1
|
Daina A, Zoete V. Testing the predictive power of reverse screening to infer drug targets, with the help of machine learning. Commun Chem 2024; 7:105. [PMID: 38724725 PMCID: PMC11082207 DOI: 10.1038/s42004-024-01179-2] [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: 04/06/2023] [Accepted: 04/16/2024] [Indexed: 05/12/2024] Open
Abstract
Estimating protein targets of compounds based on the similarity principle-similar molecules are likely to show comparable bioactivity-is a long-standing strategy in drug research. Having previously quantified this principle, we present here a large-scale evaluation of its predictive power for inferring macromolecular targets by reverse screening an unprecedented vast external test set of more than 300,000 active small molecules against another bioactivity set of more than 500,000 compounds. We show that machine-learning can predict the correct targets, with the highest probability among 2069 proteins, for more than 51% of the external molecules. The strong enrichment thus obtained demonstrates its usefulness in supporting phenotypic screens, polypharmacology, or repurposing. Moreover, we quantified the impact of the bioactivity knowledge available for proteins in terms of number and diversity of actives. Finally, we advise that developers of such approaches follow an application-oriented benchmarking strategy and use large, high-quality, non-overlapping datasets as provided here.
Collapse
Affiliation(s)
- Antoine Daina
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, CH-1015, Lausanne, Switzerland
| | - Vincent Zoete
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, CH-1015, Lausanne, Switzerland.
- Computer-Aided Molecular Engineering, Department of Oncology UNIL-CHUV, Ludwig Institute for Cancer Research Lausanne Branch, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
2
|
Durojaye OA, Okoro NO, Odiba AS, Nwanguma BC. MasitinibL shows promise as a drug-like analog of masitinib that elicits comparable SARS-Cov-2 3CLpro inhibition with low kinase preference. Sci Rep 2023; 13:6972. [PMID: 37117213 PMCID: PMC10141821 DOI: 10.1038/s41598-023-33024-2] [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: 02/03/2023] [Accepted: 04/06/2023] [Indexed: 04/30/2023] Open
Abstract
SARS-CoV-2 infection has led to several million deaths worldwide and ravaged the economies of many countries. Hence, developing therapeutics against SARS-CoV-2 remains a core priority in the fight against COVID-19. Most of the drugs that have received emergency use authorization for treating SARS-CoV-2 infection exhibit a number of limitations, including side effects and questionable efficacy. This challenge is further compounded by reinfection after vaccination and the high likelihood of mutations, as well as the emergence of viral escape mutants that render SARS-CoV-2 spike glycoprotein-targeting vaccines ineffective. Employing de novo drug synthesis or repurposing to discover broad-spectrum antivirals that target highly conserved pathways within the viral machinery is a focus of current research. In a recent drug repurposing study, masitinib, a clinically safe drug against the human coronavirus OC43 (HCoV-OC43), was identified as an antiviral agent with effective inhibitory activity against the SARS-CoV-2 3CLpro. Masitinib is currently under clinical trial in combination with isoquercetin in hospitalized patients (NCT04622865). Nevertheless, masitinib has kinase-related side effects; hence, the development of masitinib analogs with lower anti-tyrosine kinase activity becomes necessary. In this study, in an attempt to address this limitation, we executed a comprehensive virtual workflow in silico to discover drug-like compounds matching selected pharmacophore features in the SARS-CoV-2 3CLpro-bound state of masitinib. We identified a novel lead compound, "masitinibL", a drug-like analog of masitinib that demonstrated strong inhibitory properties against the SARS-CoV-2 3CLpro. In addition, masitinibL further displayed low selectivity for tyrosine kinases, which strongly suggests that masitinibL is a highly promising therapeutic that is preferable to masitinib.
Collapse
Affiliation(s)
- Olanrewaju Ayodeji Durojaye
- MOE Key Laboratory of Membraneless Organelle and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230027, Anhui, China
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Department of Chemical Sciences, Coal City University, Emene, Enugu State, Nigeria
| | - Nkwachukwu Oziamara Okoro
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, 410001, Nigeria
| | - Arome Solomon Odiba
- Department of Molecular Genetics and Biotechnology, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria.
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria.
| | - Bennett Chima Nwanguma
- Department of Molecular Genetics and Biotechnology, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria.
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria.
