1
|
Duan SF, Song L, Guo HY, Deng H, Huang X, Shen QK, Quan ZS, Yin XM. Research status of indole-modified natural products. RSC Med Chem 2023; 14:2535-2563. [PMID: 38107170 PMCID: PMC10718587 DOI: 10.1039/d3md00560g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 10/16/2023] [Indexed: 12/19/2023] Open
Abstract
Indole is a heterocyclic compound formed by the fusion of a benzene ring and pyrrole ring, which has rich biological activity. Many indole-containing compounds have been sold on the market due to their excellent pharmacological activity. For example, vincristine and reserpine have been widely used in clinical practice. The diverse structures and biological activities of natural products provide abundant resources for the development of new drugs. Therefore, this review classifies natural products by structure, and summarizes the research progress of indole-containing natural product derivatives, their biological activities, structure-activity relationship and research mechanism which has been studied in the past 13 years, so as to provide a basis for the development of new drug development.
Collapse
Affiliation(s)
- Song-Fang Duan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Lei Song
- Yanbian University Hospital, Yanbian University Yanji 133002 People's Republic of China
| | - Hong-Yan Guo
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Hao Deng
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Xing Huang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Qing-Kun Shen
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Zhe-Shan Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Xiu-Mei Yin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| |
Collapse
|
2
|
Arora R, Mirabi B, Durant AG, Bozal-Ginesta C, Marchese AD, Aspuru-Guzik A, Lautens M. Palladium-Catalyzed Synthesis of Linked Bis-Heterocycles─Synthesis and Investigation of Photophysical Properties. J Am Chem Soc 2023. [PMID: 38039391 DOI: 10.1021/jacs.3c07234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2023]
Abstract
A palladium-catalyzed domino C-N coupling/Cacchi reaction is reported. Design of photoluminescent bis-heterocycles, aided by density functional theory calculations, was performed with synthetic yields up to 98%. The photophysical properties of the products accessed via this strategy were part of a comprehensive study that led to broad emission spectra and quantum yields of up to 0.59. Mechanistic experiments confirmed bromoalkynes as competent intermediates, and a density functional theory investigation suggests a pathway involving initial oxidative addition into the cis C-Br bond of the gem-dihaloolefin.
Collapse
Affiliation(s)
- Ramon Arora
- Davenport Research Laboratories, Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Bijan Mirabi
- Davenport Research Laboratories, Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Andrew G Durant
- Davenport Research Laboratories, Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Carlota Bozal-Ginesta
- Davenport Research Laboratories, Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Austin D Marchese
- Davenport Research Laboratories, Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Alán Aspuru-Guzik
- Davenport Research Laboratories, Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Mark Lautens
- Davenport Research Laboratories, Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| |
Collapse
|
3
|
Guidetti L, Zappia A, Scalvini L, Ferrari FR, Giorgio C, Castelli R, Galvani F, Vacondio F, Rivara S, Mor M, Urbinati C, Rusnati M, Tognolini M, Lodola A. Molecular Determinants of EphA2 and EphB2 Antagonism Enable the Design of Ligands with Improved Selectivity. J Chem Inf Model 2023; 63:6900-6911. [PMID: 37910792 PMCID: PMC10647059 DOI: 10.1021/acs.jcim.3c01064] [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: 07/13/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 11/03/2023]
Abstract
With the aim of identifying novel antagonists selective for the EphA receptor family, a combined experimental and computational approach was taken to investigate the molecular basis of the recognition between a prototypical Eph-ephrin antagonist (UniPR1447) and two representative receptors of the EphA and EphB subfamilies, namely, EphA2 and EphB2 receptors. The conformational free-energy surface (FES) of the binding state of UniPR1447 within the ligand binding domain of EphA2 and EphB2, reconstructed from molecular dynamics (MD) simulations performed on the microsecond time scale, was exploited to drive the design and synthesis of a novel antagonist selective for EphA2 over the EphB2 receptor. The availability of compounds with this pharmacological profile will help discriminate the importance of these two receptors in the insurgence and progression of cancer.
Collapse
Affiliation(s)
- Lorenzo Guidetti
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I- 43124 Parma, Italy
| | - Alfonso Zappia
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I- 43124 Parma, Italy
| | - Laura Scalvini
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I- 43124 Parma, Italy
| | - Francesca Romana Ferrari
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I- 43124 Parma, Italy
| | - Carmine Giorgio
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I- 43124 Parma, Italy
| | - Riccardo Castelli
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I- 43124 Parma, Italy
| | - Francesca Galvani
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I- 43124 Parma, Italy
| | - Federica Vacondio
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I- 43124 Parma, Italy
| | - Silvia Rivara
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I- 43124 Parma, Italy
| | - Marco Mor
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I- 43124 Parma, Italy
- Microbiome
Research Hub, Università degli Studi
di Parma, Parco Area
delle scienze 11/A, I- 43124 Parma, Italy
| | - Chiara Urbinati
- Dipartimento
di Medicina Molecolare Traslazionale, Università
degli Studi di Brescia, Brescia 25121, Italy
| | - Marco Rusnati
- Dipartimento
di Medicina Molecolare Traslazionale, Università
degli Studi di Brescia, Brescia 25121, Italy
| | - Massimiliano Tognolini
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I- 43124 Parma, Italy
| | - Alessio Lodola
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I- 43124 Parma, Italy
| |
Collapse
|
4
|
Galvani F, Pala D, Cuzzolin A, Scalvini L, Lodola A, Mor M, Rizzi A. Unbinding Kinetics of Muscarinic M3 Receptor Antagonists Explained by Metadynamics Simulations. J Chem Inf Model 2023; 63:2842-2856. [PMID: 37053454 PMCID: PMC10170513 DOI: 10.1021/acs.jcim.3c00042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Indexed: 04/15/2023]
Abstract
The residence time (RT), the time for which a drug remains bound to its biological target, is a critical parameter for drug design. The prediction of this key kinetic property has been proven to be challenging and computationally demanding in the framework of atomistic simulations. In the present work, we setup and applied two distinct metadynamics protocols to estimate the RTs of muscarinic M3 receptor antagonists. In the first method, derived from the conformational flooding approach, the kinetics of unbinding is retrieved from a physics-based parameter known as the acceleration factor α (i.e., the running average over time of the potential deposited in the bound state). Such an approach is expected to recover the absolute RT value for a compound of interest. In the second method, known as the tMETA-D approach, a qualitative estimation of the RT is given by the time of simulation required to drive the ligand from the binding site to the solvent bulk. This approach has been developed to reproduce the change of experimental RTs for compounds targeting the same target. Our analysis shows that both computational protocols are able to rank compounds in agreement with their experimental RTs. Quantitative structure-kinetics relationship (SKR) models can be identified and employed to predict the impact of a chemical modification on the experimental RT once a calibration study has been performed.
