1
|
Prat A, Abdel Aty H, Bastas O, Kamuntavičius G, Paquet T, Norvaišas P, Gasparotto P, Tal R. HydraScreen: A Generalizable Structure-Based Deep Learning Approach to Drug Discovery. J Chem Inf Model 2024; 64:5817-5831. [PMID: 39037942 DOI: 10.1021/acs.jcim.4c00481] [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: 07/24/2024]
Abstract
We propose HydraScreen, a deep-learning framework for safe and robust accelerated drug discovery. HydraScreen utilizes a state-of-the-art 3D convolutional neural network designed for the effective representation of molecular structures and interactions in protein-ligand binding. We designed an end-to-end pipeline for high-throughput screening and lead optimization, targeting applications in structure-based drug design. We assessed our approach using established public benchmarks based on the CASF-2016 core set, achieving top-tier results in affinity and pose prediction (Pearson's r = 0.86, RMSE = 1.15, Top-1 = 0.95). We introduced a novel approach for interaction profiling, aimed at detecting potential biases within both the model and data sets. This approach not only enhanced interpretability but also reinforced the impartiality of our methodology. Finally, we demonstrated HydraScreen's ability to generalize effectively across novel proteins and ligands through a temporal split. We also provide insights into potential avenues for future development aimed at enhancing the robustness of machine learning scoring functions. HydraScreen (accessible at http://hydrascreen.ro5.ai/paper) provides a user-friendly GUI and a public API, facilitating the easy-access assessment of protein-ligand complexes.
Collapse
Affiliation(s)
- Alvaro Prat
- AI Chemistry, Ro5 2801 Gateway Drive, Irving, 75063 Texas, United States
| | - Hisham Abdel Aty
- AI Chemistry, Ro5 2801 Gateway Drive, Irving, 75063 Texas, United States
| | - Orestis Bastas
- AI Chemistry, Ro5 2801 Gateway Drive, Irving, 75063 Texas, United States
| | | | - Tanya Paquet
- AI Chemistry, Ro5 2801 Gateway Drive, Irving, 75063 Texas, United States
| | - Povilas Norvaišas
- AI Chemistry, Ro5 2801 Gateway Drive, Irving, 75063 Texas, United States
| | - Piero Gasparotto
- AI Chemistry, Ro5 2801 Gateway Drive, Irving, 75063 Texas, United States
| | - Roy Tal
- AI Chemistry, Ro5 2801 Gateway Drive, Irving, 75063 Texas, United States
| |
Collapse
|
2
|
Sultana F, Ghosh A. Exploring the evolutionary landscape and structural resonances of ferritin with insights into functional significance in plant. Biochimie 2024:S0300-9084(24)00173-1. [PMID: 39047810 DOI: 10.1016/j.biochi.2024.07.014] [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: 04/20/2024] [Revised: 07/04/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
The mineral iron plays a crucial role in facilitating the optimal functioning of numerous biological processes within the cellular environment. These processes involve the transportation of oxygen, energy production, immune system functioning, cognitive abilities, and muscle function. However, it is crucial to note that excessive levels of iron can result in oxidative damage within cells, primarily through Fenton reactions. Iron availability and toxicity present significant challenges that have been addressed through evolution. Ferritin is an essential protein that stores iron and is divided into different subfamilies, including DNA-binding proteins under starvation (Dps), bacterioferritin, and classical ferritin. Ferritin plays a critical role in maintaining cellular balance and protecting against oxidative damage. This study delves into ferritin's evolutionary dynamics across diverse taxa, emphasizing structural features and regulatory mechanisms. Insights into ferritin's evolution and functional diversity are gained through phylogenetic and structural analysis in bacterial Dps, bacterioferritin, and classical ferritin proteins. Additionally, the involvement of ferritin in plant stress responses and development is explored. Analysis of ferritin gene expression across various developmental stages and stress conditions provides insights into its regulatory roles. This comprehensive exploration enhances our understanding of ferritin's significance in plant biology, offering insights into its evolutionary history, structural diversity, and protective mechanisms against oxidative stress.
Collapse
Affiliation(s)
- Fahmida Sultana
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Ajit Ghosh
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh.
| |
Collapse
|
3
|
Shuvo MN, Halder SK, Alam N, Himel MK, Shil A. Developing phytocompound-based new drugs against multi-drug-resistant Staphylococcus aureus. ROYAL SOCIETY OPEN SCIENCE 2024; 11:231475. [PMID: 39050719 PMCID: PMC11265916 DOI: 10.1098/rsos.231475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 05/28/2024] [Accepted: 06/13/2024] [Indexed: 07/27/2024]
Abstract
Staphylococcus aureus, a prevalent component of the human microbiota, is associated with skin infections to life-threatening diseases, presenting challenges in treatment options and necessitating the development of effective treatments. This study integrated computational and in vitro approaches to identify promising phytocompounds with therapeutic potential. Staphopain B emerged as a target protein for its role in immune evasion, exhibiting stability during molecular dynamic simulation (MDS) with a root mean square deviation value of 2.376 Å. Screening 115 phytocompounds with antibacterial properties from the PubChem database identified 12 with drug-like properties, nine of which showed superior binding affinity to Staphopain B compared to a commercial antibiotic, doxycycline (-7.8 kcal mol-1). Notably, epoxyazadiradione and nimbolide displayed higher estimated free energy of binding scores (-7.91 and -7.93 kcal mol-1, respectively), indicating strong protein-ligand interactions. The root mean square fluctuation values for epoxyazadiradione and nimbolide were 1.097 and 1.034 Å, respectively, which was confirmed through MDS. Crude ethanolic extracts (100% and 70%) of neem (Azadirachta indica) leaves demonstrated narrow inhibition against the bacteria in comparison to doxycycline in the disc-diffusion assay. This study underscores the potential of phytocompounds as therapeutic agents against S. aureus; however, further in vitro experiments and testing of the phytocompounds in vivo are required.
Collapse
Affiliation(s)
| | | | - Nuhu Alam
- Department of Botany, Jahangirnagar University, Savar, Dhaka1342, Bangladesh
| | - Mahbubul Kabir Himel
- Department of Botany, Jahangirnagar University, Savar, Dhaka1342, Bangladesh
- Padma Bioresearch, Dhaka1342, Bangladesh
| | - Aparna Shil
- Department of Botany, Jahangirnagar University, Savar, Dhaka1342, Bangladesh
- Padma Bioresearch, Dhaka1342, Bangladesh
| |
Collapse
|
4
|
Ren L, Zhang T, Zhang J. Recent advances in dietary androgen receptor inhibitors. Med Res Rev 2024; 44:1446-1500. [PMID: 38279967 DOI: 10.1002/med.22019] [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: 10/20/2022] [Revised: 12/07/2023] [Accepted: 01/10/2024] [Indexed: 01/29/2024]
Abstract
As a nuclear transcription factor, the androgen receptor (AR) plays a crucial role not only in normal male sexual differentiation and growth of the prostate, but also in benign prostatic hyperplasia, prostatitis, and prostate cancer. Multiple population-based epidemiological studies demonstrated that prostate cancer risk was inversely associated with increased dietary intakes of green tea, soy products, tomato, and so forth. Therefore, this review aimed to summarize the structure and function of AR, and further illustrate the structural basis for antagonistic mechanisms of the currently clinically available antiandrogens. Due to the limitations of these antiandrogens, a series of natural AR inhibitors have been identified from edible plants such as fruits and vegetables, as well as folk medicines, health foods, and nutritional supplements. Hence, this review mainly focused on recent experimental, epidemiological, and clinical studies about natural AR inhibitors, particularly the association between dietary intake of natural antiandrogens and reduced risk of prostatic diseases. Since natural products offer multiple advantages over synthetic antiandrogens, this review may provide a comprehensive and updated overview of dietary-derived AR inhibitors, as well as their potential for the nutritional intervention against prostatic disorders.
Collapse
Affiliation(s)
- Li Ren
- College of Food Science and Engineering, Jilin University, Changchun, China
| | - Tiehua Zhang
- College of Food Science and Engineering, Jilin University, Changchun, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun, China
| |
Collapse
|
5
|
Gutkin E, Gusev F, Gentile F, Ban F, Koby SB, Narangoda C, Isayev O, Cherkasov A, Kurnikova MG. In silico screening of LRRK2 WDR domain inhibitors using deep docking and free energy simulations. Chem Sci 2024; 15:8800-8812. [PMID: 38873063 PMCID: PMC11168082 DOI: 10.1039/d3sc06880c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/10/2024] [Indexed: 06/15/2024] Open
Abstract
The Critical Assessment of Computational Hit-Finding Experiments (CACHE) Challenge series is focused on identifying small molecule inhibitors of protein targets using computational methods. Each challenge contains two phases, hit-finding and follow-up optimization, each of which is followed by experimental validation of the computational predictions. For the CACHE Challenge #1, the Leucine-Rich Repeat Kinase 2 (LRRK2) WD40 Repeat (WDR) domain was selected as the target for in silico hit-finding and optimization. Mutations in LRRK2 are the most common genetic cause of the familial form of Parkinson's disease. The LRRK2 WDR domain is an understudied drug target with no known molecular inhibitors. Herein we detail the first phase of our winning submission to the CACHE Challenge #1. We developed a framework for the high-throughput structure-based virtual screening of a chemically diverse small molecule space. Hit identification was performed using the large-scale Deep Docking (DD) protocol followed by absolute binding free energy (ABFE) simulations. ABFEs were computed using an automated molecular dynamics (MD)-based thermodynamic integration (TI) approach. 4.1 billion ligands from Enamine REAL were screened with DD followed by ABFEs computed by MD TI for 793 ligands. 76 ligands were prioritized for experimental validation, with 59 compounds successfully synthesized and 5 compounds identified as hits, yielding a 8.5% hit rate. Our results demonstrate the efficacy of the combined DD and ABFE approaches for hit identification for a target with no previously known hits. This approach is widely applicable for the efficient screening of ultra-large chemical libraries as well as rigorous protein-ligand binding affinity estimation leveraging modern computational resources.