| |
Collapse
|
3
|
Zhu Y, Yang H, Han L, Mervin LH, Hosseini-Gerami L, Li P, Wright P, Trapotsi MA, Liu K, Fan TP, Bender A. In silico prediction and biological assessment of novel angiogenesis modulators from traditional Chinese medicine. Front Pharmacol 2023; 14:1116081. [PMID: 36817116 PMCID: PMC9937659 DOI: 10.3389/fphar.2023.1116081] [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: 12/05/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
Uncontrolled angiogenesis is a common denominator underlying many deadly and debilitating diseases such as myocardial infarction, chronic wounds, cancer, and age-related macular degeneration. As the current range of FDA-approved angiogenesis-based medicines are far from meeting clinical demands, the vast reserve of natural products from traditional Chinese medicine (TCM) offers an alternative source for developing pro-angiogenic or anti-angiogenic modulators. Here, we investigated 100 traditional Chinese medicine-derived individual metabolites which had reported gene expression in MCF7 cell lines in the Gene Expression Omnibus (GSE85871). We extracted literature angiogenic activities for 51 individual metabolites, and subsequently analysed their predicted targets and differentially expressed genes to understand their mechanisms of action. The angiogenesis phenotype was used to generate decision trees for rationalising the poly-pharmacology of known angiogenesis modulators such as ferulic acid and curculigoside and validated by an in vitro endothelial tube formation assay and a zebrafish model of angiogenesis. Moreover, using an in silico model we prospectively examined the angiogenesis-modulating activities of the remaining 49 individual metabolites. In vitro, tetrahydropalmatine and 1 beta-hydroxyalantolactone stimulated, while cinobufotalin and isoalantolactone inhibited endothelial tube formation. In vivo, ginsenosides Rb3 and Rc, 1 beta-hydroxyalantolactone and surprisingly cinobufotalin, restored angiogenesis against PTK787-induced impairment in zebrafish. In the absence of PTK787, deoxycholic acid and ursodeoxycholic acid did not affect angiogenesis. Despite some limitations, these results suggest further refinements of in silico prediction combined with biological assessment will be a valuable platform for accelerating the research and development of natural products from traditional Chinese medicine and understanding their mechanisms of action, and also for other traditional medicines for the prevention and treatment of angiogenic diseases.
Collapse
Affiliation(s)
- Yingli Zhu
- Department of Clinical Chinese Pharmacy, School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, China,Department of Chemistry, Center for Molecular Science Informatics, University of Cambridge, Cambridge, United Kingdom,Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Hongbin Yang
- Department of Chemistry, Center for Molecular Science Informatics, University of Cambridge, Cambridge, United Kingdom
| | - Liwen Han
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan, China,School of Pharmacy and Pharmaceutical Science, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Lewis H. Mervin
- Department of Chemistry, Center for Molecular Science Informatics, University of Cambridge, Cambridge, United Kingdom
| | - Layla Hosseini-Gerami
- Department of Chemistry, Center for Molecular Science Informatics, University of Cambridge, Cambridge, United Kingdom
| | - Peihai Li
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan, China
| | - Peter Wright
- Department of Chemistry, Center for Molecular Science Informatics, University of Cambridge, Cambridge, United Kingdom
| | - Maria-Anna Trapotsi
- Department of Chemistry, Center for Molecular Science Informatics, University of Cambridge, Cambridge, United Kingdom
| | - Kechun Liu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan, China
| | - Tai-Ping Fan
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom,*Correspondence: Tai-Ping Fan, ; Andreas Bender,
| | - Andreas Bender
- Department of Chemistry, Center for Molecular Science Informatics, University of Cambridge, Cambridge, United Kingdom,*Correspondence: Tai-Ping Fan, ; Andreas Bender,
| |
Collapse
|
4
|
Bustamante C, Muskus C, Ochoa R. Rational computational approaches to predict novel drug candidates against leishmaniasis. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2022. [DOI: 10.1016/bs.armc.2022.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
5
|
Morphological profiling by means of the Cell Painting assay enables identification of tubulin-targeting compounds. Cell Chem Biol 2021; 29:1053-1064.e3. [PMID: 34968420 DOI: 10.1016/j.chembiol.2021.12.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/27/2021] [Accepted: 12/06/2021] [Indexed: 11/23/2022]
Abstract
In phenotypic compound discovery, conclusive identification of cellular targets and mode of action are often impaired by off-target binding. In particular, microtubules are frequently targeted in cellular assays. However, in vitro tubulin binding assays do not correctly reflect the cellular context, and conclusive high-throughput phenotypic assays monitoring tubulin binding are scarce, such that tubulin binding is rarely identified. We report that morphological profiling using the Cell Painting assay (CPA) can efficiently detect tubulin modulators in compound collections with a high throughput, including annotated reference compounds and unannotated compound classes with unrelated chemotypes and scaffolds. Small-molecule tubulin binders share similar CPA fingerprints, which enables prediction and experimental validation of microtubule-binding activity. Our findings suggest that CPA or a related morphological profiling approach will be an invaluable addition to small-molecule discovery programs in chemical biology and medicinal chemistry, enabling early identification of one of the most frequently observed off-target activities.
Collapse
|
6
|
Friedrich L, Cingolani G, Ko Y, Iaselli M, Miciaccia M, Perrone MG, Neukirch K, Bobinger V, Merk D, Hofstetter RK, Werz O, Koeberle A, Scilimati A, Schneider G. Learning from Nature: From a Marine Natural Product to Synthetic Cyclooxygenase-1 Inhibitors by Automated De Novo Design. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100832. [PMID: 34176236 PMCID: PMC8373093 DOI: 10.1002/advs.202100832] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/16/2021] [Indexed: 05/03/2023]
Abstract
The repertoire of natural products offers tremendous opportunities for chemical biology and drug discovery. Natural product-inspired synthetic molecules represent an ecologically and economically sustainable alternative to the direct utilization of natural products. De novo design with machine intelligence bridges the gap between the worlds of bioactive natural products and synthetic molecules. On employing the compound Marinopyrrole A from marine Streptomyces as a design template, the algorithm constructs innovative small molecules that can be synthesized in three steps, following the computationally suggested synthesis route. Computational activity prediction reveals cyclooxygenase (COX) as a putative target of both Marinopyrrole A and the de novo designs. The molecular designs are experimentally confirmed as selective COX-1 inhibitors with nanomolar potency. X-ray structure analysis reveals the binding of the most selective compound to COX-1. This molecular design approach provides a blueprint for natural product-inspired hit and lead identification for drug discovery with machine intelligence.