Collapse
Affiliation(s)
- Francesca Galvani
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I-43124 Parma, Italy
| | - Daniele Pala
- Chemistry
Research and Drug Design Department, Chiesi
Farmaceutici S.p.A., Largo F. Belloli 11/A, 43122 Parma, Italy
| | - Alberto Cuzzolin
- Chemistry
Research and Drug Design Department, Chiesi
Farmaceutici S.p.A., Largo F. Belloli 11/A, 43122 Parma, Italy
| | - Laura Scalvini
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I-43124 Parma, Italy
| | - Alessio Lodola
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I-43124 Parma, Italy
| | - Marco Mor
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I-43124 Parma, Italy
- Microbiome
Research Hub, University of Parma, Parco Area delle Scienze 11/A, I-43124 Parma, Italy
| | - Andrea Rizzi
- Chemistry
Research and Drug Design Department, Chiesi
Farmaceutici S.p.A., Largo F. Belloli 11/A, 43122 Parma, Italy
| |
Collapse
|
5
|
Djokovic N, Ruzic D, Rahnasto-Rilla M, Srdic-Rajic T, Lahtela-Kakkonen M, Nikolic K. Expanding the Accessible Chemical Space of SIRT2 Inhibitors through Exploration of Binding Pocket Dynamics. J Chem Inf Model 2022; 62:2571-2585. [PMID: 35467856 DOI: 10.1021/acs.jcim.2c00241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Considerations of binding pocket dynamics are one of the crucial aspects of the rational design of binders. Identification of alternative conformational states or cryptic subpockets could lead to the discovery of completely novel groups of the ligands. However, experimental characterization of pocket dynamics, besides being expensive, may not be able to elucidate all of the conformational states relevant for drug discovery projects. In this study, we propose the protocol for computational simulations of sirtuin 2 (SIRT2) binding pocket dynamics and its integration into the structure-based virtual screening (SBVS) pipeline. Initially, unbiased molecular dynamics simulations of SIRT2:inhibitor complexes were performed using optimized force field parameters of SIRT2 inhibitors. Time-lagged independent component analysis (tICA) was used to design pocket-related collective variables (CVs) for enhanced sampling of SIRT2 pocket dynamics. Metadynamics simulations in the tICA eigenvector space revealed alternative conformational states of the SIRT2 binding pocket and the existence of a cryptic subpocket. Newly identified SIRT2 conformational states outperformed experimentally resolved states in retrospective SBVS validation. After performing prospective SBVS, compounds from the under-represented portions of the SIRT2 inhibitor chemical space were selected for in vitro evaluation. Two compounds, NDJ18 and NDJ85, were identified as potent and selective SIRT2 inhibitors, which validated the in silico protocol and opened up the possibility for generalization and broadening of its application. The anticancer effects of the most potent compound NDJ18 were examined on the triple-negative breast cancer cell line. Results indicated that NDJ18 represents a promising structure suitable for further evaluation.
Collapse
Affiliation(s)
- Nemanja Djokovic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Dusan Ruzic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Minna Rahnasto-Rilla
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, 70210 Kuopio, Finland
| | - Tatjana Srdic-Rajic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | | | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| |
Collapse
|
6
|
Protein-Protein Interaction Inhibitors Targeting the Eph-Ephrin System with a Focus on Amino Acid Conjugates of Bile Acids. Pharmaceuticals (Basel) 2022; 15:ph15020137. [PMID: 35215250 PMCID: PMC8880657 DOI: 10.3390/ph15020137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/05/2023] Open
Abstract
The role of the Eph-ephrin system in the etiology of pathological conditions has been consolidated throughout the years. In this context, approaches directed against this signaling system, intended to modulate its activity, can be strategic therapeutic opportunities. Currently, the most promising class of compounds able to interfere with the Eph receptor-ephrin protein interaction is composed of synthetic derivatives of bile acids. In the present review, we summarize the progresses achieved, in terms of chemical expansions and structure-activity relationships, both in the steroidal core and the terminal carboxylic acid group, along with the pharmacological characterization for the most promising Eph-ephrin antagonists in in vivo settings.
