be facilitated by machine learning techniques to provide a fast and efficient method for selecting new hit candidates. Using Random Forest and k-Nearest Neighbors, two models were constructed to predict the biological activity of natural products from plants against intracellular amastigotes of T. cruzi. The diterpenoid andrographolide was identified from a virtual screening as a promising hit compound. Hereafter, it was isolated from Cymbopogon schoenanthus and chemically characterized by spectral data analysis. Andrographolide was evaluated against trypomastigote and amastigote forms of T. cruzi, showing IC50 values of 29.4 and 2.9 μM, respectively, while the standard drug benznidazole displayed IC50 values of 17.7 and 5.0 μM, respectively. Additionally, the isolated compound exhibited a reduced cytotoxicity (CC50 = 92.8 μM) against mammalian cells and afforded a selectivity index (SI) of 32, similar to that of benznidazole (SI = 39). From the in silico analyses, we can conclude that andrographolide fulfills many requirements implemented by DNDi to be a hit compound. Therefore, this work successfully obtained machine learning models capable of predicting the activity of compounds against intracellular forms of T. cruzi.
Collapse
Affiliation(s)
- Henrique Barbosa
- Center for Natural and Human Sciences, Federal University of ABC, São Paulo 09210-180, Brazil
| | | | - Maiara Amaral
- Laboratory of Pathophysiology, Butantan Institute, São Paulo 05503-900, Brazil
| | | | | | - Gabriel Correa Veríssimo
- Department of Pharmaceutical Products, Federal University of Minas Gerais, Minas Gerais, 31270-901, Brazil
| | | | | | | | - Kathia Maria Honorio
- Center for Natural and Human Sciences, Federal University of ABC, São Paulo 09210-180, Brazil
- School of Arts, Science, and Humanities, University of São Paulo, São Paulo 03828-000, Brazil
| | | |
Collapse
|
17
|
Srisongkram T, Tookkane D. Insights into the structure-activity relationship of pyrimidine-sulfonamide analogues for targeting BRAF V600E protein. Biophys Chem 2024; 307:107179. [PMID: 38241826 DOI: 10.1016/j.bpc.2024.107179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024]
Abstract
B-rapidly accelerated fibrosarcoma (BRAF) V600E plays a crucial role in the progression of cutaneous melanoma. Core structures of BRAF V600E inhibitors are based on pyrimidine-sulfonamide scaffolds. Exploring the QSAR of these structures can improve our understanding of BRAF V600E inhibitor drug design. This study utilized machine learning-based QSAR to elucidate chemical substructures of pyrimidine-sulfonamide analogues that correlated to the BRAF V600E inhibitory activity. The findings indicate that the support vector regression (SVR) combined with 15 fingerprints achieved the highest statistical performances in terms of goodness-of-fit, robustness, and predictability. Nine key fingerprints from pyrimidine-sulfonamide analogues were identified to exert the BRAF V600E inhibitory activity. These key fingerprints were validated using network-based activity cliff landscape and molecular docking. Together, the developed algorithm can serve as a screening tool for designing BRAF V600E inhibitors. To further utilize this model, we deployed our developed algorithm at https://qsarlabs.com/#braf.
Collapse
Affiliation(s)
- Tarapong Srisongkram
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, 40002, Thailand.
| | - Dheerapat Tookkane
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, 40002, Thailand
| |
Collapse
|
18
|
Purificação A, Silva-Mendonça S, Cruz LV, Sacramento CQ, Temerozo JR, Fintelman-Rodrigues N, de Freitas CS, Godoi BF, Vaidergorn MM, Leite JA, Salazar Alvarez LC, Freitas MV, Silvac MFB, Martin BA, Lopez RFV, Neves BJ, Costa FTM, Souza TML, da Silva Emery F, Andrade CH, Nonato MC. Unveiling the Antiviral Capabilities of Targeting Human Dihydroorotate Dehydrogenase against SARS-CoV-2. ACS OMEGA 2024; 9:11418-11430. [PMID: 38496952 PMCID: PMC10938441 DOI: 10.1021/acsomega.3c07845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 01/18/2024] [Accepted: 02/05/2024] [Indexed: 03/19/2024]
Abstract
The urgent need for effective treatments against emerging viral diseases, driven by drug-resistant strains and new viral variants, remains critical. We focus on inhibiting the human dihydroorotate dehydrogenase (HsDHODH), one of the main enzymes responsible for pyrimidine nucleotide synthesis. This strategy could impede viral replication without provoking resistance. We evaluated naphthoquinone fragments, discovering potent HsDHODH inhibition with IC50 ranging from 48 to 684 nM, and promising in vitro anti-SARS-CoV-2 activity with EC50 ranging from 1.2 to 2.3 μM. These compounds exhibited low toxicity, indicating potential for further development. Additionally, we employed computational tools such as molecular docking and quantitative structure-activity relationship (QSAR) models to analyze protein-ligand interactions, revealing that these naphthoquinones exhibit a protein binding pattern similar to brequinar, a potent HsDHODH inhibitor. These findings represent a significant step forward in the search for effective antiviral treatments and have great potential to impact the development of new broad-spectrum antiviral drugs.
Collapse
Affiliation(s)
- Aline
D. Purificação
- Protein
Crystallography Laboratory, Department of Biomolecular Sciences, School
of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto 05508-060, SP, Brazil
- Center
for the Research and Advancement in Fragments and molecular Targets
(CRAFT), School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto 05508-060, SP, Brazil
| | - Sabrina Silva-Mendonça
- Center
for the Research and Advancement in Fragments and molecular Targets
(CRAFT), School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto 05508-060, SP, Brazil
- Laboratory
for Molecular Modeling and Drug Design (LabMol), Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia 74605-170, GO, Brazil
| | - Luiza V. Cruz
- Center
for the Research and Advancement in Fragments and molecular Targets
(CRAFT), School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto 05508-060, SP, Brazil
- Laboratory
for Molecular Modeling and Drug Design (LabMol), Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia 74605-170, GO, Brazil
| | - Carolina Q. Sacramento
- Laboratory
of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de
Janeiro 21040-900, RJ, Brazil
- National
Institute for Science and Technology on Innovation in Diseases of
Neglected Populations (INCT/IDPN), Center for Technological Development
in Health (CDTS), Fiocruz, Rio de Janeiro 21040-900, RJ, Brazil
| | - Jairo R. Temerozo
- Laboratory
of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de
Janeiro 21040-900, RJ, Brazil
- National
Institute for Science and Technology on Innovation in Diseases of
Neglected Populations (INCT/IDPN), Center for Technological Development
in Health (CDTS), Fiocruz, Rio de Janeiro 21040-900, RJ, Brazil
- National
Institute for Science and Technology on Neuroimmunomodulation, Oswaldo
Cruz Institute, Fiocruz, Rio de Janeiro 21040-900, RJ, Brazil
| | - Natalia Fintelman-Rodrigues
- Laboratory
of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de
Janeiro 21040-900, RJ, Brazil
- National
Institute for Science and Technology on Innovation in Diseases of
Neglected Populations (INCT/IDPN), Center for Technological Development
in Health (CDTS), Fiocruz, Rio de Janeiro 21040-900, RJ, Brazil
| | - Caroline Souza de Freitas
- Laboratory
of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de
Janeiro 21040-900, RJ, Brazil
- National
Institute for Science and Technology on Innovation in Diseases of
Neglected Populations (INCT/IDPN), Center for Technological Development
in Health (CDTS), Fiocruz, Rio de Janeiro 21040-900, RJ, Brazil
| | - Bruna Fleck Godoi
- Center
for the Research and Advancement in Fragments and molecular Targets
(CRAFT), School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto 05508-060, SP, Brazil
- Laboratory
of Heterocyclic and Medicinal Chemistry (QHeteM), Department of Pharmaceutical
Sciences, School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirao Preto 05508-060, SP, Brazil
| | - Miguel Menezes Vaidergorn
- Center
for the Research and Advancement in Fragments and molecular Targets
(CRAFT), School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto 05508-060, SP, Brazil
- Laboratory
of Heterocyclic and Medicinal Chemistry (QHeteM), Department of Pharmaceutical
Sciences, School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirao Preto 05508-060, SP, Brazil
| | - Juliana Almeida Leite
- Laboratory
of Tropical Diseases, Department of Genetics, Evolution, Microbiology
and Immunology, Institute of Biology, Unicamp, Campinas 13.083-857, SP, Brazil
| | - Luis Carlos Salazar Alvarez
- Laboratory
of Tropical Diseases, Department of Genetics, Evolution, Microbiology
and Immunology, Institute of Biology, Unicamp, Campinas 13.083-857, SP, Brazil
| | - Murillo V. Freitas
- Laboratory
for Molecular Modeling and Drug Design (LabMol), Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia 74605-170, GO, Brazil
| | - Meryck F. B. Silvac
- Laboratory
for Molecular Modeling and Drug Design (LabMol), Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia 74605-170, GO, Brazil
- Laboratory
of Cheminformatics, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia 74605-170, GO, Brazil
| | - Bianca A. Martin
- Innovation
Center in Nanostructured Systems and Topical Administration (NanoTop),
School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto 05508-060, SP, Brazil
| | - Renata F. V. Lopez
- Innovation
Center in Nanostructured Systems and Topical Administration (NanoTop),
School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto 05508-060, SP, Brazil
| | - Bruno J. Neves
- Laboratory
of Cheminformatics, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia 74605-170, GO, Brazil
| | - Fabio T. M. Costa
- Laboratory
of Tropical Diseases, Department of Genetics, Evolution, Microbiology
and Immunology, Institute of Biology, Unicamp, Campinas 13.083-857, SP, Brazil
| | - Thiago M. L. Souza
- Laboratory
of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de
Janeiro 21040-900, RJ, Brazil
- National
Institute for Science and Technology on Innovation in Diseases of
Neglected Populations (INCT/IDPN), Center for Technological Development
in Health (CDTS), Fiocruz, Rio de Janeiro 21040-900, RJ, Brazil
| | - Flavio da Silva Emery
- Center
for the Research and Advancement in Fragments and molecular Targets
(CRAFT), School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto 05508-060, SP, Brazil
- Laboratory
of Heterocyclic and Medicinal Chemistry (QHeteM), Department of Pharmaceutical
Sciences, School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirao Preto 05508-060, SP, Brazil
| | - Carolina Horta Andrade
- Center
for the Research and Advancement in Fragments and molecular Targets
(CRAFT), School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto 05508-060, SP, Brazil
- Laboratory
for Molecular Modeling and Drug Design (LabMol), Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia 74605-170, GO, Brazil
- Center
for Excellence in Artificial Intelligence (CEIA), Institute of Informatics, Universidade Federal de Goiás, Goiânia 74605-170, GO, Brazil
| | - M. Cristina Nonato
- Protein
Crystallography Laboratory, Department of Biomolecular Sciences, School
of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto 05508-060, SP, Brazil
- Center
for the Research and Advancement in Fragments and molecular Targets
(CRAFT), School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto 05508-060, SP, Brazil
| |
Collapse
|
19
|
Mansouri K, Moreira-Filho JT, Lowe CN, Charest N, Martin T, Tkachenko V, Judson R, Conway M, Kleinstreuer NC, Williams AJ. Free and open-source QSAR-ready workflow for automated standardization of chemical structures in support of QSAR modeling. J Cheminform 2024; 16:19. [PMID: 38378618 PMCID: PMC10880251 DOI: 10.1186/s13321-024-00814-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/10/2024] [Indexed: 02/22/2024] Open
Abstract
The rapid increase of publicly available chemical structures and associated experimental data presents a valuable opportunity to build robust QSAR models for applications in different fields. However, the common concern is the quality of both the chemical structure information and associated experimental data. This is especially true when those data are collected from multiple sources as chemical substance mappings can contain many duplicate structures and molecular inconsistencies. Such issues can impact the resulting molecular descriptors and their mappings to experimental data and, subsequently, the quality of the derived models in terms of accuracy, repeatability, and reliability. Herein we describe the development of an automated workflow to standardize chemical structures according to a set of standard rules and generate two and/or three-dimensional "QSAR-ready" forms prior to the calculation of molecular descriptors. The workflow was designed in the KNIME workflow environment and consists of three high-level steps. First, a structure encoding is read, and then the resulting in-memory representation is cross-referenced with any existing identifiers for consistency. Finally, the structure is standardized using a series of operations including desalting, stripping of stereochemistry (for two-dimensional structures), standardization of tautomers and nitro groups, valence correction, neutralization when possible, and then removal of duplicates. This workflow was initially developed to support collaborative modeling QSAR projects to ensure consistency of the results from the different participants. It was then updated and generalized for other modeling applications. This included modification of the "QSAR-ready" workflow to generate "MS-ready structures" to support the generation of substance mappings and searches for software applications related to non-targeted analysis mass spectrometry. Both QSAR and MS-ready workflows are freely available in KNIME, via standalone versions on GitHub, and as docker container resources for the scientific community. Scientific contribution: This work pioneers an automated workflow in KNIME, systematically standardizing chemical structures to ensure their readiness for QSAR modeling and broader scientific applications. By addressing data quality concerns through desalting, stereochemistry stripping, and normalization, it optimizes molecular descriptors' accuracy and reliability. The freely available resources in KNIME, GitHub, and docker containers democratize access, benefiting collaborative research and advancing diverse modeling endeavors in chemistry and mass spectrometry.