Collapse
Affiliation(s)
- Lukas Friedrich
- Department of Chemistry and Applied BiosciencesETH ZurichVladimir‐Prelog‐Weg 4Zurich8093Switzerland
| | - Gino Cingolani
- Department of Biochemistry and Molecular BiologySidney Kimmel Cancer CenterThomas Jefferson University1020 Locust StreetPhiladelphiaPA19107USA
| | - Ying‐Hui Ko
- Department of Biochemistry and Molecular BiologySidney Kimmel Cancer CenterThomas Jefferson University1020 Locust StreetPhiladelphiaPA19107USA
| | - Mariaclara Iaselli
- Department of Pharmacy – Pharmaceutical SciencesUniversity of BariVia E. Orabona 4Bari70125Italy
| | - Morena Miciaccia
- Department of Pharmacy – Pharmaceutical SciencesUniversity of BariVia E. Orabona 4Bari70125Italy
| | - Maria Grazia Perrone
- Department of Pharmacy – Pharmaceutical SciencesUniversity of BariVia E. Orabona 4Bari70125Italy
| | - Konstantin Neukirch
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruck6020Austria
| | - Veronika Bobinger
- Department of Chemistry and Applied BiosciencesETH ZurichVladimir‐Prelog‐Weg 4Zurich8093Switzerland
| | - Daniel Merk
- Department of Chemistry and Applied BiosciencesETH ZurichVladimir‐Prelog‐Weg 4Zurich8093Switzerland
- Institute of Pharmaceutical ChemistryGoethe‐UniversityMax‐von‐Laue Straße 9Frankfurt am Main60438Germany
| | - Robert Klaus Hofstetter
- Department of Pharmaceutical/Medicinal ChemistryFriedrich‐Schiller‐University JenaPhilosophenweg 14Jena07743Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal ChemistryFriedrich‐Schiller‐University JenaPhilosophenweg 14Jena07743Germany
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruck6020Austria
| | - Antonio Scilimati
- Department of Pharmacy – Pharmaceutical SciencesUniversity of BariVia E. Orabona 4Bari70125Italy
| | - Gisbert Schneider
- Department of Chemistry and Applied BiosciencesETH ZurichVladimir‐Prelog‐Weg 4Zurich8093Switzerland
- ETH Singapore SEC Ltd1 CREATE Way, #06‐01 CREATE TowerSingapore138602Singapore
| |
Collapse
|
7
|
Ye X, Wu J, Zhang D, Lan Z, Yang S, Zhu J, Yang M, Gong Q, Zhong L. How Aconiti Radix Cocta can Treat Gouty Arthritis Based on Systematic Pharmacology and UPLC-QTOF-MS/MS. Front Pharmacol 2021; 12:618844. [PMID: 33995019 PMCID: PMC8121251 DOI: 10.3389/fphar.2021.618844] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 04/13/2021] [Indexed: 12/25/2022] Open
Abstract
Background: Gouty arthritis (GA) is a common metabolic disease caused by a long-term disorder of purine metabolism and increased serum levels of uric acid. The processed product of dried root of Aconitum carmichaeli Debeaux (Aconiti Radix cocta, ARC) is used often in traditional Chinese medicine (TCM) to treat GA, but its specific active components and mechanism of action are not clear. Methods: First, we used ultra-performance liquid chromatography-quadrupole/time-of-flight tandem mass spectrometry to identify the chemical spectrum of ARC. Based on this result, we explored the active components of ARC in GA treatment and their potential targets and pathways. Simultaneously, we used computer simulations, in vitro cell experiments and animal experiments to verify the prediction results of systems pharmacology. In vitro, we used aurantiamide acetate (AA) to treat monosodium urate (MSU)-stimulated THP-1 cells and demonstrated the reliability of the prediction by western blotting and real-time reverse transcription-quantitative polymerase chain reaction (RT-qPCR). ELISAs kit were used to measure changes in levels of proinflammatory factors in rats with GA induced by MSU to demonstrate the efficacy of ARC in GA treatment. Results: Forty-three chemical constituents in ARC were identified. ARC could regulate 65 targets through 29 active components, and then treat GA, which involved 1427 Gene Ontology (GO) terms and 146 signaling pathways. Signaling pathways such as proteoglycans in cancer, C-type lectin receptor signaling pathway, and TNF signaling pathway may have an important role in GA treatment with ARC. In silico results showed that the active components songoramine and ignavine had high binding to mitogen-activated protein kinase p38 alpha (MAPK14) and matrix metallopeptidase (MMP)9, indicating that ARC treatment of GA was through multiple components and multiple targets. In vitro experiments showed that AA in ARC could effectively reduce expression of MAPK14, MMP9, and cyclooxygenase2 (PTGS2) in THP-1 cells stimulated by MSU, whereas it could significantly inhibit the mRNA expression of Caspase-1, spleen tyrosine kinase (SYK), and PTGS2. Animal experiments showed that a ARC aqueous extract could significantly reduce expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and intereleukin (IL)-18 in the serum of GA rats stimulated by MSU. Hence, ARC may inhibit inflammation by regulating the proteoglycans in cancer-associated signaling pathways. Conclusion: ARC treatment of GA may have the following mechanisms, ARC can reduce MSU crystal-induced joint swelling, reduce synovial tissue damage, and reduce the expression of inflammatory factors in serum. AA in ARC may inhibit inflammation by regulating the protein expression of MAPK14, MMP9, and PTGS2 and the mRNA expression of caspase-1, SYK, and PTGS2.