Collapse
Affiliation(s)
- Evgeny Gutkin
- Department of Chemistry, Mellon College of Science, Carnegie Mellon University Pittsburgh PA 15213 USA
| | - Filipp Gusev
- Department of Chemistry, Mellon College of Science, Carnegie Mellon University Pittsburgh PA 15213 USA
- Computational Biology Department, School of Computer Science, Carnegie Mellon University Pittsburgh PA 15213 USA
| | - Francesco Gentile
- Department of Chemistry and Biomolecular Sciences, University of Ottawa Ottawa ON Canada
- Ottawa Institute of Systems Biology Ottawa ON Canada
| | - Fuqiang Ban
- Vancouver Prostate Centre, The University of British Columbia Vancouver BC Canada
| | - S Benjamin Koby
- Department of Chemistry, Mellon College of Science, Carnegie Mellon University Pittsburgh PA 15213 USA
| | - Chamali Narangoda
- Department of Chemistry, Mellon College of Science, Carnegie Mellon University Pittsburgh PA 15213 USA
| | - Olexandr Isayev
- Department of Chemistry, Mellon College of Science, Carnegie Mellon University Pittsburgh PA 15213 USA
- Computational Biology Department, School of Computer Science, Carnegie Mellon University Pittsburgh PA 15213 USA
| | - Artem Cherkasov
- Vancouver Prostate Centre, The University of British Columbia Vancouver BC Canada
| | - Maria G Kurnikova
- Department of Chemistry, Mellon College of Science, Carnegie Mellon University Pittsburgh PA 15213 USA
| |
Collapse
|
6
|
Stratton C, Christensen A, Jordan C, Salvatore BA, Mahdavian E. An interdisciplinary course on computer-aided drug discovery to broaden student participation in original scientific research. BIOCHEMISTRY AND MOLECULAR BIOLOGY EDUCATION : A BIMONTHLY PUBLICATION OF THE INTERNATIONAL UNION OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2024; 52:276-290. [PMID: 38308532 PMCID: PMC11251704 DOI: 10.1002/bmb.21811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 11/13/2023] [Accepted: 12/30/2023] [Indexed: 02/04/2024]
Abstract
We present a new highly interdisciplinary project-based course in computer aided drug discovery (CADD). This course was developed in response to a call for alternative pedagogical approaches during the COVID-19 pandemic, which caused the cancellation of a face-to-face summer research program sponsored by the Louisiana Biomedical Research Network (LBRN). The course integrates guided research and educational experiences for chemistry, biology, and computer science students. We implement research-based methods with publicly available tools in bioinformatics and molecular modeling to identify and prioritize promising antiviral drug candidates for COVID-19. The purpose of this course is three-fold: I. Implement an active learning and inclusive pedagogy that fosters student engagement and research mindset; II. Develop student interdisciplinary research skills that are highly beneficial in a broader scientific context; III. Demonstrate that pedagogical shifts (initially incurred during the COVID-19 pandemic) can furnish longer-term instructional benefits. The course, which has now been successfully taught a total of five times, incorporates four modules, including lectures/discussions, live demos, inquiry-based assignments, and science communication.
Collapse
Affiliation(s)
- Christopher Stratton
- Department of Biological Science, LSU Shreveport, One University Place, Shreveport, Louisiana, USA
| | - Avery Christensen
- Department of Biological Science, LSU Shreveport, One University Place, Shreveport, Louisiana, USA
| | - Chelsey Jordan
- Department of Biological Science, LSU Shreveport, One University Place, Shreveport, Louisiana, USA
| | - Brian A Salvatore
- Department of Chemistry & Physics, LSU Shreveport, One University Place, Shreveport, Louisiana, USA
| | - Elahe Mahdavian
- Department of Biological Science, LSU Shreveport, One University Place, Shreveport, Louisiana, USA
| |
Collapse
|
7
|
Smith L, Novak B, Osato M, Mobley DL, Bowman GR. PopShift: A Thermodynamically Sound Approach to Estimate Binding Free Energies by Accounting for Ligand-Induced Population Shifts from a Ligand-Free Markov State Model. J Chem Theory Comput 2024; 20:1036-1050. [PMID: 38291966 PMCID: PMC10867841 DOI: 10.1021/acs.jctc.3c00870] [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: 08/08/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 02/01/2024]
Abstract
Obtaining accurate binding free energies from in silico screens has been a long-standing goal for the computational chemistry community. However, accuracy and computational cost are at odds with one another, limiting the utility of methods that perform this type of calculation. Many methods achieve massive scale by explicitly or implicitly assuming that the target protein adopts a single structure, or undergoes limited fluctuations around that structure, to minimize computational cost. Others simulate each protein-ligand complex of interest, accepting lower throughput in exchange for better predictions of binding affinities. Here, we present the PopShift framework for accounting for the ensemble of structures a protein adopts and their relative probabilities. Protein degrees of freedom are enumerated once, and then arbitrarily many molecules can be screened against this ensemble. Specifically, we use Markov state models (MSMs) as a compressed representation of a protein's thermodynamic ensemble. We start with a ligand-free MSM and then calculate how addition of a ligand shifts the populations of each protein conformational state based on the strength of the interaction between that protein conformation and the ligand. In this work we use docking to estimate the affinity between a given protein structure and ligand, but any estimator of binding affinities could be used in the PopShift framework. We test PopShift on the classic benchmark pocket T4 Lysozyme L99A. We find that PopShift is more accurate than common strategies, such as docking to a single structure and traditional ensemble docking─producing results that compare favorably with alchemical binding free energy calculations in terms of RMSE but not correlation─and may have a more favorable computational cost profile in some applications. In addition to predicting binding free energies and ligand poses, PopShift also provides insight into how the probability of different protein structures is shifted upon addition of various concentrations of ligand, providing a platform for predicting affinities and allosteric effects of ligand binding. Therefore, we expect PopShift will be valuable for hit finding and for providing insight into phenomena like allostery.
Collapse
Affiliation(s)
- Louis
G. Smith
- Departments
of Biochemistry & Biophysics and Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Borna Novak
- Department
of Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Medical
Scientist Training Program, Washington University
in St. Louis, St. Louis, Missouri 63130, United
States
| | - Meghan Osato
- School
of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, California 92697, United States
| | - David L. Mobley
- School
of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, California 92697, United States
| | - Gregory R. Bowman
- Departments
of Biochemistry & Biophysics and Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
8
|
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
|
9
|
Pereira TO, Abbasi M, Oliveira RI, Guedes RA, Salvador JAR, Arrais JP. Artificial intelligence for prediction of biological activities and generation of molecular hits using stereochemical information. J Comput Aided Mol Des 2023; 37:791-806. [PMID: 37847342 PMCID: PMC10618333 DOI: 10.1007/s10822-023-00539-9] [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: 01/31/2023] [Accepted: 10/02/2023] [Indexed: 10/18/2023]
Abstract
In this work, we develop a method for generating targeted hit compounds by applying deep reinforcement learning and attention mechanisms to predict binding affinity against a biological target while considering stereochemical information. The novelty of this work is a deep model Predictor that can establish the relationship between chemical structures and their corresponding [Formula: see text] values. We thoroughly study the effect of different molecular descriptors such as ECFP4, ECFP6, SMILES and RDKFingerprint. Also, we demonstrated the importance of attention mechanisms to capture long-range dependencies in molecular sequences. Due to the importance of stereochemical information for the binding mechanism, this information was employed both in the prediction and generation processes. To identify the most promising hits, we apply the self-adaptive multi-objective optimization strategy. Moreover, to ensure the existence of stereochemical information, we consider all the possible enumerated stereoisomers to provide the most appropriate 3D structures. We evaluated this approach against the Ubiquitin-Specific Protease 7 (USP7) by generating putative inhibitors for this target. The predictor with SMILES notations as descriptor plus bidirectional recurrent neural network using attention mechanism has the best performance. Additionally, our methodology identify the regions of the generated molecules that are important for the interaction with the receptor's active site. Also, the obtained results demonstrate that it is possible to discover synthesizable molecules with high biological affinity for the target, containing the indication of their optimal stereochemical conformation.