Collapse
Affiliation(s)
- Kamel Mansouri
- National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA.
| | - José T Moreira-Filho
- National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Charles N Lowe
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Nathaniel Charest
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Todd Martin
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | | | - Richard Judson
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Mike Conway
- National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Nicole C Kleinstreuer
- National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Antony J Williams
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| |
Collapse
|
20
|
Andrade C, Sousa BKDP, Sigurdardóttir S, Bourgard C, Borba J, Clementino L, Salazar-Alvarez LC, Groustra S, Zigweid R, Khim M, Staker B, Costa F, Eriksson L, Sunnerhagen P. Selective Bias Virtual Screening for Discovery of Promising Antimalarial Candidates targeting Plasmodium N-Myristoyltransferase. RESEARCH SQUARE 2024:rs.3.rs-3963523. [PMID: 38463971 PMCID: PMC10925453 DOI: 10.21203/rs.3.rs-3963523/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Malaria remains a significant public health challenge, with Plasmodium vivax being the species responsible for the most prevalent form of the disease. Given the limited therapeutic options available, the search for new antimalarials against P. vivax is urgent. This study aims to identify new inhibitors for P. vivax N-myristoyltransferase (PvNMT), an essential drug target against malaria. Through a validated virtual screening campaign, we prioritized 23 candidates for further testing. In the yeast NMT system, seven compounds exhibit a potential inhibitor phenotype. In vitro antimalarial phenotypic assays confirmed the activity of four candidates while demonstrating an absence of cytotoxicity. Enzymatic assays reveal LabMol-394 as the most promising inhibitor, displaying selectivity against the parasite and a strong correlation within the yeast system. Furthermore, molecular dynamics simulations shed some light into its binding mode. This study constitutes a substantial contribution to the exploration of a selective quinoline scaffold and provides valuable insights into the development of new antimalarial candidates.
Collapse
|
21
|
Tropsha A, Isayev O, Varnek A, Schneider G, Cherkasov A. Integrating QSAR modelling and deep learning in drug discovery: the emergence of deep QSAR. Nat Rev Drug Discov 2024; 23:141-155. [PMID: 38066301 DOI: 10.1038/s41573-023-00832-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2023] [Indexed: 02/08/2024]
Abstract
Quantitative structure-activity relationship (QSAR) modelling, an approach that was introduced 60 years ago, is widely used in computer-aided drug design. In recent years, progress in artificial intelligence techniques, such as deep learning, the rapid growth of databases of molecules for virtual screening and dramatic improvements in computational power have supported the emergence of a new field of QSAR applications that we term 'deep QSAR'. Marking a decade from the pioneering applications of deep QSAR to tasks involved in small-molecule drug discovery, we herein describe key advances in the field, including deep generative and reinforcement learning approaches in molecular design, deep learning models for synthetic planning and the application of deep QSAR models in structure-based virtual screening. We also reflect on the emergence of quantum computing, which promises to further accelerate deep QSAR applications and the need for open-source and democratized resources to support computer-aided drug design.
Collapse
Affiliation(s)
| | | | | | | | - Artem Cherkasov
- University of British Columbia, Vancouver, BC, Canada.
- Photonic Inc., Coquitlam, BC, Canada.
| |
Collapse
|
22
|
Das A, Mathur P, Agarwal SM. Machine Learning, Molecular Docking, and Dynamics-Based Computational Identification of Potential Inhibitors against Lung Cancer. ACS OMEGA 2024; 9:4528-4539. [PMID: 38313551 PMCID: PMC10831845 DOI: 10.1021/acsomega.3c07338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 02/06/2024]
Abstract
Lung cancer is the most prevalent cause of cancer deaths worldwide. However, its treatment faces a significant hurdle due to the development of resistance. Phytomolecules are an important source of new chemical entities due to their rich chemical diversity. Therefore, a machine learning (ML) model was developed to computationally identify potential inhibitors using a curated data set of 649 phytomolecules with inhibitory activity against lung cancer cell lines. Four distinct ML approaches, including k-nearest neighbor, random forest, support vector machine, and extreme gradient boosting, were used in conjugation with MACCS and Morgan2 fingerprints to generate the models. It was observed that the random forest model developed by using the MACCS fingerprint shows the best performance. To further explore the chemical space and feature importance, k-means clustering, t-SNE analysis, and mean decrease in impurity had been calculated. Simultaneously, ∼400 000 natural products (NPs) retrieved from the COCONUT database were filtered for pharmacokinetic properties and taken for a multistep screening using docking against epidermal growth factor receptor (EGFR) mutant, a therapeutic drug target of lung cancer. Thereafter, the best-performing random forest model was used to predict the antilung cancer potential of the NPs having binding affinity better than the cocrystal ligand. This allowed the identification of 205 potential inhibitors, wherein the molecules with an indolocarbazole scaffold were enriched in top-scoring molecules. The top three indolocarbazole molecules with the lowest binding energy were further evaluated through 100 ns molecular dynamics (MD) simulations, which suggested that these molecules are strong binders. Also, structural similarity analysis against known drugs revealed that these NPs are similar to staurosporine, which demonstrates potent and selective activity against EGFR mutants. Thereby, the consensus analysis employing ML, molecular docking, and dynamics revealed that the molecules having an indolocarbazole scaffold are the most promising NPs that can act as potential inhibitors against lung cancer.
Collapse
Affiliation(s)
- Agneesh
Pratim Das
- Bioinformatics
Division, ICMR-National Institute of Cancer
Prevention and Research, I-7, Sector-39, Noida 201301, Uttar Pradesh, India
- Amity
Institute of Biotechnology, Amity University Uttar Pradesh, Sector-125, Noida 201313, Uttar
Pradesh, India
| | - Puniti Mathur
- Amity
Institute of Biotechnology, Amity University Uttar Pradesh, Sector-125, Noida 201313, Uttar
Pradesh, India
| | - Subhash M. Agarwal
- Bioinformatics
Division, ICMR-National Institute of Cancer
Prevention and Research, I-7, Sector-39, Noida 201301, Uttar Pradesh, India
| |
Collapse
|
23
|
Siramshetty VB, Xu X, Shah P. Artificial Intelligence in ADME Property Prediction. Methods Mol Biol 2024; 2714:307-327. [PMID: 37676606 DOI: 10.1007/978-1-0716-3441-7_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Absorption, distribution, metabolism, excretion (ADME) are key properties of a small molecule that govern pharmacokinetic profiles and impact its efficacy and safety. Computational methods such as machine learning and artificial intelligence have gained significant interest in both academic and industrial settings to predict pharmacokinetic properties of small molecules. These methods are applied in drug discovery to optimize chemical libraries, prioritize hits from biological screens, and optimize ADME properties of lead molecules. In the recent years, the drug discovery community witnessed the use of a range of neural network architectures such as deep neural networks, recurrent neural networks, graph neural networks, and transformer neural networks, which marked a paradigm shift in computer-aided drug design and development. This chapter discusses recent developments with an emphasis on their application to predict ADME properties.
Collapse
Affiliation(s)
- Vishal B Siramshetty
- National Center for Advancing Translational Sciences, Rockville, MD, USA
- Department of Safety Assessment, Genentech, Inc., South San Francisco, CA, USA
| | - Xin Xu
- National Center for Advancing Translational Sciences, Rockville, MD, USA
| | - Pranav Shah
- National Center for Advancing Translational Sciences, Rockville, MD, USA.
| |
Collapse
|
24
|
Srisongkram T. Ensemble Quantitative Read-Across Structure-Activity Relationship Algorithm for Predicting Skin Cytotoxicity. Chem Res Toxicol 2023; 36:1961-1972. [PMID: 38047785 DOI: 10.1021/acs.chemrestox.3c00238] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Read-across (RA) and quantitative structure-activity relationship (QSAR) are two alternative methods commonly used to fill data gaps in chemical registrations. These approaches use physicochemical properties or molecular fingerprints of source substances to predict the properties of unknown substances that have similar chemical structures or physicochemical properties. Research on RA and QSAR is essential to minimize the time, money, and animal testing needed to determine biological properties that are not currently known. This study developed a stacked ensemble quantitative read-across structure-activity relationship algorithm (enQRASAR) for predicting skin irritation toxicity based on negative log cell viability inhibition concentration at 50% (pIC50) against skin keratinocytes as the end point. The goodness-of-fit and predictability of this algorithm were validated using leave-one-out cross-validation and external test data sets. The results obtained were statistically reliable in terms of goodness-of-fit, robustness, and predictability metrics. Additionally, the developed model demonstrated a low prediction error when predicting FDA-approved drugs. These results confirm that the enQRASAR algorithm can be used to predict skin cytotoxicity of chemicals. Therefore, this model was publicly available to further facilitate toxicity predictions of unknown compounds in chemical registrations.