Collapse
Affiliation(s)
- Xietao Ye
- Pharmacy College, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Jianxiong Wu
- Pharmacy College, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Dayong Zhang
- Sichuan New Lotus Chinese Herbal Medicine, Chengdu, China
| | - Zelun Lan
- Sichuan New Lotus Chinese Herbal Medicine, Chengdu, China
| | - Songhong Yang
- Pharmacy College, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Jing Zhu
- Pharmacy College, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Ming Yang
- Pharmacy College, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Qianfeng Gong
- Pharmacy College, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Lingyun Zhong
- Pharmacy College, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| |
Collapse
|
8
|
Trapotsi MA, Mervin LH, Afzal AM, Sturm N, Engkvist O, Barrett IP, Bender A. Comparison of Chemical Structure and Cell Morphology Information for Multitask Bioactivity Predictions. J Chem Inf Model 2021; 61:1444-1456. [PMID: 33661004 DOI: 10.1021/acs.jcim.0c00864] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The understanding of the mechanism-of-action (MoA) of compounds and the prediction of potential drug targets play an important role in small-molecule drug discovery. The aim of this work was to compare chemical and cell morphology information for bioactivity prediction. The comparison was performed using bioactivity data from the ExCAPE database, image data (in the form of CellProfiler features) from the Cell Painting data set (the largest publicly available data set of cell images with ∼30,000 compound perturbations), and extended connectivity fingerprints (ECFPs) using the multitask Bayesian matrix factorization (BMF) approach Macau. We found that the BMF Macau and random forest (RF) performance were overall similar when ECFPs were used as compound descriptors. However, BMF Macau outperformed RF in 159 out of 224 targets (71%) when image data were used as compound information. Using BMF Macau, 100 (corresponding to about 45%) and 90 (about 40%) of the 224 targets were predicted with high predictive performance (AUC > 0.8) with ECFP data and image data as side information, respectively. There were targets better predicted by image data as side information, such as β-catenin, and others better predicted by fingerprint-based side information, such as proteins belonging to the G-protein-Coupled Receptor 1 family, which could be rationalized from the underlying data distributions in each descriptor domain. In conclusion, both cell morphology changes and chemical structure information contain information about compound bioactivity, which is also partially complementary, and can hence contribute to in silico MoA analysis.
Collapse
Affiliation(s)
- Maria-Anna Trapotsi
- Department of Chemistry, Centre for Molecular Informatics, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - Lewis H Mervin
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Avid M Afzal
- Data Sciences & Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Noé Sturm
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Gothenburg SE-43183, Sweden
| | - Ola Engkvist
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Gothenburg SE-43183, Sweden
| | - Ian P Barrett
- Data Sciences & Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Andreas Bender
- Department of Chemistry, Centre for Molecular Informatics, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| |
Collapse
|
9
|
Ziegler S, Sievers S, Waldmann H. Morphological profiling of small molecules. Cell Chem Biol 2021; 28:300-319. [PMID: 33740434 DOI: 10.1016/j.chembiol.2021.02.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/22/2021] [Accepted: 02/17/2021] [Indexed: 12/30/2022]
Abstract
Profiling approaches such as gene expression or proteome profiling generate small-molecule bioactivity profiles that describe a perturbed cellular state in a rather unbiased manner and have become indispensable tools in the evaluation of bioactive small molecules. Automated imaging and image analysis can record morphological alterations that are induced by small molecules through the detection of hundreds of morphological features in high-throughput experiments. Thus, morphological profiling has gained growing attention in academia and the pharmaceutical industry as it enables detection of bioactivity in compound collections in a broader biological context in the early stages of compound development and the drug-discovery process. Profiling may be used successfully to predict mode of action or targets of unexplored compounds and to uncover unanticipated activity for already characterized small molecules. Here, we review the reported approaches to morphological profiling and the kind of bioactivity that can be detected so far and, thus, predicted.
