1
|
Cozzini P, Agosta F. The Potential of Molecular Docking for Predictive Toxicology. Methods Mol Biol 2025; 2834:171-180. [PMID: 39312165 DOI: 10.1007/978-1-0716-4003-6_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Molecular modeling techniques are widely used in medicinal chemistry for the study of biological targets, the rational design of new drugs, or the investigation of their mechanism of action.They are also applied in toxicology to identify chemical potential harmful effects.Molecular docking is a computational technique to predict the ligand binding mode and evaluate the interaction energy with a biological target.This chapter describes a computational workflow to predict possible endocrine disruptors on peroxisome proliferator-activated receptor alpha (PPARα), a nuclear receptor involved in glucose and lipid metabolism. The analyzed compounds are food contact chemicals, natural or synthetic substances intentionally added to food or released from the package or during production or technological processes.
Collapse
Affiliation(s)
- Pietro Cozzini
- Molecular Modeling Lab. Food and Drug Department, University of Parma, Parma, Italy.
| | - Federica Agosta
- Molecular Modeling Lab. Food and Drug Department, University of Parma, Parma, Italy
| |
Collapse
|
2
|
Aggarwal R, R Koes D. PharmRL: pharmacophore elucidation with deep geometric reinforcement learning. BMC Biol 2024; 22:301. [PMID: 39736736 DOI: 10.1186/s12915-024-02096-5] [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: 09/04/2024] [Accepted: 12/16/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND Molecular interactions between proteins and their ligands are important for drug design. A pharmacophore consists of favorable molecular interactions in a protein binding site and can be utilized for virtual screening. Pharmacophores are easiest to identify from co-crystal structures of a bound protein-ligand complex. However, designing a pharmacophore in the absence of a ligand is a much harder task. RESULTS In this work, we develop a deep learning method that can identify pharmacophores in the absence of a ligand. Specifically, we train a CNN model to identify potential favorable interactions in the binding site, and develop a deep geometric Q-learning algorithm that attempts to select an optimal subset of these interaction points to form a pharmacophore. With this algorithm, we show better prospective virtual screening performance, in terms of F1 scores, on the DUD-E dataset than random selection of ligand-identified features from co-crystal structures. We also conduct experiments on the LIT-PCBA dataset and show that it provides efficient solutions for identifying active molecules. Finally, we test our method by screening the COVID moonshot dataset and show that it would be effective in identifying prospective lead molecules even in the absence of fragment screening experiments. CONCLUSIONS PharmRL addresses the need for automated methods in pharmacophore design, particularly in cases where a cognate ligand is unavailable. Experimental results demonstrate that PharmRL generates functional pharmacophores. Additionally, we provide a Google Colab notebook to facilitate the use of this method.
Collapse
Affiliation(s)
- Rishal Aggarwal
- Joint PhD Program in Computational Biology, Carnegie Mellon University-University of Pittsburgh, Pittsburgh, PA, USA
- Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - David R Koes
- Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Szalai T, Bajusz D, Börzsei R, Zsidó BZ, Ilaš J, Ferenczy GG, Hetényi C, Keserű GM. Effect of Water Networks On Ligand Binding: Computational Predictions vs Experiments. J Chem Inf Model 2024; 64:8980-8998. [PMID: 39576659 PMCID: PMC11632780 DOI: 10.1021/acs.jcim.4c01291] [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: 07/23/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 12/10/2024]
Abstract
Rational drug design focuses on the explanation and prediction of complex formation between therapeutic targets and small-molecule ligands. As a third and often overlooked interacting partner, water molecules play a critical role in the thermodynamics of protein-ligand binding, impacting both the entropy and enthalpy components of the binding free energy and by extension, on-target affinity and bioactivity. The community has realized the importance of binding site waters, as evidenced by the number of computational tools to predict the structure and thermodynamics of their networks. However, quantitative experimental characterization of relevant protein-ligand-water systems, and consequently the validation of these modeling methods, remains challenging. Here, we investigated the impact of solvent exchange from light (H2O) to heavy water (D2O) to provide complete thermodynamic profiling of these ternary systems. Utilizing the solvent isotope effects, we gain a deeper understanding of the energetic contributions of various components. Specifically, we conducted isothermal titration calorimetry experiments on trypsin with a series of p-substituted benzamidines, as well as carbonic anhydrase II (CAII) with a series of aromatic sulfonamides. Significant differences in binding enthalpies found between light vs heavy water indicate a substantial role of the binding site water network in protein-ligand binding. Next, we challenged two conceptually distinct modeling methods, the grid-based WaterFLAP and the molecular dynamics-based MobyWat, by predicting and scoring relevant water networks. The predicted water positions accurately reproduce those in available high-resolution X-ray and neutron diffraction structures of the relevant protein-ligand complexes. Estimated energetic contributions of the identified water networks were corroborated by the experimental thermodynamics data. Besides providing a direct validation for the predictive power of these methods, our findings confirmed the importance of considering binding site water networks in computational ligand design.
Collapse
Affiliation(s)
- Tibor
Viktor Szalai
- Medicinal
Chemistry Research Group, Drug Innovation Centre, HUN-REN Research
Centre for Natural Sciences, Magyar tudósok krt. 2, Budapest 1117, Hungary
- Department
of Inorganic and Analytical Chemistry, Faculty of Chemical Technology
and Biotechnology, Budapest University of
Technology and Economics, Műegyetem rkp. 3, Budapest H-1111, Hungary
- National
Drug Research and Development Laboratory, Magyar tudósok krt. 2, Budapest 1117, Hungary
| | - Dávid Bajusz
- Medicinal
Chemistry Research Group, Drug Innovation Centre, HUN-REN Research
Centre for Natural Sciences, Magyar tudósok krt. 2, Budapest 1117, Hungary
- National
Drug Research and Development Laboratory, Magyar tudósok krt. 2, Budapest 1117, Hungary
| | - Rita Börzsei
- National
Drug Research and Development Laboratory, Magyar tudósok krt. 2, Budapest 1117, Hungary
- Pharmacoinformatics
Unit, Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti út 12, Pécs H-7624, Hungary
| | - Balázs Zoltán Zsidó
- National
Drug Research and Development Laboratory, Magyar tudósok krt. 2, Budapest 1117, Hungary
- Pharmacoinformatics
Unit, Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti út 12, Pécs H-7624, Hungary
| | - Janez Ilaš
- Medicinal
Chemistry Research Group, Drug Innovation Centre, HUN-REN Research
Centre for Natural Sciences, Magyar tudósok krt. 2, Budapest 1117, Hungary
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - György G. Ferenczy
- Medicinal
Chemistry Research Group, Drug Innovation Centre, HUN-REN Research
Centre for Natural Sciences, Magyar tudósok krt. 2, Budapest 1117, Hungary
- National
Drug Research and Development Laboratory, Magyar tudósok krt. 2, Budapest 1117, Hungary
| | - Csaba Hetényi
- National
Drug Research and Development Laboratory, Magyar tudósok krt. 2, Budapest 1117, Hungary
- Pharmacoinformatics
Unit, Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti út 12, Pécs H-7624, Hungary
| | - György M. Keserű
- Medicinal
Chemistry Research Group, Drug Innovation Centre, HUN-REN Research
Centre for Natural Sciences, Magyar tudósok krt. 2, Budapest 1117, Hungary
- National
Drug Research and Development Laboratory, Magyar tudósok krt. 2, Budapest 1117, Hungary
- Department
of Organic Chemistry and Technology, Faculty of Chemical Technology
and Biotechnology, Budapest University of
Technology and Economics, Műegyetem rkp. 3, Budapest H-1111, Hungary
| |
Collapse
|
4
|
Peter S, Siragusa L, Thomas M, Palomba T, Cross S, O’Boyle NM, Bajusz D, Ferenczy GG, Keserű GM, Bottegoni G, Bender B, Chen I, De Graaf C. Comparative Study of Allosteric GPCR Binding Sites and Their Ligandability Potential. J Chem Inf Model 2024; 64:8176-8192. [PMID: 39441864 PMCID: PMC11558664 DOI: 10.1021/acs.jcim.4c00819] [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: 05/10/2024] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024]
Abstract
The steadily growing number of experimental G-protein-coupled receptor (GPCR) structures has revealed diverse locations of allosteric modulation, and yet few drugs target them. This gap highlights the need for a deeper understanding of allosteric modulation in GPCR drug discovery. The current work introduces a systematic annotation scheme to structurally classify GPCR binding sites based on receptor class, transmembrane helix contacts, and, for membrane-facing sites, membrane sublocation. This GPCR specific annotation scheme was applied to 107 GPCR structures bound by small molecules contributing to 24 distinct allosteric binding sites for comparative evaluation of three binding site detection methods (BioGPS, SiteMap, and FTMap). BioGPS identified the most in 22 of 24 sites. In addition, our property analysis showed that extrahelical allosteric ligands and binding sites represent a distinct chemical space characterized by shallow pockets with low volume, and the corresponding allosteric ligands showed an enrichment of halogens. Furthermore, we demonstrated that combining receptor and ligand similarity can be a viable method for ligandability assessment. One challenge regarding site prediction is the ligand shaping effect on the observed binding site, especially for extrahelical sites where the ligand-induced effect was most pronounced. To our knowledge, this is the first study presenting a binding site annotation scheme standardized for GPCRs, and it allows a comparison of allosteric binding sites across different receptors in an objective way. The insight from this study provides a framework for future GPCR binding site studies and highlights the potential of targeting allosteric sites for drug development.
Collapse
Affiliation(s)
- Sonja Peter
- Computational
Chemistry, Nxera Pharma U.K., Steinmetz Building, Granta Park, Cambridge CB21 6DG, United Kingdom
- Department
of Biomolecular Sciences, University of
Urbino Carlo Bo, Piazza Rinascimento 6, Urbino 61029, Italy
| | - Lydia Siragusa
- Kinetic Business
Centre, Molecular Discovery Ltd., Theobald Street, Elstree, Borehamwood, Hertfordshire WD6 4PJ, United
Kingdom
- Molecular
Horizon srl, via Montelino
30, Bettona, PG 06084, Italy
| | - Morgan Thomas
- Computational
Chemistry, Nxera Pharma U.K., Steinmetz Building, Granta Park, Cambridge CB21 6DG, United Kingdom
- Yusuf Hamied
Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Tommaso Palomba
- Kinetic Business
Centre, Molecular Discovery Ltd., Theobald Street, Elstree, Borehamwood, Hertfordshire WD6 4PJ, United
Kingdom
| | - Simon Cross
- Kinetic Business
Centre, Molecular Discovery Ltd., Theobald Street, Elstree, Borehamwood, Hertfordshire WD6 4PJ, United
Kingdom
| | - Noel M. O’Boyle
- Computational
Chemistry, Nxera Pharma U.K., Steinmetz Building, Granta Park, Cambridge CB21 6DG, United Kingdom
| | - Dávid Bajusz
- Medicinal
Chemistry Research Group and Drug Innovation Centre, HUN-REN Research Centre for Natural Sciences, Magyar tudósok krt. 2, Budapest 1117, Hungary
| | - György G. Ferenczy
- Medicinal
Chemistry Research Group and Drug Innovation Centre, HUN-REN Research Centre for Natural Sciences, Magyar tudósok krt. 2, Budapest 1117, Hungary
| | - György M. Keserű
- Medicinal
Chemistry Research Group and Drug Innovation Centre, HUN-REN Research Centre for Natural Sciences, Magyar tudósok krt. 2, Budapest 1117, Hungary
| | - Giovanni Bottegoni
- Department
of Biomolecular Sciences, University of
Urbino Carlo Bo, Piazza Rinascimento 6, Urbino 61029, Italy
- Institute
of Clinical Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Brian Bender
- Computational
Chemistry, Nxera Pharma U.K., Steinmetz Building, Granta Park, Cambridge CB21 6DG, United Kingdom
| | - Ijen Chen
- Computational
Chemistry, Nxera Pharma U.K., Steinmetz Building, Granta Park, Cambridge CB21 6DG, United Kingdom
| | - Chris De Graaf
- Computational
Chemistry, Nxera Pharma U.K., Steinmetz Building, Granta Park, Cambridge CB21 6DG, United Kingdom
| |
Collapse
|
5
|
Milon TI, Wang Y, Fontenot RL, Khajouie P, Villinger F, Raghavan V, Xu W. Development of a novel representation of drug 3D structures and enhancement of the TSR-based method for probing drug and target interactions. Comput Biol Chem 2024; 112:108117. [PMID: 38852360 PMCID: PMC11390338 DOI: 10.1016/j.compbiolchem.2024.108117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/13/2024] [Accepted: 05/31/2024] [Indexed: 06/11/2024]
Abstract
Understanding the mechanisms underlying interactions between drugs and target proteins is critical for drug discovery. In our earlier studies, we introduced the Triangular Spatial Relationship (TSR)-based algorithm, which enables the representation of a protein's 3D structure as a vector of integers (TSR keys). These TSR keys correspond to substructures of the 3D structure of a protein and are computed based on the triangles constructed by all possible triples of Cα atoms within the protein. In this study, we report on a new TSR-based algorithm for probing drug and target interactions. Specifically, we have extended the previous algorithm in three novel directions: TSR keys for representing the 3D structure of a drug or a ligand, cross TSR keys between drugs and their targets and intra-residual TSR keys for phosphorylated amino acids. The outcomes illustrate the key contributions as follows: (i) The TSR-based method, which uses the TSR keys as features, is unique in its capability to interpret hierarchical relationships of drugs as well as drug - target complexes using common and specific TSR keys. (ii) The method can distinguish not only the binding sites from the rest of the protein structures, but also the binding sites of primary targets from those of off-targets. (iii) The method has the potential to correlate the 3D structures of drugs with their functions. (iv) Representation of 3D structures by TSR keys has its unique advantage in terms of ease of making searching for similar substructures across structure datasets easier. In summary, this study presents a novel computational methodology, with significant advantages, for providing insights into the mechanism underlying drug and target interactions.
