1
|
Zdrazil B. Fifteen years of ChEMBL and its role in cheminformatics and drug discovery. J Cheminform 2025; 17:32. [PMID: 40065463 PMCID: PMC11895189 DOI: 10.1186/s13321-025-00963-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 01/20/2025] [Indexed: 03/14/2025] Open
Abstract
In October 2024 we celebrated the 15th anniversary of the first launch of ChEMBL, Europe's most impactful, open-access drug discovery database, hosted by EMBL's European Bioinformatics Institute (EMBL-EBI). This is a good moment to reflect on ChEMBL's history, the role that ChEMBL plays in Cheminformatics and Drug Discovery as well as innovations accelerated using data extracted from it. The review closes by discussing current challenges and possible directions that need to be taken to guarantee that ChEMBL continues to be the pioneering resource for highly curated, open bioactivity data on the European continent and beyond.
Collapse
Affiliation(s)
- Barbara Zdrazil
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridgeshire, CB101SD, UK.
| |
Collapse
|
2
|
Nada H, Meanwell NA, Gabr MT. Virtual screening: hope, hype, and the fine line in between. Expert Opin Drug Discov 2025; 20:145-162. [PMID: 39862145 PMCID: PMC11844436 DOI: 10.1080/17460441.2025.2458666] [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: 12/11/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
INTRODUCTION Technological advancements in virtual screening (VS) have rapidly accelerated its application in drug discovery, as reflected by the exponential growth in VS-related publications. However, a significant gap remains between the volume of computational predictions and their experimental validation. This discrepancy has led to a rise in the number of unverified 'claimed' hits which impedes the drug discovery efforts. AREAS COVERED This perspective examines the current VS landscape, highlighting essential practices and identifying critical challenges, limitations, and common pitfalls. Using case studies and practices, this perspective aims to highlight strategies that can effectively mitigate or overcome these challenges. Furthermore, the perspective explores common approaches for addressing pharmacodynamic and pharmacokinetic issues in optimizing VS hits. EXPERT OPINION VS has become a tried-and-true technique of drug discovery due to the rapid advances in computational methods and machine learning (ML) over the past two decades. Although each VS workflow varies depending on the chosen approach and methodology, integrated strategies that combine biological and in silico data have consistently yielded higher success rates. Moreover, the widespread adoption of ML has enhanced the integration of VS into the drug discovery pipeline. However, the absence of standardized evaluation criteria hinders the objective assessment of VS studies' success and the identification of optimal adoption methods.
Collapse
Affiliation(s)
- Hossam Nada
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY 10065, USA
| | - Nicholas A. Meanwell
- Baruch S. Blumberg Institute, Doylestown, PA, USA; School of Pharmacy, University of Michigan, Ann Arbor, MI, USA
- Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Moustafa T. Gabr
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
3
|
Uncovering Quercetin’s Effects against Influenza A Virus Using Network Pharmacology and Molecular Docking. Processes (Basel) 2021. [DOI: 10.3390/pr9091627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
(1) Background: Re-emerging influenza threats continue to challenge medical and public health systems. Quercetin is a ubiquitous flavonoid found in food and is recognized to possess antioxidant, anti-inflammatory, antiviral, and anticancer activities. (2) Methods: To elucidate the targets and mechanisms underlying the action of quercetin as a therapeutic agent for influenza, network pharmacology and molecular docking were employed. Biological targets of quercetin and target genes associated with influenza were retrieved from public databases. Compound–disease target (C-D) networks were constructed, and targets were further analyzed using KEGG pathway analysis. Potent target genes were retrieved from the compound–disease–pathway (C-D-P) and protein–protein interaction (PPI) networks. The binding affinities between quercetin and the targets were identified using molecular docking. (3) Results: The pathway study revealed that quercetin-associated influenza targets were mainly involved in viral diseases, inflammation-associated pathways, and cancer. Four targets, MAPK1, NFKB1, RELA, and TP53, were identified to be involved in the inhibitory effects of quercetin on influenza. Using the molecular docking method, we evaluated the binding affinity of each ligand (quercetin)–target and discovered that quercetin and MAPK1 showed the strongest calculated binding energy among the four ligand–target complexes. (4) Conclusion: These findings identified potential targets of quercetin and suggest quercetin as a potential drug for influenza treatment.