Collapse
Affiliation(s)
- Tarapong Srisongkram
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40000, Thailand
| |
Collapse
|
25
|
Jia X, Wang T, Zhu H. Advancing Computational Toxicology by Interpretable Machine Learning. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:17690-17706. [PMID: 37224004 PMCID: PMC10666545 DOI: 10.1021/acs.est.3c00653] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/05/2023] [Accepted: 05/05/2023] [Indexed: 05/26/2023]
Abstract
Chemical toxicity evaluations for drugs, consumer products, and environmental chemicals have a critical impact on human health. Traditional animal models to evaluate chemical toxicity are expensive, time-consuming, and often fail to detect toxicants in humans. Computational toxicology is a promising alternative approach that utilizes machine learning (ML) and deep learning (DL) techniques to predict the toxicity potentials of chemicals. Although the applications of ML- and DL-based computational models in chemical toxicity predictions are attractive, many toxicity models are "black boxes" in nature and difficult to interpret by toxicologists, which hampers the chemical risk assessments using these models. The recent progress of interpretable ML (IML) in the computer science field meets this urgent need to unveil the underlying toxicity mechanisms and elucidate the domain knowledge of toxicity models. In this review, we focused on the applications of IML in computational toxicology, including toxicity feature data, model interpretation methods, use of knowledge base frameworks in IML development, and recent applications. The challenges and future directions of IML modeling in toxicology are also discussed. We hope this review can encourage efforts in developing interpretable models with new IML algorithms that can assist new chemical assessments by illustrating toxicity mechanisms in humans.
Collapse
Affiliation(s)
- Xuelian Jia
- Department
of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey 08028, United States
| | - Tong Wang
- Department
of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey 08028, United States
| | - Hao Zhu
- Department
of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey 08028, United States
| |
Collapse
|
26
|
Moreira-Filho JT, Neves BJ, Cajas RA, Moraes JD, Andrade CH. Artificial intelligence-guided approach for efficient virtual screening of hits against Schistosoma mansoni. Future Med Chem 2023; 15:2033-2050. [PMID: 37937522 DOI: 10.4155/fmc-2023-0152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/06/2023] [Indexed: 11/09/2023] Open
Abstract
Background: The impact of schistosomiasis, which affects over 230 million people, emphasizes the urgency of developing new antischistosomal drugs. Artificial intelligence is vital in accelerating the drug discovery process. Methodology & results: We developed classification and regression machine learning models to predict the schistosomicidal activity of compounds not experimentally tested. The prioritized compounds were tested on schistosomula and adult stages of Schistosoma mansoni. Four compounds demonstrated significant activity against schistosomula, with 50% effective concentration values ranging from 9.8 to 32.5 μM, while exhibiting no toxicity in animal and human cell lines. Conclusion: These findings represent a significant step forward in the discovery of antischistosomal drugs. Further optimization of these active compounds can pave the way for their progression into preclinical studies.
Collapse
Affiliation(s)
- José Teófilo Moreira-Filho
- Laboratory of Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, 74605-170, Brazil
| | - Bruno Junior Neves
- Laboratory of Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, 74605-170, Brazil
| | - Rayssa Araujo Cajas
- Research Center on Neglected Diseases (NPDN), Universidade Guarulhos, Guarulhos, 07023-070, Brazil
| | - Josué de Moraes
- Research Center on Neglected Diseases (NPDN), Universidade Guarulhos, Guarulhos, 07023-070, Brazil
| | - Carolina Horta Andrade
- Laboratory of Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, 74605-170, Brazil
- Center for the Research and Advancement in Fragments and molecular Targets (CRAFT), School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
27
|
Júnior MA, Silva LC, Rocha OB, Oliveira AA, Portis IG, Alonso A, Alonso L, Silva KS, Gomes MN, Andrade CH, Soares CM, Pereira M. Proteomic identification of metabolic changes in Paracoccidioides brasiliensis induced by a nitroheteroarylchalcone. Future Microbiol 2023; 18:1077-1093. [PMID: 37424510 DOI: 10.2217/fmb-2022-0150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023] Open
Abstract
Aim: To access the metabolic changes caused by a chalcone derivative (LabMol-75) through a proteomic approach. Methods: Proteomic analysis was performed after 9 h of Paracoccidioides brasiliensis yeast (Pb18) cell incubation with the LabMol-75 at MIC. The proteomic findings were validated through in vitro and in silico assays. Results: Exposure to the compound led to the downregulation of proteins associated with glycolysis and gluconeogenesis, β-oxidation, the citrate cycle and the electron transport chain. Conclusion: LabMol-75 caused an energetic imbalance in the fungus metabolism and deep oxidative stress. Additionally, the in silico molecular docking approach pointed to this molecule as a putative competitive inhibitor of DHPS.
Collapse
Affiliation(s)
- Marcos Abc Júnior
- Laboratory of Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Lívia C Silva
- Laboratory of Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Olivia B Rocha
- Laboratory of Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Amanda A Oliveira
- Laboratory of Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Igor G Portis
- Laboratory of Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Antonio Alonso
- Institute of Physics, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Lais Alonso
- Institute of Physics, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Kleber Sf Silva
- Laboratory of Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Marcelo N Gomes
- InsiChem, Goiás State University, Anápolis, Goiás, Brazil
- Faculdade Metropolitana de Anápolis, Anápolis, Goiás, Brazil
| | - Carolina H Andrade
- Laboratory for Molecular Modeling & Drug Design, Faculty of Pharmacy, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Célia Ma Soares
- Laboratory of Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Maristela Pereira
- Laboratory of Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, Brazil
| |
Collapse
|
28
|
Srisongkram T, Syahid NF, Tookkane D, Weerapreeyakul N, Puthongking P. Stacked ensemble learning on HaCaT cytotoxicity for skin irritation prediction: A case study on dipterocarpol. Food Chem Toxicol 2023; 181:114115. [PMID: 37863382 DOI: 10.1016/j.fct.2023.114115] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/26/2023] [Accepted: 10/17/2023] [Indexed: 10/22/2023]
Abstract
Skin irritation is an adverse effect associated with various substances, including chemicals, drugs, or natural products. Dipterocarpol, extracted from Dipterocarpus alatus, contains several skin benefits notably anticancer, wound healing, and antibacterial properties. However, the skin irritation of dipterocarpol remains unassessed. Quantitative structure-activity relationship (QSAR) is a recommended tool for toxicity assessment involving less time, money, and animal testing to access unavailable acute toxicity data. Therefore, our study aimed to develop a highly accurate machine learning-based QSAR model for predicting skin irritation. We utilized a stacked ensemble learning model with 1064 chemicals. We also adhered to the recommendations from the OECD for QSAR validation. Subsequently, we used the proposed model to explore the cytotoxicity of dipterocarpol on keratinocytes. Our findings indicate that the model displayed promising statistical quality in terms of accuracy, precision, and recall in both 10-fold cross-validation and test datasets. Moreover, the model predicted that dipterocarpol does not have skin irritation, which was confirmed by the cell-based assay. In conclusion, our proposed model can be applied for the risk assessment of skin irritation in untested compounds that fall within its applicability domain. The web application of this model is available at https://qsarlabs.com/#stackhacat.
Collapse
Affiliation(s)
- Tarapong Srisongkram
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, 40002, Thailand; Human High Performance and Health Promotion Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand.
| | - Nur Fadhilah Syahid
- Graduate School in the Program of Pharmaceutical Chemistry and Natural Products, Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Khon Kaen University, 40002, Thailand
| | - Dheerapat Tookkane
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, 40002, Thailand
| | - Natthida Weerapreeyakul
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, 40002, Thailand; Human High Performance and Health Promotion Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Ploenthip Puthongking
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, 40002, Thailand
| |
Collapse
|
29
|
Alves VM, Yasgar A, Wellnitz J, Rai G, Rath M, Braga RC, Capuzzi SJ, Simeonov A, Muratov EN, Zakharov AV, Tropsha A. Lies and Liabilities: Computational Assessment of High-Throughput Screening Hits to Identify Artifact Compounds. J Med Chem 2023; 66:12828-12839. [PMID: 37677128 DOI: 10.1021/acs.jmedchem.3c00482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Hits from high-throughput screening (HTS) of chemical libraries are often false positives due to their interference with assay detection technology. In response, we generated the largest publicly available library of chemical liabilities and developed "Liability Predictor," a free web tool to predict HTS artifacts. More specifically, we generated, curated, and integrated HTS data sets for thiol reactivity, redox activity, and luciferase (firefly and nano) activity and developed and validated quantitative structure-interference relationship (QSIR) models to predict these nuisance behaviors. The resulting models showed 58-78% external balanced accuracy for 256 external compounds per assay. QSIR models developed and validated herein identify nuisance compounds among experimental hits more reliably than do popular PAINS filters. Both the models and the curated data sets were implemented in "Liability Predictor," publicly available at https://liability.mml.unc.edu/. "Liability Predictor" may be used as part of chemical library design or for triaging HTS hits.
Collapse
Affiliation(s)
- Vinicius M Alves
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Adam Yasgar
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - James Wellnitz
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Ganesha Rai
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Marielle Rath
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | | | - Stephen J Capuzzi
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Anton Simeonov
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Eugene N Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
- Department of Pharmaceutical Sciences, Federal University of Paraiba, Joao Pessoa, PB 58059, Brazil
| | - Alexey V Zakharov
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
30
|
Martin HJ, Melo-Filho CC, Korn D, Eastman RT, Rai G, Simeonov A, Zakharov AV, Muratov E, Tropsha A. Small molecule antiviral compound collection (SMACC): A comprehensive, highly curated database to support the discovery of broad-spectrum antiviral drug molecules. Antiviral Res 2023; 217:105620. [PMID: 37169224 PMCID: PMC11069349 DOI: 10.1016/j.antiviral.2023.105620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/13/2023] [Accepted: 04/27/2023] [Indexed: 05/13/2023]
Abstract
Diseases caused by new viruses cost thousands if not millions of human lives and trillions of dollars. We have identified, collected, curated, and integrated all chemogenomics data from ChEMBL for 13 emerging viruses that hold the greatest potential threat to global human health. By identifying and solving several challenges related to data annotation accuracy, we developed a highly curated and thoroughly annotated database of compounds tested in both phenotypic and target-based assays for these viruses that we dubbed SMACC (Small Molecule Antiviral Compound Collection). The pilot version of the SMACC database contains over 32,500 entries for 13 viruses. By analyzing data in SMACC, we have identified ∼50 compounds with polyviral inhibition profile, mostly covering flavi- and coronaviruses. The SMACC database may serve as a reference for virologists and medicinal chemists working on the development of novel BSA agents in preparation for future viral outbreaks. SMACC is publicly available at https://smacc.mml.unc.edu.