Collapse
Affiliation(s)
- Tiago O Pereira
- Centre for Informatics and Systems, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal.
| | - Maryam Abbasi
- Centre for Informatics and Systems, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal
- Applied Research Institute, Polytechnic Institute of Coimbra, Coimbra, Portugal
- Research Centre for Natural Resources Environment and Society (CERNAS), Polytechnic Institute of Coimbra, Coimbra, Portugal
| | - Rita I Oliveira
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, Coimbra, Portugal
| | - Romina A Guedes
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, Coimbra, Portugal
| | - Jorge A R Salvador
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, Coimbra, Portugal
| | - Joel P Arrais
- Centre for Informatics and Systems, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
10
|
Gupta SRR, Mittal P, Kundu B, Singh A, Singh IK. Silibinin: an inhibitor for a high-expressed BCL-2A1/BFL1 protein, linked with poor prognosis in breast cancer. J Biomol Struct Dyn 2023:1-11. [PMID: 37837418 DOI: 10.1080/07391102.2023.2268176] [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: 03/14/2023] [Accepted: 09/29/2023] [Indexed: 10/16/2023]
Abstract
Breast cancer (BC) accounts for 30% of all diagnosed cases of cancer in women and remains a leading cause of cancer-related deaths among women worldwide. The current study looks for a protein from the anti-apoptotic/pro-survival BCL-2 family whose overexpression reduces survivability in BC patients and a potential inhibitor for the protein. We found BCL-2A1/BFL1 protein with high expression linked to low survivability in BC. The protein shows prognosis in 8 out of 29 categories, whereas no other family member manifests this property. Out of 7379 compounds, three small molecules (CHEMBL9509, CHEMBL2104550 and CHEMBL3545011) form an H-bond with BCL-2A1/BFL1 protein's unique residue Cys55. Of the three small molecules, we found CHEMBL9509 (Silibinin) to be a potent inhibitor. The compound forms a stable H-bond with the residue Cys55 with the lowest binding energy compared to the other two compounds. It remains stable in the BH3 binding region for more than 100 ns, whereas the other two detach from the region. Additionally, the compound is found to be better than Venetoclax and Nematoclax. We firmly believe in the compound CHEMBL9509 potency to halt BC's progression by inhibiting the BCL-2A1/BFL1 protein, increasing patients' survivability.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shradheya R R Gupta
- Molecular Biology Research Laboratory, Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, India
| | - Pooja Mittal
- Molecular Biology Research Laboratory, Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, India
- Norris Comprehensive Cancer Center, Division of Medical Oncology, University of Southern California, Los Angeles, USA
| | - Bishwajit Kundu
- Kusuma School of Biological Science, Indian Institute of Technology Delhi, New Delhi, India
| | - Archana Singh
- Department of Plant Molecular Biology, University of Delhi (South Campus), New Delhi, India
| | - Indrakant K Singh
- Molecular Biology Research Laboratory, Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, India
- Norris Comprehensive Cancer Center, Division of Medical Oncology, University of Southern California, Los Angeles, USA
- Institute of Eminence, Delhi School of Public Health, University of Delhi, Delhi, India
| |
Collapse
|
11
|
Gupta SRR, Ta TM, Khan M, Singh A, Singh IK, Peethambaran B. Identification and validation of a small molecule targeting ROR1 for the treatment of triple negative breast cancer. Front Cell Dev Biol 2023; 11:1243763. [PMID: 37779899 PMCID: PMC10534069 DOI: 10.3389/fcell.2023.1243763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/11/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction: Breast cancer is the most common cancer in women, with roughly 10-15% of new cases classified as triple-negative breast cancer (TNBC). Traditional chemotherapies are often toxic to normal cells. Therefore, it is important to discover new anticancer compounds that target TNBC while causing minimal damage to normal cells. Receptor tyrosine kinase-like Orphan Receptor 1 (ROR1) is an oncofetal protein overexpressed in numerous human malignancies, including TNBC. This study investigated potential small molecules targeting ROR1. Methodology: Using AutoDock Vina and Glide, we screened 70,000 chemicals for our investigation. We obtained 10 representative compounds via consensus voting, deleting structural alerts, and clustering. After manual assessment, compounds 2 and 4 were chosen for MD simulation and cell viability experiment. Compound 4 showed promising results in the viability assay, which led us to move further with the apoptosis assay and immunoblotting. Results: Compound 4 (CID1261330) had docking scores of -6.635 and -10.8. It fits into the pocket and shows interactions with GLU64, ASP174, and PHE93. Its RMSD fluctuates around 0.20 nm and forms two stable H-bonds indicating compound 4 stability. It inhibits cell proliferation in MDA-MB-231, HCC1937, and HCC1395 cell lines, with IC50 values of approximately 2 μM to 10 μM, respectively. Compound 4 did not kill non-malignant epithelial breast cells MCF-10A (IC50 > 27 μM). These results were confirmed by the significant number of apoptotic cells in MDA-MB-231 cells (47.6%) but not in MCF-10A cells (7.3%). Immunoblot analysis provided additional support in the same direction. Discussion: These findings collectively suggest that compound 4 has the potential to effectively eliminate TNBC cells while causing minimal harm to normal breast cells. The promising outcomes of this study lay the groundwork for further testing of compound 4 in other malignancies characterized by ROR1 upregulation, serving as a proof-of-concept for its broader applicability.
Collapse
Affiliation(s)
- Shradheya R. R. Gupta
- Molecular Biology Research Laboratory, Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, India
| | - Tram M. Ta
- Department of Biology, Saint Joseph’s University, Philadelphia, PA, United States
| | - Maryam Khan
- Department of Biology, Saint Joseph’s University, Philadelphia, PA, United States
| | - Archana Singh
- Department of Botany, Hans Raj College, University of Delhi, New Delhi, India
| | - Indrakant K. Singh
- Molecular Biology Research Laboratory, Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, India
- Delhi School of Public Health, Institute of Eminence, University of Delhi, New Delhi, India
- Norris Comprehensive Cancer Center, Division of Medical Oncology, University of Southern California, Los Angeles, CA, United States
| | - Bela Peethambaran
- Department of Biology, Saint Joseph’s University, Philadelphia, PA, United States
| |
Collapse
|
12
|
Kamerlin SCL. Progress in using deep learning to treat cancer. NATURE COMPUTATIONAL SCIENCE 2023; 3:739-740. [PMID: 38177785 DOI: 10.1038/s43588-023-00514-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
|
13
|
Riley CM, Elwood JML, Henry MC, Hunter I, Daniel Lopez-Fernandez J, McEwan IJ, Jamieson C. Current and emerging approaches to noncompetitive AR inhibition. Med Res Rev 2023; 43:1701-1747. [PMID: 37062876 DOI: 10.1002/med.21961] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/14/2023] [Accepted: 03/28/2023] [Indexed: 04/18/2023]
Abstract
The androgen receptor (AR) has been shown to be a key determinant in the pathogenesis of castration-resistant prostate cancer (CRPC). The current standard of care therapies targets the ligand-binding domain of the receptor and can afford improvements to life expectancy often only in the order of months before resistance occurs. Emerging preclinical and clinical compounds that inhibit receptor activity via differentiated mechanisms of action which are orthogonal to current antiandrogens show promise for overcoming treatment resistance. In this review, we present an authoritative summary of molecules that noncompetitively target the AR. Emerging small molecule strategies for targeting alternative domains of the AR represent a promising area of research that shows significant potential for future therapies. The overall quality of lead candidates in the area of noncompetitive AR inhibition is discussed, and it identifies the key chemotypes and associated properties which are likely to be, or are currently, positioned to be first in human applications.
Collapse
Affiliation(s)
- Christopher M Riley
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Jessica M L Elwood
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Martyn C Henry
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Irene Hunter
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | | | - Iain J McEwan
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Craig Jamieson
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| |
Collapse
|
14
|
Smith LG, Novak B, Osato M, Mobley DL, Bowman GR. PopShift: A thermodynamically sound approach to estimate binding free energies by accounting for ligand-induced population shifts from a ligand-free MSM. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.14.549110. [PMID: 37503302 PMCID: PMC10370083 DOI: 10.1101/2023.07.14.549110] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Obtaining accurate binding free energies from in silico screens has been a longstanding goal for the computational chemistry community. However, accuracy and computational cost are at odds with one another, limiting the utility of methods that perform this type of calculation. Many methods achieve massive scale by explicitly or implicitly assuming that the target protein adopts a single structure, or undergoes limited fluctuations around that structure, to minimize computational cost. Others simulate each protein-ligand complex of interest, accepting lower throughput in exchange for better predictions of binding affinities. Here, we present the PopShift framework for accounting for the ensemble of structures a protein adopts and their relative probabilities. Protein degrees of freedom are enumerated once, and then arbitrarily many molecules can be screened against this ensemble. Specifically, we use Markov state models (MSMs) as a compressed representation of a protein's thermodynamic ensemble. We start with a ligand-free MSM and then calculate how addition of a ligand shifts the populations of each protein conformational state based on the strength of the interaction between that protein conformation and the ligand. In this work we use docking to estimate the affinity between a given protein structure and ligand, but any estimator of binding affinities could be used in the PopShift framework. We test PopShift on the classic benchmark pocket T4 Lysozyme L99A. We find that PopShift is more accurate than common strategies, such as docking to a single structure and traditional ensemble docking-producing results that compare favorably with alchemical binding free energy calculations in terms of RMSE but not correlation - and may have a more favorable computational cost profile in some applications. In addition to predicting binding free energies and ligand poses, PopShift also provides insight into how the probability of different protein structures is shifted upon addition of various concentrations of ligand, providing a platform for predicting affinities and allosteric effects of ligand binding. Therefore, we expect PopShift will be valuable for hit finding and for providing insight into phenomena like allostery.