Collapse
|
7
|
Ferlenghi F, Giorgio C, Incerti M, Guidetti L, Chiodelli P, Rusnati M, Tognolini M, Vacondio F, Mor M, Lodola A. Metabolic Soft Spot and Pharmacokinetics: Functionalization of C-3 Position of an Eph-Ephrin Antagonist Featuring a Bile Acid Core as an Effective Strategy to Obtain Oral Bioavailability in Mice. Pharmaceuticals (Basel) 2021; 15:ph15010041. [PMID: 35056098 PMCID: PMC8779995 DOI: 10.3390/ph15010041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 11/16/2022] Open
Abstract
UniPR129, an L-β-homotryptophan conjugate of the secondary bile acid lithocholic acid (LCA), acts as an effective protein-protein interaction (PPI) inhibitor of the Eph-ephrin system but suffers from a poor oral bioavailability in mice. To improve UniPR129 bioavailability, a metabolic soft spot, i.e., the 3α-hydroxyl group on the LCA steroidal ring, was functionalized to 3-hydroxyimine. In vitro metabolism of UniPR129 and 3-hydroxyimine derivative UniPR500 was compared in mouse liver subcellular fractions, and main metabolites were profiled by high resolution (HR-MS) and tandem (MS/MS) mass spectrometry. In mouse liver microsomes (MLM), UniPR129 was converted into several metabolites: M1 derived from the oxidation of the 3-hydroxy group to 3-oxo, M2-M7, mono-hydroxylated metabolites, M8-M10, di-hydroxylated metabolites, and M11, a mono-hydroxylated metabolite of M1. Phase II reactions were only minor routes of in vitro biotransformation. UniPR500 shared several metabolic pathways with parent UniPR129, but it showed higher stability in MLM, with a half-life (t1/2) of 60.4 min, if compared to a t1/2 = 16.8 min for UniPR129. When orally administered to mice at the same dose, UniPR500 showed an increased systemic exposure, maintaining an in vitro valuable pharmacological profile as an EphA2 receptor antagonist and an overall improvement in its physico-chemical profile (solubility, lipophilicity), if compared to UniPR129. The present work highlights an effective strategy for the pharmacokinetic optimization of aminoacid conjugates of bile acids as small molecule Eph-ephrin antagonists.
Collapse
Affiliation(s)
- Francesca Ferlenghi
- Food and Drug Department, University of Parma, Viale delle Scienze 27/A, 43124 Parma, Italy; (F.F.); (C.G.); (M.I.); (L.G.); (M.T.); (A.L.)
| | - Carmine Giorgio
- Food and Drug Department, University of Parma, Viale delle Scienze 27/A, 43124 Parma, Italy; (F.F.); (C.G.); (M.I.); (L.G.); (M.T.); (A.L.)
| | - Matteo Incerti
- Food and Drug Department, University of Parma, Viale delle Scienze 27/A, 43124 Parma, Italy; (F.F.); (C.G.); (M.I.); (L.G.); (M.T.); (A.L.)
| | - Lorenzo Guidetti
- Food and Drug Department, University of Parma, Viale delle Scienze 27/A, 43124 Parma, Italy; (F.F.); (C.G.); (M.I.); (L.G.); (M.T.); (A.L.)
| | - Paola Chiodelli
- Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (P.C.); (M.R.)
| | - Marco Rusnati
- Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (P.C.); (M.R.)
| | - Massimiliano Tognolini
- Food and Drug Department, University of Parma, Viale delle Scienze 27/A, 43124 Parma, Italy; (F.F.); (C.G.); (M.I.); (L.G.); (M.T.); (A.L.)
| | - Federica Vacondio
- Food and Drug Department, University of Parma, Viale delle Scienze 27/A, 43124 Parma, Italy; (F.F.); (C.G.); (M.I.); (L.G.); (M.T.); (A.L.)
- Correspondence: (F.V.); (M.M.); Tel.: +39-0521-905076 (F.V.); +39-0521-905059 (M.M.)
| | - Marco Mor
- Food and Drug Department, University of Parma, Viale delle Scienze 27/A, 43124 Parma, Italy; (F.F.); (C.G.); (M.I.); (L.G.); (M.T.); (A.L.)
- Correspondence: (F.V.); (M.M.); Tel.: +39-0521-905076 (F.V.); +39-0521-905059 (M.M.)
| | - Alessio Lodola
- Food and Drug Department, University of Parma, Viale delle Scienze 27/A, 43124 Parma, Italy; (F.F.); (C.G.); (M.I.); (L.G.); (M.T.); (A.L.)