Collapse
Affiliation(s)
- Holli-Joi Martin
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Cleber C Melo-Filho
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Daniel Korn
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Richard T Eastman
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, 20850, USA
| | - Ganesha Rai
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, 20850, USA
| | - Anton Simeonov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, 20850, USA
| | - Alexey V Zakharov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, 20850, USA.
| | - Eugene Muratov
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| | - Alexander Tropsha
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
31
|
de Oliveira AA, Carmo Silva LD, Neves BJ, Fiaia Costa VA, Muratov EN, Andrade CH, de Almeida Soares CM, Alves VM, Pereira M. Cheminformatics-driven discovery of hit compounds against Paracoccidioides spp. Future Med Chem 2023; 15:1553-1567. [PMID: 37727967 DOI: 10.4155/fmc-2022-0288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023] Open
Abstract
Aims: The development of safe and effective therapies for treating paracoccidioidomycosis using computational strategies were employed to discover anti-Paracoccidioides compounds. Materials & methods: We 1) collected, curated and integrated the largest library of compounds tested against Paracoccidioides spp.; 2) employed a similarity search to virtually screen the ChemBridge database and select nine compounds for experimental evaluation; 3) performed an experimental evaluation to determine the minimum inhibitory concentration and minimum fungicidal concentration as well as cytotoxicity; and 4) employed computational tools to identify potential targets for the most active compounds. Seven compounds presented activity against Paracoccidioides spp. Conclusion: These compounds are new hits with a predicted mechanisms of action, making them potentially attractive to develop new compounds.
Collapse
Affiliation(s)
- Amanda Alves de Oliveira
- Institute of Tropical Pathology & Public Health, Federal University of Goiás, Goiânia, 74690-900, Brazil
- Laboratory for Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, 74690-900, Brazil
| | - Lívia do Carmo Silva
- Laboratory for Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, 74690-900, Brazil
| | - Bruno Junior Neves
- Laboratory of Cheminformatics, Faculty of Pharmacy, Federal University of Goiás, 74690-900, Brazil
| | | | - Eugene N Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology & Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
- Department of Pharmaceutical Sciences, Federal University of Paraiba, Joao Pessoa, 58051-900, Brazil
| | - Carolina Horta Andrade
- Laboratory for Molecular Modeling & Design, Faculty of Pharmacy, Federal University of Goiás, 74690-900, Brazil
| | | | - Vinicius M Alves
- Laboratory for Molecular Modeling, Division of Chemical Biology & Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
- Laboratory for Molecular Modeling & Design, Faculty of Pharmacy, Federal University of Goiás, 74690-900, Brazil
| | - Maristela Pereira
- Laboratory for Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, 74690-900, Brazil
| |
Collapse
|
32
|
Mohiuddin A, Mondal S. Advancement of Computational Design Drug Delivery System in COVID-19: Current Updates and Future Crosstalk- A Critical update. Infect Disord Drug Targets 2023; 23:IDDT-EPUB-133706. [PMID: 37584349 PMCID: PMC11348471 DOI: 10.2174/1871526523666230816151614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/22/2023] [Accepted: 07/17/2023] [Indexed: 08/17/2023]
Abstract
Positive strides have been achieved in developing vaccines to combat the coronavirus-2019 infection (COVID-19) pandemic. Still, the outline of variations, particularly the most current delta divergent, has posed significant health encounters for people. Therefore, developing strong treatment strategies, such as an anti-COVID-19 medicine plan, may help deal with the pandemic more effectively. During the COVID-19 pandemic, some drug design techniques were effectively used to develop and substantiate relevant critical medications. Extensive research, both experimental and computational, has been dedicated to comprehending and characterizing the devastating COVID-19 disease. The urgency of the situation has led to the publication of over 130,000 COVID-19-related research papers in peer-reviewed journals and preprint servers. A significant focus of these efforts has been the identification of novel drug candidates and the repurposing of existing drugs to combat the virus. Many projects have utilized computational or computer-aided approaches to facilitate their studies. In this overview, we will explore the key computational methods and their applications in the discovery of small-molecule therapeutics for COVID-19, as reported in the research literature. We believe that the true effectiveness of computational tools lies in their ability to provide actionable and experimentally testable hypotheses, which in turn facilitate the discovery of new drugs and combinations thereof. Additionally, we recognize that open science and the rapid sharing of research findings are vital in expediting the development of much-needed therapeutics for COVID-19.
Collapse
Affiliation(s)
- Abu Mohiuddin
- Department of Pharmaceutical Science, GITAM School of Pharmacy, GITAM (Deemed to be University), Visakhapatnam-530045, A.P., India
| | - Sumanta Mondal
- Department of Pharmaceutical Science, GITAM School of Pharmacy, GITAM (Deemed to be University), Visakhapatnam-530045, A.P., India
| |
Collapse
|
33
|
Lemos JM, Brito da Silva MF, Dos Santos Carvalho AM, Vicente Gil HP, Fiaia Costa VA, Andrade CH, Braga RC, Grellier P, Muratov EN, Charneau S, Moreira-Filho JT, Dourado Bastos IM, Neves BJ. Multitask learning-driven identification of novel antitrypanosomal compounds. Future Med Chem 2023; 15:1449-1467. [PMID: 37701989 DOI: 10.4155/fmc-2023-0074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023] Open
Abstract
Background: Chagas disease and human African trypanosomiasis cause substantial death and morbidity, particularly in low- and middle-income countries, making the need for novel drugs urgent. Methodology & results: Therefore, an explainable multitask pipeline to profile the activity of compounds against three trypanosomes (Trypanosoma brucei brucei, Trypanosoma brucei rhodesiense and Trypanosoma cruzi) were created. These models successfully discovered four new experimental hits (LC-3, LC-4, LC-6 and LC-15). Among them, LC-6 showed promising results, with IC50 values ranging 0.01-0.072 μM and selectivity indices >10,000. Conclusion: These results demonstrate that the multitask protocol offers predictivity and interpretability in the virtual screening of new antitrypanosomal compounds and has the potential to improve hit rates in Chagas and human African trypanosomiasis projects.
Collapse
Affiliation(s)
- Jade Milhomem Lemos
- LabChem - Laboratory of Cheminformatics, Faculty of Pharmacy, Federal University of Goiás, Goiânia,74605-170, GO, Brazil
| | - Meryck Felipe Brito da Silva
- LabChem - Laboratory of Cheminformatics, Faculty of Pharmacy, Federal University of Goiás, Goiânia,74605-170, GO, Brazil
| | - Alexandra Maria Dos Santos Carvalho
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia, 70910-900, DF, Brazil
| | - Henric Pietro Vicente Gil
- LabChem - Laboratory of Cheminformatics, Faculty of Pharmacy, Federal University of Goiás, Goiânia,74605-170, GO, Brazil
| | - Vinícius Alexandre Fiaia Costa
- LabChem - Laboratory of Cheminformatics, Faculty of Pharmacy, Federal University of Goiás, Goiânia,74605-170, GO, Brazil
| | - Carolina Horta Andrade
- LabMol - Laboratory for Molecular Modeling & Drug Design, Faculty of Pharmacy, Federal University of Goiás, Goiânia, 74605-170, GO, Brazil
| | - Rodolpho Campos Braga
- InsilicAll Ltda, Av. Eng. Luis Carlos Berrini,1748 - Itaim Bibi, 04571-010, Sao Paulo, SP, Brazil
| | - Philippe Grellier
- UMR 7245 Molécules de Communication et Adaptation des Micro-organismes, Muséum National d'Histoire Naturelle, Équipe Parasites et Protistes Libres, Paris, 0575231, France
| | - Eugene N Muratov
- Laboratory for Molecular Modeling, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA
- Department of Pharmaceutical Sciences, Federal University of Paraiba, Joao Pessoa, 58059-900, PB, Brazil
| | - Sébastien Charneau
- Department of Cell Biology, Laboratory of Biochemistry & Protein Chemistry, Institute of Biological Sciences, University of Brasilia, Brasilia, 70910-900, DF, Brazil
| | - José Teófilo Moreira-Filho
- LabMol - Laboratory for Molecular Modeling & Drug Design, Faculty of Pharmacy, Federal University of Goiás, Goiânia, 74605-170, GO, Brazil
| | - Izabela Marques Dourado Bastos
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia, 70910-900, DF, Brazil
| | - Bruno Junior Neves
- LabChem - Laboratory of Cheminformatics, Faculty of Pharmacy, Federal University of Goiás, Goiânia,74605-170, GO, Brazil
| |
Collapse
|
34
|
Bhat-Ambure J, Ambure P, Serrano-Candelas E, Galiana-Roselló C, Gil-Martínez A, Guerrero M, Martin M, González-García J, García-España E, Gozalbes R. G4-QuadScreen: A Computational Tool for Identifying Multi-Target-Directed Anticancer Leads against G-Quadruplex DNA. Cancers (Basel) 2023; 15:3817. [PMID: 37568632 PMCID: PMC10416877 DOI: 10.3390/cancers15153817] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/16/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
The study presents 'G4-QuadScreen', a user-friendly computational tool for identifying MTDLs against G4s. Also, it offers a few hit MTDLs based on in silico and in vitro approaches. Multi-tasking QSAR models were developed using linear discriminant analysis and random forest machine learning techniques for predicting the responses of interest (G4 interaction, G4 stabilization, G4 selectivity, and cytotoxicity) considering the variations in the experimental conditions (e.g., G4 sequences, endpoints, cell lines, buffers, and assays). A virtual screening with G4-QuadScreen and molecular docking using YASARA (AutoDock-Vina) was performed. G4 activities were confirmed via FRET melting, FID, and cell viability assays. Validation metrics demonstrated the high discriminatory power and robustness of the models (the accuracy of all models is ~>90% for the training sets and ~>80% for the external sets). The experimental evaluations showed that ten screened MTDLs have the capacity to selectively stabilize multiple G4s. Three screened MTDLs induced a strong inhibitory effect on various human cancer cell lines. This pioneering computational study serves a tool to accelerate the search for new leads against G4s, reducing false positive outcomes in the early stages of drug discovery. The G4-QuadScreen tool is accessible on the ChemoPredictionSuite website.