Collapse
Affiliation(s)
- Louis G Smith
- University of Pennsylvania, Depts. of Biochemistry & Biophysics and Bioengineering
| | - Borna Novak
- Washington University in St. Louis, Department of Biochemistry and Molecular Biophysics
- Medical Scientist Training Program, Washington University in St. Louis
| | - Meghan Osato
- University of California Irvine, School of Pharmacy and Pharmaceutical Sciences
| | - David L Mobley
- University of California Irvine, School of Pharmacy and Pharmaceutical Sciences
| | - Gregory R Bowman
- University of Pennsylvania, Depts. of Biochemistry & Biophysics and Bioengineering
| |
Collapse
|
15
|
Radaeva M, Morin H, Pandey M, Ban F, Guo M, LeBlanc E, Lallous N, Cherkasov A. Novel Inhibitors of androgen receptor's DNA binding domain identified using an ultra-large virtual screening. Mol Inform 2023; 42:e2300026. [PMID: 37193651 DOI: 10.1002/minf.202300026] [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: 01/27/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/18/2023]
Abstract
Androgen receptor (AR) inhibition remains the primary strategy to combat the progression of prostate cancer (PC). However, all clinically used AR inhibitors target the ligand-binding domain (LBD), which is highly susceptible to truncations through splicing or mutations that confer drug resistance. Thus, there exists an urgent need for AR inhibitors with novel modes of action. We thus launched a virtual screening of an ultra-large chemical library to find novel inhibitors of the AR DNA-binding domain (DBD) at two sites: protein-DNA interface (P-box) and dimerization site (D-box). The compounds selected through vigorous computational filtering were then experimentally validated. We identified several novel chemotypes that effectively suppress transcriptional activity of AR and its splice variant V7. The identified compounds represent previously unexplored chemical scaffolds with a mechanism of action that evades the conventional drug resistance manifested through LBD mutations. Additionally, we describe the binding features required to inhibit AR DBD at both P-box and D-box target sites.
Collapse
Affiliation(s)
- Mariia Radaeva
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia, Canada, V6H 3Z6
| | - Helene Morin
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia, Canada, V6H 3Z6
| | - Mohit Pandey
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia, Canada, V6H 3Z6
| | - Fuqiang Ban
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia, Canada, V6H 3Z6
| | - Maria Guo
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia, Canada, V6H 3Z6
| | - Eric LeBlanc
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia, Canada, V6H 3Z6
| | - Nada Lallous
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia, Canada, V6H 3Z6
| | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia, Canada, V6H 3Z6
| |
Collapse
|
16
|
Takada Y, Kaneko K. Automated machine learning approach for developing a quantitative structure-activity relationship model for cardiac steroid inhibition of Na +/K +-ATPase. Pharmacol Rep 2023:10.1007/s43440-023-00508-x. [PMID: 37354314 DOI: 10.1007/s43440-023-00508-x] [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: 03/27/2023] [Revised: 06/09/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND Quantitative structure-activity relationship (QSAR) modeling is a method of characterizing the relationship between chemical structures and biological activity. Automated machine learning enables computers to learn from large datasets and can be used for chemoinformatics. Cardiac steroids (CSs) inhibit the activity of Na+/K+-ATPase (NKA) in several species, including humans, since the binding pocket in which NKA binds to CSs is highly conserved. CSs are used to treat heart disease and have been developed into anticancer drugs for use in clinical trials. Novel CSs are, therefore, frequently synthesized and their activities evaluated. The purpose of this study is to develop a QSAR model via automated machine learning to predict the potential inhibitory activity of compounds without performing experiments. METHODS The chemical structures and inhibitory activities of 215 CS derivatives were obtained from the scientific literature. Predictive QSAR models were constructed using molecular descriptors, fingerprints, and biological activities. RESULTS The best predictive QSAR models were selected based on the LogLoss value. Using these models, the Matthews correlation coefficient, F1 score, and area under the curve of the test dataset were 0.6729, 0.8813, and 0.8812, respectively. Next, we showed automated construction of the predictive models for CS derivatives, which may be useful for identifying novel CSs suitable for candidate drug development. CONCLUSION The automated machine learning-based QSAR method developed here should be applicable for the time-efficient construction of predictive models using only a small number of compounds.
Collapse
Affiliation(s)
- Yohei Takada
- Corporate Planning Department, Otsuka Holdings Co., Ltd, Shinagawa Grand Central Tower 2-16-4 Konan, Minato-ku, Tokyo, 108-8241, Japan.
| | - Kazuhiro Kaneko
- Headquarters of Clinical Development, Otsuka Pharmaceutical Co., Ltd, Shinagawa Grand Central Tower 2-16-4 Konan, Minato-ku, Tokyo, 108-8241, Japan
| |
Collapse
|
17
|
Jat S, Bhatt M, Roychowdhury S, Dixit VA, Pawar SD, Kulhari H, Alexander A, Kumar P. Preparation and characterization of amoxapine- and naringin-loaded solid lipid nanoparticles: drug-release and molecular-docking studies. Nanomedicine (Lond) 2023; 17:2133-2144. [PMID: 36786368 DOI: 10.2217/nnm-2022-0167] [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: 02/15/2023] Open
Abstract
Aim: Amoxapine (AMX) has been reported to be metabolized by CYP3A4 and CYP2D6. Naringin (NG) has been reported to inhibit CYP enzymes. Therefore, the current work was designed to develop AMX solid lipid nanoparticles (AMX-SLNs) and NG-SLNs for better therapeutic performance. Materials & methods: AMX-SLNs and NG-SLNs were prepared and characterized. AMX and NG interactions with CYP450s were studied with molecular docking to rationalize the effectiveness of the combination. Results: AMX-SLNs and NG-SLNs showed nanometric size with a sustained in vitro drug-release profile. NG showed a higher predicted binding affinity for CYP3A4 and CYP2D6, suggesting the potential for inhibition. Conclusion: The developed formulations were thoroughly characterized along with molecular docking data indicating promising AMX and NG combinations that may show good therapeutic activity.
Collapse
Affiliation(s)
- Sandeep Jat
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education & Research, Guwahati, Sila Katamur (Halugurisuk), Changsari, Dist. Kamrup, Assam, 781101, India
| | - Manini Bhatt
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education & Research, Guwahati, Sila Katamur (Halugurisuk), Changsari, Dist. Kamrup, Assam, 781101, India
| | - Sanjana Roychowdhury
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education & Research, Guwahati, Sila Katamur (Halugurisuk), Changsari, Dist. Kamrup, Assam, 78110, India
| | - Vaibhav A Dixit
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education & Research, Guwahati, Sila Katamur (Halugurisuk), Changsari, Dist. Kamrup, Assam, 78110, India
| | - Sachin Dattram Pawar
- School of Nano Sciences, Central University of Gujarat, Gandhinagar, 382030, India
| | - Hitesh Kulhari
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education & Research, Guwahati, Sila Katamur (Halugurisuk), Changsari, Dist. Kamrup, Assam, 781101, India.,School of Nano Sciences, Central University of Gujarat, Gandhinagar, 382030, India
| | - Amit Alexander
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research, Guwahati, Sila Katamur (Halugurisuk), Changsari, Dist. Kamrup, Assam, 781101, India
| | - Pramod Kumar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education & Research, Guwahati, Sila Katamur (Halugurisuk), Changsari, Dist. Kamrup, Assam, 781101, India
| |
Collapse
|
18
|
Abstract
In the wake of recent COVID-19 pandemics scientists around the world rushed to deliver numerous CADD (Computer-Aided Drug Discovery) methods and tools that could be reliably used to discover novel drug candidates against the SARS-CoV-2 virus. With that, there emerged a trend of a significant democratization of CADD that contributed to the rapid development of various COVID-19 drug candidates currently undergoing different stages of validation. On the other hand, this democratization also inadvertently led to the surge rapidly performed molecular docking studies to nominate multiple scores of novel drug candidates supported by computational arguments only. Albeit driven by best intentions, most of such studies also did not follow best practices in the field that require experience and expertise learned through multiple rigorously designed benchmarking studies and rigorous experimental validation. In this Viewpoint we reflect on recent disbalance between small number of rigorous and comprehensive studies and the proliferation of purely computational studies enabled by the ease of docking software availability. We further elaborate on the hyped oversale of CADD methods' ability to rapidly yield viable drug candidates and reiterate the critical importance of rigor and adherence to the best practices of CADD in view of recent emergence of AI and Big Data in the field.
Collapse
Affiliation(s)
- F Gentile
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada
| | - T I Oprea
- Roivant Sciences Inc, 451 D Street, Boston, MA, USA
| | - A Tropsha
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - A Cherkasov
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
19
|
The 'Big Bang' of the chemical universe. Nat Chem Biol 2023; 19:667-668. [PMID: 36646955 DOI: 10.1038/s41589-022-01233-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
20
|
Cytotoxic Potential of Bioactive Compounds from Aspergillus flavus, an Endophytic Fungus Isolated from Cynodon dactylon, against Breast Cancer: Experimental and Computational Approach. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248814. [PMID: 36557944 PMCID: PMC9784115 DOI: 10.3390/molecules27248814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
Endophytic fungi are a diverse group of microorganisms that colonize the inter- or intracellular spaces of plants and exhibit mutual benefits. Their interactions with the host plant and other microbiomes are multidimensional and play a crucial role in the production of secondary metabolites. We screened bioactive compounds present in the extracts of Aspergillus flavus, an endophytic fungus isolated from the roots of the medicinal grass Cynodon dactylon, for its anticancer potential. An in vitro analysis of the Ethyl acetate extract from A. flavus showed significant cytostatic effects (IC50: 16.25 μg/mL) against breast cancer cells (MCF-7). A morphological analysis of the cells and a flow cytometry of the cells with annexin V/Propidium Iodide suggested that the extract induced apoptosis in the MCF-7 cells. The extract of A. flavus increased reactive oxygen species (ROS) generation and caused a loss of mitochondrial membrane potential in MCF-7 cells. To identify the metabolites that might be responsible for the anticancer effect, the extract was subjected to a gas chromatography-mass spectrometry (GC-MS) analysis. Interestingly, nine phytochemicals that induced cytotoxicity in the breast cancer cell line were found in the extract. The in silico molecular docking and molecular dynamics simulation studies revealed that two compounds, 2,4,7-trinitrofluorenone and 3α, 5 α-cyclo-ergosta-7,9(11), 22t-triene-6beta-ol exhibited significant binding affinities (-9.20, and -9.50 Kcal/mol, respectively) against Bcl-2, along with binding stability and intermolecular interactions of its ligand-Bcl-2 complexes. Overall, the study found that the endophytic A. flavus from C. dactylon contains plant-like bioactive compounds that have a promising effect in breast cancer.