| |
Collapse
|
8
|
Rocha REO, Chaves EJF, Fischer PHC, Costa LSC, Grillo IB, da Cruz LEG, Guedes FC, da Silveira CH, Scotti MT, Camargo AD, Machado KS, Werhli AV, Ferreira RS, Rocha GB, de Lima LHF. A higher flexibility at the SARS-CoV-2 main protease active site compared to SARS-CoV and its potentialities for new inhibitor virtual screening targeting multi-conformers. J Biomol Struct Dyn 2021; 40:9214-9234. [PMID: 33970798 PMCID: PMC8127201 DOI: 10.1080/07391102.2021.1924271] [Citation(s) in RCA: 2] [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: 03/26/2021] [Accepted: 04/26/2021] [Indexed: 12/14/2022]
Abstract
The main-protease (Mpro) catalyzes a crucial step for the SARS-CoV-2 life cycle. The recent SARS-CoV-2 presents the main protease (MCoV2pro) with 12 mutations compared to SARS-CoV (MCoV1pro). Recent studies point out that these subtle differences lead to mobility variances at the active site loops with functional implications. We use metadynamics simulations and a sort of computational analysis to probe the dynamic, pharmacophoric and catalytic environment differences between the monomers of both enzymes. So, we verify how much intrinsic distinctions are preserved in the functional dimer of MCoV2pro, as well as its implications for ligand accessibility and optimized drug screening. We find a significantly higher accessibility to open binding conformers in the MCoV2pro monomer compared to MCoV1pro. A higher hydration propensity for the MCoV2pro S2 loop with the A46S substitution seems to exercise a key role. Quantum calculations suggest that the wider conformations for MCoV2pro are less catalytically active in the monomer. However, the statistics for contacts involving the N-finger suggest higher maintenance of this activity at the dimer. Docking analyses suggest that the ability to vary the active site width can be important to improve the access of the ligand to the active site in different ways. So, we carry out a multiconformational virtual screening with different ligand bases. The results point to the importance of taking into account the protein conformational multiplicity for new promissors anti MCoV2pro ligands. We hope these results will be useful in prospecting, repurposing and/or designing new anti SARS-CoV-2 drugs.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rafael E. O. Rocha
- Laboratory of Molecular Modeling and Drug Design, Department of Biochemistry and Imunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Laboratory of Structural Biology, Department of Molecular Biology, Universität Salzburg, Salzburg, Austria
| | - Elton J. F. Chaves
- Laboratory of Computational and Quantum Chemistry, Department of Chemistry, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Pedro H. C. Fischer
- Laboratory of Molecular Modeling and Bioinformatics, Department of Exact and Biological Sciences, Universidade Federal de São João Del Rei, Sete Lagoas, Brazil
| | - Leon S. C. Costa
- Comp. Modeling Coordination, Laboratório Nacional de Computação Científica, Petrópolis, Brazil
| | - Igor Barden Grillo
- Laboratory of Computational and Quantum Chemistry, Department of Chemistry, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Luiz E. G. da Cruz
- Laboratory of Computational and Quantum Chemistry, Department of Chemistry, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Fabiana C. Guedes
- Structural Bioinformatic Laboratory, Institute of Technological Sciences, Universidade Federal de Itajubá, Itabira, Brazil
| | - Carlos H. da Silveira
- Structural Bioinformatic Laboratory, Institute of Technological Sciences, Universidade Federal de Itajubá, Itabira, Brazil
| | - Marcus T. Scotti
- Graduate Program in Natural and Synthetic Bioactive Products; Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Alex D. Camargo
- Computational Biology Laboratory, Center for computational sciences, Universidade Federal do Rio Grande, Rio Grande, Brazil
| | - Karina S. Machado
- Computational Biology Laboratory, Center for computational sciences, Universidade Federal do Rio Grande, Rio Grande, Brazil
| | - Adriano V. Werhli
- Computational Biology Laboratory, Center for computational sciences, Universidade Federal do Rio Grande, Rio Grande, Brazil
| | - Rafaela S. Ferreira
- Laboratory of Molecular Modeling and Drug Design, Department of Biochemistry and Imunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Gerd B. Rocha
- Laboratory of Computational and Quantum Chemistry, Department of Chemistry, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Leonardo H. F. de Lima
- Laboratory of Molecular Modeling and Bioinformatics, Department of Exact and Biological Sciences, Universidade Federal de São João Del Rei, Sete Lagoas, Brazil
| |
Collapse
|
9
|
Hsu K, Middlemiss S, Saletta F, Gottschalk S, McCowage GB, Kramer B. Chimeric Antigen Receptor-modified T cells targeting EphA2 for the immunotherapy of paediatric bone tumours. Cancer Gene Ther 2021; 28:321-334. [PMID: 32873870 PMCID: PMC8057949 DOI: 10.1038/s41417-020-00221-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/14/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023]
Abstract
Chimeric Antigen Receptor (CAR) T-cell therapy, as an approved treatment option for patients with B cell malignancies, demonstrates that genetic modification of autologous immune cells is an effective anti-cancer regimen. Erythropoietin-producing Hepatocellular receptor tyrosine kinase class A2 (EphA2) is a tumour associated antigen expressed on a range of sarcomas, including paediatric osteosarcoma (OS) and Ewing sarcoma (ES). We tested human EphA2 directed CAR T cells for their capacity to target and kill human OS and ES tumour cells using in vitro and in vivo assays, demonstrating that EphA2 CAR T cells have potent anti-tumour efficacy in vitro and can eliminate established OS and ES tumours in vivo in a dose and delivery route dependent manner. Next, in an aggressive metastatic OS model we demonstrated that systemically infused EphA2 CAR T cells can traffic to and eradicate tumour deposits in murine livers and lungs. These results support further pre-clinical evaluation of EphA2 CAR T cells to inform the design of early phase clinical trial protocols to test the feasibility and safety of this immune cell therapy in paediatric bone sarcoma patients.