Collapse
Affiliation(s)
| | - Pravin Ambure
- ProtoQSAR SL, Centro Europeo de Empresas Innovadoras (CEEI), Parque Tecnológico de Valencia, 46980 Valencia, Spain; (P.A.); (E.S.-C.)
| | - Eva Serrano-Candelas
- ProtoQSAR SL, Centro Europeo de Empresas Innovadoras (CEEI), Parque Tecnológico de Valencia, 46980 Valencia, Spain; (P.A.); (E.S.-C.)
| | - Cristina Galiana-Roselló
- Department of Inorganic Chemistry, Institute of Molecular Science, University of Valencia, 46980 Valencia, Spain; (C.G.-R.); (A.G.-M.); (J.G.-G.); (E.G.-E.)
| | - Ariadna Gil-Martínez
- Department of Inorganic Chemistry, Institute of Molecular Science, University of Valencia, 46980 Valencia, Spain; (C.G.-R.); (A.G.-M.); (J.G.-G.); (E.G.-E.)
| | - Mario Guerrero
- Biochemistry and Molecular Biology Unit, Biomedicine Department, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (M.G.); (M.M.)
| | - Margarita Martin
- Biochemistry and Molecular Biology Unit, Biomedicine Department, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (M.G.); (M.M.)
- Clinical and Experimental Respiratory Immunoallergy (IRCE), Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Jorge González-García
- Department of Inorganic Chemistry, Institute of Molecular Science, University of Valencia, 46980 Valencia, Spain; (C.G.-R.); (A.G.-M.); (J.G.-G.); (E.G.-E.)
| | - Enrique García-España
- Department of Inorganic Chemistry, Institute of Molecular Science, University of Valencia, 46980 Valencia, Spain; (C.G.-R.); (A.G.-M.); (J.G.-G.); (E.G.-E.)
| | - Rafael Gozalbes
- MolDrug AI Systems SL, c/Olimpia Arozena Torres, 46018 Valencia, Spain;
- ProtoQSAR SL, Centro Europeo de Empresas Innovadoras (CEEI), Parque Tecnológico de Valencia, 46980 Valencia, Spain; (P.A.); (E.S.-C.)
| |
Collapse
|
35
|
Syahid NF, Weerapreeyakul N, Srisongkram T. StackBRAF: A Large-Scale Stacking Ensemble Learning for BRAF Affinity Prediction. ACS OMEGA 2023; 8:20881-20891. [PMID: 37332807 PMCID: PMC10268632 DOI: 10.1021/acsomega.3c01641] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023]
Abstract
The B-rapidly accelerated fibrosarcoma (BRAF) is a proto-oncogene that plays a vital role in cell signaling and growth regulation. Identifying a potent BRAF inhibitor can enhance therapeutic success in high-stage cancers, particularly metastatic melanoma. In this study, we proposed a stacking ensemble learning framework for the accurate prediction of BRAF inhibitors. We obtained 3857 curated molecules with BRAF inhibitory activity expressed as a predicted half-maximal inhibitory concentration value (pIC50) from the ChEMBL database. Twelve molecular fingerprints from PaDeL-Descriptor were calculated for model training. Three machine learning algorithms including extreme gradient boosting, support vector regression, and multilayer perceptron were utilized for constructing new predictive features (PFs). The meta-ensemble random forest regression, called StackBRAF, was created based on the 36 PFs. The StackBRAF model achieves lower mean absolute error (MAE) and higher coefficient of determination (R2 and Q2) than the individual baseline models. The stacking ensemble learning model provides good y-randomization results, indicating a strong correlation between molecular features and pIC50. An applicability domain of the model with an acceptable Tanimoto similarity score was also defined. Moreover, a large-scale high-throughput screening of 2123 FDA-approved drugs against the BRAF protein was successfully demonstrated using the StackBRAF algorithm. Thus, the StackBRAF model proved beneficial as a drug design algorithm for BRAF inhibitor drug discovery and drug development.
Collapse
Affiliation(s)
- Nur Fadhilah Syahid
- Graduate
School in the Program of Pharmaceutical Chemistry and Natural Products,
Faculty of Pharmaceutical Sciences, Khon
Kaen University, Khon Kaen 40002, Thailand
| | - Natthida Weerapreeyakul
- Division
of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
- Human
High Performance and Health Promotion Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Tarapong Srisongkram
- Division
of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
- Human
High Performance and Health Promotion Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
36
|
Luna IS, Souza TAD, da Silva MS, Franca Rodrigues KAD, Scotti L, Scotti MT, Mendonça-Junior FJB. Computer-Aided drug design of new 2-amino-thiophene derivatives as anti-leishmanial agents. Eur J Med Chem 2023; 250:115223. [PMID: 36848847 DOI: 10.1016/j.ejmech.2023.115223] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 02/23/2023]
Abstract
The leishmaniasis is a neglected disease caused by a group of protozoan parasites from the genus Leishmania whose treatment is limited, obsolete, toxic, and ineffective in certain cases. These characteristics motivate researchers worldwide to plan new therapeutic alternatives for the treatment of leishmaniasis, where the use of cheminformatics tools applied to computer-assisted drug design has allowed research to make great advances in the search for new drugs candidates. In this study, a series of 2-amino-thiophene (2-AT) derivatives was screened virtually using QSAR tools, ADMET filters and prediction models, allowing direct the synthesis of compounds, which were evaluated in vitro against promastigotes and axenic amastigotes of Leishmania amazonensis. The combination of different descriptors and machine learning methods led to obtaining robust and predictive QSAR models, which was obtained from a dataset composed of 1862 compounds extracted from the ChEMBL database, with correct classification rates ranging from 0.53 (for amastigotes) to 0.91 (for promastigotes), allowing to select eleven 2-AT derivatives, which do not violate Lipinski's rules, exhibit good druglikeness, and with probability ≤70% of potential activity against the two evolutionary forms of the parasite. All compounds were properly synthesized and 8 of them were shown to be active at least against one of the evolutionary forms of the parasite with IC50 values lower than 10 μM, being more active than the reference drug meglumine antimoniate, and showing low or no citotoxicity against macrophage J774.A1 for the most part. Compounds 8CN and DCN-83, respectively, are the most active against promastigote and amastigote forms, with IC50 values of 1.20 and 0.71 μM, and selectivity indexes (SI) of 36.58 and 119.33. Structure Activity Relationship (SAR) study was carried out and allowed to identify some favorable and/or essential substitution patterns for the leishmanial activity of 2-AT derivatives. Taken together, these findings demonstrate that the use of ligand-based virtual screening proved to be quite effective and saved time, effort, and money in the selection of potential anti-leishmanial agents, and confirm, once again that 2-AT derivatives are promising hit compounds for the development of new anti-leishmanial agents.
Collapse
Affiliation(s)
- Isadora Silva Luna
- Laboratory of Synthesis and Drug Delivery, State University of Paraiba, João Pessoa, PB, Brazil; Post-Graduation Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, João Pessoa, PB, Brazil
| | - Thalisson Amorim de Souza
- Multiuser Laboratory Center of Characterization and Analysis, Federal University of Paraiba, João Pessoa, PB, Brazil
| | - Marcelo Sobral da Silva
- Multiuser Laboratory Center of Characterization and Analysis, Federal University of Paraiba, João Pessoa, PB, Brazil
| | | | - Luciana Scotti
- Post-Graduation Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, João Pessoa, PB, Brazil
| | - Marcus Tullius Scotti
- Post-Graduation Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, João Pessoa, PB, Brazil
| | - Francisco Jaime Bezerra Mendonça-Junior
- Laboratory of Synthesis and Drug Delivery, State University of Paraiba, João Pessoa, PB, Brazil; Post-Graduation Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, João Pessoa, PB, Brazil.
| |
Collapse
|
37
|
de Sousa NF, da Silva Souza HD, de Menezes RPB, da Silva Alves F, Acevedo CAH, de Lima Nunes TA, Sessions ZL, Scotti L, Muratov EN, Mendonça-Junior FJB, da Franca Rodrigues KA, de Athayde Filho PF, Scotti MT. Selene-Ethylenelacticamides and N-Aryl-Propanamides as Broad-Spectrum Leishmanicidal Agents. Pathogens 2023; 12:136. [PMID: 36678484 PMCID: PMC9860784 DOI: 10.3390/pathogens12010136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/20/2022] [Accepted: 12/26/2022] [Indexed: 01/18/2023] Open
Abstract
The World Health Organization classifies Leishmania as one of the 17 “neglected diseases” that burden tropical and sub-tropical climate regions with over half a million diagnosed cases each year. Despite this, currently available anti-leishmania drugs have high toxicity and the potential to be made obsolete by parasite drug resistance. We chose to analyze organoselenides for leishmanicidal potential given the reduced toxicity inherent to selenium and the displayed biological activity of organoselenides against Leishmania. Thus, the biological activities of 77 selenoesters and their N-aryl-propanamide derivatives were predicted using robust in silico models of Leishmania infantum, Leishmania amazonensis, Leishmania major, and Leishmania (Viannia) braziliensis. The models identified 28 compounds with >60% probability of demonstrating leishmanicidal activity against L. infantum, and likewise, 26 for L. amazonesis, 25 for L. braziliensis, and 23 for L. major. The in silico prediction of ADMET properties suggests high rates of oral absorption and good bioavailability for these compounds. In the in silico toxicity evaluation, only seven compounds showed signs of toxicity in up to one or two parameters. The methodology was corroborated with the ensuing experimental validation, which evaluated the inhibition of the Promastigote form of the Leishmania species under study. The activity of the molecules was determined by the IC50 value (µM); IC50 values < 20 µM indicated better inhibition profiles. Sixteen compounds were synthesized and tested for their activity. Eight molecules presented IC50 values < 20 µM for at least one of the Leishmania species under study, with compound NC34 presenting the strongest parasite inhibition profile. Furthermore, the methodology used was effective, as many of the compounds with the highest probability of activity were confirmed by the in vitro tests performed.
Collapse
Affiliation(s)
- Natália Ferreira de Sousa
- Post-Graduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| | | | | | - Francinara da Silva Alves
- Post-Graduate Program in Chemistry, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| | - Chonny Alexander Herrera Acevedo
- Post-Graduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| | - Thaís Amanda de Lima Nunes
- Infectious Diseases Laboratory, Federal University of Delta of Parnaíba, Av. São Sebastião, nº 2819-Nossa Sra. de Fátima, Parnaíba 64202-020, PI, Brazil
| | - Zoe L. Sessions
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Luciana Scotti
- Post-Graduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| | - Eugene N. Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | - Klinger Antônio da Franca Rodrigues
- Infectious Diseases Laboratory, Federal University of Delta of Parnaíba, Av. São Sebastião, nº 2819-Nossa Sra. de Fátima, Parnaíba 64202-020, PI, Brazil
| | | | - Marcus Tullius Scotti
- Post-Graduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| |
Collapse
|
38
|
Bahia MS, Kaspi O, Touitou M, Binayev I, Dhail S, Spiegel J, Khazanov N, Yosipof A, Senderowitz H. A comparison between 2D and 3D descriptors in QSAR modeling based on bio-active conformations. Mol Inform 2023; 42:e2200186. [PMID: 36617991 DOI: 10.1002/minf.202200186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/10/2023]
Abstract
QSAR models are widely and successfully used in many research areas. The success of such models highly depends on molecular descriptors typically classified as 1D, 2D, 3D, or 4D. While 3D information is likely important, e. g., for modeling ligand-protein binding, previous comparisons between the performances of 2D and 3D descriptors were inconclusive. Yet in such comparisons the modeled ligands were not necessarily represented by their bioactive conformations. With this in mind, we mined the PDB for sets of protein-ligand complexes sharing the same protein for which uniform activity data were reported. The results, totaling 461 structures spread across six series were compiled into a carefully curated, first of its kind dataset in which each ligand is represented by its bioactive conformation. Next, each set was characterized by 2D, 3D and 2D + 3D descriptors and modeled using three machine learning algorithms, namely, k-Nearest Neighbors, Random Forest and Lasso Regression. Models' performances were evaluated on external test sets derived from the parent datasets either randomly or in a rational manner. We found that many more significant models were obtained when combining 2D and 3D descriptors. We attribute these improvements to the ability of 2D and 3D descriptors to code for different, yet complementary molecular properties.