Collapse
|
21
|
Design, Synthesis and Anticancer Screening of Cu-Catalyzed SnAr Substituted Pyridine Bridged Ring Systems. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
22
|
Patil P, N NB, Satyanarayan ND, Pore S, Zond R, Hangirgekar AGS, Sankpal S. Design, synthesis, docking studies and anticancer evaluation of spiro[indoline-3,4′-pyrano[2,3-c]pyrazole] derivatives on MIN-6 cancer cell line. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
23
|
Choi SYC, Ribeiro CF, Wang Y, Loda M, Plymate SR, Uo T. Druggable Metabolic Vulnerabilities Are Exposed and Masked during Progression to Castration Resistant Prostate Cancer. Biomolecules 2022; 12:1590. [PMID: 36358940 PMCID: PMC9687810 DOI: 10.3390/biom12111590] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 08/27/2023] Open
Abstract
There is an urgent need for exploring new actionable targets other than androgen receptor to improve outcome from lethal castration-resistant prostate cancer. Tumor metabolism has reemerged as a hallmark of cancer that drives and supports oncogenesis. In this regard, it is important to understand the relationship between distinctive metabolic features, androgen receptor signaling, genetic drivers in prostate cancer, and the tumor microenvironment (symbiotic and competitive metabolic interactions) to identify metabolic vulnerabilities. We explore the links between metabolism and gene regulation, and thus the unique metabolic signatures that define the malignant phenotypes at given stages of prostate tumor progression. We also provide an overview of current metabolism-based pharmacological strategies to be developed or repurposed for metabolism-based therapeutics for castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Stephen Y. C. Choi
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Caroline Fidalgo Ribeiro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY 10021, USA
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY 10021, USA
- New York Genome Center, New York, NY 10013, USA
| | - Stephen R. Plymate
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, 850 Republican St., Seattle, WA 98109, USA
- Geriatrics Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Takuma Uo
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, 850 Republican St., Seattle, WA 98109, USA
| |
Collapse
|
24
|
Design and synthesis of novel tetrazolo quinoline bridged isatin derivatives as potential anticancer leads against MIA PaCa-2 human pancreatic cancer cell line. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
25
|
Kallert E, Fischer TR, Schneider S, Grimm M, Helm M, Kersten C. Protein-Based Virtual Screening Tools Applied for RNA-Ligand Docking Identify New Binders of the preQ 1-Riboswitch. J Chem Inf Model 2022; 62:4134-4148. [PMID: 35994617 DOI: 10.1021/acs.jcim.2c00751] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Targeting RNA with small molecules is an emerging field. While several ligands for different RNA targets are reported, structure-based virtual screenings (VSs) against RNAs are still rare. Here, we elucidated the general capabilities of protein-based docking programs to reproduce native binding modes of small-molecule RNA ligands and to discriminate known binders from decoys by the scoring function. The programs were found to perform similar compared to the RNA-based docking tool rDOCK, and the challenges faced during docking, namely, protomer and tautomer selection, target dynamics, and explicit solvent, do not largely differ from challenges in conventional protein-ligand docking. A prospective VS with the Bacillus subtilis preQ1-riboswitch aptamer domain performed with FRED, HYBRID, and FlexX followed by microscale thermophoresis assays identified six active compounds out of 23 tested VS hits with potencies between 29.5 nM and 11.0 μM. The hits were selected not solely based on their docking score but for resembling key interactions of the native ligand. Therefore, this study demonstrates the general feasibility to perform structure-based VSs against RNA targets, while at the same time it highlights pitfalls and their potential solutions when executing RNA-ligand docking.
Collapse
Affiliation(s)
- Elisabeth Kallert
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - Tim R Fischer
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - Simon Schneider
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - Maike Grimm
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - Mark Helm
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - Christian Kersten
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| |
Collapse
|
26
|
Pang JP, Shen C, Zhou WF, Wang YX, Shan LH, Chai X, Shao Y, Hu XP, Zhu F, Zhu DY, Xiao L, Xu L, Xu XH, Li D, Hou TJ. Discovery of novel antagonists targeting the DNA binding domain of androgen receptor by integrated docking-based virtual screening and bioassays. Acta Pharmacol Sin 2022; 43:229-239. [PMID: 33767381 PMCID: PMC8724294 DOI: 10.1038/s41401-021-00632-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/24/2021] [Indexed: 02/01/2023] Open
Abstract
Androgen receptor (AR), a ligand-activated transcription factor, is a master regulator in the development and progress of prostate cancer (PCa). A major challenge for the clinically used AR antagonists is the rapid emergence of resistance induced by the mutations at AR ligand binding domain (LBD), and therefore the discovery of novel anti-AR therapeutics that can combat mutation-induced resistance is quite demanding. Therein, blocking the interaction between AR and DNA represents an innovative strategy. However, the hits confirmed targeting on it so far are all structurally based on a sole chemical scaffold. In this study, an integrated docking-based virtual screening (VS) strategy based on the crystal structure of the DNA binding domain (DBD) of AR was conducted to search for novel AR antagonists with new scaffolds and 2-(2-butyl-1,3-dioxoisoindoline-5-carboxamido)-4,5-dimethoxybenzoicacid (Cpd39) was identified as a potential hit, which was competent to block the binding of AR DBD to DNA and showed decent potency against AR transcriptional activity. Furthermore, Cpd39 was safe and capable of effectively inhibiting the proliferation of PCa cell lines (i.e., LNCaP, PC3, DU145, and 22RV1) and reducing the expression of the genes regulated by not only the full-length AR but also the splice variant AR-V7. The novel AR DBD-ARE blocker Cpd39 could serve as a starting point for the development of new therapeutics for castration-resistant PCa.
Collapse
Affiliation(s)
- Jin-Ping Pang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chao Shen
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wen-Fang Zhou
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yun-Xia Wang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Lu-Hu Shan
- Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Xin Chai
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ying Shao
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xue-Ping Hu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Feng Zhu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Dan-Yan Zhu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Li Xiao
- School of Life Science, Huzhou University, Huzhou, 313000, China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, 213001, China
| | - Xiao-Hong Xu
- Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Dan Li
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Ting-Jun Hou
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- State Key Lab of CAD & CG, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
27
|
Gentile F, Fernandez M, Ban F, Ton AT, Mslati H, Perez CF, Leblanc E, Yaacoub JC, Gleave J, Stern A, Wong B, Jean F, Strynadka N, Cherkasov A. Automated discovery of noncovalent inhibitors of SARS-CoV-2 main protease by consensus Deep Docking of 40 billion small molecules. Chem Sci 2021; 12:15960-15974. [PMID: 35024120 PMCID: PMC8672713 DOI: 10.1039/d1sc05579h] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 11/12/2021] [Indexed: 12/24/2022] Open
Abstract
Recent explosive growth of 'make-on-demand' chemical libraries brought unprecedented opportunities but also significant challenges to the field of computer-aided drug discovery. To address this expansion of the accessible chemical universe, molecular docking needs to accurately rank billions of chemical structures, calling for the development of automated hit-selecting protocols to minimize human intervention and error. Herein, we report the development of an artificial intelligence-driven virtual screening pipeline that utilizes Deep Docking with Autodock GPU, Glide SP, FRED, ICM and QuickVina2 programs to screen 40 billion molecules against SARS-CoV-2 main protease (Mpro). This campaign returned a significant number of experimentally confirmed inhibitors of Mpro enzyme, and also enabled to benchmark the performance of twenty-eight hit-selecting strategies of various degrees of stringency and automation. These findings provide new starting scaffolds for hit-to-lead optimization campaigns against Mpro and encourage the development of fully automated end-to-end drug discovery protocols integrating machine learning and human expertise.