Collapse
Affiliation(s)
- Slava Ziegler
- Max-Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany.
| | - Sonja Sievers
- Max-Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Herbert Waldmann
- Max-Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany; Technical University Dortmund, Faculty of Chemistry and Chemical Biology, Otto-Hahn-Strasse 6, 44227 Dortmund, Germany.
| |
Collapse
|
10
|
Espinoza-Culupú A, Vázquez-Ramírez R, Farfán-López M, Mendes E, Notomi Sato M, da Silva Junior PI, Borges MM. Acylpolyamine Mygalin as a TLR4 Antagonist Based on Molecular Docking and In Vitro Analyses. Biomolecules 2020; 10:E1624. [PMID: 33271940 PMCID: PMC7761503 DOI: 10.3390/biom10121624] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/19/2020] [Accepted: 11/25/2020] [Indexed: 01/18/2023] Open
Abstract
Toll-like receptors (TLRs) are transmembrane proteins that are key regulators of innate and adaptive immune responses, particularly TLR4, and they have been identified as potential drug targets for the treatment of disease. Several low-molecular-weight compounds are being considered as new drug targets for various applications, including as immune modulators. Mygalin, a 417 Da synthetic bis-acylpolyamine, is an analog of spermidine that has microbicidal activity. In this study, we investigated the effect of mygalin on the innate immune response based on a virtual screening (VS) and molecular docking analysis. Bone marrow-derived macrophages and the cell lines J774A.1 and RAW 264.7 stimulated with lipopolysaccharide (LPS) were used to confirm the data obtained in silico. Virtual screening and molecular docking suggested that mygalin binds to TLR4 via the protein myeloid differentiation factor 2 (MD-2) and LPS. Macrophages stimulated by mygalin plus LPS showed suppressed gene expression of tumor necrosis factor (TNF-α), interleukine 6 (IL-6), cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS), as well as inhibition of signaling protein p65 of the nuclear factor κB (NF-κB), resulting in decreased production of nitric oxide (NO) and TNF-α. These results indicate that mygalin has anti-inflammatory potential, being an attractive option to be explored. In addition, we reinforce the importance of virtual screening analysis to assist in the discovery of new drugs.
Collapse
Affiliation(s)
- Abraham Espinoza-Culupú
- Interunits Graduate Program in Biotechnology, USP/IBu/IPT, São Paulo 01000-000, Brazil; (A.E.-C.); (P.I.d.S.J.)
- Bacteriology Laboratory, Butantan Institute, São Paulo 01000-000, Brazil;
| | - Ricardo Vázquez-Ramírez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 00-16, Mexico;
| | - Mariella Farfán-López
- Microbiology Molecular and Biotechnology Laboratory, Universidad Nacional Mayor de San Marcos, Lima District 15081, Peru;
| | - Elizabeth Mendes
- Bacteriology Laboratory, Butantan Institute, São Paulo 01000-000, Brazil;
| | - Maria Notomi Sato
- Laboratory of Dermatology and Immunodeficiencies, Medical School, University of São Paulo, São Paulo 01000-000, Brazil;
| | - Pedro Ismael da Silva Junior
- Interunits Graduate Program in Biotechnology, USP/IBu/IPT, São Paulo 01000-000, Brazil; (A.E.-C.); (P.I.d.S.J.)
- Laboratory for Applied Toxinology (LETA), Butantan Institute, São Paulo 01000-000, Brazil
| | - Monamaris Marques Borges
- Interunits Graduate Program in Biotechnology, USP/IBu/IPT, São Paulo 01000-000, Brazil; (A.E.-C.); (P.I.d.S.J.)
- Bacteriology Laboratory, Butantan Institute, São Paulo 01000-000, Brazil;
| |
Collapse
|
11
|
Schneidewind T, Brause A, Pahl A, Burhop A, Mejuch T, Sievers S, Waldmann H, Ziegler S. Morphological Profiling Identifies a Common Mode of Action for Small Molecules with Different Targets. Chembiochem 2020; 21:3197-3207. [PMID: 32618075 PMCID: PMC7754162 DOI: 10.1002/cbic.202000381] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/02/2020] [Indexed: 12/24/2022]
Abstract
Unbiased morphological profiling of bioactivity, for example, in the cell painting assay (CPA), enables the identification of a small molecule's mode of action based on its similarity to the bioactivity of reference compounds, irrespective of the biological target or chemical similarity. This is particularly important for small molecules with nonprotein targets as these are rather difficult to identify with widely employed target-identification methods. We employed morphological profiling using the CPA to identify compounds that are biosimilar to the iron chelator deferoxamine. Structurally different compounds with different annotated cellular targets provoked a shared physiological response, thereby defining a cluster based on their morphological fingerprints. This cluster is based on a shared mode of action and not on a shared target, that is, cell-cycle modulation in the S or G2 phase. Hierarchical clustering of morphological fingerprints revealed subclusters that are based on the mechanism of action and could be used to predict target-related bioactivity.