Collapse
Affiliation(s)
| | - Omer Kaspi
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Meir Touitou
- School of Cancer and Pharmaceutical Sciences, King's College London, London, 150 Stamford Street, SE1 9NH, United Kingdom
| | - Idan Binayev
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Seema Dhail
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Jacob Spiegel
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Netaly Khazanov
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Abraham Yosipof
- Department of Information Systems, College of Law & Business, Ramat-Gan, P.O. Box 852, Bnei Brak, 5110801, Israel
| | - Hanoch Senderowitz
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| |
Collapse
|
39
|
Machine Learning Prediction of Mycobacterial Cell Wall Permeability of Drugs and Drug-like Compounds. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020633. [PMID: 36677691 PMCID: PMC9863426 DOI: 10.3390/molecules28020633] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 12/30/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023]
Abstract
The cell wall of Mycobacterium tuberculosis and related organisms has a very complex and unusual organization that makes it much less permeable to nutrients and antibiotics, leading to the low activity of many potential antimycobacterial drugs against whole-cell mycobacteria compared to their isolated molecular biotargets. The ability to predict and optimize the cell wall permeability could greatly enhance the development of novel antitubercular agents. Using an extensive structure-permeability dataset for organic compounds derived from published experimental big data (5371 compounds including 2671 penetrating and 2700 non-penetrating compounds), we have created a predictive classification model based on fragmental descriptors and an artificial neural network of a novel architecture that provides better accuracy (cross-validated balanced accuracy 0.768, sensitivity 0.768, specificity 0.769, area under ROC curve 0.911) and applicability domain compared with the previously published results.
Collapse
|
40
|
Silva LC, Dos Santos Filho RF, de Oliveira AA, Martins FT, Cunha S, de Almeida Soares CM, Pereira M. 3-phenacylideneoxindoles as a new class of antifungal compounds against Paracoccidioides spp. Future Microbiol 2023; 18:93-105. [PMID: 36661071 DOI: 10.2217/fmb-2022-0133] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Aims: Considering the need to identify new compounds with antifungal action, the activity of five 3-phenacylideneoxindoles compounds was evaluated. Materials & methods: The compounds were synthesized, and their antifungal activity was elucidated through minimum inhibitory concentration tests and interaction assay with other antifungals. Potential targets of compounds were predicted in silico. Results: 3-phenacylideneoxindoles compounds inhibited fungal growth with minimum inhibitory concentration and minimum fungicidal concentration ranging from 3.05 to 12.26 μM. The compounds demonstrated high selectivity index and presented a synergistic effect with itraconazole. In silico prediction revealed the pentafunctional AROM polypeptide, enolase, superoxide dismutase, catalase and kinases as proteins targets of the compound 4a. Conclusion: The results demonstrate that 3-phenacylideneoxindoles is a potential new class of antifungal compounds for paracoccidioidomycosis treatment.
Collapse
Affiliation(s)
- Lívia C Silva
- Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
| | | | - Amanda A de Oliveira
- Institute of Tropical Pathology & Public Health, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
| | - Felipe T Martins
- Chemistry institute, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
| | - Silvio Cunha
- Chemistry institute, Federal University of Bahia, Salvador, Bahia, 40170-115, Brazil
| | | | - Maristela Pereira
- Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
| |
Collapse
|
41
|
D'Souza S, Balaji S, K V P. QSAR, molecular docking, molecular dynamics and MM-GBSA approach for identification of prospective benzotriazole-based SARS-CoV 3CL protease inhibitors. J Biomol Struct Dyn 2022; 40:14247-14261. [PMID: 34877897 DOI: 10.1080/07391102.2021.2002718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The 3CL Protease of severe acute respiratory syndrome coronavirus (SARS-CoV), responsible for viral replication, has emerged as an essential target for designing anti-coronaviral inhibitors in drug discovery. In recent years, small molecule and peptidomimetic inhibitors have been used to target the inhibition of SARS-CoV 3CL Protease. In this study, we have developed 2D and 3D Quantitative structure activity relationship (QSAR) models on 3CL protease inhibitors with good predictive capability to propose inhibitors with improved affinities. Based on the 3 D contour maps, three new inhibitors were designed in silico, which were further subjected to molecular docking to explore their binding modes. The newly designed compounds showed improved interaction energies toward SARS-CoV-3CLPro due to additional interactions with the active site residues. The molecular docking studies of the most potent compounds revealed specific interactions with Glu 166 and Cys 145. Furthermore, absorption, distribution, metabolism, elimination (ADME) and drug-likeness evaluation revealed improved pharmacokinetic properties for these compounds. The molecular dynamics simulations confirmed the stability of the interactions identified by docking. The results presented would guide the development of new 3CL protease inhibitors with improved affinities in the future.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sofia D'Souza
- Department of Computer Science and Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - S Balaji
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Prema K V
- Department of Computer Science and Engineering, Manipal Institute of Technology Bengaluru, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
42
|
Valencia J, Rubio V, Puerto G, Vasquez L, Bernal A, Mora JR, Cuesta SA, Paz JL, Insuasty B, Abonia R, Quiroga J, Insuasty A, Coneo A, Vidal O, Márquez E, Insuasty D. QSAR Studies, Molecular Docking, Molecular Dynamics, Synthesis, and Biological Evaluation of Novel Quinolinone-Based Thiosemicarbazones against Mycobacterium tuberculosis. Antibiotics (Basel) 2022; 12:antibiotics12010061. [PMID: 36671262 PMCID: PMC9854539 DOI: 10.3390/antibiotics12010061] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
In this study, a series of novel quinolinone-based thiosemicarbazones were designed in silico and their activities tested in vitro against Mycobacterium tuberculosis (M. tuberculosis). Quantitative structure-activity relationship (QSAR) studies were performed using quinolinone and thiosemicarbazide as pharmacophoric nuclei; the best model showed statistical parameters of R2 = 0.83; F = 47.96; s = 0.31, and was validated by several different methods. The van der Waals volume, electron density, and electronegativity model results suggested a pivotal role in antituberculosis (anti-TB) activity. Subsequently, from this model a new series of quinolinone-thiosemicarbazone 11a-e was designed and docked against two tuberculosis protein targets: enoyl-acyl carrier protein reductase (InhA) and decaprenylphosphoryl-β-D-ribose-2'-oxidase (DprE1). Molecular dynamics simulation over 200 ns showed a binding energy of -71.3 to -12.7 Kcal/mol, suggesting likely inhibition. In vitro antimycobacterial activity of quinolinone-thiosemicarbazone for 11a-e was evaluated against M. bovis, M. tuberculosis H37Rv, and six different strains of drug-resistant M. tuberculosis. All compounds exhibited good to excellent activity against all the families of M. tuberculosis. Several of the here synthesized compounds were more effective than the standard drugs (isoniazid, oxafloxacin), 11d and 11e being the most active products. The results suggest that these compounds may contribute as lead compounds in the research of new potential antimycobacterial agents.
Collapse
Affiliation(s)
- Jhesua Valencia
- Grupo de Investigación en Química y Biología, Universidad del Norte, Km 5 vía Puerto Colombia, Barranquilla 081007, Colombia
| | - Vivian Rubio
- Grupo de Micobacterias, Red TB Colombia, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud, Bogotá 111321, Colombia
| | - Gloria Puerto
- Grupo de Micobacterias, Red TB Colombia, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud, Bogotá 111321, Colombia
| | - Luisa Vasquez
- Grupo de Micobacterias, Red TB Colombia, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud, Bogotá 111321, Colombia
| | - Anthony Bernal
- Grupo de Investigación en Química y Biología, Universidad del Norte, Km 5 vía Puerto Colombia, Barranquilla 081007, Colombia
| | - José R. Mora
- Grupo de Química Computacional y Teórica (QCT-USFQ), Departamento de Ingeniería Química, Universidad San Francisco de Quito, Diego de Robles y Vía Interoceánica, Quito 170157, Ecuador
| | - Sebastian A. Cuesta
- Grupo de Química Computacional y Teórica (QCT-USFQ), Departamento de Ingeniería Química, Universidad San Francisco de Quito, Diego de Robles y Vía Interoceánica, Quito 170157, Ecuador
- Department of Chemistry, Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
| | - José Luis Paz
- Departamento Académico de Química Inorgánica, Facultad de Química e Ingeniería Química, Universidad Nacional Mayor de San Marcos, Cercado de Lima 15081, Peru
| | - Braulio Insuasty
- Research Group of Heterocyclic Compounds, Department of Chemistry, Universidad del Valle, A. A., Cali 25360, Colombia
| | - Rodrigo Abonia
- Research Group of Heterocyclic Compounds, Department of Chemistry, Universidad del Valle, A. A., Cali 25360, Colombia
| | - Jairo Quiroga
- Research Group of Heterocyclic Compounds, Department of Chemistry, Universidad del Valle, A. A., Cali 25360, Colombia
| | - Alberto Insuasty
- Grupo de Investigación en Materiales Funcionales Nanoestructurados, Universidad CESMAG, Pasto 520003, Colombia
| | - Andres Coneo
- Medicine Department, Division of Health Sciences, Universidad del Norte, Barranquilla 081007, Colombia
| | - Oscar Vidal
- Medicine Department, Division of Health Sciences, Universidad del Norte, Barranquilla 081007, Colombia
| | - Edgar Márquez
- Grupo de Investigación en Química y Biología, Universidad del Norte, Km 5 vía Puerto Colombia, Barranquilla 081007, Colombia
- Correspondence: (E.M.); (D.I.)
| | - Daniel Insuasty
- Grupo de Investigación en Química y Biología, Universidad del Norte, Km 5 vía Puerto Colombia, Barranquilla 081007, Colombia
- Correspondence: (E.M.); (D.I.)
| |
Collapse
|
43
|
Scalfani VF, Patel VD, Fernandez AM. Visualizing chemical space networks with RDKit and NetworkX. J Cheminform 2022; 14:87. [PMID: 36578091 PMCID: PMC9798653 DOI: 10.1186/s13321-022-00664-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 11/27/2022] [Indexed: 12/29/2022] Open
Abstract
This article demonstrates how to create Chemical Space Networks (CSNs) using a Python RDKit and NetworkX workflow. CSNs are a type of network visualization that depict compounds as nodes connected by edges, defined as a pairwise relationship such as a 2D fingerprint similarity value. A step by step approach is presented for creating two different CSNs in this manuscript, one based on RDKit 2D fingerprint Tanimoto similarity values, and another based on maximum common substructure similarity values. Several different CSN visualization features are included in the tutorial including methods to represent nodes with color based on bioactivity attribute value, edges with different line styles based on similarity value, as well as replacing the circle nodes with 2D structure depictions. Finally, some common network property and analysis calculations are presented including the clustering coefficient, degree assortativity, and modularity. All code is provided in the form of Jupyter Notebooks and is available on GitHub with a permissive BSD-3 open-source license: https://github.com/vfscalfani/CSN_tutorial.