Collapse
Affiliation(s)
- Tarikul I Milon
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 44370, Lafayette, LA 70504, USA
| | - Yuhong Wang
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Ryan L Fontenot
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 44370, Lafayette, LA 70504, USA
| | - Poorya Khajouie
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 44370, Lafayette, LA 70504, USA; The Center for Advanced Computer Studies, University of Louisiana at Lafayette, LA 70504, USA
| | - Francois Villinger
- Department of Biology, University of Louisiana at Lafayette, New Iberia, LA 70560, USA
| | - Vijay Raghavan
- The Center for Advanced Computer Studies, University of Louisiana at Lafayette, LA 70504, USA
| | - Wu Xu
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 44370, Lafayette, LA 70504, USA.
| |
Collapse
|
6
|
Wahl J. PheSA: An Open-Source Tool for Pharmacophore-Enhanced Shape Alignment. J Chem Inf Model 2024; 64:5944-5953. [PMID: 39092495 DOI: 10.1021/acs.jcim.4c00516] [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: 08/04/2024]
Abstract
PheSA is an open-source pharmacophore- and shape-based screening and molecular alignment tool that is fully open-source as part of OpenChemLib. Supporting standard ligand-based screening, flexible refinement of alignments, and receptor-guided shape docking, PheSA is a very flexible tool and can be used for different use cases in structure-based drug design. We present the algorithm and different benchmark studies that investigate the screening performance and also the quality of the generated alignments and the pose prediction performance of the receptor-guided PheSA algorithm. An important finding is the effect of the type of similarity metric used for measuring screening enrichment (symmetric Tanimoto versus asymmetric Tversky), whereby we could observe improved enrichment rates by using Tversky. PheSA exhibits enrichments on the DUD-E that are on par with commercial methods.
Collapse
Affiliation(s)
- Joel Wahl
- Scientific Computing Drug Discovery, Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| |
Collapse
|
7
|
Desantis J, Bazzacco A, Eleuteri M, Tuci S, Bianconi E, Macchiarulo A, Mercorelli B, Loregian A, Goracci L. Design, synthesis, and biological evaluation of first-in-class indomethacin-based PROTACs degrading SARS-CoV-2 main protease and with broad-spectrum antiviral activity. Eur J Med Chem 2024; 268:116202. [PMID: 38394929 DOI: 10.1016/j.ejmech.2024.116202] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024]
Abstract
To date, Proteolysis Targeting Chimera (PROTAC) technology has been successfully applied to mediate proteasomal-induced degradation of several pharmaceutical targets mainly related to oncology, immune disorders, and neurodegenerative diseases. On the other hand, its exploitation in the field of antiviral drug discovery is still in its infancy. Recently, we described two indomethacin (INM)-based PROTACs displaying broad-spectrum antiviral activity against coronaviruses. Here, we report the design, synthesis, and characterization of a novel series of INM-based PROTACs that recruit either Von-Hippel Lindau (VHL) or cereblon (CRBN) E3 ligases. The panel of INM-based PROTACs was also enlarged by varying the linker moiety. The antiviral activity resulted very susceptible to this modification, particularly for PROTACs hijacking VHL as E3 ligase, with one piperazine-based compound (PROTAC 6) showing potent anti-SARS-CoV-2 activity in infected human lung cells. Interestingly, degradation assays in both uninfected and virus-infected cells with the most promising PROTACs emerged so far (PROTACs 5 and 6) demonstrated that INM-PROTACs do not degrade human PGES-2 protein, as initially hypothesized, but induce the concentration-dependent degradation of SARS-CoV-2 main protease (Mpro) both in Mpro-transfected and in SARS-CoV-2-infected cells. Importantly, thanks to the target degradation, INM-PROTACs exhibited a considerable enhancement in antiviral activity with respect to indomethacin, with EC50 values in the low-micromolar/nanomolar range. Finally, kinetic solubility as well as metabolic and chemical stability were measured for PROTACs 5 and 6. Altogether, the identification of INM-based PROTACs as the first class of SARS-CoV-2 Mpro degraders demonstrating activity also in SARS-CoV-2-infected cells represents a significant advance in the development of effective, broad-spectrum anti-coronavirus strategies.
Collapse
Affiliation(s)
- Jenny Desantis
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Italy
| | | | - Michela Eleuteri
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Italy
| | - Sara Tuci
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Elisa Bianconi
- Department of Pharmaceutical Science, University of Perugia, Italy
| | | | | | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, Padua, Italy.
| | - Laura Goracci
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Italy.
| |
Collapse
|
8
|
Palomba T, Baroni M, Cross S, Cruciani G, Siragusa L. ELIOT: A platform to navigate the E3 pocketome and aid the design of new PROTACs. Chem Biol Drug Des 2023; 101:69-86. [PMID: 35857806 DOI: 10.1111/cbdd.14123] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/11/2022] [Accepted: 07/17/2022] [Indexed: 12/15/2022]
Abstract
Proteolysis-targeting chimeras (PROTACs) are novel therapeutics for the treatment of human disease. They exploit the enormous potential of the E3 ligases, a class of proteins that mark a target protein for degradation via the ubiquitin-proteasome system. Despite the existence of several E3 ligase-related databases, the choice of the functioning ligase is limited to only 1.6% of those available, probably due to the fragmentary understanding of their structures and their known ligands; in fact, none of the existing databases report detailed studies covering their 3D structure or their pockets. Here, we report ELIOT (E3 LIgase pocketOme navigaTor), an accurate and complete platform containing the E3 ligase pocketome to enable navigation and selection of new E3 ligases and new ligands for the design of new PROTACs. All E3 ligase pockets were characterized with innovative 3D descriptors including their PROTAC-ability score, and similarity analyses between E3 pockets are presented. Tissue specificity and their degree of involvement in patients with specific cancer types are also annotated for each E3 ligase, enabling appropriate selection for the design of a PROTAC with improved specificity. All data are available at https://eliot.moldiscovery.com.
Collapse
Affiliation(s)
- Tommaso Palomba
- Laboratory for Chemometrics and Molecular Modeling, Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy
| | - Massimo Baroni
- Molecular Discovery Ltd., The Kinetic Centre, Hertfordshire, UK
| | - Simon Cross
- Molecular Discovery Ltd., The Kinetic Centre, Hertfordshire, UK
| | - Gabriele Cruciani
- Laboratory for Chemometrics and Molecular Modeling, Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy
| | - Lydia Siragusa
- Molecular Discovery Ltd., The Kinetic Centre, Hertfordshire, UK.,Molecular Horizon Srl, Bettona, Italy
| |
Collapse
|
9
|
Costantino L, Ferrari S, Santucci M, Salo-Ahen OMH, Carosati E, Franchini S, Lauriola A, Pozzi C, Trande M, Gozzi G, Saxena P, Cannazza G, Losi L, Cardinale D, Venturelli A, Quotadamo A, Linciano P, Tagliazucchi L, Moschella MG, Guerrini R, Pacifico S, Luciani R, Genovese F, Henrich S, Alboni S, Santarem N, da Silva Cordeiro A, Giovannetti E, Peters GJ, Pinton P, Rimessi A, Cruciani G, Stroud RM, Wade RC, Mangani S, Marverti G, D'Arca D, Ponterini G, Costi MP. Destabilizers of the thymidylate synthase homodimer accelerate its proteasomal degradation and inhibit cancer growth. eLife 2022; 11:e73862. [PMID: 36475542 PMCID: PMC9831607 DOI: 10.7554/elife.73862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
Drugs that target human thymidylate synthase (hTS), a dimeric enzyme, are widely used in anticancer therapy. However, treatment with classical substrate-site-directed TS inhibitors induces over-expression of this protein and development of drug resistance. We thus pursued an alternative strategy that led us to the discovery of TS-dimer destabilizers. These compounds bind at the monomer-monomer interface and shift the dimerization equilibrium of both the recombinant and the intracellular protein toward the inactive monomers. A structural, spectroscopic, and kinetic investigation has provided evidence and quantitative information on the effects of the interaction of these small molecules with hTS. Focusing on the best among them, E7, we have shown that it inhibits hTS in cancer cells and accelerates its proteasomal degradation, thus causing a decrease in the enzyme intracellular level. E7 also showed a superior anticancer profile to fluorouracil in a mouse model of human pancreatic and ovarian cancer. Thus, over sixty years after the discovery of the first TS prodrug inhibitor, fluorouracil, E7 breaks the link between TS inhibition and enhanced expression in response, providing a strategy to fight drug-resistant cancers.
Collapse
Affiliation(s)
- Luca Costantino
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Stefania Ferrari
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Matteo Santucci
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Outi MH Salo-Ahen
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical StudiesHeidelbergGermany
| | - Emanuele Carosati
- Department of Chemistry, Biology and Biotechnology, University of PerugiaPerugiaItaly
| | - Silvia Franchini
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Angela Lauriola
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Cecilia Pozzi
- Department of Biotechnology, Chemistry and Pharmacy, University of SienaSienaItaly
| | - Matteo Trande
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Gaia Gozzi
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Puneet Saxena
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Giuseppe Cannazza
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Lorena Losi
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Daniela Cardinale
- Respiratory, Critical Care & Anesthesia UCL Great Ormond Street Institute of Child HealthLondonUnited Kingdom
| | - Alberto Venturelli
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Antonio Quotadamo
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Pasquale Linciano
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| | | | - Maria Gaetana Moschella
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, ItalyModenaItaly
| | - Remo Guerrini
- Department of Chemical and Pharmaceutical Science, University of FerraraFerraraItaly
| | - Salvatore Pacifico
- Department of Chemical and Pharmaceutical Science, University of FerraraFerraraItaly
| | - Rosaria Luciani
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Filippo Genovese
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Stefan Henrich
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical StudiesHeidelbergGermany
| | - Silvia Alboni
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| | | | - Anabela da Silva Cordeiro
- IBMC I3SPortoPortugal
- Department of Biological Sciences, Faculty of Pharmacy, University of PortoPortoPortugal
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, 1081HV, Vrije Universiteit AmsterdamAmsterdamNetherlands
- CancerPharmacology Lab, Fondazione Pisana per la ScienzaPisaItaly
| | - Godefridus J Peters
- Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, 1081HV, Vrije Universiteit AmsterdamAmsterdamNetherlands
| | - Paolo Pinton
- Dept. of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of FerraraFerraraItaly
| | - Alessandro Rimessi
- Dept. of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of FerraraFerraraItaly
| | - Gabriele Cruciani
- Department of Chemistry, Biology and Biotechnology, University of PerugiaPerugiaItaly
| | - Robert M Stroud
- Biochemistry and Biophysics Department, University of California San FranciscoSan FranciscoUnited States
| | - Rebecca C Wade
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical StudiesHeidelbergGermany
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg UniversityHeidelbergGermany
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg UniversityHeidelbergGermany
| | - Stefano Mangani
- Department of Biotechnology, Chemistry and Pharmacy, University of SienaSienaItaly
| | - Gaetano Marverti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Domenico D'Arca
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Glauco Ponterini
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| | - Maria Paola Costi
- Department of Life Sciences, University of Modena and Reggio EmiliaModenaItaly
| |
Collapse
|
10
|
Exploiting ELIOT for Scaffold-Repurposing Opportunities: TRIM33 a Possible Novel E3 Ligase to Expand the Toolbox for PROTAC Design. Int J Mol Sci 2022; 23:ijms232214218. [PMID: 36430693 PMCID: PMC9698485 DOI: 10.3390/ijms232214218] [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: 10/26/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
The field of targeted protein degradation, through the control of the ubiquitin-proteasome system (UPS), is progressing considerably; to exploit this new therapeutic modality, the proteolysis targeting chimera (PROTAC) technology was born. The opportunity to use PROTACs engaging of new E3 ligases that can hijack and control the UPS system could greatly extend the applicability of degrading molecules. To this end, here we show a potential application of the ELIOT (E3 LIgase pocketOme navigaTor) platform, previously published by this group, for a scaffold-repurposing strategy to identify new ligands for a novel E3 ligase, such as TRIM33. Starting from ELIOT, a case study of the cross-relationship using GRID Molecular Interaction Field (MIF) similarities between TRIM24 and TRIM33 binding sites was selected. Based on the assumption that similar pockets could bind similar ligands and considering that TRIM24 has 12 known co-crystalised ligands, we applied a scaffold-repurposing strategy for the identification of TRIM33 ligands exploiting the scaffold of TRIM24 ligands. We performed a deeper computational analysis to identify pocket similarities and differences, followed by docking and water analysis; selected ligands were synthesised and subsequently tested against TRIM33 via HTRF binding assay, and we obtained the first-ever X-ray crystallographic complexes of TRIM33α with three of the selected compounds.