Collapse
Affiliation(s)
- Tabea Schneidewind
- Max-Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyOtto-Hahn-Strasse 11Dortmund44227Germany
- Technical University DortmundFaculty of Chemistry and Chemical BiologyOtto-Hahn-Strasse 6Dortmund44227Germany
| | - Alexandra Brause
- Max-Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyOtto-Hahn-Strasse 11Dortmund44227Germany
| | - Axel Pahl
- Max-Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyOtto-Hahn-Strasse 11Dortmund44227Germany
| | - Annina Burhop
- Max-Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyOtto-Hahn-Strasse 11Dortmund44227Germany
| | - Tom Mejuch
- Max-Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyOtto-Hahn-Strasse 11Dortmund44227Germany
| | - Sonja Sievers
- Max-Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyOtto-Hahn-Strasse 11Dortmund44227Germany
| | - Herbert Waldmann
- Max-Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyOtto-Hahn-Strasse 11Dortmund44227Germany
- Technical University DortmundFaculty of Chemistry and Chemical BiologyOtto-Hahn-Strasse 6Dortmund44227Germany
| | - Slava Ziegler
- Max-Planck Institute of Molecular PhysiologyDepartment of Chemical BiologyOtto-Hahn-Strasse 11Dortmund44227Germany
| |
Collapse
|
12
|
Deshapriya US, Dinuka DLS, Ratnaweera PB, Ratnaweera CN. In silico study for prediction of novel bioactivities of the endophytic fungal alkaloid, mycoleptodiscin B for human targets. J Mol Graph Model 2020; 102:107767. [PMID: 33130394 DOI: 10.1016/j.jmgm.2020.107767] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/23/2020] [Accepted: 09/28/2020] [Indexed: 11/28/2022]
Abstract
Mycoleptodiscin B is a natural product extracted from the endophytic fungus Mycoleptodiscus sp. found in Sri Lanka and Panama with experimentally unexplored activities for human targets. In this study, a computational methodology was applied to determine druggable targets of mycoleptodiscin B. According to the computational toxicity and pharmacokinetics assessment, mycoleptodiscin B was proven to be a suitable drug candidate. Druggable targets for this compound, aromatase, acidic plasma glycoprotein and androgen receptor, were predicted using reverse docking. A two-step validation of those targets was performed using conventional molecular docking and molecular dynamic (MD) simulations, resulting in aromatase being determined as the potential therapeutic target. Based on molecular mechanics/Generalized Born Surface Area (GBSA) free energies and ligand stability inside the active site cavity during its 120 ns MD run, it can be concluded that mycoleptodiscin B is a potent aromatase inhibitor and could be subjected to further in vitro and in vivo experiments in the drug development pipeline. Consequently, natural product chemists can quickly identify the hidden medicinal properties of their miracle compounds using the computational approach applied in this research.
Collapse
Affiliation(s)
- Uthpala S Deshapriya
- College of Chemical Sciences, Institute of Chemistry Ceylon, Rajagiriya, Sri Lanka; Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - D L Senal Dinuka
- College of Chemical Sciences, Institute of Chemistry Ceylon, Rajagiriya, Sri Lanka; Department of Chemistry, Mississippi State University, Mississippi State, USA
| | - Pamoda B Ratnaweera
- Department of Science and Technology, Faculty of Applied Sciences, Uva Wellassa University, Badulla, Sri Lanka
| | - Chinthaka N Ratnaweera
- College of Chemical Sciences, Institute of Chemistry Ceylon, Rajagiriya, Sri Lanka; Department of Chemistry, University of Ruhuna, Matara, Sri Lanka.
| |
Collapse
|
13
|
Agamah FE, Mazandu GK, Hassan R, Bope CD, Thomford NE, Ghansah A, Chimusa ER. Computational/in silico methods in drug target and lead prediction. Brief Bioinform 2020; 21:1663-1675. [PMID: 31711157 PMCID: PMC7673338 DOI: 10.1093/bib/bbz103] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/17/2019] [Accepted: 07/18/2019] [Indexed: 01/10/2023] Open
Abstract
Drug-like compounds are most of the time denied approval and use owing to the unexpected clinical side effects and cross-reactivity observed during clinical trials. These unexpected outcomes resulting in significant increase in attrition rate centralizes on the selected drug targets. These targets may be disease candidate proteins or genes, biological pathways, disease-associated microRNAs, disease-related biomarkers, abnormal molecular phenotypes, crucial nodes of biological network or molecular functions. This is generally linked to several factors, including incomplete knowledge on the drug targets and unpredicted pharmacokinetic expressions upon target interaction or off-target effects. A method used to identify targets, especially for polygenic diseases, is essential and constitutes a major bottleneck in drug development with the fundamental stage being the identification and validation of drug targets of interest for further downstream processes. Thus, various computational methods have been developed to complement experimental approaches in drug discovery. Here, we present an overview of various computational methods and tools applied in predicting or validating drug targets and drug-like molecules. We provide an overview on their advantages and compare these methods to identify effective methods which likely lead to optimal results. We also explore major sources of drug failure considering the challenges and opportunities involved. This review might guide researchers on selecting the most efficient approach or technique during the computational drug discovery process.