Collapse
Affiliation(s)
- Vincent F. Scalfani
- grid.411015.00000 0001 0727 7545University Libraries, Rodgers Library for Science and Engineering, The University of Alabama, Tuscaloosa, AL 35487 USA
| | - Vishank D. Patel
- grid.411015.00000 0001 0727 7545University Libraries, Rodgers Library for Science and Engineering, The University of Alabama, Tuscaloosa, AL 35487 USA
| | - Avery M. Fernandez
- grid.411015.00000 0001 0727 7545University Libraries, Rodgers Library for Science and Engineering, The University of Alabama, Tuscaloosa, AL 35487 USA
| |
Collapse
|
44
|
Mottin M, de Paula Sousa BK, de Moraes Roso Mesquita NC, de Oliveira KIZ, Noske GD, Sartori GR, de Oliveira Albuquerque A, Urbina F, Puhl AC, Moreira-Filho JT, Souza GE, Guido RV, Muratov E, Neves BJ, da Silva JHM, Clark AE, Siqueira-Neto JL, Perryman AL, Oliva G, Ekins S, Andrade CH. Discovery of New Zika Protease and Polymerase Inhibitors through the Open Science Collaboration Project OpenZika. J Chem Inf Model 2022; 62:6825-6843. [PMID: 36239304 PMCID: PMC9923514 DOI: 10.1021/acs.jcim.2c00596] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The Zika virus (ZIKV) is a neurotropic arbovirus considered a global threat to public health. Although there have been several efforts in drug discovery projects for ZIKV in recent years, there are still no antiviral drugs approved to date. Here, we describe the results of a global collaborative crowdsourced open science project, the OpenZika project, from IBM's World Community Grid (WCG), which integrates different computational and experimental strategies for advancing a drug candidate for ZIKV. Initially, molecular docking protocols were developed to identify potential inhibitors of ZIKV NS5 RNA-dependent RNA polymerase (NS5 RdRp), NS3 protease (NS2B-NS3pro), and NS3 helicase (NS3hel). Then, a machine learning (ML) model was built to distinguish active vs inactive compounds for the cytoprotective effect against ZIKV infection. We performed three independent target-based virtual screening campaigns (NS5 RdRp, NS2B-NS3pro, and NS3hel), followed by predictions by the ML model and other filters, and prioritized a total of 61 compounds for further testing in enzymatic and phenotypic assays. This yielded five non-nucleoside compounds which showed inhibitory activity against ZIKV NS5 RdRp in enzymatic assays (IC50 range from 0.61 to 17 μM). Two compounds thermally destabilized NS3hel and showed binding affinity in the micromolar range (Kd range from 9 to 35 μM). Moreover, the compounds LabMol-301 inhibited both NS5 RdRp and NS2B-NS3pro (IC50 of 0.8 and 7.4 μM, respectively) and LabMol-212 thermally destabilized the ZIKV NS3hel (Kd of 35 μM). Both also protected cells from death induced by ZIKV infection in in vitro cell-based assays. However, while eight compounds (including LabMol-301 and LabMol-212) showed a cytoprotective effect and prevented ZIKV-induced cell death, agreeing with our ML model for prediction of this cytoprotective effect, no compound showed a direct antiviral effect against ZIKV. Thus, the new scaffolds discovered here are promising hits for future structural optimization and for advancing the discovery of further drug candidates for ZIKV. Furthermore, this work has demonstrated the importance of the integration of computational and experimental approaches, as well as the potential of large-scale collaborative networks to advance drug discovery projects for neglected diseases and emerging viruses, despite the lack of available direct antiviral activity and cytoprotective effect data, that reflects on the assertiveness of the computational predictions. The importance of these efforts rests with the need to be prepared for future viral epidemic and pandemic outbreaks.
Collapse
Affiliation(s)
- Melina Mottin
- Laboratory of Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, 74605-170, Brazil
- Pathogen-Host Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia, 70910-900, Brazil
| | - Bruna Katiele de Paula Sousa
- Laboratory of Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, 74605-170, Brazil
| | | | | | - Gabriela Dias Noske
- São Carlos Institute of Physics, University of São Paulo, Avenida João Dagnone, 1100, São Carlos, São Paulo, 13563-120, Brazil
| | | | | | - Fabio Urbina
- Collaborations Pharmaceuticals, Inc., Raleigh, NC, 27606, USA
| | - Ana C. Puhl
- Collaborations Pharmaceuticals, Inc., Raleigh, NC, 27606, USA
| | - José Teófilo Moreira-Filho
- Laboratory of Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, 74605-170, Brazil
| | - Guilherme E. Souza
- São Carlos Institute of Physics, University of São Paulo, Avenida João Dagnone, 1100, São Carlos, São Paulo, 13563-120, Brazil
| | - Rafael V.C. Guido
- São Carlos Institute of Physics, University of São Paulo, Avenida João Dagnone, 1100, São Carlos, São Paulo, 13563-120, Brazil
| | - Eugene Muratov
- University of North Carolina - University of North Carolina at Chapel Hill, 27599, USA
- Universidade Federal de Paraíba, Joao Pessoa, PB, 58051-900, Brazil
| | - Bruno Junior Neves
- Laboratory of Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, 74605-170, Brazil
| | | | - Alex E. Clark
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, 92093, USA
| | - Jair L. Siqueira-Neto
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, 92093, USA
| | - Alexander L. Perryman
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University–New Jersey Medical School, Newark, NJ 07103, United States
- Repare Therapeutics, 7210 Rue Frederick-Banting, Suite 100, Montreal, QC, H4S 2A1, Canada
| | - Glaucius Oliva
- São Carlos Institute of Physics, University of São Paulo, Avenida João Dagnone, 1100, São Carlos, São Paulo, 13563-120, Brazil
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., Raleigh, NC, 27606, USA
| | - Carolina Horta Andrade
- Laboratory of Molecular Modeling and Drug Design (LabMol), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, 74605-170, Brazil
| |
Collapse
|
45
|
Palazzotti D, Fiorelli M, Sabatini S, Massari S, Barreca ML, Astolfi A. Q-raKtion: A Semiautomated KNIME Workflow for Bioactivity Data Points Curation. J Chem Inf Model 2022; 62:6309-6315. [PMID: 36442071 PMCID: PMC9795488 DOI: 10.1021/acs.jcim.2c01199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The recent increase of bioactivity data freely available to the scientific community and stored as activity data points in chemogenomic repositories provides a huge amount of ready-to-use information to support the development of predictive models. However, the benefits provided by the availability of such a vast amount of accessible information are strongly counteracted by the lack of uniformity and consistency of data from multiple sources, requiring a process of integration and harmonization. While different automated pipelines for processing and assessing chemical data have emerged in the last years, the curation of bioactivity data points is a less investigated topic, with useful concepts provided but no tangible tools available. In this context, the present work represents a first step toward the filling of this gap, by providing a tool to meet the needs of end-user in building proprietary high-quality data sets for further studies. Specifically, we herein describe Q-raKtion, a systematic, semiautomated, flexible, and, above all, customizable KNIME workflow that effectively aggregates information on biological activities of compounds retrieved by two of the most comprehensive and widely used repositories, PubChem and ChEMBL.
Collapse
|
46
|
Cheminformatics analysis of chemicals that increase estrogen and progesterone synthesis for a breast cancer hazard assessment. Sci Rep 2022; 12:20647. [PMID: 36450809 PMCID: PMC9712655 DOI: 10.1038/s41598-022-24889-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Factors that increase estrogen or progesterone (P4) action are well-established as increasing breast cancer risk, and many first-line treatments to prevent breast cancer recurrence work by blocking estrogen synthesis or action. In previous work, using data from an in vitro steroidogenesis assay developed for the U.S. Environmental Protection Agency (EPA) ToxCast program, we identified 182 chemicals that increased estradiol (E2up) and 185 that increased progesterone (P4up) in human H295R adrenocortical carcinoma cells, an OECD validated assay for steroidogenesis. Chemicals known to induce mammary effects in vivo were very likely to increase E2 or P4 synthesis, further supporting the importance of these pathways for breast cancer. To identify additional chemical exposures that may increase breast cancer risk through E2 or P4 steroidogenesis, we developed a cheminformatics approach to identify structural features associated with these activities and to predict other E2 or P4 steroidogens from their chemical structures. First, we used molecular descriptors and physicochemical properties to cluster the 2,012 chemicals screened in the steroidogenesis assay using a self-organizing map (SOM). Structural features such as triazine, phenol, or more broadly benzene ramified with halide, amine or alcohol, are enriched for E2 or P4up chemicals. Among E2up chemicals, phenol and benzenone are found as significant substructures, along with nitrogen-containing biphenyls. For P4up chemicals, phenol and complex aromatic systems ramified with oxygen-based groups such as flavone or phenolphthalein are significant substructures. Chemicals that are active for both E2up and P4up are enriched with substructures such as dihydroxy phosphanedithione or are small chemicals that contain one benzene ramified with chlorine, alcohol, methyl or primary amine. These results are confirmed with a chemotype ToxPrint analysis. Then, we used machine learning and artificial intelligence algorithms to develop and validate predictive classification QSAR models for E2up and P4up chemicals. These models gave reasonable external prediction performances (balanced accuracy ~ 0.8 and Matthews Coefficient Correlation ~ 0.5) on an external validation. The QSAR models were enriched by adding a confidence score that considers the chemical applicability domain and a ToxPrint assessment of the chemical. This profiling and these models may be useful to direct future testing and risk assessments for chemicals related to breast cancer and other hormonally-mediated outcomes.