Collapse
Affiliation(s)
- Kenneth Hsu
- Children's Cancer Research Unit, Kid's Research, The Children's Hospital at Westmead, Westmead, NSW, 2145, Australia
| | - Shiloh Middlemiss
- Children's Cancer Research Unit, Kid's Research, The Children's Hospital at Westmead, Westmead, NSW, 2145, Australia
| | - Federica Saletta
- Children's Cancer Research Unit, Kid's Research, The Children's Hospital at Westmead, Westmead, NSW, 2145, Australia
| | - Stephen Gottschalk
- Department of Bone Marrow Transplant and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Geoffrey B McCowage
- Children's Cancer Centre, The Children's Hospital at Westmead, Westmead, NSW, 2145, Australia
| | - Belinda Kramer
- Children's Cancer Research Unit, Kid's Research, The Children's Hospital at Westmead, Westmead, NSW, 2145, Australia.
| |
Collapse
|
10
|
Evaluation of the Anti-Tumor Activity of Small Molecules Targeting Eph/Ephrins in APC min/J Mice. Pharmaceuticals (Basel) 2020; 13:ph13040069. [PMID: 32316101 PMCID: PMC7243115 DOI: 10.3390/ph13040069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/12/2020] [Accepted: 04/14/2020] [Indexed: 12/09/2022] Open
Abstract
The Eph receptors are the largest receptors tyrosine kinases (RTKs) family in humans and together with ephrin ligands constitute a complex cellular communication system often dysregulated in many tumors. The role of the Eph-ephrin system in colorectal cancer (CRC) has been investigated and different expression of Eph receptors have been associated with tumor development and progression. In light of this evidence, we investigated if a pharmacological approach aimed at inhibiting Eph/ephrin interaction through small molecules could prevent tumor growth in APC min/J mice. The 8-week treatment with the Eph-ephrin antagonist UniPR129 significantly reduced the number of adenomas in the ileum and decreased the diameter of adenomas in the same region. Overall our data suggested as UniPR129 could be able to slow down the tumor development in APC min/J mice. These results further confirm literature data about Eph kinases as a new valuable target in the intestinal cancer and for the first time showed the feasibility of the Eph-ephrin inhibition as a useful pharmacological approach against the intestinal tumorigenesis. In conclusion this work paves the way for further studies with Eph-ephrin inhibitors in order to confirm the Eph antagonism as innovative pharmacological approach with preventive benefit in the intestinal tumor development.
Collapse
|
11
|
Giorgio C, Zanotti I, Lodola A, Tognolini M. Ephrin or not? Six tough questions on Eph targeting. Expert Opin Ther Targets 2020; 24:403-415. [PMID: 32197575 DOI: 10.1080/14728222.2020.1745187] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Introduction: The Eph-ephrin is a cell-cell communication system generating a forward signal in cell expressing Eph receptors and a reverse signal in ephrin-ligand expressing cells. While clearly involved in the insurgence and progression of cancer, the understanding of the molecular mechanisms regulated by this system needs development; this is a hurdle to the development of therapeutic strategies that can target the Eph receptors and/or their ephrin ligands.Areas covered: We have taken the opportunity to share some key questions on the most effective strategies to target the Eph-ephrin system. This article is based on our experience of the field and therefore is a Perspective and not comprehensive examination of the literature.Expert opinion: Targeting of the Eph-ephrin system has emerged as a potentially valuable approach for cancer therapy. Pharmacological tools have been reported in the last 15 years and these include forward signaling blockers such as kinases inhibitors and antagonists of forward and reverse signaling. Also, biologics including antibodies and recombinant proteins have been developed and some have reached early clinical stages. Data deem the Eph-ephrin system as a signaling axis that is an elusive target. A better understanding of the basic pharmacology behind the activity of available agents and a comprehensive knowledge of the ephrin biology are necessary. We are looking forward to knowing the opinion of the readers.
Collapse
Affiliation(s)
- Carmine Giorgio
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| | - Ilaria Zanotti
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| | - Alessio Lodola
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| | | |
Collapse
|
12
|
Fusani L, Palmer DS, Somers DO, Wall ID. Exploring Ligand Stability in Protein Crystal Structures Using Binding Pose Metadynamics. J Chem Inf Model 2020; 60:1528-1539. [PMID: 31910338 PMCID: PMC7145342 DOI: 10.1021/acs.jcim.9b00843] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
![]()
Identification of
correct protein–ligand binding poses is
important in structure-based drug design and crucial for the evaluation
of protein–ligand binding affinity. Protein–ligand coordinates are commonly obtained from
crystallography experiments that provide a static model of an ensemble
of conformations. Binding pose metadynamics (BPMD) is an enhanced
sampling method that allows for an efficient assessment of ligand
stability in solution. Ligand poses that are unstable under the bias
of the metadynamics simulation are expected to be infrequently occupied
in the energy landscape, thus making minimal contributions to the
binding affinity. Here, the robustness of the method is studied using
crystal structures with ligands known to be incorrectly modeled, as
well as 63 structurally diverse crystal structures with ligand fit
to electron density from the Twilight database. Results show that
BPMD can successfully differentiate compounds whose binding pose is
not supported by the electron density from those with well-defined
electron density.