Collapse
Affiliation(s)
- Francesco Gentile
- Vancouver Prostate Centre, Department of Urologic Sciences, The University of British Columbia 2660 Oak Street Vancouver BC V6H3Z6 Canada
| | - Michael Fernandez
- Vancouver Prostate Centre, Department of Urologic Sciences, The University of British Columbia 2660 Oak Street Vancouver BC V6H3Z6 Canada
| | - Fuqiang Ban
- Vancouver Prostate Centre, Department of Urologic Sciences, The University of British Columbia 2660 Oak Street Vancouver BC V6H3Z6 Canada
| | - Anh-Tien Ton
- Vancouver Prostate Centre, Department of Urologic Sciences, The University of British Columbia 2660 Oak Street Vancouver BC V6H3Z6 Canada
| | - Hazem Mslati
- Vancouver Prostate Centre, Department of Urologic Sciences, The University of British Columbia 2660 Oak Street Vancouver BC V6H3Z6 Canada
| | - Carl F Perez
- Vancouver Prostate Centre, Department of Urologic Sciences, The University of British Columbia 2660 Oak Street Vancouver BC V6H3Z6 Canada
| | - Eric Leblanc
- Vancouver Prostate Centre, Department of Urologic Sciences, The University of British Columbia 2660 Oak Street Vancouver BC V6H3Z6 Canada
| | - Jean Charle Yaacoub
- Vancouver Prostate Centre, Department of Urologic Sciences, The University of British Columbia 2660 Oak Street Vancouver BC V6H3Z6 Canada
| | - James Gleave
- Vancouver Prostate Centre, Department of Urologic Sciences, The University of British Columbia 2660 Oak Street Vancouver BC V6H3Z6 Canada
| | | | | | - François Jean
- Department of Microbiology and Immunology, The University of British Columbia Vancouver BC Canada
| | - Natalie Strynadka
- Department of Biochemistry and Molecular Biology, The University of British Columbia Vancouver BC Canada
| | - Artem Cherkasov
- Vancouver Prostate Centre, Department of Urologic Sciences, The University of British Columbia 2660 Oak Street Vancouver BC V6H3Z6 Canada
| |
Collapse
|
28
|
[MerDABCO-SO3H]Cl catalyzed synthesis, antimicrobial and antioxidant evaluation and molecular docking study of pyrazolopyranopyrimidines. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130672] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
Taxifolin Targets PI3K and mTOR and Inhibits Glioblastoma Multiforme. JOURNAL OF ONCOLOGY 2021; 2021:5560915. [PMID: 34462635 PMCID: PMC8403040 DOI: 10.1155/2021/5560915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 07/31/2021] [Indexed: 01/12/2023]
Abstract
Glioblastoma multiforme (GBM), the most common malignant primary brain tumor, has a very poor prognosis. With increasing knowledge of tumor molecular biology, targeted therapies are becoming increasingly integral to comprehensive GBM treatment strategies. mTOR is a key downstream molecule of the PI3K/Akt signaling pathway, integrating input signals from growth factors, nutrients, and energy sources to regulate cell growth and cell proliferation through multiple cellular responses. mTOR/PI3K dual-targeted therapy has shown promise in managing various cancers. Here, we report that taxifolin, a flavanone commonly found in milk thistle, inhibited mTOR/PI3K, promoted autophagy, and suppressed lipid synthesis in GBM. In silico analysis showed that taxifolin can bind to the rapamycin binding site of mTOR and the catalytic site of PI3K (p110α). In in vitro experiments, taxifolin inhibited mTOR and PI3K activity in five different glioma cell lines. Lastly, we showed that taxifolin suppressed tumors in mice; stimulated expression of autophagy-related genes LC3B-II, Atg7, atg12, and Beclin-1; and inhibited expression of fatty acid synthesis-related genes C/EBPα, PPARγ, FABP4, and FAS. Our observations suggest that taxifolin is potentially a valuable drug for treating GBM.
Collapse
|
30
|
Sisquellas M, Cecchini M. PrepFlow: A Toolkit for Chemical Library Preparation and Management for Virtual Screening. Mol Inform 2021; 40:e2100139. [PMID: 34448369 DOI: 10.1002/minf.202100139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/13/2021] [Indexed: 11/10/2022]
Abstract
In the era of big data in Chemistry, the need for automated tools for virtual screening is compelling. Here, we present PrepFlow a toolkit for chemical library preparation and management. Starting from a list of compounds in SMILES or 2D molecular format, PrepFlow outputs a set of 3D molecular structures ready for use in subsequent drug discovery projects. Our development stands out for speed and robustness of execution, the efficient exploitation of HPC resources, and the implementation of an archiving strategy to save computer time, storage, and human intervention. Using a random selection of 600 compounds from available drug banks, we show that the preparation time per ligand on a desktop computer is 6.6 s. Thanks to these performances and the automatic parallelization on HPC, a chemical library of the size of ChEMBL (2 M) was prepared in around 3 days on a computer cluster. PrepFlow is freely distributed at the following link: https://ifm.chimie.unistra.fr/prepflow.
Collapse
Affiliation(s)
- Marion Sisquellas
- Institut de Chimie de Strasbourg, UMR7177, CNRS, Université de Strasbourg, F-67083, Strasbourg Cedex, France
| | - Marco Cecchini
- Institut de Chimie de Strasbourg, UMR7177, CNRS, Université de Strasbourg, F-67083, Strasbourg Cedex, France
| |
Collapse
|
31
|
Muratov EN, Amaro R, Andrade CH, Brown N, Ekins S, Fourches D, Isayev O, Kozakov D, Medina-Franco JL, Merz KM, Oprea TI, Poroikov V, Schneider G, Todd MH, Varnek A, Winkler DA, Zakharov AV, Cherkasov A, Tropsha A. A critical overview of computational approaches employed for COVID-19 drug discovery. Chem Soc Rev 2021; 50:9121-9151. [PMID: 34212944 PMCID: PMC8371861 DOI: 10.1039/d0cs01065k] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Indexed: 01/18/2023]
Abstract
COVID-19 has resulted in huge numbers of infections and deaths worldwide and brought the most severe disruptions to societies and economies since the Great Depression. Massive experimental and computational research effort to understand and characterize the disease and rapidly develop diagnostics, vaccines, and drugs has emerged in response to this devastating pandemic and more than 130 000 COVID-19-related research papers have been published in peer-reviewed journals or deposited in preprint servers. Much of the research effort has focused on the discovery of novel drug candidates or repurposing of existing drugs against COVID-19, and many such projects have been either exclusively computational or computer-aided experimental studies. Herein, we provide an expert overview of the key computational methods and their applications for the discovery of COVID-19 small-molecule therapeutics that have been reported in the research literature. We further outline that, after the first year the COVID-19 pandemic, it appears that drug repurposing has not produced rapid and global solutions. However, several known drugs have been used in the clinic to cure COVID-19 patients, and a few repurposed drugs continue to be considered in clinical trials, along with several novel clinical candidates. We posit that truly impactful computational tools must deliver actionable, experimentally testable hypotheses enabling the discovery of novel drugs and drug combinations, and that open science and rapid sharing of research results are critical to accelerate the development of novel, much needed therapeutics for COVID-19.
Collapse
Affiliation(s)
- Eugene N. Muratov
- UNC Eshelman School of Pharmacy, University of North CarolinaChapel HillNCUSA
| | - Rommie Amaro
- University of California in San DiegoSan DiegoCAUSA
| | | | | | - Sean Ekins
- Collaborations PharmaceuticalsRaleighNCUSA
| | - Denis Fourches
- Department of Chemistry, North Carolina State UniversityRaleighNCUSA
| | - Olexandr Isayev
- Department of Chemistry, Carnegie Melon UniversityPittsburghPAUSA
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook UniversityStony BrookNYUSA
| | | | - Kenneth M. Merz
- Department of Chemistry, Michigan State UniversityEast LansingMIUSA
| | - Tudor I. Oprea
- Department of Internal Medicine and UNM Comprehensive Cancer Center, University of New Mexico, AlbuquerqueNMUSA
- Department of Rheumatology and Inflammation Research, Gothenburg UniversitySweden
- Novo Nordisk Foundation Center for Protein Research, University of CopenhagenDenmark
| | | | - Gisbert Schneider
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of TechnologyZurichSwitzerland
| | | | - Alexandre Varnek
- Department of Chemistry, University of StrasbourgStrasbourgFrance
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido UniversitySapporoJapan
| | - David A. Winkler
- Monash Institute of Pharmaceutical Sciences, Monash UniversityMelbourneVICAustralia
- School of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe UniversityBundooraAustralia
- School of Pharmacy, University of NottinghamNottinghamUK
| | | | - Artem Cherkasov
- Vancouver Prostate Centre, University of British ColumbiaVancouverBCCanada
| | - Alexander Tropsha
- UNC Eshelman School of Pharmacy, University of North CarolinaChapel HillNCUSA
| |
Collapse
|
32
|
Computational Chemistry in the Undergraduate Classroom – Pedagogical Considerations and Teaching Challenges. Isr J Chem 2021. [DOI: 10.1002/ijch.202100042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
33
|
Iusupov IR, Curreli F, Spiridonov EA, Markov PO, Ahmed S, Belov DS, Manasova EV, Altieri A, Kurkin AV, Debnath AK. Design of gp120 HIV-1 entry inhibitors by scaffold hopping via isosteric replacements. Eur J Med Chem 2021; 224:113681. [PMID: 34246921 DOI: 10.1016/j.ejmech.2021.113681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 10/20/2022]
Abstract
We present the development of alternative scaffolds and validation of their synthetic pathways as a tool for the exploration of new HIV gp120 inhibitors based on the recently discovered inhibitor of this class, NBD-14136. The new synthetic routes were based on isosteric replacements of the amine and acid precursors required for the synthesis of NBD-14136, guided by molecular modeling and chemical feasibility analysis. To ensure that these synthetic tools and new scaffolds had the potential for further exploration, we eventually tested few representative compounds from each newly designed scaffold against the gp120 inhibition assay and cell viability assays.
Collapse
Affiliation(s)
- Ildar R Iusupov
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia
| | - Francesca Curreli
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute, New York Blood Center, 310 E 67th Street, New York, 10065, New York, United States
| | - Evgeniy A Spiridonov
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia
| | - Pavel O Markov
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia
| | - Shahad Ahmed
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute, New York Blood Center, 310 E 67th Street, New York, 10065, New York, United States
| | - Dmitry S Belov
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia
| | - Ekaterina V Manasova
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia
| | - Andrea Altieri
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia.
| | - Alexander V Kurkin
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia.
| | - Asim K Debnath
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute, New York Blood Center, 310 E 67th Street, New York, 10065, New York, United States.
| |
Collapse
|
34
|
Temml V, Kutil Z. Structure-based molecular modeling in SAR analysis and lead optimization. Comput Struct Biotechnol J 2021; 19:1431-1444. [PMID: 33777339 PMCID: PMC7979990 DOI: 10.1016/j.csbj.2021.02.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
In silico methods like molecular docking and pharmacophore modeling are established strategies in lead identification. Their successful application for finding new active molecules for a target is reported by a plethora of studies. However, once a potential lead is identified, lead optimization, with the focus on improving potency, selectivity, or pharmacokinetic parameters of a parent compound, is a much more complex task. Even though in silico molecular modeling methods could contribute a lot of time and cost-saving by rationally filtering synthetic optimization options, they are employed less widely in this stage of research. In this review, we highlight studies that have successfully used computer-aided SAR analysis in lead optimization and want to showcase sound methodology and easily accessible in silico tools for this purpose.