Collapse
|
4
|
Titov IY, Stroylov VS, Rusina P, Svitanko IV. Preliminary modelling as the first stage of targeted organic synthesis. RUSSIAN CHEMICAL REVIEWS 2021. [DOI: 10.1070/rcr5012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The review aims to present a classification and applicability analysis of methods for preliminary molecular modelling for targeted organic, catalytic and biocatalytic synthesis. The following three main approaches are considered as a primary classification of the methods: modelling of the target – ligand coordination without structural information on both the target and the resulting complex; calculations based on experimentally obtained structural information about the target; and dynamic simulation of the target – ligand complex and the reaction mechanism with calculation of the free energy of the reaction. The review is meant for synthetic chemists to be used as a guide for building an algorithm for preliminary modelling and synthesis of structures with specified properties.
The bibliography includes 353 references.
Collapse
|
5
|
Saini R, Fatima S, Agarwal SM. TMLRpred: A machine learning classification model to distinguish reversible EGFR double mutant inhibitors. Chem Biol Drug Des 2021; 96:921-930. [PMID: 33058464 DOI: 10.1111/cbdd.13697] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 03/26/2020] [Accepted: 04/03/2020] [Indexed: 12/26/2022]
Abstract
The EGFR is a clinically important therapeutic drug target in lung cancer. The first-generation tyrosine kinase inhibitors used in clinics are effective against L858R-mutated EGFR. However, relapse of the disease due to the presence of resistant mutation (T790M) makes these inhibitors ineffective. This has necessitated the need to identify new potent EGFR inhibitors against the resistant double mutants. Therefore, various machine learning techniques ((instance-based learner (IBK), naïve Bayesian (NB), sequential minimal optimization (SMO), and random forest (RF)) were employed to develop twelve classification models on three different datasets (high, moderate, and weakly active inhibitors). The models were validated using fivefold cross-validation and independent validation datasets. It was observed that the random forest-based models showed best performance. Also, functional groups, PubChem fingerprints, and substructure of highly active inhibitors were compared to inactive to identify structural features which are important for activity. To promote open-source drug discovery, a tool has been developed, which incorporates the best performing models and allows users to predict the potential of chemical molecules as anti-TMLR inhibitor. It is expected that the machine learning classification models developed in this study will pave way for identifying novel inhibitors against the resistant EGFR double mutants.
Collapse
Affiliation(s)
- Ravi Saini
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, India
| | - Shehnaz Fatima
- Bioinformatics Division, ICMR-National Institute of Cancer Prevention and Research, Noida, India
| | - Subhash Mohan Agarwal
- Bioinformatics Division, ICMR-National Institute of Cancer Prevention and Research, Noida, India
| |
Collapse
|
6
|
Radchenko DS, Naumchyk VS, Dziuba I, Kyrylchuk AA, Gubina KE, Moroz YS, Grygorenko OO. One-pot parallel synthesis of 1,3,5-trisubstituted 1,2,4-triazoles. Mol Divers 2021; 26:993-1004. [PMID: 33797670 DOI: 10.1007/s11030-021-10218-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 03/24/2021] [Indexed: 11/24/2022]
Abstract
An implementation of the three-component one-pot approach to unsymmetrical 1,3,5-trisubstituted-1,2,4-triazoles into combinatorial chemistry is described. The procedure is based on the coupling of amidines with carboxylic acids and subsequent cyclization with hydrazines. After the preliminary assessment of the reagent scope, the method had 81% success rate in parallel synthesis. It was shown that over a billion-sized chemical space of readily accessible ("REAL") compounds may be generated based on the proposed methodology. Analysis of physicochemical parameters shows that the library contains significant fractions of both drug-like and "beyond-rule-of-five" members. More than 10 million of accessible compounds meet the strictest lead-likeness criteria. Additionally, 195 Mln of sp3-enriched compounds can be produced. This makes the proposed approach a valuable tool in medicinal chemistry.