Collapse
Affiliation(s)
- Lucia Fusani
- Molecular Design UK, GSK Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K.,Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G11XL, U.K
| | - David S Palmer
- Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G11XL, U.K
| | - Don O Somers
- Protein, Cellular and Structural Sciences, GSK Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Ian D Wall
- Molecular Design UK, GSK Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| |
Collapse
|
13
|
Incerti M, Russo S, Corrado M, Giorgio C, Ballabeni V, Chiodelli P, Rusnati M, Scalvini L, Callegari D, Castelli R, Vacondio F, Ferlenghi F, Tognolini M, Lodola A. Optimization of EphA2 antagonists based on a lithocholic acid core led to the identification of UniPR505, a new 3α-carbamoyloxy derivative with antiangiogenetic properties. Eur J Med Chem 2020; 189:112083. [PMID: 32000051 DOI: 10.1016/j.ejmech.2020.112083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 01/16/2020] [Accepted: 01/19/2020] [Indexed: 11/24/2022]
Abstract
The EphA2 receptor has been validated in animal models as new target for treating tumors depending on angiogenesis and vasculogenic mimicry. In the present work, we extended our current knowledge on structure-activity relationship (SAR) data of two related classes of antagonists of the EphA2 receptor, namely 5β-cholan-24-oic acids and 5β-cholan-24-oyl l-β-homotryptophan conjugates, with the aim to develop new antiangiogenic compounds able to efficiently prevent the formation of blood vessels. As a result of our exploration, we identified UniPR505, N-[3α-(Ethylcarbamoyl)oxy-5β-cholan-24-oyl]-l-β-homo-tryptophan (compound 14), as a submicromolar antagonist of the EphA2 receptor capable to block EphA2 phosphorylation and to inhibit neovascularization in a chorioallantoic membrane (CAM) assay.
Collapse
Affiliation(s)
- Matteo Incerti
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | - Simonetta Russo
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | - Miriam Corrado
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | - Carmine Giorgio
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | - Vigilio Ballabeni
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | - Paola Chiodelli
- Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| | - Marco Rusnati
- Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| | - Laura Scalvini
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | | | - Riccardo Castelli
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | - Federica Vacondio
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | | | | | - Alessio Lodola
- Department of Food and Drug, University of Parma, 43124, Parma, Italy.
| |
Collapse
|
14
|
Ferlenghi F, Castelli R, Scalvini L, Giorgio C, Corrado M, Tognolini M, Mor M, Lodola A, Vacondio F. Drug-gut microbiota metabolic interactions: the case of UniPR1331, selective antagonist of the Eph-ephrin system, in mice. J Pharm Biomed Anal 2019; 180:113067. [PMID: 31891876 DOI: 10.1016/j.jpba.2019.113067] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/24/2022]
Abstract
The interest on the role of gut microbiota in the biotransformation of drugs and xenobiotics has grown over the last decades and a deeper understanding of the mutual interactions is expected to help future improvements in the fields of drug development, toxicological risk assessment and precision medicine. In this paper, a microbiome drug metabolism case is presented, involving a lipophilic small molecule, N-(3β-hydroxy-Δ5-cholen-24-oyl)-l-tryptophan, UniPR1331, active as antagonist of the Eph-ephrin system and effective in vivo in a murine orthotopic model of glioblastoma multiforme (GBM). Following the administration of a single 30 mg/kg dose (p.o.) to mice, maximal plasma levels were reached 30 min after dosing and rapidly declined thereafter. To explain the observed in vivo behaviour, in vitro phase I and II metabolism assays were conducted employing mouse and human liver subcellular fractions and profiling main metabolites by means of tandem (HPLC-ESI-MS/MS) and high resolution mass spectrometry (HPLC-ESI-HR-MS). In the presence of in vitro mouse liver fractions, UniPR1331 showed a low phase I metabolic clearance, despite the identification of a 3-oxo and several hydroxylated metabolites. Conversely, after oral administration of UniPR1331 to mice, a novel isobaric metabolite was detected that (i) was subjected, as parent UniPR1331, to enterohepatic circulation (ii) had not been previously identified in vitro in mouse liver microsomes and (iii) was not observed forming after intraperitoneal (i.p.) administration of UniPR1331. An in vitro faecal fermentation assay produced the same chemical entity supporting a major role of gut microbiota in the in vivo clearance of UniPR1331.
Collapse
Affiliation(s)
- Francesca Ferlenghi
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/a, 43124, Parma, Italy
| | - Riccardo Castelli
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/a, 43124, Parma, Italy
| | - Laura Scalvini
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/a, 43124, Parma, Italy
| | - Carmine Giorgio
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/a, 43124, Parma, Italy
| | - Miriam Corrado
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/a, 43124, Parma, Italy
| | - Massimiliano Tognolini
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/a, 43124, Parma, Italy
| | - Marco Mor
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/a, 43124, Parma, Italy
| | - Alessio Lodola
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/a, 43124, Parma, Italy.
| | - Federica Vacondio
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/a, 43124, Parma, Italy.
| |
Collapse
|
15
|
Zhang H, Kai ELJ, Lu L. Investigating the stability of dengue virus envelope protein dimer using well-tempered metadynamics simulations. Proteins 2019; 88:643-653. [PMID: 31697409 DOI: 10.1002/prot.25844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/24/2019] [Accepted: 11/03/2019] [Indexed: 12/21/2022]
Abstract
We explored the stability of the dengue virus envelope (E) protein dimer since it is widely assumed that the E protein dimer is stabilized by drug ligands or antibodies in an acidic environment, neutralizing the virus's ability to fuse with human cells. During this process, a large conformational change of the E protein dimer is required. We performed Molecular Dynamics simulations to mimic the conformational change and stability of the dimer in neutral and acidic conditions with the well-tempered metadynamics method. Furthermore, as a few neutralizing antibodies discovered from dengue patients were reported, we used the same simulation method to examine the influence of a selected antibody on the dimer stability in both neutral and acidic conditions. We also investigated the antibody's influence on a point-mutated E protein that had been reported to interrupt the protein-antibody interaction and result in more than 95% loss of the antibody's binding ability. Our simulation results are highly consistent with the experimental conclusion that binding of the antibody to the E protein dimer neutralizes the virus, especially in a low pH condition, while the mutation of W101A or N153A significantly reduces the antibody's ability in stabilizing the E protein dimer. We demonstrate that well-tempered metadynamics can be used to accurately explore the antibody's interaction on large protein complexes such as the E protein dimer, and the computational approach in this work is promising in future antibody development.