Collapse
|
11
|
Gado F, Ferrisi R, Polini B, Mohamed KA, Ricardi C, Lucarini E, Carpi S, Domenichini F, Stevenson LA, Rapposelli S, Saccomanni G, Nieri P, Ortore G, Pertwee RG, Ghelardini C, Di Cesare Mannelli L, Chiellini G, Laprairie RB, Manera C. Design, Synthesis, and Biological Activity of New CB2 Receptor Ligands: from Orthosteric and Allosteric Modulators to Dualsteric/Bitopic Ligands. J Med Chem 2022; 65:9918-9938. [PMID: 35849804 PMCID: PMC10168668 DOI: 10.1021/acs.jmedchem.2c00582] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The design of dualsteric/bitopic agents as single chemical entities able to simultaneously interact with both the orthosteric and an allosteric binding site represents a novel approach in medicinal chemistry. Biased dualsteric/bitopic agents could enhance certain signaling pathways while diminishing the others that cause unwanted side effects. We have designed, synthesized, and functionally characterized the first CB2R heterobivalent bitopic ligands. In contrast to the parent orthosteric compound, our bitopic ligands selectively target CB2R versus CB1R and show a functional selectivity for the cAMP signaling pathway versus βarrestin2 recruitment. Moreover, the most promising bitopic ligand FD-22a displayed anti-inflammatory activity in a human microglial cell inflammatory model and antinociceptive activity in vivo in an experimental mouse model of neuropathic pain. Finally, computational studies clarified the binding mode of these compounds inside the CB2R, further confirming their bitopic nature.
Collapse
Affiliation(s)
- Francesca Gado
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy
| | - Rebecca Ferrisi
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy
| | - Beatrice Polini
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy.,Department of Pathology, University of Pisa, Pisa 56126, Italy
| | - Kawthar A Mohamed
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon SK S7N 5E5, Canada
| | | | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Section of Pharmacology and Toxicology, University of Florence, Florence 50139, Italy
| | - Sara Carpi
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy.,NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro, Pisa 56126, Italy
| | | | - Lesley A Stevenson
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, U.K
| | - Simona Rapposelli
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy.,CISUP, Centre for Instrumentation Sharing Pisa University, Lungarno Pacinotti 43, Pisa 56126, Italy
| | | | - Paola Nieri
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy
| | | | - Roger G Pertwee
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, U.K
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Section of Pharmacology and Toxicology, University of Florence, Florence 50139, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health, Section of Pharmacology and Toxicology, University of Florence, Florence 50139, Italy
| | - Grazia Chiellini
- Department of Pathology, University of Pisa, Pisa 56126, Italy.,CISUP, Centre for Instrumentation Sharing Pisa University, Lungarno Pacinotti 43, Pisa 56126, Italy
| | - Robert B Laprairie
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon SK S7N 5E5, Canada.,Department of Pharmacology, College of Medicine, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Clementina Manera
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy.,CISUP, Centre for Instrumentation Sharing Pisa University, Lungarno Pacinotti 43, Pisa 56126, Italy
| |
Collapse
|
12
|
Discovery of new chemotypes of dual 5-HT 2A/D 2 receptor antagonists with a strategy of drug design methodologies. Future Med Chem 2022; 14:963-989. [PMID: 35674007 DOI: 10.4155/fmc-2021-0340] [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: 11/17/2022] Open
Abstract
Aim: Through the application of structure- and ligand-based methods, the authors aimed to create an integrative approach to developing a computational protocol for the rational drug design of potent dual 5-HT2A/D2 receptor antagonists without off-target activities on H1 receptors. Materials & methods: Molecular dynamics and virtual docking methods were used to identify key interactions of the structurally diverse antagonists in the binding sites of the studied targets, and to generate their bioactive conformations for further 3D-quantitative structure-activity relationship modeling. Results & conclusion: Toward the goal of finding multi-potent drugs with a more effective and safer profile, the obtained results led to the design of a new set of dual antagonists and opened a new perspective on the therapy for complex brain diseases.
Collapse
|
13
|
Smilova MD, Curran PR, Radoux CJ, von Delft F, Cole JC, Bradley AR, Marsden BD. Fragment Hotspot Mapping to Identify Selectivity-Determining Regions between Related Proteins. J Chem Inf Model 2022; 62:284-294. [PMID: 35020376 PMCID: PMC8790751 DOI: 10.1021/acs.jcim.1c00823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
![]()
Selectivity is a
crucial property in small molecule development.
Binding site comparisons within a protein family are a key piece of
information when aiming to modulate the selectivity profile of a compound.
Binding site differences can be exploited to confer selectivity for
a specific target, while shared areas can provide insights into polypharmacology.
As the quantity of structural data grows, automated methods are needed
to process, summarize, and present these data to users. We present
a computational method that provides quantitative and data-driven
summaries of the available binding site information from an ensemble
of structures of the same protein. The resulting ensemble maps identify
the key interactions important for ligand binding in the ensemble.
The comparison of ensemble maps of related proteins enables the identification
of selectivity-determining regions within a protein family. We applied
the method to three examples from the well-researched human bromodomain
and kinase families, demonstrating that the method is able to identify
selectivity-determining regions that have been used to introduce selectivity
in past drug discovery campaigns. We then illustrate how the resulting
maps can be used to automate comparisons across a target protein family.
Collapse
Affiliation(s)
- Mihaela D Smilova
- Centre for Medicines Discovery, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, U.K
| | - Peter R Curran
- The Cambridge Crystallographic Data Centre (CCDC), Cambridge CB2 1EZ, U.K.,Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Chris J Radoux
- Exscientia Ltd., The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, U.K
| | - Frank von Delft
- Centre for Medicines Discovery, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, U.K.,Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot OX11 0DE, U.K.,Research Complex at Harwell. Harwell Science and Innovation Campus, Didcot OX11 0FA, U.K.,Department of Biochemistry, University of Johannesburg, Auckland Park 2006, South Africa
| | - Jason C Cole
- The Cambridge Crystallographic Data Centre (CCDC), Cambridge CB2 1EZ, U.K
| | - Anthony R Bradley
- Exscientia Ltd., The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, U.K
| | - Brian D Marsden
- Centre for Medicines Discovery, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, U.K.,Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7DQ, U.K
| |
Collapse
|
14
|
Gado F, Ceni C, Ferrisi R, Sbrana G, Stevenson LA, Macchia M, Pertwee RG, Bertini S, Manera C, Ortore G. CB1 receptor binding sites for NAM and PAM: A first approach for studying, new n‑butyl‑diphenylcarboxamides as allosteric modulators. Eur J Pharm Sci 2021; 169:106088. [PMID: 34863873 DOI: 10.1016/j.ejps.2021.106088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 01/01/2023]
Abstract
The development of cannabinoid receptor type-1 (CB1R) modulators has been implicated in multiple pathophysiological events ranging from memory deficits to neurodegenerative disorders among others, even if their central psychiatric side effects such as depression, anxiety, and suicidal tendencies, have limited their clinical use. Thus, the identification of ligands which selectively act on peripheral CB1Rs, is becoming more interesting. A recent study reported a class of peripheral CB1R selective antagonists, characterized by a 5-aryl substituted nicotinamide core. These derivatives have structural similarities with the biphenyl compounds, endowed with CB2R antagonist activity, previously synthesized by our research group. In this work we combined the pharmacophoric portion of both classes, in order to obtain novel CBR antagonists. Among the synthesized compounds rather unexpectedly two compounds of this series, C7 and C10, did not show the radioligand ([3H]CP55940) displacement on CB1R but increased binding (∼ 150%), suggesting a possible allosteric behavior. Computational studies were performed to investigate the role of these compounds in CB1R modulation. The analysis of their binding poses in two different binding cavities of the CB1R surface, revealed a preferred interaction with the experimental binding site for negative allosteric modulators.
Collapse
Affiliation(s)
- Francesca Gado
- Department of Pharmacy, University of Pisa, 56126 Pisa Italy
| | - Costanza Ceni
- Department of Pharmacy, University of Pisa, 56126 Pisa Italy; Doctoral school in Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Rebecca Ferrisi
- Department of Pharmacy, University of Pisa, 56126 Pisa Italy
| | - Giulia Sbrana
- Department of Pharmacy, University of Pisa, 56126 Pisa Italy
| | - Lesley A Stevenson
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, AB25 2ZD Aberdeen, Scotland, UK
| | - Marco Macchia
- Department of Pharmacy, University of Pisa, 56126 Pisa Italy
| | - Roger G Pertwee
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, AB25 2ZD Aberdeen, Scotland, UK
| | - Simone Bertini
- Department of Pharmacy, University of Pisa, 56126 Pisa Italy
| | | | | |
Collapse
|
15
|
Altalib MK, Salim N. Similarity-Based Virtual Screen Using Enhanced Siamese Multi-Layer Perceptron. Molecules 2021; 26:6669. [PMID: 34771076 PMCID: PMC8588560 DOI: 10.3390/molecules26216669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/24/2021] [Accepted: 11/01/2021] [Indexed: 11/30/2022] Open
Abstract
Traditional drug development is a slow and costly process that leads to the production of new drugs. Virtual screening (VS) is a computational procedure that measures the similarity of molecules as one of its primary tasks. Many techniques for capturing the biological similarity between a test compound and a known target ligand have been established in ligand-based virtual screens (LBVSs). However, despite the good performances of the above methods compared to their predecessors, especially when dealing with molecules that have structurally homogenous active elements, they are not satisfied when dealing with molecules that are structurally heterogeneous. The main aim of this study is to improve the performance of similarity searching, especially with molecules that are structurally heterogeneous. The Siamese network will be used due to its capability to deal with complicated data samples in many fields. The Siamese multi-layer perceptron architecture will be enhanced by using two similarity distance layers with one fused layer, then multiple layers will be added after the fusion layer, and then the nodes of the model that contribute less or nothing during inference according to their signal-to-noise ratio values will be pruned. Several benchmark datasets will be used, which are: the MDL Drug Data Report (MDDR-DS1, MDDR-DS2, and MDDR-DS3), the Maximum Unbiased Validation (MUV), and the Directory of Useful Decoys (DUD). The results show the outperformance of the proposed method on standard Tanimoto coefficient (TAN) and other methods. Additionally, it is possible to reduce the number of nodes in the Siamese multilayer perceptron model while still keeping the effectiveness of recall on the same level.