Collapse
Affiliation(s)
- Dmytro S Radchenko
- Enamine Ltd., Chervonotkatska Street 78, Kyiv, 02094, Ukraine.,Taras Shevchenko National University of Kyiv, Volodymyrska Street 60, Kyiv, 01601, Ukraine
| | | | - Igor Dziuba
- Chemspace, Chervonotkatska Street 78, Kyiv, 02094, Ukraine
| | - Andrii A Kyrylchuk
- Enamine Ltd., Chervonotkatska Street 78, Kyiv, 02094, Ukraine.,Institute of Organic Chemistry, National Academy of Sciences of Ukraine, Murmanska Street 5, Kyiv, 02094, Ukraine
| | - Kateryna E Gubina
- Taras Shevchenko National University of Kyiv, Volodymyrska Street 60, Kyiv, 01601, Ukraine
| | - Yurii S Moroz
- Taras Shevchenko National University of Kyiv, Volodymyrska Street 60, Kyiv, 01601, Ukraine.,Chemspace, Chervonotkatska Street 78, Kyiv, 02094, Ukraine
| | - Oleksandr O Grygorenko
- Enamine Ltd., Chervonotkatska Street 78, Kyiv, 02094, Ukraine. .,Taras Shevchenko National University of Kyiv, Volodymyrska Street 60, Kyiv, 01601, Ukraine.
| |
Collapse
|
7
|
Zabolotna Y, Lin A, Horvath D, Marcou G, Volochnyuk DM, Varnek A. Chemography: Searching for Hidden Treasures. J Chem Inf Model 2020; 61:179-188. [PMID: 33334102 DOI: 10.1021/acs.jcim.0c00936] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The days when medicinal chemistry was limited to a few series of compounds of therapeutic interest are long gone. Nowadays, no human may succeed to acquire a complete overview of more than a billion existing or feasible compounds within which the potential "blockbuster drugs" are well hidden and yet only a few mouse clicks away. To reach these "hidden treasures", we adapted the generative topographic mapping method to enable efficient navigation through the chemical space, from a global overview to a structural pattern detection, covering, for the first time, the complete ZINC library of purchasable compounds, relative to 1.6 million biologically relevant ChEMBL molecules. About 40 000 hierarchical maps of the chemical space were constructed. Structural motifs inherent to only one library were identified. Roughly 20 000 off-market ChEMBL compound families represent incentives to enrich commercial catalogs. Alternatively, 125 000 ZINC-specific compound classes, absent in structure-activity bases, are novel paths to explore in medicinal chemistry. The complete list of these chemotypes can be downloaded using the link https://forms.gle/B6bUJj82t9EfmttV6.
Collapse
Affiliation(s)
- Yuliana Zabolotna
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081 France
| | - Arkadii Lin
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081 France
| | - Dragos Horvath
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081 France
| | - Gilles Marcou
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081 France
| | - Dmitriy M Volochnyuk
- Institute of Organic Chemistry National Academy of Sciences of Ukraine, Murmanska Street 5, Kyiv 02660, Ukraine.,Enamine Ltd., Chervonotkatska Street 78, Kyiv 02094, Ukraine
| | - Alexandre Varnek
- University of Strasbourg, Laboratoire de Chemoinformatique, 4, rue B. Pascal, Strasbourg 67081 France
| |
Collapse
|
8
|
Lane TR, Foil DH, Minerali E, Urbina F, Zorn KM, Ekins S. Bioactivity Comparison across Multiple Machine Learning Algorithms Using over 5000 Datasets for Drug Discovery. Mol Pharm 2020; 18:403-415. [PMID: 33325717 DOI: 10.1021/acs.molpharmaceut.0c01013] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Machine learning methods are attracting considerable attention from the pharmaceutical industry for use in drug discovery and applications beyond. In recent studies, we and others have applied multiple machine learning algorithms and modeling metrics and, in some cases, compared molecular descriptors to build models for individual targets or properties on a relatively small scale. Several research groups have used large numbers of datasets from public databases such as ChEMBL in order to evaluate machine learning methods of interest to them. The largest of these types of studies used on the order of 1400 datasets. We have now extracted well over 5000 datasets from CHEMBL for use with the ECFP6 fingerprint and in comparison of our proprietary software Assay Central with random forest, k-nearest neighbors, support vector classification, naïve Bayesian, AdaBoosted decision trees, and deep neural networks (three layers). Model performance was assessed using an array of fivefold cross-validation metrics including area-under-the-curve, F1 score, Cohen's kappa, and Matthews correlation coefficient. Based on ranked normalized scores for the metrics or datasets, all methods appeared comparable, while the distance from the top indicated that Assay Central and support vector classification were comparable. Unlike prior studies which have placed considerable emphasis on deep neural networks (deep learning), no advantage was seen in this case. If anything, Assay Central may have been at a slight advantage as the activity cutoff for each of the over 5000 datasets representing over 570,000 unique compounds was based on Assay Central performance, although support vector classification seems to be a strong competitor. We also applied Assay Central to perform prospective predictions for the toxicity targets PXR and hERG to further validate these models. This work appears to be the largest scale comparison of these machine learning algorithms to date. Future studies will likely evaluate additional databases, descriptors, and machine learning algorithms and further refine the methods for evaluating and comparing such models.