Collapse
Affiliation(s)
- Haiping Zhang
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,Joint Engineering Research Center for Health Big Data Intelligent Analysis Technology, Shenzhen Institutes of Advanced Technology, Shenzhen, Guangdong Province, People's Republic of China
| | - Eric L J Kai
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Lanyuan Lu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
16
|
Provasi D. Ligand-Binding Calculations with Metadynamics. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2019; 2022:233-253. [PMID: 31396906 DOI: 10.1007/978-1-4939-9608-7_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
All-atom molecular dynamics simulations can capture the dynamic degrees of freedom that characterize molecular recognition, the knowledge of which constitutes the cornerstone of rational approaches to drug design and optimization. In particular, enhanced sampling algorithms, such as metadynamics, are powerful tools to dramatically reduce the computational cost required for a mechanistic description of the binding process. Here, we describe the essential details characterizing these simulation strategies, focusing on the critical step of identifying suitable reaction coordinates, as well as on the different analysis algorithms to estimate binding affinity and residence times. We conclude with a survey of published applications that provides explicit examples of successful simulations for several targets.
Collapse
Affiliation(s)
- Davide Provasi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
17
|
Giorgio C, Incerti M, Pala D, Russo S, Chiodelli P, Rusnati M, Cantoni A, Di Lecce R, Barocelli E, Bertoni S, Ravassard P, Manenti F, Piemonti L, Ferlenghi F, Lodola A, Tognolini M. Inhibition of Eph/ephrin interaction with the small molecule UniPR500 improves glucose tolerance in healthy and insulin-resistant mice. Pharmacol Res 2019; 141:319-330. [DOI: 10.1016/j.phrs.2019.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 11/05/2018] [Accepted: 01/04/2019] [Indexed: 01/22/2023]
|
18
|
Mohammadi M, Mohammadiarani H, Shaw VS, Neubig RR, Vashisth H. Interplay of cysteine exposure and global protein dynamics in small-molecule recognition by a regulator of G-protein signaling protein. Proteins 2018; 87:146-156. [PMID: 30521141 DOI: 10.1002/prot.25642] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 11/07/2018] [Accepted: 11/29/2018] [Indexed: 02/06/2023]
Abstract
Regulator of G protein signaling (RGS) proteins play a pivotal role in regulation of G protein-coupled receptor (GPCR) signaling and are therefore becoming an increasingly important therapeutic target. Recently discovered thiadiazolidinone (TDZD) compounds that target cysteine residues have shown different levels of specificities and potencies for the RGS4 protein, thereby suggesting intrinsic differences in dynamics of this protein upon binding of these compounds. In this work, we investigated using atomistic molecular dynamics (MD) simulations the effect of binding of several small-molecule inhibitors on perturbations and dynamical motions in RGS4. Specifically, we studied two conformational models of RGS4 in which a buried cysteine residue is solvent-exposed due to side-chain motions or due to flexibility in neighboring helices. We found that TDZD compounds with aromatic functional groups perturb the RGS4 structure more than compounds with aliphatic functional groups. Moreover, small-molecules with aromatic functional groups but lacking sulfur atoms only transiently reside within the protein and spontaneously dissociate to the solvent. We further measured inhibitory effects of TDZD compounds using a protein-protein interaction assay on a single-cysteine RGS4 protein showing trends in potencies of compounds consistent with our simulation studies. Thermodynamic analyses of RGS4 conformations in the apo-state and on binding to TDZD compounds revealed links between both conformational models of RGS4. The exposure of cysteine side-chains appears to facilitate initial binding of TDZD compounds followed by migration of the compound into a bundle of four helices, thereby causing allosteric perturbations in the RGS/Gα protein-protein interface.