Collapse
Affiliation(s)
- Mohammed Khaldoon Altalib
- School of Computing, Universiti Teknologi Malaysia, Johor Bahru 81310, Malaysia
- Computer Science Department, Education for Pure Science College, University of Mosul, Mosul 41002, Iraq
| | - Naomie Salim
- School of Computing, Universiti Teknologi Malaysia, Johor Bahru 81310, Malaysia
| |
Collapse
|
16
|
Khoshbin Z, Housaindokht MR, Izadyar M, Bozorgmehr MR, Verdian A. Recent advances in computational methods for biosensor design. Biotechnol Bioeng 2020; 118:555-578. [PMID: 33135778 DOI: 10.1002/bit.27618] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/25/2020] [Accepted: 10/29/2020] [Indexed: 01/20/2023]
Abstract
Biosensors are analytical tools with a great application in healthcare, food quality control, and environmental monitoring. They are of considerable interest to be designed by using cost-effective and efficient approaches. Designing biosensors with improved functionality or application in new target detection has been converted to a fast-growing field of biomedicine and biotechnology branches. Experimental efforts have led to valuable successes in the field of biosensor design; however, some deficiencies restrict their utilization for this purpose. Computational design of biosensors is introduced as a promising key to eliminate the gap. A set of reliable structure prediction of the biosensor segments, their stability, and accurate descriptors of molecular interactions are required to computationally design biosensors. In this review, we provide a comprehensive insight into the progress of computational methods to guide the design and development of biosensors, including molecular dynamics simulation, quantum mechanics calculations, molecular docking, virtual screening, and a combination of them as the hybrid methodologies. By relying on the recent advances in the computational methods, an opportunity emerged for them to be complementary or an alternative to the experimental methods in the field of biosensor design.
Collapse
Affiliation(s)
- Zahra Khoshbin
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | - Mohammad Izadyar
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | - Asma Verdian
- Department of Food Safety and Quality Control, Research Institute of Food Science and Technology (RIFST), Mashhad, Iran
| |
Collapse
|
17
|
Vázquez J, López M, Gibert E, Herrero E, Luque FJ. Merging Ligand-Based and Structure-Based Methods in Drug Discovery: An Overview of Combined Virtual Screening Approaches. Molecules 2020; 25:E4723. [PMID: 33076254 PMCID: PMC7587536 DOI: 10.3390/molecules25204723] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/06/2020] [Accepted: 10/11/2020] [Indexed: 12/20/2022] Open
Abstract
Virtual screening (VS) is an outstanding cornerstone in the drug discovery pipeline. A variety of computational approaches, which are generally classified as ligand-based (LB) and structure-based (SB) techniques, exploit key structural and physicochemical properties of ligands and targets to enable the screening of virtual libraries in the search of active compounds. Though LB and SB methods have found widespread application in the discovery of novel drug-like candidates, their complementary natures have stimulated continued efforts toward the development of hybrid strategies that combine LB and SB techniques, integrating them in a holistic computational framework that exploits the available information of both ligand and target to enhance the success of drug discovery projects. In this review, we analyze the main strategies and concepts that have emerged in the last years for defining hybrid LB + SB computational schemes in VS studies. Particularly, attention is focused on the combination of molecular similarity and docking, illustrating them with selected applications taken from the literature.
Collapse
Affiliation(s)
- Javier Vázquez
- Pharmacelera, Plaça Pau Vila, 1, Sector C 2a, Edificio Palau de Mar, 08039 Barcelona, Spain;
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), and Institute of Theoretical and Computational Chemistry (IQTC-UB), University of Barcelona, Av. Prat de la Riba 171, E-08921 Santa Coloma de Gramanet, Spain
| | - Manel López
- AB Science, Parc Scientifique de Luminy, Zone Luminy Enterprise, Case 922, 163 Av. de Luminy, 13288 Marseille, France;
| | - Enric Gibert
- Pharmacelera, Plaça Pau Vila, 1, Sector C 2a, Edificio Palau de Mar, 08039 Barcelona, Spain;
| | - Enric Herrero
- Pharmacelera, Plaça Pau Vila, 1, Sector C 2a, Edificio Palau de Mar, 08039 Barcelona, Spain;
| | - F. Javier Luque
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), and Institute of Theoretical and Computational Chemistry (IQTC-UB), University of Barcelona, Av. Prat de la Riba 171, E-08921 Santa Coloma de Gramanet, Spain
| |
Collapse
|
18
|
Sandomenico A, Caporale A, Doti N, Cross S, Cruciani G, Chambery A, De Falco S, Ruvo M. Synthetic Peptide Libraries: From Random Mixtures to In Vivo Testing. Curr Med Chem 2020; 27:997-1016. [PMID: 30009695 DOI: 10.2174/0929867325666180716110833] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 06/22/2018] [Accepted: 06/29/2018] [Indexed: 01/13/2023]
Abstract
Combinatorially generated molecular repertoires have been largely used to identify novel bioactive compounds. Ever more sophisticated technological solutions have been proposed to simplify and speed up such process, expanding the chemical diversity space and increasing the prospect to select new molecular entities with specific and potent activities against targets of therapeutic relevance. In this context, random mixtures of oligomeric peptides were originally used and since 25 years they represent a continuous source of bioactive molecules with potencies ranging from the sub-nM to microM concentration. Synthetic peptide libraries are still employed as starting "synthetic broths" of structurally and chemically diversified molecular fragments from which lead compounds can be extracted and further modified. Thousands of studies have been reported describing the application of combinatorial mixtures of synthetic peptides with different complexity and engrafted on diverse structural scaffolds for the identification of new compounds which have been further developed and also tested in in vivo models of relevant diseases. We briefly review some of the most used methodologies for library preparation and screening and the most recent case studies appeared in the literature where compounds have reached at least in vivo testing in animal or similar models. Recent technological advancements in biotechnology, engineering and computer science have suggested new options to facilitate the discovery of new bioactive peptides. In this instance, we anticipate here a new approach for the design of simple but focused tripeptide libraries against druggable cavities of therapeutic targets and its complementation with existing approaches.
Collapse
Affiliation(s)
- Annamaria Sandomenico
- Istituto di Biostrutture e Bioimmagini del CNR and CIRPeB, Universita Federico II di Napoli, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Andrea Caporale
- Istituto di Biostrutture e Bioimmagini del CNR and CIRPeB, Universita Federico II di Napoli, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Nunzianna Doti
- Istituto di Biostrutture e Bioimmagini del CNR and CIRPeB, Universita Federico II di Napoli, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Simon Cross
- Molecular Discovery Ltd, Unit 501 Centennial Park, Centennial Avenue Elstree, Borehamwood, Hertfordshire WD6 3FG, United Kingdom
| | - Gabriele Cruciani
- Molecular Discovery Ltd, Unit 501 Centennial Park, Centennial Avenue Elstree, Borehamwood, Hertfordshire WD6 3FG, United Kingdom.,Dipartimento di Chimica, Biologia e Biotecnologia, Via Elce di Sotto 8, 06123 Perugia, Italy
| | - Angela Chambery
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università della Campania "Luigi Vanvitelli", via Vivaldi, 43, 81100 Caserta, Italy
| | - Sandro De Falco
- Istituto di Genetica e Biofisica del CNR, via Pietro Castellino, 111, 80131, Napoli, Italy
| | - Menotti Ruvo
- Istituto di Biostrutture e Bioimmagini del CNR and CIRPeB, Universita Federico II di Napoli, via Mezzocannone, 16, 80134 Napoli, Italy
| |
Collapse
|
19
|
Ibrahim RS, Mahrous RSR, Fathy HM, Omar AA, Abu EL-Khair RM. Anticoagulant activity screening of an in-house database of natural compounds for discovering novel selective factor Xa inhibitors; a combined in silico and in vitro approach. Med Chem Res 2020. [DOI: 10.1007/s00044-020-02516-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
20
|
A platform for target prediction of phenotypic screening hit molecules. J Mol Graph Model 2019; 95:107485. [PMID: 31836397 PMCID: PMC6983931 DOI: 10.1016/j.jmgm.2019.107485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 09/25/2019] [Accepted: 10/21/2019] [Indexed: 01/09/2023]
Abstract
Many drug discovery programmes, particularly for infectious diseases, are conducted phenotypically. Identifying the targets of phenotypic screening hits experimentally can be complex, time-consuming, and expensive. However, it would be valuable to know what the molecular target(s) is, as knowledge of the binding pose of the hit molecule in the binding site can facilitate the compound optimisation. Furthermore, knowing the target would allow de-prioritisation of less attractive chemical series or molecular targets. To generate target-hypotheses for phenotypic active compounds, an in silico platform was developed that utilises both ligand and protein-structure information to generate a ranked set of predicted molecular targets. As a result of the web-based workflow the user obtains a set of 3D structures of the predicted targets with the active molecule bound. The platform was exemplified using Mycobacterium tuberculosis, the causative organism of tuberculosis. In a test that we performed, the platform was able to predict the targets of 60% of compounds investigated, where there was some similarity to a ligand in the protein database. An algorithm to predict the molecular target(s) of phenotypic hits against TB. Uses information based on the ligand and protein structure. Allow visualisation of proposed binding pose. Web interface developed.
Collapse
|
21
|
Vázquez J, Deplano A, Herrero A, Ginex T, Gibert E, Rabal O, Oyarzabal J, Herrero E, Luque FJ. Development and Validation of Molecular Overlays Derived from Three-Dimensional Hydrophobic Similarity with PharmScreen. J Chem Inf Model 2018; 58:1596-1609. [PMID: 30010337 DOI: 10.1021/acs.jcim.8b00216] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Molecular alignment is a standard procedure for three-dimensional (3D) similarity measurements and pharmacophore elucidation. This process is influenced by several factors, such as the physicochemical descriptors utilized to account for the molecular determinants of biological activity and the reference templates. Relying on the hypothesis that the maximal achievable binding affinity for a drug-like molecule is largely due to desolvation, we explore a novel strategy for 3D molecular overlays that exploits the partitioning of molecular hydrophobicity into atomic contributions in conjunction with information about the distribution of hydrogen-bond (HB) donor/acceptor groups. A brief description of the method, as implemented in the software package PharmScreen, including the derivation of the fractional hydrophobic contributions within the quantum mechanical version of the Miertus-Scrocco-Tomasi (MST) continuum model, and the procedure utilized for the optimal superposition between molecules, is presented. The computational procedure is calibrated by using a data set of 402 molecules pertaining to 14 distinct targets taken from the literature and validated against the AstraZeneca test, which comprises 121 experimentally derived sets of molecular overlays. The results point out the suitability of the MST-based hydrophobic parameters for generating molecular overlays, as correct predictions were obtained for 94%, 79%, and 54% of the molecules classified into easy, moderate, and hard sets, respectively. Moreover, the results point out that this accuracy is attained at a much lower degree of identity between the templates used by hydrophobic/HB fields and electrostatic/steric ones. These findings support the usefulness of the hydrophobic/HB descriptors to generate complementary overlays that may be valuable to rationalize structure-activity relationships and for virtual screening campaigns.