Collapse
Affiliation(s)
- Thomas R Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Daniel H Foil
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Eni Minerali
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Fabio Urbina
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7545, United States
| | - Kimberley M Zorn
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
9
|
Lin A, Baskin II, Marcou G, Horvath D, Beck B, Varnek A. Parallel Generative Topographic Mapping: An Efficient Approach for Big Data Handling. Mol Inform 2020; 39:e2000009. [PMID: 32347666 PMCID: PMC7757192 DOI: 10.1002/minf.202000009] [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: 01/16/2020] [Accepted: 04/10/2020] [Indexed: 11/12/2022]
Abstract
Generative Topographic Mapping (GTM) can be efficiently used to visualize, analyze and model large chemical data. The GTM manifold needs to span the chemical space deemed relevant for a given problem. Therefore, the Frame set (FS) of compounds used for the manifold construction must well cover a given chemical space. Intuitively, the FS size must raise with the size and diversity of the target library. At the same time, the GTM training can be very slow or even becomes technically impossible at FS sizes of the order of 105 compounds - which is a very small number compared to today's commercially accessible compounds, and, especially, to the theoretically feasible molecules. In order to solve this problem, we propose a Parallel GTM algorithm based on the merging of "intermediate" manifolds constructed in parallel for different subsets of molecules. An ensemble of these subsets forms a FS for the "final" manifold. In order to assess the efficiency of the new algorithm, 80 GTMs were built on the FSs of different sizes ranging from 10 to 1.8 M compounds selected from the ChEMBL database. Each GTM was challenged to build classification models for up to 712 biological activities (depending on the FS size). With the novel parallel GTM procedure, we could thus cover the entire spectrum of possible FS sizes, whereas previous studies were forced to rely on the working hypothesis that FS sizes of few thousands of compounds are sufficient to describe the ChEMBL chemical space. In fact, this study formally proves this to be true: a FS containing only 5000 randomly picked compounds is sufficient to represent the entire ChEMBL collection (1.8 M molecules), in the sense that a further increase of FS compound numbers has no benefice impact on the predictive propensity of the above-mentioned 712 activity classification models. Parallel GTM may, however, be required to generate maps based on very large FS, that might improve chemical space cartography of big commercial and virtual libraries, approaching billions of compounds.
Collapse
Affiliation(s)
- Arkadii Lin
- University of StrasbourgLaboratory of Chemoinformatics, Faculty of Chemistry4, Blaise Pascal str.67081StrasbourgFrance
| | - Igor I. Baskin
- Faculty of PhysicsLomonosov Moscow State University1/2, Leninskie Gory str.119991MoscowRussia
| | - Gilles Marcou
- University of StrasbourgLaboratory of Chemoinformatics, Faculty of Chemistry4, Blaise Pascal str.67081StrasbourgFrance
| | - Dragos Horvath
- University of StrasbourgLaboratory of Chemoinformatics, Faculty of Chemistry4, Blaise Pascal str.67081StrasbourgFrance
| | - Bernd Beck
- Department of Medicinal ChemistryBoehringer Ingelheim Pharma GmbH & Co. KG65, Birkendorfer str.88397Biberach an der RissGermany
| | - Alexandre Varnek
- University of StrasbourgLaboratory of Chemoinformatics, Faculty of Chemistry4, Blaise Pascal str.67081StrasbourgFrance
| |
Collapse
|
10
|
Duncan KK, Rudnicki DD, Austin CP, Tagle DA. Exploring Novel Biologically-Relevant Chemical Space Through Artificial Intelligence: The NCATS ASPIRE Program. Front Robot AI 2020; 6:143. [PMID: 33501158 PMCID: PMC7805902 DOI: 10.3389/frobt.2019.00143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 12/09/2019] [Indexed: 11/13/2022] Open
Abstract
In recent years, artificial intelligence (AI)/machine learning (ML; a subset of AI) have become increasingly important to the biomedical research community. These technologies, coupled to big data and cheminformatics, have tremendous potential to improve the design of novel therapeutics and to provide safe and effective drugs to patients. A National Center for Advancing Translational Sciences (NCATS) program called A Specialized Platform for Innovative Research Exploration (ASPIRE) leverages advances in AI/ML, automated synthetic chemistry, and high-throughput biology, and seeks to enable translation and drug development by catalyzing exploration of biologically active chemical space. Here we discuss the opportunities and challenges surrounding the application of AI/ML to the exploration of novel biologically relevant chemical space as part of ASPIRE.