Collapse
Affiliation(s)
- Francis E Agamah
- Division of Human Genetics, Department of Pathology, University of Cape Town, Observatory 7925, South Africa
| | - Gaston K Mazandu
- Division of Human Genetics, Department of Pathology, University of Cape Town, Observatory 7925, South Africa
- African Institute for Mathematical Sciences, Muizenberg, Cape Town 7945, South Africa
| | - Radia Hassan
- Division of Human Genetics, Department of Pathology, University of Cape Town, Observatory 7925, South Africa
| | - Christian D Bope
- Division of Human Genetics, Department of Pathology, University of Cape Town, Observatory 7925, South Africa
- Faculty of Sciences, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Nicholas E Thomford
- Division of Human Genetics, Department of Pathology, University of Cape Town, Observatory 7925, South Africa
- School of Medical Sciences, University of Cape Coast, PMB, Cape Coast, Ghana
| | - Anita Ghansah
- Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, PO Box LG 581, Legon, Ghana
| | - Emile R Chimusa
- Division of Human Genetics, Department of Pathology, University of Cape Town, Observatory 7925, South Africa
| |
Collapse
|
14
|
Wilkinson IVL, Terstappen GC, Russell AJ. Combining experimental strategies for successful target deconvolution. Drug Discov Today 2020; 25:S1359-6446(20)30373-1. [PMID: 32971235 DOI: 10.1016/j.drudis.2020.09.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/10/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023]
Abstract
Investment in phenotypic drug discovery has led to increased demand for rapid and robust target deconvolution to aid successful drug development. Although methods for target identification and mechanism of action (MoA) discovery are flourishing, they typically lead to lists of putative targets. Validating which target(s) are involved in the therapeutic mechanism of a compound poses a significant challenge, requiring direct binding, target engagement, and functional studies in relevant physiological contexts. A combination of orthogonal approaches can allow target identification beyond the proteome as well as aid prioritisation for resource-intensive target validation studies.
Collapse
Affiliation(s)
- Isabel V L Wilkinson
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | - Georg C Terstappen
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3PQ, UK
| | - Angela J Russell
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK; Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3PQ, UK.
| |
Collapse
|
15
|
Ekanem TI, Tsai WL, Lin YH, Tan WQ, Chang HY, Huang TC, Chen HY, Lee KH. Identification of the Effects of Aspirin and Sulindac Sulfide on the Inhibition of HMGA2-Mediated Oncogenic Capacities in Colorectal Cancer. Molecules 2020; 25:molecules25173826. [PMID: 32842685 PMCID: PMC7504004 DOI: 10.3390/molecules25173826] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/17/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
Distant metastatic colorectal cancer (CRC) is present in approximately 25% of patients at initial diagnosis, and eventually half of CRC patients will develop metastatic disease. The 5-year survival rate for patients with metastatic CRC is a mere 12.5%; thus, there is an urgent need to investigate the molecular mechanisms of cancer progression in CRC. High expression of human high-mobility group A2 (HMGA2) is related to tumor progression, a poor prognosis, and a poor response to therapy for CRC. Therefore, HMGA2 is an attractive target for cancer therapy. In this study, we identified aspirin and sulindac sulfide as novel potential inhibitors of HMGA2 using a genome-wide mRNA signature-based approach. In addition, aspirin and sulindac sulfide induced cytotoxicity of CRC cells stably expressing HMGA2 by inhibiting cell proliferation and migration. Moreover, a gene set enrichment analysis (GSEA) revealed that gene sets related to inflammation were positively correlated with HMGA2 and that the main molecular function of these genes was categorized as a G-protein-coupled receptor (GPCR) activity event. Collectively, this is the first study to report that aspirin and sulindac sulfide are novel potential inhibitors of HMGA2, which can induce cytotoxicity of CRC cells stably expressing HMGA2 by inhibiting cell proliferation and migration through influencing inflammatory-response genes, the majority of which are involved in GPCR signaling.
Collapse
Affiliation(s)
- Titus Ime Ekanem
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan;
- Department of Hematology, University of Uyo, Uyo 520271, Nigeria
| | - Wei-Lun Tsai
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (W.-L.T.); (W.-Q.T.); (T.-C.H.)
| | - Yi-Hsuan Lin
- Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan;
| | - Wan-Qian Tan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (W.-L.T.); (W.-Q.T.); (T.-C.H.)
| | - Hsin-Yi Chang
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan;
| | - Tsui-Chin Huang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (W.-L.T.); (W.-Q.T.); (T.-C.H.)
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsin-Yi Chen
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (W.-L.T.); (W.-Q.T.); (T.-C.H.)
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (H.-Y.C.); (K.-H.L.); Tel.: +886-2-26972035 (H.-Y.C.); +886-2-26972035 (K.-H.L.); Fax: +886-2-66387537 (H.-Y.C.); +886-2-66387537 (K.-H.L.)
| | - Kuen-Haur Lee
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (W.-L.T.); (W.-Q.T.); (T.-C.H.)
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Cancer Center, Wan Fang Hospital, Taipei Medical University 11696, Taipei, Taiwan
- Correspondence: (H.-Y.C.); (K.-H.L.); Tel.: +886-2-26972035 (H.-Y.C.); +886-2-26972035 (K.-H.L.); Fax: +886-2-66387537 (H.-Y.C.); +886-2-66387537 (K.-H.L.)