Collapse
Affiliation(s)
- Javier Vázquez
- Pharmacelera , Plaça Pau Vila, 1, Sector C 2a , Edifici Palau de Mar, Barcelona 08039 , Spain.,Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), and Institute of Theoretical and Computational Chemistry (IQTC-UB) , University of Barcelona , Av. Prat de la Riba 171 , Santa Coloma de Gramenet E-08921 , Spain
| | - Alessandro Deplano
- Pharmacelera , Plaça Pau Vila, 1, Sector C 2a , Edifici Palau de Mar, Barcelona 08039 , Spain
| | - Albert Herrero
- Pharmacelera , Plaça Pau Vila, 1, Sector C 2a , Edifici Palau de Mar, Barcelona 08039 , Spain
| | - Tiziana Ginex
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), and Institute of Theoretical and Computational Chemistry (IQTC-UB) , University of Barcelona , Av. Prat de la Riba 171 , Santa Coloma de Gramenet E-08921 , Spain
| | - Enric Gibert
- Pharmacelera , Plaça Pau Vila, 1, Sector C 2a , Edifici Palau de Mar, Barcelona 08039 , Spain
| | - Obdulia Rabal
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA) , University of Navarra , Avda. Pio XII 55 , Pamplona E-31008 , Spain
| | - Julen Oyarzabal
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA) , University of Navarra , Avda. Pio XII 55 , Pamplona E-31008 , Spain
| | - Enric Herrero
- Pharmacelera , Plaça Pau Vila, 1, Sector C 2a , Edifici Palau de Mar, Barcelona 08039 , Spain
| | - F Javier Luque
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), and Institute of Theoretical and Computational Chemistry (IQTC-UB) , University of Barcelona , Av. Prat de la Riba 171 , Santa Coloma de Gramenet E-08921 , Spain
| |
Collapse
|
22
|
Poli G, Seidel T, Langer T. Conformational Sampling of Small Molecules With iCon: Performance Assessment in Comparison With OMEGA. Front Chem 2018; 6:229. [PMID: 29971231 PMCID: PMC6018197 DOI: 10.3389/fchem.2018.00229] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/31/2018] [Indexed: 11/21/2022] Open
Abstract
Herein we present the algorithm and performance assessment of our newly developed conformer generator iCon that was implemented in LigandScout 4.0. Two data sets of high-quality X-ray structures of drug-like small molecules originating from the Protein Data Bank (200 ligands) and the Cambridge Structural Database (481 molecules) were used to validate iCon's performance in the reproduction of experimental conformations. OpenEye's conformer generator OMEGA was subjected to the same evaluation and served as a reference software in this analysis. We tested several setting patterns in order to identify the most suitable and efficient ones for conformational sampling with iCon; equivalent settings were also tested on OMEGA in order to compare the results obtained from the two programs and better assess iCon's performance. Overall, this study proved that iCon is able to generate reliable representative conformational ensembles of drug-like small molecules, yielding results comparable to those showed by OMEGA, and thus is ready to serve as a valuable tool for computer-aided drug design.
Collapse
Affiliation(s)
- Giulio Poli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Thomas Seidel
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Thierry Langer
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| |
Collapse
|
23
|
Bocci G, Moreau A, Vayer P, Denizot C, Fardel O, Parmentier Y. New insights in the in vitro characterisation and molecular modelling of the P-glycoprotein inhibitory promiscuity. Eur J Pharm Sci 2018; 121:85-94. [PMID: 29709579 DOI: 10.1016/j.ejps.2018.04.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/18/2018] [Accepted: 04/26/2018] [Indexed: 12/28/2022]
Abstract
The presence of several binding sites for both substrates and inhibitors is yet a poorly explored thematic concerning the assessment of the drug-drug interactions risk due to interactions of multiple drugs with the human transport protein P-glycoprotein (P-gp or MDR1, gene ABCB1). In this study we measured the inhibitory behaviour of a set of known drugs towards P-gp by using three different probe substrates (digoxin, Hoechst 33,342 and rhodamine 123). A structure-based model was built to unravel the different substrates binding sites and to rationalize the cases where drugs were not inhibiting all the substrates. A separate set of experiments was used to validate the model and confirmed its suitability to either detect the substrate-dependent P-gp inhibition and to anticipate proper substrates for in vitro experiments case by case. The modelling strategy described can be applied for either design safer drugs (P-gp as antitarget) or to target specific sub-site inhibitors towards other drugs (P-gp as target).
Collapse
Affiliation(s)
- Giovanni Bocci
- Laboratory of Chemometrics, Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
| | - Amélie Moreau
- Technologie Servier, 25-27 rue Eugène Vignat, BP 11749, 45007 Orléans cedex 1, France
| | - Philippe Vayer
- Technologie Servier, 25-27 rue Eugène Vignat, BP 11749, 45007 Orléans cedex 1, France.
| | - Claire Denizot
- Technologie Servier, 25-27 rue Eugène Vignat, BP 11749, 45007 Orléans cedex 1, France
| | - Olivier Fardel
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 2 Avenue du Pr Léon Bernard, F-35043 Rennes, France
| | - Yannick Parmentier
- Technologie Servier, 25-27 rue Eugène Vignat, BP 11749, 45007 Orléans cedex 1, France
| |
Collapse
|
24
|
Abstract
The Warburg effect describes how most cancer cells exhibit higher-than-normal glucose consumption, not only under hypoxic conditions, but also when normal oxygen levels are present. Although glucose transporter 1 (GLUT1) has been found to play a key role in the cellular uptake of glucose, especially in cancer cells, where it is generally overexpressed, it has not been given consideration as a suitable target for the development of anticancer drugs. In this chapter, an example of molecular design and realization of novel GLUT1 inhibitors, including in silico modeling, chemical synthesis, and biological characterization, is provided. This process started with the identification of a focused series of oxime derivatives, originally designed as estrogen receptor (ER) ligands, which were structurally optimized in order to direct their activity towards GLUT1 and to minimize their binding to the ERs, leading to the production of efficient and selective inhibitors of glucose uptake in cancer cells.
Collapse
Affiliation(s)
- Carlotta Granchi
- Department of Pharmacy, University of Pisa, Via Bonanno 33, 56126, Pisa, Italy
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, Via Bonanno 33, 56126, Pisa, Italy
| | - Filippo Minutolo
- Department of Pharmacy, University of Pisa, Via Bonanno 33, 56126, Pisa, Italy.
| |
Collapse
|
25
|
Catto M, Trisciuzzi D, Alberga D, Mangiatordi GF, Nicolotti O. Multitarget Drug Design for Neurodegenerative Diseases. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2018. [DOI: 10.1007/7653_2018_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
26
|
Perrone MG, Vitale P, Ferorelli S, Boccarelli A, Coluccia M, Pannunzio A, Campanella F, Di Mauro G, Bonaccorso C, Fortuna CG, Scilimati A. Effect of mofezolac-galactose distance in conjugates targeting cyclooxygenase (COX)-1 and CNS GLUT-1 carrier. Eur J Med Chem 2017; 141:404-416. [DOI: 10.1016/j.ejmech.2017.09.066] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/25/2017] [Accepted: 09/29/2017] [Indexed: 01/04/2023]
|
27
|
Sirci F, Napolitano F, Pisonero-Vaquero S, Carrella D, Medina DL, di Bernardo D. Comparing structural and transcriptional drug networks reveals signatures of drug activity and toxicity in transcriptional responses. NPJ Syst Biol Appl 2017; 3:23. [PMID: 28861278 PMCID: PMC5572457 DOI: 10.1038/s41540-017-0022-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/27/2017] [Accepted: 07/07/2017] [Indexed: 02/07/2023] Open
Abstract
We performed an integrated analysis of drug chemical structures and drug-induced transcriptional responses. We demonstrated that a network representing three-dimensional structural similarities among 5452 compounds can be used to automatically group together drugs with similar scaffolds, physicochemical parameters and mode-of-action. We compared the structural network to a network representing transcriptional similarities among a subset of 1309 drugs for which transcriptional response were available in the Connectivity Map data set. Analysis of structurally similar, but transcriptionally different drugs sharing the same MOA enabled us to detect and remove weak and noisy transcriptional responses, greatly enhancing the reliability of transcription-based approaches to drug discovery and drug repositioning. Cardiac glycosides exhibited the strongest transcriptional responses with a significant induction of pathways related to epigenetic regulation, which suggests an epigenetic mechanism of action for these drugs. Drug classes with the weakest transcriptional responses tended to induce expression of cytochrome P450 enzymes, hinting at drug-induced drug resistance. Analysis of transcriptionally similar, but structurally different drugs with unrelated MOA, led us to the identification of a 'toxic' transcriptional signature indicative of lysosomal stress (lysosomotropism) and lipid accumulation (phospholipidosis) partially masking the target-specific transcriptional effects of these drugs. We found that this transcriptional signature is shared by 258 compounds and it is associated to the activation of the transcription factor TFEB, a master regulator of lysosomal biogenesis and autophagy. Finally, we built a predictive Random Forest model of these 258 compounds based on 128 physicochemical parameters, which should help in the early identification of potentially toxic drug candidates. Transcriptional responses to drug treatment can reveal mechanism of action and off-target effects thus enabling drug repositioning, but only if measured in the appropriate cells at clinically relevant concentrations. A team led by Diego di Bernardo and Diego Medina generated a network representing structural similarities among compounds to automatically group together drugs with similar scaffolds and MOA. By comparing the structural drug network with a transcriptional drug network based on similarities in transcriptional response, the team observed broad differences between the two. This observation led to the identification of a transcriptional signature related lysosomal stress and phospholipidosis, and a physicochemical model to identify such compounds. These results provide general guidelines to prevent erroneous conclusion when using transcriptional responses of small molecules for drug discovery and drug repositioning
Collapse
Affiliation(s)
- Francesco Sirci
- Telethon Institute of Genetics and Medicine (TIGEM), System Biology and Bioinformatics lab. and High Content Screening facility, Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy
| | - Francesco Napolitano
- Telethon Institute of Genetics and Medicine (TIGEM), System Biology and Bioinformatics lab. and High Content Screening facility, Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy
| | - Sandra Pisonero-Vaquero
- Telethon Institute of Genetics and Medicine (TIGEM), System Biology and Bioinformatics lab. and High Content Screening facility, Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy
| | - Diego Carrella
- Telethon Institute of Genetics and Medicine (TIGEM), System Biology and Bioinformatics lab. and High Content Screening facility, Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy
| | - Diego L Medina
- Telethon Institute of Genetics and Medicine (TIGEM), System Biology and Bioinformatics lab. and High Content Screening facility, Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy
| | - Diego di Bernardo
- Telethon Institute of Genetics and Medicine (TIGEM), System Biology and Bioinformatics lab. and High Content Screening facility, Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy.,Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Piazzale Tecchio 80, 80125 Naples, Italy
| |
Collapse
|
28
|
Dobričić V, Jaćević V, Vučićević J, Nikolic K, Vladimirov S, Čudina O. Evaluation of Biological Activity and Computer-Aided Design of New Soft Glucocorticoids. Arch Pharm (Weinheim) 2017; 350. [PMID: 28418199 DOI: 10.1002/ardp.201600383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 03/21/2017] [Accepted: 03/27/2017] [Indexed: 12/12/2022]
Abstract
Soft glucocorticoids are compounds that are biotransformed to inactive and non-toxic metabolites and have fewer side effects than traditional glucocorticoids. A new class of 17β-carboxamide steroids has been recently introduced by our group. In this study, local anti-inflammatory activity of these derivatives was evaluated by use of the croton oil-induced ear edema test. Glucocorticoids with the highest maximal edema inhibition (MEI) were pointed out, and the systemic side effects of those with the lowest EC50 values were significantly lower in comparison to dexamethasone. A 3D-QSAR model was created and employed for the design of 27 compounds. By use of the sequential combination of ligand-based and structure-based virtual screening, three compounds were selected from the ChEMBL library and used as a starting point for the design of 15 derivatives. Molecular docking analysis of the designed derivatives with the highest predicted MEI and relative glucocorticoid receptor binding affinity (20, 22, 24-1, 25-1, 27, VS7, VS13, and VS14) confirmed the presence of interactions with the glucocorticoid receptor that are important for the activity.
Collapse
Affiliation(s)
- Vladimir Dobričić
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Belgrade, Belgrade, Serbia
| | - Vesna Jaćević
- National Poison Control Centre, Department of Experimental Toxicology and Pharmacology, Military Medical Academy, Belgrade, Serbia.,Medical Faculty of the Military Medical Academy, University of Defence, Belgrade, Serbia.,Faculty of Science, Department of Chemistry, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jelica Vučićević
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Belgrade, Belgrade, Serbia
| | - Katarina Nikolic
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Belgrade, Belgrade, Serbia
| | - Sote Vladimirov
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Belgrade, Belgrade, Serbia
| | - Olivera Čudina
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
29
|
Structure-metabolism relationships in human-AOX: Chemical insights from a large database of aza-aromatic and amide compounds. Proc Natl Acad Sci U S A 2017; 114:E3178-E3187. [PMID: 28373537 DOI: 10.1073/pnas.1618881114] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Aldehyde oxidase (AOX) is a metabolic enzyme catalyzing the oxidation of aldehyde and aza-aromatic compounds and the hydrolysis of amides, moieties frequently shared by the majority of drugs. Despite its key role in human metabolism, to date only fragmentary information about the chemical features responsible for AOX susceptibility are reported and only "very local" structure-metabolism relationships based on a small number of similar compounds have been developed. This study reports a more comprehensive coverage of the chemical space of structures with a high risk of AOX phase I metabolism in humans. More than 270 compounds were studied to identify the site of metabolism and the metabolite(s). Both electronic [supported by density functional theory (DFT) calculations] and exposure effects were considered when rationalizing the structure-metabolism relationship.