Collapse
Affiliation(s)
| | | | | | - Danilo A. Tagle
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
11
|
Horvath D, Marcou G, Varnek A. Generative topographic mapping in drug design. DRUG DISCOVERY TODAY. TECHNOLOGIES 2019; 32-33:99-107. [PMID: 33386101 DOI: 10.1016/j.ddtec.2020.06.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/10/2020] [Accepted: 06/18/2020] [Indexed: 06/12/2023]
Abstract
This is a review article of Generative Topographic Mapping (GTM) - a non-linear dimensionality reduction technique producing generative 2D maps of high-dimensional vector spaces - and its specific applications in Drug Design (chemical space cartography, compound library design and analysis, virtual screening, pharmacological profiling, de novo drug design, conformational space & docking interaction cartography, etc.) Written by chemoinformaticians for potential users among medicinal chemists and biologists, the article purposely avoids all underlying mathematics. First, the GTM concept is intuitively explained, based on the strong analogies with the rather popular Self-Organizing Maps (SOMs), which are well established library analysis tools. GTM is basically a fuzzy-logics-based generalization of SOMs. The second part of the review, some of published GTM applications in drug design are briefly revisited.
Collapse
Affiliation(s)
- Dragos Horvath
- Laboratory of Chemoinformatics, UMR 7140 University of Strasbourg/CNRS, 4 rue Blaise Pascal, 67000 Strasbourg, France.
| | - Gilles Marcou
- Laboratory of Chemoinformatics, UMR 7140 University of Strasbourg/CNRS, 4 rue Blaise Pascal, 67000 Strasbourg, France
| | - Alexandre Varnek
- Laboratory of Chemoinformatics, UMR 7140 University of Strasbourg/CNRS, 4 rue Blaise Pascal, 67000 Strasbourg, France.
| |
Collapse
|
12
|
Zhi Y, Wang S, Huang W, Zeng S, Liang M, Zhang C, Ma Z, Wang Z, Zhang Z, Shen Z. Novel phenanthridin-6(5H)-one derivatives as potent and selective BET bromodomain inhibitors: Rational design, synthesis and biological evaluation. Eur J Med Chem 2019; 179:502-514. [PMID: 31276895 DOI: 10.1016/j.ejmech.2019.06.067] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/12/2019] [Accepted: 06/24/2019] [Indexed: 12/17/2022]
Abstract
Inhibition of BET family of bromodomain is an appealing intervention strategy for several cancers and inflammatory diseases. This article highlights our work toward the identification of potent, selective, and efficacious BET inhibitors using a structure-based approach focused on improving potency. Our medicinal chemistry efforts led to the identification of compound 24, a novel phenanthridin-6(5H)-one derivative, as a potent (IC50 = 0.24 μM) and selective BET inhibitor with excellent cancer cell lines inhibitory activities and favorable oral pharmacokinetic properties.
Collapse
Affiliation(s)
- Yanle Zhi
- College of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, 450046, Henan Province, PR China
| | - Shu Wang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, 310013, PR China
| | - Wenhai Huang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, 310013, PR China
| | - Shenxin Zeng
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, 310013, PR China
| | - Meihao Liang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, 310013, PR China
| | - Chixiao Zhang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, 310013, PR China
| | - Zhen Ma
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, 310013, PR China
| | - Zunyuan Wang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, 310013, PR China
| | - Zhimin Zhang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, 310013, PR China.
| | - Zhengrong Shen
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, 310013, PR China.
| |
Collapse
|