Collapse
Affiliation(s)
| | | | - Vincent S Shaw
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Richard R Neubig
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Harish Vashisth
- Department of Chemical Engineering, University of New Hampshire, Durham, New Hampshire
| |
Collapse
|
19
|
Iglesias J, Saen‐oon S, Soliva R, Guallar V. Computational structure‐based drug design: Predicting target flexibility. WILEY INTERDISCIPLINARY REVIEWS-COMPUTATIONAL MOLECULAR SCIENCE 2018. [DOI: 10.1002/wcms.1367] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
| | | | | | - Victor Guallar
- Life Science DepartmentBarcelonaSpain
- ICREA, Passeig Lluís Companys 23BarcelonaSpain
| |
Collapse
|
20
|
Berthonneau C, Nun P, Rivière M, Pauvert M, Dénès F, Lebreton J. Hemisynthesis of 2,3,4-13C3-1,4-Androstadien-3,17-dione: A Key Precursor for the Synthesis of 13C3-Androstanes and 13C3-Estranes. J Org Chem 2018; 83:3727-3737. [DOI: 10.1021/acs.joc.7b03216] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Clément Berthonneau
- Université de Nantes, Chimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM), UMR CNRS 6230, 2, rue de la Houssinière BP 92208, Nantes Cedex 3 44322, France
- Atlanchim Pharma, 3, Rue Aronnax, Saint Herblain 44800, France
| | - Pierrick Nun
- Université de Nantes, Chimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM), UMR CNRS 6230, 2, rue de la Houssinière BP 92208, Nantes Cedex 3 44322, France
| | - Matthieu Rivière
- Université de Nantes, Chimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM), UMR CNRS 6230, 2, rue de la Houssinière BP 92208, Nantes Cedex 3 44322, France
| | - Mickael Pauvert
- Atlanchim Pharma, 3, Rue Aronnax, Saint Herblain 44800, France
| | - Fabrice Dénès
- Université de Nantes, Chimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM), UMR CNRS 6230, 2, rue de la Houssinière BP 92208, Nantes Cedex 3 44322, France
| | - Jacques Lebreton
- Université de Nantes, Chimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM), UMR CNRS 6230, 2, rue de la Houssinière BP 92208, Nantes Cedex 3 44322, France
- Atlanchim Pharma, 3, Rue Aronnax, Saint Herblain 44800, France
| |
Collapse
|
21
|
Giorgio C, Incerti M, Corrado M, Rusnati M, Chiodelli P, Russo S, Callegari D, Ferlenghi F, Ballabeni V, Barocelli E, Lodola A, Tognolini M. Pharmacological evaluation of new bioavailable small molecules targeting Eph/ephrin interaction. Biochem Pharmacol 2017; 147:21-29. [PMID: 29129483 DOI: 10.1016/j.bcp.2017.11.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/07/2017] [Indexed: 11/30/2022]
Abstract
Eph/ephrin system is an emerging target for cancer therapy but the lack of potent, stable and orally bioavailable compounds is impairing the development of the field. Since 2009 our research group has been devoted to the discovery and development of small molecules targeting Eph/ephrin system and our research culminated with the synthesis of UniPR129, a potent but problematic Eph/ephrin antagonist. Herein, we describe the in vitro pharmacological properties of two derivatives (UniPR139 and UniPR502) stemmed from structure of UniPR129. These two compounds acted as competitive and reversible antagonists of all Eph receptors reducing both ephrin-A1 and -B1 binding to EphAs and EphBs receptors in the low micromolar range. The compounds acted as antagonists inhibiting ephrin-A1-dependent EphA2 activation and UniPR139 exerted an anti-angiogenic effect, inhibiting HUVEC tube formation in vitro and VEGF-induced vessel formation in the chick chorioallantoic membrane assay. Finally, the oral bioavailability of UniPR139 represents a step forward in the search of molecules targeting the Eph/ephrin system and offers a new pharmacological tool useful for future in vivo studies.
Collapse
Affiliation(s)
- Carmine Giorgio
- Department of Food and Drugs, University of Parma, 43124 Parma, Italy
| | - Matteo Incerti
- Department of Food and Drugs, University of Parma, 43124 Parma, Italy
| | - Miriam Corrado
- Department of Food and Drugs, University of Parma, 43124 Parma, Italy
| | - Marco Rusnati
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Paola Chiodelli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Simonetta Russo
- Department of Food and Drugs, University of Parma, 43124 Parma, Italy
| | | | | | - Vigilio Ballabeni
- Department of Food and Drugs, University of Parma, 43124 Parma, Italy
| | | | - Alessio Lodola
- Department of Food and Drugs, University of Parma, 43124 Parma, Italy.
| | | |
Collapse
|
22
|
Targeting Eph/ephrin system in cancer therapy. Eur J Med Chem 2017; 142:152-162. [PMID: 28780190 DOI: 10.1016/j.ejmech.2017.07.029] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/12/2017] [Accepted: 07/16/2017] [Indexed: 12/19/2022]
Abstract
It is well established that the Eph/ephrin system plays a central role in the embryonic development, with minor implications in the physiology of the adult. However, it is overexpressed and deregulated in a variety of tumors, with a primary involvement in tumorigenesis, tumor angiogenesis, metastasis development, and cancer stem cell regeneration. Targeting the Eph/ephrin system with biologicals, including antibodies and recombinant proteins, reduces tumor growth in animal models of hematological malignancies, breast, prostate, colon, head and neck cancers and glioblastoma. Currently, some of these biopharmaceutical agents are under investigations in phase I or phase II clinical trials. Peptides and small molecules targeting protein-protein-interaction (PPI) are in the late preclinical phase where they are showing promising activity in models of glioblastoma, ovarian and lung cancer. The present review summarizes the most critical findings proposing the Eph/ephrin signaling system as a new target in molecularly targeted oncology.
Collapse
|
23
|
De Vivo M, Cavalli A. Recent advances in dynamic docking for drug discovery. WILEY INTERDISCIPLINARY REVIEWS-COMPUTATIONAL MOLECULAR SCIENCE 2017. [DOI: 10.1002/wcms.1320] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Marco De Vivo
- Laboratory of Molecular Modeling and Drug DiscoveryIstituto Italiano di TecnologiaGenoaItaly
- IAS‐S/INM‐9 Computational BiomedicineForschungszentrum JülichJülichGermany
| | - Andrea Cavalli
- CompunetIstituto Italiano di TecnologiaGenoaItaly
- Department of Pharmacy and Biotechnology, Alma Mater StudiorumUniversity of BolognaBolognaItaly
| |
Collapse
|