Collapse
|
30
|
Chaudhari R, Tan Z, Huang B, Zhang S. Computational polypharmacology: a new paradigm for drug discovery. Expert Opin Drug Discov 2017; 12:279-291. [PMID: 28067061 PMCID: PMC7241838 DOI: 10.1080/17460441.2017.1280024] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Over the past couple of years, the cost of drug development has sharply increased along with the high rate of clinical trial failures. Such increase in expenses is partially due to the inability of the "one drug - one target" approach to predict drug side effects and toxicities. To tackle this issue, an alternative approach, known as polypharmacology, is being adopted to study small molecule interactions with multiple targets. Apart from developing more potent and effective drugs, this approach allows for studies of off-target activities and the facilitation of drug repositioning. Although exhaustive polypharmacology studies in-vitro or in-vivo are not practical, computational methods of predicting unknown targets or side effects are being developed. Areas covered: This article describes various computational approaches that have been developed to study polypharmacology profiles of small molecules. It also provides a brief description of the algorithms used in these state-of-the-art methods. Expert opinion: Recent success in computational prediction of multi-targeting drugs has established polypharmacology as a promising alternative approach to tackle some of the daunting complications in drug discovery. This will not only help discover more effective agents, but also present tremendous opportunities to study novel target pharmacology and facilitate drug repositioning efforts in the pharmaceutical industry.
Collapse
Affiliation(s)
- Rajan Chaudhari
- Integrated Molecular Discovery Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Zhi Tan
- Integrated Molecular Discovery Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030
| | - Beibei Huang
- Integrated Molecular Discovery Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Shuxing Zhang
- Integrated Molecular Discovery Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030
| |
Collapse
|
31
|
Buonerba F, Lepri S, Goracci L, Schindler BD, Seo SM, Kaatz GW, Cruciani G. Improved Potency of Indole-Based NorA Efflux Pump Inhibitors: From Serendipity toward Rational Design and Development. J Med Chem 2016; 60:517-523. [PMID: 27977195 DOI: 10.1021/acs.jmedchem.6b01281] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The NorA efflux pump is a potential drug target for reversal of resistance to selected antibacterial agents, and recently we described indole-based inhibitor candidates. Herein we report a second class of inhibitors derived from them but with significant differences in shape and size. In particular, compounds 13 and 14 are very potent inhibitors in that they demonstrated the lowest IC50 values (2 μM) ever observed among all indole-based compounds we have evaluated.
Collapse
Affiliation(s)
- Federica Buonerba
- Department of Chemistry, Biology and Biotechnology, University of Perugia , 06123 Perugia, Italy
| | - Susan Lepri
- Department of Chemistry, Biology and Biotechnology, University of Perugia , 06123 Perugia, Italy
| | - Laura Goracci
- Department of Chemistry, Biology and Biotechnology, University of Perugia , 06123 Perugia, Italy
| | - Bryan D Schindler
- The John D. Dingell Department of Veterans Affairs Medical Center , Detroit, Michigan 48201, United States
| | - Susan M Seo
- The John D. Dingell Department of Veterans Affairs Medical Center , Detroit, Michigan 48201, United States
| | - Glenn W Kaatz
- The John D. Dingell Department of Veterans Affairs Medical Center , Detroit, Michigan 48201, United States.,Department of Internal Medicine, Division of Infectious Diseases, Wayne State University School of Medicine , Detroit, Michigan 48201, United States
| | - Gabriele Cruciani
- Department of Chemistry, Biology and Biotechnology, University of Perugia , 06123 Perugia, Italy
| |
Collapse
|
32
|
Babajide Mustapha I, Saeed F. Bioactive Molecule Prediction Using Extreme Gradient Boosting. Molecules 2016; 21:molecules21080983. [PMID: 27483216 PMCID: PMC6273295 DOI: 10.3390/molecules21080983] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 07/19/2016] [Accepted: 07/22/2016] [Indexed: 01/29/2023] Open
Abstract
Following the explosive growth in chemical and biological data, the shift from traditional methods of drug discovery to computer-aided means has made data mining and machine learning methods integral parts of today's drug discovery process. In this paper, extreme gradient boosting (Xgboost), which is an ensemble of Classification and Regression Tree (CART) and a variant of the Gradient Boosting Machine, was investigated for the prediction of biological activity based on quantitative description of the compound's molecular structure. Seven datasets, well known in the literature were used in this paper and experimental results show that Xgboost can outperform machine learning algorithms like Random Forest (RF), Support Vector Machines (LSVM), Radial Basis Function Neural Network (RBFN) and Naïve Bayes (NB) for the prediction of biological activities. In addition to its ability to detect minority activity classes in highly imbalanced datasets, it showed remarkable performance on both high and low diversity datasets.
Collapse
Affiliation(s)
- Ismail Babajide Mustapha
- UTM Big Data Centre, Ibnu Sina Institute for Scientific and Industrial Research, Universiti Teknologi Malaysia, Skudai, Johor 81310, Malaysia.
| | - Faisal Saeed
- Information Systems Department, Faculty of Computing, Universiti Teknologi Malaysia, Skudai, Johor 81310, Malaysia.
| |
Collapse
|
33
|
Synthesis of the vitamin E amino acid esters with an enhanced anticancer activity and in silico screening for new antineoplastic drugs. Eur J Pharm Sci 2016; 88:59-69. [DOI: 10.1016/j.ejps.2016.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/05/2016] [Accepted: 04/05/2016] [Indexed: 12/18/2022]
|
34
|
Vucicevic J, Srdic-Rajic T, Pieroni M, Laurila JMM, Perovic V, Tassini S, Azzali E, Costantino G, Glisic S, Agbaba D, Scheinin M, Nikolic K, Radi M, Veljkovic N. A combined ligand- and structure-based approach for the identification of rilmenidine-derived compounds which synergize the antitumor effects of doxorubicin. Bioorg Med Chem 2016; 24:3174-83. [PMID: 27265687 DOI: 10.1016/j.bmc.2016.05.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 05/17/2016] [Accepted: 05/20/2016] [Indexed: 10/21/2022]
Abstract
The clonidine-like central antihypertensive agent rilmenidine, which has high affinity for I1-type imidazoline receptors (I1-IR) was recently found to have cytotoxic effects on cultured cancer cell lines. However, due to its pharmacological effects resulting also from α2-adrenoceptor activation, rilmenidine cannot be considered a suitable anticancer drug candidate. Here, we report the identification of novel rilmenidine-derived compounds with anticancer potential and devoid of α2-adrenoceptor effects by means of ligand- and structure-based drug design approaches. Starting from a large virtual library, eleven compounds were selected, synthesized and submitted to biological evaluation. The most active compound 5 exhibited a cytotoxic profile similar to that of rilmenidine, but without appreciable affinity to α2-adrenoceptors. In addition, compound 5 significantly enhanced the apoptotic response to doxorubicin, and may thus represent an important tool for the development of better adjuvant chemotherapeutic strategies for doxorubicin-insensitive cancers.
Collapse
Affiliation(s)
- Jelica Vucicevic
- Institute of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Tatjana Srdic-Rajic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Marco Pieroni
- P4T Group, Dipartimento di Farmacia, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124 Parma, Italy
| | - Jonne M M Laurila
- Department of Pharmacology, Drug Development and Therapeutics, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| | - Vladimir Perovic
- Center for Multidisciplinary Research, Institute of Nuclear Sciences Vinca, University of Belgrade, POB 522, Mihaila Petrovica Alasa 14, 11001 Belgrade, Serbia
| | - Sabrina Tassini
- P4T Group, Dipartimento di Farmacia, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124 Parma, Italy
| | - Elisa Azzali
- P4T Group, Dipartimento di Farmacia, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124 Parma, Italy
| | - Gabriele Costantino
- P4T Group, Dipartimento di Farmacia, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124 Parma, Italy
| | - Sanja Glisic
- Center for Multidisciplinary Research, Institute of Nuclear Sciences Vinca, University of Belgrade, POB 522, Mihaila Petrovica Alasa 14, 11001 Belgrade, Serbia
| | - Danica Agbaba
- Institute of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Mika Scheinin
- Department of Pharmacology, Drug Development and Therapeutics, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland; Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Katarina Nikolic
- Institute of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia.
| | - Marco Radi
- P4T Group, Dipartimento di Farmacia, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124 Parma, Italy.
| | - Nevena Veljkovic
- Center for Multidisciplinary Research, Institute of Nuclear Sciences Vinca, University of Belgrade, POB 522, Mihaila Petrovica Alasa 14, 11001 Belgrade, Serbia.
| |
Collapse
|
35
|
|
36
|
Volkamer A, Eid S, Turk S, Rippmann F, Fulle S. Identification and Visualization of Kinase-Specific Subpockets. J Chem Inf Model 2016; 56:335-46. [PMID: 26735903 DOI: 10.1021/acs.jcim.5b00627] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The identification and design of selective compounds is important for the reduction of unwanted side effects as well as for the development of tool compounds for target validation studies. This is, in particular, true for therapeutically important protein families that possess conserved folds and have numerous members such as kinases. To support the design of selective kinase inhibitors, we developed a novel approach that allows identification of specificity determining subpockets between closely related kinases solely based on their three-dimensional structures. To account for the intrinsic flexibility of the proteins, multiple X-ray structures of the target protein of interest as well as of unwanted off-target(s) are taken into account. The binding pockets of these protein structures are calculated and fused to a combined target and off-target pocket, respectively. Subsequently, shape differences between these two combined pockets are identified via fusion rules. The approach provides a user-friendly visualization of target-specific areas in a binding pocket which should be explored when designing selective compounds. Furthermore, the approach can be easily combined with in silico alanine mutation studies to identify selectivity determining residues. The potential impact of the approach is demonstrated in four retrospective experiments on closely related kinases, i.e., p38α vs Erk2, PAK1 vs PAK4, ITK vs AurA, and BRAF vs VEGFR2. Overall, the presented approach does not require any profiling data for training purposes, provides an intuitive visualization of a large number of protein structures at once, and could also be applied to other target classes.
Collapse
Affiliation(s)
- Andrea Volkamer
- BioMed X Innovation Center , Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Sameh Eid
- BioMed X Innovation Center , Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Samo Turk
- BioMed X Innovation Center , Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Friedrich Rippmann
- Global Computational Chemistry, Merck KGaA , Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Simone Fulle
- BioMed X Innovation Center , Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| |
Collapse
|
37
|
Shin WH, Bures MG, Kihara D. PatchSurfers: Two methods for local molecular property-based binding ligand prediction. Methods 2016; 93:41-50. [PMID: 26427548 PMCID: PMC4718779 DOI: 10.1016/j.ymeth.2015.09.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 09/27/2015] [Accepted: 09/28/2015] [Indexed: 01/09/2023] Open
Abstract
Protein function prediction is an active area of research in computational biology. Function prediction can help biologists make hypotheses for characterization of genes and help interpret biological assays, and thus is a productive area for collaboration between experimental and computational biologists. Among various function prediction methods, predicting binding ligand molecules for a target protein is an important class because ligand binding events for a protein are usually closely intertwined with the proteins' biological function, and also because predicted binding ligands can often be directly tested by biochemical assays. Binding ligand prediction methods can be classified into two types: those which are based on protein-protein (or pocket-pocket) comparison, and those that compare a target pocket directly to ligands. Recently, our group proposed two computational binding ligand prediction methods, Patch-Surfer, which is a pocket-pocket comparison method, and PL-PatchSurfer, which compares a pocket to ligand molecules. The two programs apply surface patch-based descriptions to calculate similarity or complementarity between molecules. A surface patch is characterized by physicochemical properties such as shape, hydrophobicity, and electrostatic potentials. These properties on the surface are represented using three-dimensional Zernike descriptors (3DZD), which are based on a series expansion of a 3 dimensional function. Utilizing 3DZD for describing the physicochemical properties has two main advantages: (1) rotational invariance and (2) fast comparison. Here, we introduce Patch-Surfer and PL-PatchSurfer with an emphasis on PL-PatchSurfer, which is more recently developed. Illustrative examples of PL-PatchSurfer performance on binding ligand prediction as well as virtual drug screening are also provided.