Collapse
|
47
|
Liu Z, Zubatiuk T, Roitberg A, Isayev O. Auto3D: Automatic Generation of the Low-Energy 3D Structures with ANI Neural Network Potentials. J Chem Inf Model 2022; 62:5373-5382. [PMID: 36112860 DOI: 10.1021/acs.jcim.2c00817] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Computational programs accelerate the chemical discovery processes but often need proper three-dimensional molecular information as part of the input. Getting optimal molecular structures is challenging because it requires enumerating and optimizing a huge space of stereoisomers and conformers. We developed the Python-based Auto3D package for generating the low-energy 3D structures using SMILES as the input. Auto3D is based on state-of-the-art algorithms and can automatize the isomer enumeration and duplicate filtering process, 3D building process, geometry optimization, and ranking process. Tested on 50 molecules with multiple unspecified stereocenters, Auto3D is guaranteed to find the stereoconfiguration that yields the lowest-energy conformer. With Auto3D, we provide an extension of the ANI model. The new model, dubbed ANI-2xt, is trained on a tautomer-rich data set. ANI-2xt is benchmarked with DFT methods on geometry optimization and electronic and Gibbs free energy calculations. Compared with ANI-2x, ANI-2xt provides a 42% error reduction for tautomeric reaction energy calculations when using the gold-standard coupled-cluster calculation as the reference. ANI-2xt can accurately predict the energies and is several orders of magnitude faster than DFT methods.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Chemistry, Mellon College of Science, Carnegie Mellon University, Pittsburgh, Pennsylvania15213, United States
| | - Tetiana Zubatiuk
- Department of Chemistry, Mellon College of Science, Carnegie Mellon University, Pittsburgh, Pennsylvania15213, United States
| | - Adrian Roitberg
- Department of Chemistry, University of Florida, Gainesville, Florida32611, United States
| | - Olexandr Isayev
- Department of Chemistry, Mellon College of Science, Carnegie Mellon University, Pittsburgh, Pennsylvania15213, United States
| |
Collapse
|
48
|
Ahmadi S, Abdolmaleki A, Jebeli Javan M. In silico study of natural antioxidants. VITAMINS AND HORMONES 2022; 121:1-43. [PMID: 36707131 DOI: 10.1016/bs.vh.2022.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Antioxidants are the body's defense system against the damage of reactive oxygen species, which are usually produced in the body through various physiological processes. There are various sources of these antioxidants such as endogenous antioxidants in the body and exogenous food sources. This chapter provides important information on methods used to investigate antioxidant activity and sources of plant antioxidants. Over the past two decades, numerous studies have demonstrated the importance of in silico research in the development of novel natural and synthesized antioxidants. In silico methods such as quantitative structure-activity relationships (QSAR), pharmacophore, docking, and virtual screenings are play critical roles in designing effective antioxidants that may be synthesized and tested later. This chapter introduces the available in silico approaches for different classes of antioxidants. Many successful applications of in silico methods in the development and design of novel antioxidants are thoroughly discussed. The QSAR, pharmacophore, molecular docking techniques, and virtual screenings process summarized here would help readers to find out the proper mechanism for the interaction between the free radicals and antioxidant compounds. Furthermore, this chapter focuses on introducing new QSAR models in combination with other in silico methods to predict antioxidants activity and design more active antioxidants. In silico studies are essential to explore largely unknown plant tissue, food sources for antioxidant synthesis, as well as saving time and money in such studies.
Collapse
Affiliation(s)
- Shahin Ahmadi
- Department of Chemistry, Faculty of Pharmaceutical Chemistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Azizeh Abdolmaleki
- Department of Chemistry, Tuyserkan Branch, Islamic Azad University, Tuyserkan, Iran
| | - Marjan Jebeli Javan
- Department of Chemistry, Faculty of Pharmaceutical Chemistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
49
|
Abrahamsson D, Siddharth A, Robinson JF, Soshilov A, Elmore S, Cogliano V, Ng C, Khan E, Ashton R, Chiu WA, Fung J, Zeise L, Woodruff TJ. Modeling the transplacental transfer of small molecules using machine learning: a case study on per- and polyfluorinated substances (PFAS). JOURNAL OF EXPOSURE SCIENCE & ENVIRONMENTAL EPIDEMIOLOGY 2022; 32:808-819. [PMID: 36207486 PMCID: PMC9742309 DOI: 10.1038/s41370-022-00481-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 05/10/2023]
Abstract
BACKGROUND Despite their large numbers and widespread use, very little is known about the extent to which per- and polyfluoroalkyl substances (PFAS) can cross the placenta and expose the developing fetus. OBJECTIVE The aim of our study is to develop a computational approach that can be used to evaluate the of extend to which small molecules, and in particular PFAS, can cross to cross the placenta and partition to cord blood. METHODS We collected experimental values of the concentration ratio between cord and maternal blood (RCM) for 260 chemical compounds and calculated their physicochemical descriptors using the cheminformatics package Mordred. We used the compiled database to, train and test an artificial neural network (ANN). And then applied the best performing model to predict RCM for a large dataset of PFAS chemicals (n = 7982). We, finally, examined the calculated physicochemical descriptors of the chemicals to identify which properties correlated significantly with RCM. RESULTS We determined that 7855 compounds were within the applicability domain and 127 compounds are outside the applicability domain of our model. Our predictions of RCM for PFAS suggested that 3623 compounds had a log RCM > 0 indicating preferable partitioning to cord blood. Some examples of these compounds were bisphenol AF, 2,2-bis(4-aminophenyl)hexafluoropropane, and nonafluoro-tert-butyl 3-methylbutyrate. SIGNIFICANCE These observations have important public health implications as many PFAS have been shown to interfere with fetal development. In addition, as these compounds are highly persistent and many of them can readily cross the placenta, they are expected to remain in the population for a long time as they are being passed from parent to offspring. IMPACT Understanding the behavior of chemicals in the human body during pregnancy is critical in preventing harmful exposures during critical periods of development. Many chemicals can cross the placenta and expose the fetus, however, the mechanism by which this transport occurs is not well understood. In our study, we developed a machine learning model that describes the transplacental transfer of chemicals as a function of their physicochemical properties. The model was then used to make predictions for a set of 7982 per- and polyfluorinated alkyl substances that are listed on EPA's CompTox Chemicals Dashboard. The model can be applied to make predictions for other chemical categories of interest, such as plasticizers and pesticides. Accurate predictions of RCM can help scientists and regulators to prioritize chemicals that have the potential to cause harm by exposing the fetus.
Collapse
Affiliation(s)
- Dimitri Abrahamsson
- Department of Obstetrics, Gynecology and Reproductive Sciences, Program on Reproductive Health and the Environment, University of California, San Francisco, 490 Illinois Street, San Francisco, CA, 94143, USA.
| | - Adi Siddharth
- Department of Obstetrics, Gynecology and Reproductive Sciences, Program on Reproductive Health and the Environment, University of California, San Francisco, 490 Illinois Street, San Francisco, CA, 94143, USA
| | - Joshua F Robinson
- Department of Obstetrics, Gynecology and Reproductive Sciences, Program on Reproductive Health and the Environment, University of California, San Francisco, 490 Illinois Street, San Francisco, CA, 94143, USA
| | - Anatoly Soshilov
- California Environmental Protection Agency, Office of Environmental Health Hazard Assessment, 1001 I St, Sacramento, CA, 95814, USA
- California Environmental Protection Agency, Office of Environmental Health Hazard Assessment, 1515 Clay St, Oakland, CA, 94612, USA
| | - Sarah Elmore
- California Environmental Protection Agency, Office of Environmental Health Hazard Assessment, 1001 I St, Sacramento, CA, 95814, USA
- California Environmental Protection Agency, Office of Environmental Health Hazard Assessment, 1515 Clay St, Oakland, CA, 94612, USA
| | - Vincent Cogliano
- California Environmental Protection Agency, Office of Environmental Health Hazard Assessment, 1001 I St, Sacramento, CA, 95814, USA
- California Environmental Protection Agency, Office of Environmental Health Hazard Assessment, 1515 Clay St, Oakland, CA, 94612, USA
| | - Carla Ng
- Department of Civil and Environmental Engineering, University of Pittsburgh, 3700 O'Hara St, Pittsburgh, PA, 15261, USA
| | - Elaine Khan
- California Environmental Protection Agency, Office of Environmental Health Hazard Assessment, 1001 I St, Sacramento, CA, 95814, USA
- California Environmental Protection Agency, Office of Environmental Health Hazard Assessment, 1515 Clay St, Oakland, CA, 94612, USA
| | - Randolph Ashton
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, 330 N Orchard St, Madison, WI, 53715, USA
- The Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
- Department of Biomedical Engineering, University of Wisconsin - Madison, 1550 Engineering Drive, Madison, WI, 53706, USA
| | - Weihsueh A Chiu
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Jennifer Fung
- Department of Obstetrics, Gynecology, and Reproductive Science and the Center of Reproductive Science, University of California, San Francisco, San Francisco, CA, 94143-2240, USA
| | - Lauren Zeise
- California Environmental Protection Agency, Office of Environmental Health Hazard Assessment, 1001 I St, Sacramento, CA, 95814, USA
- California Environmental Protection Agency, Office of Environmental Health Hazard Assessment, 1515 Clay St, Oakland, CA, 94612, USA
| | - Tracey J Woodruff
- Department of Obstetrics, Gynecology and Reproductive Sciences, Program on Reproductive Health and the Environment, University of California, San Francisco, 490 Illinois Street, San Francisco, CA, 94143, USA.
| |
Collapse
|
50
|
Stolbov LA, Filimonov DA, Poroikov VV. SAR based on self consistent classifier. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2022; 33:793-804. [PMID: 36369710 DOI: 10.1080/1062936x.2022.2139751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/20/2022] [Indexed: 06/16/2023]
Abstract
The accuracy and performance of (Q)SAR models depend significantly on the data used for training. Datasets prepared on the basis of publicly available databases contain structures belonging to different chemical classes and have a highly imbalanced actives/inactives ratio. Currently, hundreds of structural descriptors are used in (Q)SAR studies. The abundance of structural descriptors gives rise to the problem of the constructed (Q)SAR models stability. The methods frequently used for the selection of a small fraction of the 'best' descriptors usually do not have sufficient mathematical justification. We propose a new approach to a self-consistent classifier for SAR analysis in order to overcome these problems. Logistic (SCLC) and extreme (SCEC) extensions of self-consistent regression (SCR) were implemented to enhance the classification capabilities of SCR. The approach was applied to classification models' development for inhibiting activity endpoints in HIV-1-related data and toxicity endpoints with subsequent fivefold cross-validation to estimate the models' performance. Comparison of the proposed SCLC and SCEC models with those developed using the original SCR and support vector machine demonstrated the comparable accuracy. Advantages in feature selection using our approach provide more generalizable (Q)SAR models. In particular, the crucial factors responsible for the observed value are determined unambiguously.
Collapse
Affiliation(s)
- L A Stolbov
- Laboratory of Structure-Function Based Drug Design, Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow, Russian Federation
| | - D A Filimonov
- Laboratory of Structure-Function Based Drug Design, Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow, Russian Federation
| | - V V Poroikov
- Laboratory of Structure-Function Based Drug Design, Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow, Russian Federation
| |
Collapse
|