Collapse
Affiliation(s)
- Veronika Temml
- Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria
| | - Zsofia Kutil
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| |
Collapse
|
35
|
Guha R, Willighagen E, Zdrazil B, Jeliazkova N. What is the role of cheminformatics in a pandemic? J Cheminform 2021; 13:16. [PMID: 33653411 PMCID: PMC7922726 DOI: 10.1186/s13321-021-00491-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 01/22/2021] [Indexed: 11/10/2022] Open
Affiliation(s)
- Rajarshi Guha
- Vertex Pharmaceuticals, 50 Northern Ave, Boston, MA, 02210, USA.
| | - Egon Willighagen
- Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, Netherlands
| | - Barbara Zdrazil
- University of Vienna, Althanstraße 14, 1090, Vienna, Austria
| | | |
Collapse
|
36
|
Development of Novel Inhibitors Targeting the D-Box of the DNA Binding Domain of Androgen Receptor. Int J Mol Sci 2021; 22:ijms22052493. [PMID: 33801338 PMCID: PMC7958344 DOI: 10.3390/ijms22052493] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/25/2021] [Accepted: 02/27/2021] [Indexed: 01/01/2023] Open
Abstract
The inhibition of the androgen receptor (AR) is an established strategy in prostate cancer (PCa) treatment until drug resistance develops either through mutations in the ligand-binding domain (LBD) portion of the receptor or its deletion. We previously identified a druggable pocket on the DNA binding domain (DBD) dimerization surface of the AR and reported several potent inhibitors that effectively disrupted DBD-DBD interactions and consequently demonstrated certain antineoplastic activity. Here we describe further development of small molecule inhibitors of AR DBD dimerization and provide their broad biological characterization. The developed compounds demonstrate improved activity in the mammalian two-hybrid assay, enhanced inhibition of AR-V7 transcriptional activity, and improved microsomal stability. These findings position us for the development of AR inhibitors with entirely novel mechanisms of action that would bypass most forms of PCa treatment resistance, including the truncation of the LBD of the AR.
Collapse
|
37
|
Matias-Barrios VM, Radaeva M, Song Y, Alperstein Z, Lee AR, Schmitt V, Lee J, Ban F, Xie N, Qi J, Lallous N, Gleave ME, Cherkasov A, Dong X. Discovery of New Catalytic Topoisomerase II Inhibitors for Anticancer Therapeutics. Front Oncol 2021; 10:633142. [PMID: 33598437 PMCID: PMC7883873 DOI: 10.3389/fonc.2020.633142] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 12/15/2020] [Indexed: 01/23/2023] Open
Abstract
Poison inhibitors of DNA topoisomerase II (TOP2) are clinically used drugs that cause cancer cell death by inducing DNA damage, which mechanism of action is also associated with serious side effects such as secondary malignancy and cardiotoxicity. In contrast, TOP2 catalytic inhibitors induce limited DNA damage, have low cytotoxicity, and are effective in suppressing cancer cell proliferation. They have been sought after to be prospective anticancer therapies. Herein the discovery of new TOP2 catalytic inhibitors is described. A new druggable pocket of TOP2 protein at its DNA binding domain was used as a docking site to virtually screen ~6 million molecules from the ZINC15 library. The lead compound, T60, was characterized to be a catalytic TOP2 inhibitor that binds TOP2 protein and disrupts TOP2 from interacting with DNA, resulting in no DNA cleavage. It has low cytotoxicity, but strongly inhibits cancer cell proliferation and xenograft growth. T60 also inhibits androgen receptor activity and prostate cancer cell growth. These results indicate that T60 is a promising candidate compound that can be further developed into new anticancer drugs.
Collapse
Affiliation(s)
- Victor M Matias-Barrios
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Mariia Radaeva
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Yi Song
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada.,Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zaccary Alperstein
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ahn R Lee
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Veronika Schmitt
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Joseph Lee
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Fuqiang Ban
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ning Xie
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jianfei Qi
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Nada Lallous
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Martin E Gleave
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Artem Cherkasov
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Xuesen Dong
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
38
|
Chemoinformatics and QSAR. Adv Bioinformatics 2021. [DOI: 10.1007/978-981-33-6191-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
39
|
Macari G, Toti D, Pasquadibisceglie A, Polticelli F. DockingApp RF: A State-of-the-Art Novel Scoring Function for Molecular Docking in a User-Friendly Interface to AutoDock Vina. Int J Mol Sci 2020; 21:ijms21249548. [PMID: 33333976 PMCID: PMC7765429 DOI: 10.3390/ijms21249548] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 11/28/2022] Open
Abstract
Motivation: Bringing a new drug to the market is expensive and time-consuming. To cut the costs and time, computer-aided drug design (CADD) approaches have been increasingly included in the drug discovery pipeline. However, despite traditional docking tools show a good conformational space sampling ability, they are still unable to produce accurate binding affinity predictions. This work presents a novel scoring function for molecular docking seamlessly integrated into DockingApp, a user-friendly graphical interface for AutoDock Vina. The proposed function is based on a random forest model and a selection of specific features to overcome the existing limits of Vina’s original scoring mechanism. A novel version of DockingApp, named DockingApp RF, has been developed to host the proposed scoring function and to automatize the rescoring procedure of the output of AutoDock Vina, even to nonexpert users. Results: By coupling intermolecular interaction, solvent accessible surface area features and Vina’s energy terms, DockingApp RF’s new scoring function is able to improve the binding affinity prediction of AutoDock Vina. Furthermore, comparison tests carried out on the CASF-2013 and CASF-2016 datasets demonstrate that DockingApp RF’s performance is comparable to other state-of-the-art machine-learning- and deep-learning-based scoring functions. The new scoring function thus represents a significant advancement in terms of the reliability and effectiveness of docking compared to AutoDock Vina’s scoring function. At the same time, the characteristics that made DockingApp appealing to a wide range of users are retained in this new version and have been complemented with additional features.
Collapse
Affiliation(s)
- Gabriele Macari
- Department of Sciences, Roma Tre University, 00146 Rome, Italy; (G.M.); (A.P.)
| | - Daniele Toti
- Faculty of Mathematical, Physical and Natural Sciences, Catholic University of the Sacred Heart, 25121 Brescia, Italy;
| | | | - Fabio Polticelli
- Department of Sciences, Roma Tre University, 00146 Rome, Italy; (G.M.); (A.P.)
- National Institute of Nuclear Physics, Roma Tre Section, 00146 Rome, Italy
- Correspondence:
| |
Collapse
|
40
|
Mermer A. Design, synthesize and antiurease activity of novel thiazole derivatives: Machine learning, molecular docking and biological investigation. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
41
|
Huang M, Bolin S, Miller H, Ng HL. RORγ Structural Plasticity and Druggability. Int J Mol Sci 2020; 21:ijms21155329. [PMID: 32727079 PMCID: PMC7432406 DOI: 10.3390/ijms21155329] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/08/2020] [Accepted: 07/15/2020] [Indexed: 12/19/2022] Open
Abstract
Retinoic acid receptor-related orphan receptor γ (RORγ) is a transcription factor regulating the expression of the pro-inflammatory cytokine IL-17 in human T helper 17 (Th17) cells. Activating RORγ can induce multiple IL-17-mediated autoimmune diseases but may also be useful for anticancer therapy. Its deep immunological functions make RORɣ an attractive drug target. Over 100 crystal structures have been published describing atomic interactions between RORɣ and agonists and inverse agonists. In this review, we focus on the role of dynamic properties and plasticity of the RORɣ orthosteric and allosteric binding sites by examining structural information from crystal structures and simulated models. We discuss the possible influences of allosteric ligands on the orthosteric binding site. We find that high structural plasticity favors the druggability of RORɣ, especially for allosteric ligands.
Collapse
Affiliation(s)
- Mian Huang
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA; (M.H.); (H.M.)
| | - Shelby Bolin
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA;
| | - Hannah Miller
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA; (M.H.); (H.M.)
| | - Ho Leung Ng
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA; (M.H.); (H.M.)
- Correspondence:
| |
Collapse
|
42
|
Radaeva M, Dong X, Cherkasov A. The Use of Methods of Computer-Aided Drug Discovery in the Development of Topoisomerase II Inhibitors: Applications and Future Directions. J Chem Inf Model 2020; 60:3703-3721. [DOI: 10.1021/acs.jcim.0c00325] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Mariia Radaeva
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Xuesen Dong
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| |
Collapse
|
43
|
Gentile F, Agrawal V, Hsing M, Ton AT, Ban F, Norinder U, Gleave ME, Cherkasov A. Deep Docking: A Deep Learning Platform for Augmentation of Structure Based Drug Discovery. ACS CENTRAL SCIENCE 2020; 6:939-949. [PMID: 32607441 PMCID: PMC7318080 DOI: 10.1021/acscentsci.0c00229] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Indexed: 05/06/2023]
Abstract
Drug discovery is a rigorous process that requires billion dollars of investments and decades of research to bring a molecule "from bench to a bedside". While virtual docking can significantly accelerate the process of drug discovery, it ultimately lags the current rate of expansion of chemical databases that already exceed billions of molecular records. This recent surge of small molecules availability presents great drug discovery opportunities, but also demands much faster screening protocols. In order to address this challenge, we herein introduce Deep Docking (DD), a novel deep learning platform that is suitable for docking billions of molecular structures in a rapid, yet accurate fashion. The DD approach utilizes quantitative structure-activity relationship (QSAR) deep models trained on docking scores of subsets of a chemical library to approximate the docking outcome for yet unprocessed entries and, therefore, to remove unfavorable molecules in an iterative manner. The use of DD methodology in conjunction with the FRED docking program allowed rapid and accurate calculation of docking scores for 1.36 billion molecules from the ZINC15 library against 12 prominent target proteins and demonstrated up to 100-fold data reduction and 6000-fold enrichment of high scoring molecules (without notable loss of favorably docked entities). The DD protocol can readily be used in conjunction with any docking program and was made publicly available.