Collapse
Affiliation(s)
- Woong-Hee Shin
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Mark Gregory Bures
- Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Daisuke Kihara
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
38
|
Navakauskienė R, Mori M, Christodoulou MS, Zentelytė A, Botta B, Via LD, Ricci F, Damia G, Passarella D, Zilio C, Martinet N. Histone demethylating agents as potential S-adenosyl-l-methionine-competitors. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00170j] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Histone H3 methylation on K9 and/or K27 depends on histone lysine methyltransferases (KMTs).
Collapse
Affiliation(s)
- R. Navakauskienė
- Department of Molecular Cell Biology
- Institute of Biochemistry
- Vilnius University
- LT-08662 Vilnius
- Lithuania
| | - M. Mori
- Center for Life Nano Science@Sapienza
- Istituto Italiano di Tecnologia
- 00161 Rome
- Italy
| | | | - A. Zentelytė
- Department of Molecular Cell Biology
- Institute of Biochemistry
- Vilnius University
- LT-08662 Vilnius
- Lithuania
| | - B. Botta
- Dipartimento di Chimica e Tecnologie del Farmaco
- Università degli Studi di Roma “La Sapienza”
- 00185 Roma
- Italy
| | - L. Dalla Via
- Dipartimento di Scienze del Farmaco
- Università degli Studi di Padova
- 35131 Padova
- Italy
| | - F. Ricci
- Mario Negri Institute for Pharmacological Research
- 20156 Milano
- Italy
| | - G. Damia
- Mario Negri Institute for Pharmacological Research
- 20156 Milano
- Italy
| | - D. Passarella
- Dipartimento di Chimica
- Università degli Studi di Milano
- 20133 Milano
- Italy
| | - C. Zilio
- Institute of Chemistry
- UMR CNRS 7272
- Nice 06108
- France
| | - N. Martinet
- Institute of Chemistry
- UMR CNRS 7272
- Nice 06108
- France
| |
Collapse
|
39
|
Al-Dabbagh MM, Salim N, Himmat M, Ahmed A, Saeed F. A Quantum-Based Similarity Method in Virtual Screening. Molecules 2015; 20:18107-27. [PMID: 26445039 DOI: 10.3390/molecules201018107] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 11/16/2022] Open
Abstract
One of the most widely-used techniques for ligand-based virtual screening is similarity searching. This study adopted the concepts of quantum mechanics to present as state-of-the-art similarity method of molecules inspired from quantum theory. The representation of molecular compounds in mathematical quantum space plays a vital role in the development of quantum-based similarity approach. One of the key concepts of quantum theory is the use of complex numbers. Hence, this study proposed three various techniques to embed and to re-represent the molecular compounds to correspond with complex numbers format. The quantum-based similarity method that developed in this study depending on complex pure Hilbert space of molecules called Standard Quantum-Based (SQB). The recall of retrieved active molecules were at top 1% and top 5%, and significant test is used to evaluate our proposed methods. The MDL drug data report (MDDR), maximum unbiased validation (MUV) and Directory of Useful Decoys (DUD) data sets were used for experiments and were represented by 2D fingerprints. Simulated virtual screening experiment show that the effectiveness of SQB method was significantly increased due to the role of representational power of molecular compounds in complex numbers forms compared to Tanimoto benchmark similarity measure.
Collapse
Affiliation(s)
| | - Naomie Salim
- Faculty of Computing, Universiti Teknologi Malaysia, Skudia 81310, Malaysia.
| | - Mubarak Himmat
- Faculty of Computing, Universiti Teknologi Malaysia, Skudia 81310, Malaysia.
| | - Ali Ahmed
- Faculty of Computing, Universiti Teknologi Malaysia, Skudia 81310, Malaysia.
- Faculty of Engineering, Karary University, Khartoum 12304, Sudan.
| | - Faisal Saeed
- Faculty of Computing, Universiti Teknologi Malaysia, Skudia 81310, Malaysia.
| |
Collapse
|
40
|
Spyrakis F, Benedetti P, Decherchi S, Rocchia W, Cavalli A, Alcaro S, Ortuso F, Baroni M, Cruciani G. A Pipeline To Enhance Ligand Virtual Screening: Integrating Molecular Dynamics and Fingerprints for Ligand and Proteins. J Chem Inf Model 2015; 55:2256-74. [DOI: 10.1021/acs.jcim.5b00169] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Francesca Spyrakis
- Department of Life
Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Paolo Benedetti
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
| | - Sergio Decherchi
- CONCEPT Lab, Italian Institute of Technology, via Morego 30, 16163 Genova, Italy
- BiKi Technologies s.r.l., via XX Settembre 33, 16121 Genova, Italy
| | - Walter Rocchia
- CONCEPT Lab, Italian Institute of Technology, via Morego 30, 16163 Genova, Italy
| | - Andrea Cavalli
- CompuNet, Italian Institute of Technology, via Morego 30, 16163 Genova, Italy
- Department of Pharmacy
and Biotechnology, University of Bologna, via Belmeloro 6, 40126 Bologna, Italy
| | - Stefano Alcaro
- Department of Health Science, University Magna Graecia of Catanzaro, Campus “S Venuta”, Viale Europa 88100, Catanzaro, Italy
| | - Francesco Ortuso
- Department of Health Science, University Magna Graecia of Catanzaro, Campus “S Venuta”, Viale Europa 88100, Catanzaro, Italy
| | - Massimo Baroni
- Molecular Discovery Limited, 215
Marsh Road, Pinner Middlesex, London HA5-5NE, United Kingdom
| | - Gabriele Cruciani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
| |
Collapse
|
41
|
Carosati E, van den Höfel N, Reif M, Randazzo GM, Stanitzki B, Stevens J, Gabbert HE, Cruciani G, Mannhold R, Mahotka C. Discovery of Novel, Potent, and Specific Cell-Death Inducers in the Jurkat Acute Lymphoblastic Leukemia Cell Line. ChemMedChem 2015; 10:1700-6. [PMID: 26267799 DOI: 10.1002/cmdc.201500245] [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: 06/03/2015] [Indexed: 11/06/2022]
Abstract
The limited clinical efficacy of many cancer therapeutics has initiated intense research efforts toward the discovery of novel chemical entities in this field. In this study, 31 hit candidates were selected from nearly 800,000 database compounds in a ligand-based virtual screening campaign. In turn, three of these hits were found to have (sub)micromolar potencies in proliferation assays with the Jurkat acute lymphatic leukemic cell line. In this assay, the three hits were found to exhibit higher potency than clinically tested cell-death inducers (GDC-0152, AT-406, and birinapant). Importantly, antiproliferative activity toward non-cancer peripheral blood mononuclear cells (PBMCs) was found to be marginal. Further biological characterization demonstrated the cell-death-inducing properties of these compounds. Biological testing of hit congeners excluded a nonspecific, toxic effect of the novel structures. Altogether, these findings may have profound relevance for the development of clinical candidates in tumor therapy.
Collapse
Affiliation(s)
- Emanuele Carosati
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Via Elce di Sotto 10, 06123 Perugia (Italy).
| | - Natascha van den Höfel
- Department of Pathology, Medical Faculty, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf (Germany)
| | - Manuela Reif
- Department of Pathology, Medical Faculty, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf (Germany)
| | - Giuseppe Marco Randazzo
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Via Elce di Sotto 10, 06123 Perugia (Italy)
| | - Bettina Stanitzki
- Department of Pathology, Medical Faculty, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf (Germany)
| | - Julia Stevens
- Department of Pathology, Medical Faculty, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf (Germany)
| | - Helmut E Gabbert
- Department of Pathology, Medical Faculty, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf (Germany)
| | - Gabriele Cruciani
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Via Elce di Sotto 10, 06123 Perugia (Italy)
| | - Raimund Mannhold
- Molecular Drug Research Group, Medical Faculty, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf (Germany)
| | - Csaba Mahotka
- Department of Pathology, Medical Faculty, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf (Germany).
| |
Collapse
|
42
|
Shin WH, Zhu X, Bures MG, Kihara D. Three-dimensional compound comparison methods and their application in drug discovery. Molecules 2015; 20:12841-62. [PMID: 26193243 PMCID: PMC5005041 DOI: 10.3390/molecules200712841] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 07/07/2015] [Accepted: 07/13/2015] [Indexed: 11/16/2022] Open
Abstract
Virtual screening has been widely used in the drug discovery process. Ligand-based virtual screening (LBVS) methods compare a library of compounds with a known active ligand. Two notable advantages of LBVS methods are that they do not require structural information of a target receptor and that they are faster than structure-based methods. LBVS methods can be classified based on the complexity of ligand structure information utilized: one-dimensional (1D), two-dimensional (2D), and three-dimensional (3D). Unlike 1D and 2D methods, 3D methods can have enhanced performance since they treat the conformational flexibility of compounds. In this paper, a number of 3D methods will be reviewed. In addition, four representative 3D methods were benchmarked to understand their performance in virtual screening. Specifically, we tested overall performance in key aspects including the ability to find dissimilar active compounds, and computational speed.
Collapse
Affiliation(s)
- Woong-Hee Shin
- Department of Biological Science, Purdue University, West Lafayette, IN 47907, USA.
| | - Xiaolei Zhu
- School of Life Science, Anhui University, Hefei 230601, China.
| | - Mark Gregory Bures
- Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, IN 46285, USA.
| | - Daisuke Kihara
- Department of Biological Science, Purdue University, West Lafayette, IN 47907, USA.
- Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
43
|
Kaalia R, Kumar A, Srinivasan A, Ghosh I. An Ab Initio Method for Designing Multi-Target Specific Pharmacophores using Complementary Interaction Field of Aspartic Proteases. Mol Inform 2015; 34:380-93. [PMID: 27490384 DOI: 10.1002/minf.201400157] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 02/09/2014] [Indexed: 11/09/2022]
Abstract
For past few decades, key objectives of rational drug discovery have been the designing of specific and selective ligands for target proteins. Infectious diseases like malaria are continuously becoming resistant to traditional medicines, which inculcates need for new approaches to design inhibitors for antimalarial targets. A novel method for ab initio designing of multi target specific pharmacophores using the interaction field maps of active sites of multiple proteins has been developed to design 'specificity' pharmacophores for aspartic proteases. The molecular interaction field grid maps of active sites of aspartic proteases (plasmepsin II & IV from Plasmodium falciparum, plasmepsin from Plasmodium vivax, pepsin & cathepsin D from human) are calculated and common pharmacophoric features for favourable binding spots in active sites are extracted in the form of cliques of graphs using inductive logic programming (ILP). The two pharmacophore ensembles are constructed from largest common cliques by imposing size of receptor active site (L) and domain-specific receptor-ligand information (S). The overlap of chemical space between two ensembles and the results of virtual screening of inhibitor database with known activities show that this method can design efficient pharmacophores with no prior ligand information.