| |
Collapse
|
16
|
Daina A, Michielin O, Zoete V. SwissTargetPrediction: updated data and new features for efficient prediction of protein targets of small molecules. Nucleic Acids Res 2020; 47:W357-W364. [PMID: 31106366 PMCID: PMC6602486 DOI: 10.1093/nar/gkz382] [Citation(s) in RCA: 1794] [Impact Index Per Article: 358.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/26/2019] [Accepted: 05/01/2019] [Indexed: 12/14/2022] Open
Abstract
SwissTargetPrediction is a web tool, on-line since 2014, that aims to predict the most probable protein targets of small molecules. Predictions are based on the similarity principle, through reverse screening. Here, we describe the 2019 version, which represents a major update in terms of underlying data, backend and web interface. The bioactivity data were updated, the model retrained and similarity thresholds redefined. In the new version, the predictions are performed by searching for similar molecules, in 2D and 3D, within a larger collection of 376 342 compounds known to be experimentally active on an extended set of 3068 macromolecular targets. An efficient backend implementation allows to speed up the process that returns results for a druglike molecule on human proteins in 15-20 s. The refreshed web interface enhances user experience with new features for easy input and improved analysis. Interoperability capacity enables straightforward submission of any input or output molecule to other on-line computer-aided drug design tools, developed by the SIB Swiss Institute of Bioinformatics. High levels of predictive performance were maintained despite more extended biological and chemical spaces to be explored, e.g. achieving at least one correct human target in the top 15 predictions for >70% of external compounds. The new SwissTargetPrediction is available free of charge (www.swisstargetprediction.ch).
Collapse
Affiliation(s)
- Antoine Daina
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, University of Lausanne, Quartier UNIL-Sorge, Bâtiment Amphipôle, CH-1015 Lausanne, Switzerland
| | - Olivier Michielin
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, University of Lausanne, Quartier UNIL-Sorge, Bâtiment Amphipôle, CH-1015 Lausanne, Switzerland.,Department of Oncology, University Hospital of Lausanne, Ludwig Cancer Research - Lausanne Branch, CH-1011 Lausanne, Switzerland
| | - Vincent Zoete
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, University of Lausanne, Quartier UNIL-Sorge, Bâtiment Amphipôle, CH-1015 Lausanne, Switzerland.,Department of Fundamental Oncology, University of Lausanne, Ludwig Cancer Research - Lausanne Branch, Route de la Corniche 9A, CH-1066 Epalinges, Switzerland
| |
Collapse
|
17
|
Jenkinson S, Schmidt F, Rosenbrier Ribeiro L, Delaunois A, Valentin JP. A practical guide to secondary pharmacology in drug discovery. J Pharmacol Toxicol Methods 2020; 105:106869. [PMID: 32302774 DOI: 10.1016/j.vascn.2020.106869] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/21/2020] [Accepted: 04/03/2020] [Indexed: 01/29/2023]
Abstract
Secondary pharmacological profiling is increasingly applied in pharmaceutical drug discovery to address unwanted pharmacological side effects of drug candidates before entering the clinic. Regulators, drug makers and patients share a demand for deep characterization of secondary pharmacology effects of novel drugs and their metabolites. The scope of such profiling has therefore expanded substantially in the past two decades, leading to the implementation of broad in silico profiling methods and focused in vitro off-target screening panels, to identify liabilities, but also opportunities, as early as possible. The pharmaceutical industry applies such panels at all stages of drug discovery routinely up to early development. Nevertheless, target composition, screening technologies, assay formats, interpretation and scheduling of panels can vary significantly between companies in the absence of dedicated guidelines. To contribute towards best practices in secondary pharmacology profiling, this review aims to summarize the state-of-the art in this field. Considerations are discussed with respect to panel design, screening strategy, implementation and interpretation of the data, including regulatory perspectives. The cascaded, or integrated, use of in silico and off-target profiling allows to exploit synergies for comprehensive safety assessment of drug candidates.
Collapse
Affiliation(s)
- Stephen Jenkinson
- Drug Safety Research and Development, Pfizer Inc., La Jolla, CA 92121, United States of America.
| | - Friedemann Schmidt
- Sanofi, R&D Preclinical Safety, Industriepark Höchst, 65926 Frankfurt/Main, Germany
| | - Lyn Rosenbrier Ribeiro
- Medicines Discovery Catapult, Block 35, Mereside, Alderley Park, Alderley Edge, SK10 4TG, United Kingdom
| | - Annie Delaunois
- UCB BioPharma SRL, Early Solutions, Development Science, Non-Clinical Safety, 1420 Braine L'Alleud, Walloon Region, Belgium
| | - Jean-Pierre Valentin
- UCB BioPharma SRL, Early Solutions, Development Science, Non-Clinical Safety, 1420 Braine L'Alleud, Walloon Region, Belgium
| |
Collapse
|
18
|
Custodio JMF, Moura AF, de Moraes MO, Perez CN, Napolitano HB. On the in silico and in vitro anticancer activity of sulfonamide chalcones: potential JNKK3 inhibitors. NEW J CHEM 2020. [DOI: 10.1039/c9nj05612b] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although many compound classes have been studied as JNK inhibitors, we are interested in using chalcones for this purpose. Do different groups drive to different bindings modes to JNK?
Collapse
Affiliation(s)
- Jean M. F. Custodio
- Department of Chemistry and Biochemistry
- University of Notre Dame
- Notre Dame
- USA
- Instituto de Química
| | - Andrea F. Moura
- Núcleo de Pesquisas e Desenvolvimento de Medicamentos
- Universidade Federal do Ceará
- Fortaleza
- Brazil
- Núcleo de Pesquisa em Biotecnologia e Biodiversidade – BIOTEC
| | - Manoel O. de Moraes
- Núcleo de Pesquisas e Desenvolvimento de Medicamentos
- Universidade Federal do Ceará
- Fortaleza
- Brazil
| | | | | |
Collapse
|
19
|
|