Collapse
Affiliation(s)
- Francesco Gentile
- Vancouver
Prostate Centre, University of British Columbia, Vancouver, British Columbia V6H3Z6, Canada
| | - Vibudh Agrawal
- Vancouver
Prostate Centre, University of British Columbia, Vancouver, British Columbia V6H3Z6, Canada
| | - Michael Hsing
- Vancouver
Prostate Centre, University of British Columbia, Vancouver, British Columbia V6H3Z6, Canada
| | - Anh-Tien Ton
- Vancouver
Prostate Centre, University of British Columbia, Vancouver, British Columbia V6H3Z6, Canada
| | - Fuqiang Ban
- Vancouver
Prostate Centre, University of British Columbia, Vancouver, British Columbia V6H3Z6, Canada
| | - Ulf Norinder
- Swetox,
Unit of Toxicology Sciences, Karolinska
Institutet, Forskargatan
20, SE-151 36 Södertalje, Sweden
- Department
of Computer and Systems Sciences, Stockholm
University, Box 7003, SE-164
07 Kista, Sweden
| | - Martin E. Gleave
- Vancouver
Prostate Centre, University of British Columbia, Vancouver, British Columbia V6H3Z6, Canada
| | - Artem Cherkasov
- Vancouver
Prostate Centre, University of British Columbia, Vancouver, British Columbia V6H3Z6, Canada
| |
Collapse
|
44
|
Bafna D, Ban F, Rennie PS, Singh K, Cherkasov A. Computer-Aided Ligand Discovery for Estrogen Receptor Alpha. Int J Mol Sci 2020; 21:E4193. [PMID: 32545494 PMCID: PMC7352601 DOI: 10.3390/ijms21124193] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/30/2020] [Accepted: 06/09/2020] [Indexed: 02/08/2023] Open
Abstract
Breast cancer (BCa) is one of the most predominantly diagnosed cancers in women. Notably, 70% of BCa diagnoses are Estrogen Receptor α positive (ERα+) making it a critical therapeutic target. With that, the two subtypes of ER, ERα and ERβ, have contrasting effects on BCa cells. While ERα promotes cancerous activities, ERβ isoform exhibits inhibitory effects on the same. ER-directed small molecule drug discovery for BCa has provided the FDA approved drugs tamoxifen, toremifene, raloxifene and fulvestrant that all bind to the estrogen binding site of the receptor. These ER-directed inhibitors are non-selective in nature and may eventually induce resistance in BCa cells as well as increase the risk of endometrial cancer development. Thus, there is an urgent need to develop novel drugs with alternative ERα targeting mechanisms that can overcome the limitations of conventional anti-ERα therapies. Several functional sites on ERα, such as Activation Function-2 (AF2), DNA binding domain (DBD), and F-domain, have been recently considered as potential targets in the context of drug research and discovery. In this review, we summarize methods of computer-aided drug design (CADD) that have been employed to analyze and explore potential targetable sites on ERα, discuss recent advancement of ERα inhibitor development, and highlight the potential opportunities and challenges of future ERα-directed drug discovery.
Collapse
Affiliation(s)
| | | | | | | | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada; (D.B.); (F.B.); (P.S.R.); (K.S.)
| |
Collapse
|
45
|
Muratov EN, Bajorath J, Sheridan RP, Tetko IV, Filimonov D, Poroikov V, Oprea TI, Baskin II, Varnek A, Roitberg A, Isayev O, Curtarolo S, Fourches D, Cohen Y, Aspuru-Guzik A, Winkler DA, Agrafiotis D, Cherkasov A, Tropsha A. QSAR without borders. Chem Soc Rev 2020; 49:3525-3564. [PMID: 32356548 PMCID: PMC8008490 DOI: 10.1039/d0cs00098a] [Citation(s) in RCA: 338] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Prediction of chemical bioactivity and physical properties has been one of the most important applications of statistical and more recently, machine learning and artificial intelligence methods in chemical sciences. This field of research, broadly known as quantitative structure-activity relationships (QSAR) modeling, has developed many important algorithms and has found a broad range of applications in physical organic and medicinal chemistry in the past 55+ years. This Perspective summarizes recent technological advances in QSAR modeling but it also highlights the applicability of algorithms, modeling methods, and validation practices developed in QSAR to a wide range of research areas outside of traditional QSAR boundaries including synthesis planning, nanotechnology, materials science, biomaterials, and clinical informatics. As modern research methods generate rapidly increasing amounts of data, the knowledge of robust data-driven modelling methods professed within the QSAR field can become essential for scientists working both within and outside of chemical research. We hope that this contribution highlighting the generalizable components of QSAR modeling will serve to address this challenge.
Collapse
Affiliation(s)
- Eugene N Muratov
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Hu X, Chai X, Wang X, Duan M, Pang J, Fu W, Li D, Hou T. Advances in the computational development of androgen receptor antagonists. Drug Discov Today 2020; 25:1453-1461. [PMID: 32439609 DOI: 10.1016/j.drudis.2020.04.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/16/2020] [Accepted: 04/04/2020] [Indexed: 12/18/2022]
Abstract
The androgen receptor is a ligand-dependent transcriptional factor and an essential therapeutic target for prostate cancer. Competitive binding of antagonists to the androgen receptor can alleviate aberrant activation of the androgen receptor in prostate cancer. In recent years, computer-aided drug design has played an essential part in the discovery of novel androgen receptor antagonists. This review summarizes the recent advances in the discovery of novel androgen receptor antagonists through computer-aided drug design approaches; and discusses the applications of molecular modeling techniques to understand the resistance mechanisms of androgen receptor antagonists at the molecular level.
Collapse
Affiliation(s)
- Xueping Hu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; State Key Lab of CAD&CG, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xin Chai
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xuwen Wang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Mojie Duan
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| | - Jinping Pang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Weitao Fu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Dan Li
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Tingjun Hou
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; State Key Lab of CAD&CG, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
47
|
Kumar A, Gatto G, Delogu F, Pilia L. DFT study of [Pt(Cl)2L] complex (L = rubeanic acid) and its derived compounds with DNA purine bases. Chem Phys 2020. [DOI: 10.1016/j.chemphys.2019.110646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
48
|
Plescia J, Dufresne C, Janmamode N, Wahba AS, Mittermaier AK, Moitessier N. Discovery of covalent prolyl oligopeptidase boronic ester inhibitors. Eur J Med Chem 2020; 185:111783. [DOI: 10.1016/j.ejmech.2019.111783] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 01/22/2023]
|
49
|
Preto J, Gentile F. Assessing and improving the performance of consensus docking strategies using the DockBox package. J Comput Aided Mol Des 2019; 33:817-829. [DOI: 10.1007/s10822-019-00227-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/26/2019] [Indexed: 10/25/2022]
|
50
|
Agrawal V, Su M, Huang Y, Hsing M, Cherkasov A, Zhou Y. Computer-Aided Discovery of Small Molecule Inhibitors of Thymocyte Selection-Associated High Mobility Group Box Protein (TOX) as Potential Therapeutics for Cutaneous T-Cell Lymphomas. Molecules 2019; 24:molecules24193459. [PMID: 31554191 PMCID: PMC6803922 DOI: 10.3390/molecules24193459] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/16/2019] [Accepted: 09/20/2019] [Indexed: 01/22/2023] Open
Abstract
Cutaneous T-cell lymphomas (CTCL) are the most common primary lymphomas of the skin. We have previously identified thymocyte selection-associated high mobility group (HMG) box protein (TOX) as a promising drug target in CTCL; however, there are currently no small molecules able to directly inhibit TOX. We aimed to address this unmet opportunity by developing anti-TOX therapeutics with the use of computer-aided drug discovery methods. The available NMR-resolved structure of the TOX protein was used to model its DNA-binding HMG-box domain. To investigate the druggability of the corresponding protein–DNA interface on TOX, we performed a pilot virtual screening of 200,000 small molecules using in silico docking and identified ‘hot spots’ for drug-binding on the HMG-box domain. We then performed a large-scale virtual screening of 7.6 million drug-like compounds that were available from the ZINC15 database. As a result, a total of 140 top candidate compounds were selected for subsequent in vitro validation. Of those, 18 small molecules have been characterized as selective TOX inhibitors.
Collapse
Affiliation(s)
- Vibudh Agrawal
- Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
- The Bioinformatics Graduate Program, University of British Columbia, Vancouver, BC V5T 4S6, Canada.
| | - Mingwan Su
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada.
| | - Yuanshen Huang
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada.
| | - Michael Hsing
- Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Artem Cherkasov
- Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Youwen Zhou
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada.
- Dermatologic oncology program, BC Cancer, Vancouver, BC V5Z 1L3, Canada.
| |
Collapse
|