Collapse
Affiliation(s)
- Rama Kaalia
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi-110067, India phone/fax:9971287771
| | - Amit Kumar
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi-110067, India phone/fax:9971287771
| | - Ashwin Srinivasan
- Indraprastha Institute of Information Technology, New Delhi-110020, India.,Current address: Department of Computer Science, BITS-Pilani, Goa-403726, India
| | - Indira Ghosh
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi-110067, India phone/fax:9971287771.
| |
Collapse
|
44
|
Siragusa L, Cross S, Baroni M, Goracci L, Cruciani G. BioGPS: Navigating biological space to predict polypharmacology, off-targeting, and selectivity. Proteins 2015; 83:517-32. [DOI: 10.1002/prot.24753] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 12/09/2014] [Accepted: 12/13/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Lydia Siragusa
- Laboratory for Chemometrics and Molecular Modeling, Department of Chemistry, Biology and Biotechnology; University of Perugia; Perugia 06123 Italy
| | - Simon Cross
- Molecular Discovery Limited; Pinner, Middlesex, London HA5 5NE United Kingdom
| | - Massimo Baroni
- Molecular Discovery Limited; Pinner, Middlesex, London HA5 5NE United Kingdom
| | - Laura Goracci
- Laboratory for Chemometrics and Molecular Modeling, Department of Chemistry, Biology and Biotechnology; University of Perugia; Perugia 06123 Italy
| | - Gabriele Cruciani
- Laboratory for Chemometrics and Molecular Modeling, Department of Chemistry, Biology and Biotechnology; University of Perugia; Perugia 06123 Italy
| |
Collapse
|
45
|
Fortuna CG, Berardozzi R, Bonaccorso C, Caltabiano G, Di Bari L, Goracci L, Guarcello A, Pace A, Palumbo Piccionello A, Pescitelli G, Pierro P, Lonati E, Bulbarelli A, Cocuzza CE, Musumarra G, Musumeci R. New potent antibacterials against Gram-positive multiresistant pathogens: Effects of side chain modification and chirality in linezolid-like 1,2,4-oxadiazoles. Bioorg Med Chem 2014; 22:6814-25. [DOI: 10.1016/j.bmc.2014.10.037] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/20/2014] [Accepted: 10/24/2014] [Indexed: 01/25/2023]
|
46
|
Spinelli D, Budriesi R, Cosimelli B, Severi E, Micucci M, Baroni M, Fusi F, Ioan P, Cross S, Frosini M, Saponara S, Matucci R, Rosano C, Viale M, Chiarini A, Carosati E. Playing with opening and closing of heterocycles: using the cusmano-ruccia reaction to develop a novel class of oxadiazolothiazinones, active as calcium channel modulators and P-glycoprotein inhibitors. Molecules 2014; 19:16543-72. [PMID: 25317581 PMCID: PMC6271282 DOI: 10.3390/molecules191016543] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 08/15/2014] [Accepted: 09/04/2014] [Indexed: 11/23/2022] Open
Abstract
As a result of the ring-into-ring conversion of nitrosoimidazole derivatives, we obtained a molecular scaffold that, when properly decorated, is able to decrease inotropy by blocking L-type calcium channels. Previously, we used this scaffold to develop a quantitative structure-activity relationship (QSAR) model, and we used the most potent oxadiazolothiazinone as a template for ligand-based virtual screening. Here, we enlarge the diversity of chemical decorations, present the synthesis and in vitro data for 11 new derivatives, and develop a new 3D-QSAR model with recent in silico techniques. We observed a key role played by the oxadiazolone moiety: given the presence of positively charged calcium ions in the transmembrane channel protein, we hypothesize the formation of a ternary complex between the oxadiazolothiazinone, the Ca2+ ion and the protein. We have supported this hypothesis by means of pharmacophore generation and through the docking of the pharmacophore into a homology model of the protein. We also studied with docking experiments the interaction with a homology model of P-glycoprotein, which is inhibited by this series of molecules, and provided further evidence toward the relevance of this scaffold in biological interactions.
Collapse
Affiliation(s)
- Domenico Spinelli
- Dipartimento di Chimica "G. Ciamician", Alma Mater Studiorum-Università di Bologna, Via F. Selmi 2, Bologna 40126, Italy.
| | - Roberta Budriesi
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum-Università di Bologna, Via Belmeloro 6, Bologna 40126, Italy.
| | - Barbara Cosimelli
- Dipartimento di Farmacia, Università di Napoli "Federico II", Via D. Montesano 49, Napoli 80131, Italy.
| | - Elda Severi
- Dipartimento di Farmacia, Università di Napoli "Federico II", Via D. Montesano 49, Napoli 80131, Italy.
| | - Matteo Micucci
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum-Università di Bologna, Via Belmeloro 6, Bologna 40126, Italy.
| | - Massimo Baroni
- Molecular Discovery Ltd., 215 Marsh Road, Pinner, Middlesex HA5 5NE, UK.
| | - Fabio Fusi
- Dipartimento di Scienze della Vita, Università degli Studi di Siena, Via A. Moro 2, Siena 53100, Italy.
| | - Pierfranco Ioan
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum-Università di Bologna, Via Belmeloro 6, Bologna 40126, Italy.
| | - Simon Cross
- Molecular Discovery Ltd., 215 Marsh Road, Pinner, Middlesex HA5 5NE, UK.
| | - Maria Frosini
- Dipartimento di Scienze della Vita, Università degli Studi di Siena, Via A. Moro 2, Siena 53100, Italy.
| | - Simona Saponara
- Dipartimento di Scienze della Vita, Università degli Studi di Siena, Via A. Moro 2, Siena 53100, Italy.
| | - Rosanna Matucci
- Dipartimento di Neuroscienze, Area del Farmaco e Salute del Bambino (NEUROFARBA) Viale Pieraccini 6, Firenze 50139, Italy.
| | - Camillo Rosano
- IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, U.O.S. Biopolimeri e Proteomica, L.go R. Benzi, 10, Genova 16132, Italy.
| | - Maurizio Viale
- IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, U.O.C. Bioterapie, L.go R. Benzi, 10, Genova 16132, Italy.
| | - Alberto Chiarini
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum-Università di Bologna, Via Belmeloro 6, Bologna 40126, Italy.
| | - Emanuele Carosati
- Dipartimento di Chimica, Biologia e Biotecnologie, Università di Perugia, Via Elce di Sotto 10, Perugia 06123, Italy.
| |
Collapse
|
47
|
Siragusa L, Spyrakis F, Goracci L, Cross S, Cruciani G. BioGPS: The Music for the Chemo- and Bioinformatics Walzer. Mol Inform 2014; 33:446-53. [DOI: 10.1002/minf.201400028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/19/2014] [Indexed: 01/09/2023]
|
48
|
Wang Q, Park J, Devkota AK, Cho EJ, Dalby KN, Ren P. Identification and validation of novel PERK inhibitors. J Chem Inf Model 2014; 54:1467-75. [PMID: 24745945 PMCID: PMC4038368 DOI: 10.1021/ci500114r] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Indexed: 01/09/2023]
Abstract
PERK, as one of the principle unfolded protein response signal transducers, is believed to be associated with many human diseases, such as cancer and type-II diabetes. There has been increasing effort to discover potent PERK inhibitors due to its potential therapeutic interest. In this study, a computer-based virtual screening approach is employed to discover novel PERK inhibitors, followed by experimental validation. Using a focused library, we show that a consensus approach, combining pharmacophore modeling and docking, can be more cost-effective than using either approach alone. It is also demonstrated that the conformational flexibility near the active site is an important consideration in structure-based docking and can be addressed by using molecular dynamics. The consensus approach has further been applied to screen the ZINC lead-like database, resulting in the identification of 10 active compounds, two of which show IC50 values that are less than 10 μM in a dose-response assay.
Collapse
Affiliation(s)
- Qiantao Wang
- Division
of Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
- Department
of Biomedical Engineering, The University
of Texas at Austin, Austin, Texas 78712, United States
| | - Jihyun Park
- Division
of Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Ashwini K. Devkota
- Texas
Screening Alliance for Cancer Therapeutics, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Eun Jeong Cho
- Texas
Screening Alliance for Cancer Therapeutics, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Kevin N. Dalby
- Division
of Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
- Texas
Screening Alliance for Cancer Therapeutics, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Pengyu Ren
- Department
of Biomedical Engineering, The University
of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
49
|
Ahmed A, Saeed F, Salim N, Abdo A. Condorcet and borda count fusion method for ligand-based virtual screening. J Cheminform 2014; 6:19. [PMID: 24883114 PMCID: PMC4026830 DOI: 10.1186/1758-2946-6-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 04/23/2014] [Indexed: 11/14/2022] Open
Abstract
Background It is known that any individual similarity measure will not always give the best recall of active molecule structure for all types of activity classes. Recently, the effectiveness of ligand-based virtual screening approaches can be enhanced by using data fusion. Data fusion can be implemented using two different approaches: group fusion and similarity fusion. Similarity fusion involves searching using multiple similarity measures. The similarity scores, or ranking, for each similarity measure are combined to obtain the final ranking of the compounds in the database. Results The Condorcet fusion method was examined. This approach combines the outputs of similarity searches from eleven association and distance similarity coefficients, and then the winner measure for each class of molecules, based on Condorcet fusion, was chosen to be the best method of searching. The recall of retrieved active molecules at top 5% and significant test are used to evaluate our proposed method. The MDL drug data report (MDDR), maximum unbiased validation (MUV) and Directory of Useful Decoys (DUD) data sets were used for experiments and were represented by 2D fingerprints. Conclusions Simulated virtual screening experiments with the standard two data sets show that the use of Condorcet fusion provides a very simple way of improving the ligand-based virtual screening, especially when the active molecules being sought have a lowest degree of structural heterogeneity. However, the effectiveness of the Condorcet fusion was increased slightly when structural sets of high diversity activities were being sought.
Collapse
Affiliation(s)
- Ali Ahmed
- Soft Computing Research Group, Faculty of Computing, Universiti Teknologi Malaysia, Skudai 81310, Malaysia ; Faculty of Engineering, Karary University, Khartoum 12304, Sudan
| | - Faisal Saeed
- Soft Computing Research Group, Faculty of Computing, Universiti Teknologi Malaysia, Skudai 81310, Malaysia
| | - Naomie Salim
- Soft Computing Research Group, Faculty of Computing, Universiti Teknologi Malaysia, Skudai 81310, Malaysia
| | - Ammar Abdo
- Computer Science Department, Hodeidah University, Hodeidah, Yemen
| |
Collapse
|
50
|
Spyrakis F, Singh R, Cozzini P, Campanini B, Salsi E, Felici P, Raboni S, Benedetti P, Cruciani G, Kellogg GE, Cook PF, Mozzarelli A. Isozyme-specific ligands for O-acetylserine sulfhydrylase, a novel antibiotic target. PLoS One 2013; 8:e77558. [PMID: 24167577 PMCID: PMC3805590 DOI: 10.1371/journal.pone.0077558] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 09/03/2013] [Indexed: 01/06/2023] Open
Abstract
The last step of cysteine biosynthesis in bacteria and plants is catalyzed by O-acetylserine sulfhydrylase. In bacteria, two isozymes, O-acetylserine sulfhydrylase-A and O-acetylserine sulfhydrylase-B, have been identified that share similar binding sites, although the respective specific functions are still debated. O-acetylserine sulfhydrylase plays a key role in the adaptation of bacteria to the host environment, in the defense mechanisms to oxidative stress and in antibiotic resistance. Because mammals synthesize cysteine from methionine and lack O-acetylserine sulfhydrylase, the enzyme is a potential target for antimicrobials. With this aim, we first identified potential inhibitors of the two isozymes via a ligand- and structure-based in silico screening of a subset of the ZINC library using FLAP. The binding affinities of the most promising candidates were measured in vitro on purified O-acetylserine sulfhydrylase-A and O-acetylserine sulfhydrylase-B from Salmonella typhimurium by a direct method that exploits the change in the cofactor fluorescence. Two molecules were identified with dissociation constants of 3.7 and 33 µM for O-acetylserine sulfhydrylase-A and O-acetylserine sulfhydrylase-B, respectively. Because GRID analysis of the two isoenzymes indicates the presence of a few common pharmacophoric features, cross binding titrations were carried out. It was found that the best binder for O-acetylserine sulfhydrylase-B exhibits a dissociation constant of 29 µM for O-acetylserine sulfhydrylase-A, thus displaying a limited selectivity, whereas the best binder for O-acetylserine sulfhydrylase-A exhibits a dissociation constant of 50 µM for O-acetylserine sulfhydrylase-B and is thus 8-fold selective towards the former isozyme. Therefore, isoform-specific and isoform-independent ligands allow to either selectively target the isozyme that predominantly supports bacteria during infection and long-term survival or to completely block bacterial cysteine biosynthesis.
Collapse
Affiliation(s)
| | - Ratna Singh
- Department of Pharmacy, University of Parma, Parma, Italy
| | - Pietro Cozzini
- Department of Food Sciences, University of Parma, Parma, Italy
- National Institute of Biostructures and Biosystems, Rome, Italy
| | - Barbara Campanini
- Department of Pharmacy, University of Parma, Parma, Italy
- * E-mail: (BC); (AM)
| | - Enea Salsi
- Department of Pharmacy, University of Parma, Parma, Italy
| | - Paolo Felici
- Department of Pharmacy, University of Parma, Parma, Italy
| | - Samanta Raboni
- Department of Pharmacy, University of Parma, Parma, Italy
| | | | | | - Glen E. Kellogg
- Department of Medicinal Chemistry and Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Paul F. Cook
- Department of Biochemistry, University of Oklahoma, Norman, Oklahoma, United States of America
| | - Andrea Mozzarelli
- Department of Pharmacy, University of Parma, Parma, Italy
- National Institute of Biostructures and Biosystems, Rome, Italy
- * E-mail: (BC); (AM)
| |
Collapse
|