1
|
Acharya A, Yadav M, Nagpure M, Kumaresan S, Guchhait SK. Molecular medicinal insights into scaffold hopping-based drug discovery success. Drug Discov Today 2024; 29:103845. [PMID: 38013043 DOI: 10.1016/j.drudis.2023.103845] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 11/29/2023]
Abstract
In both academia and the pharmaceutical industry, innovative hypotheses, methodologies and technologies that can shorten the drug research and development, leading to higher success rates, are vital. In this review, we demonstrate how innovative variations of the scaffold-hopping strategy have been used to create new druggable molecular spaces, drugs, clinical candidates, preclinical candidates, and bioactive agents. We also analyze molecular modulations that enabled improvements of the pharmacodynamic (PD), physiochemical, and pharmacokinetic (PK) properties (P3 properties) of the drugs resulting from these scaffold-hopping strategies.
Collapse
Affiliation(s)
- Ayan Acharya
- National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab 160062, India
| | - Mukul Yadav
- National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab 160062, India
| | - Mithilesh Nagpure
- National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab 160062, India
| | - Sanathanalaxmi Kumaresan
- National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab 160062, India; National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab 160062, India
| | - Sankar K Guchhait
- National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab 160062, India.
| |
Collapse
|
2
|
Computational Drug Repurposing Resources and Approaches for Discovering Novel Antifungal Drugs against Candida albicans N-Myristoyl Transferase. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2021. [DOI: 10.22207/jpam.15.2.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Candida albicans is a yeast that is an opportunistic fungal pathogen and also identified as ubiquitous polymorphic species that is mainly linked with major fungal infections in humans, particularly in the immunocompromised patients including transplant recipients, chemotherapy patients, HIV-infected patients as well as in low-birth-weight infants. Systemic Candida infections have a high mortality rate of around 29 to 76%. For reducing its infection, limited drugs are existing such as caspofungin, fluconazole, terbinafine, and amphotericin B, etc. which contain unlikable side effects and also toxic. This review intends to utilize advanced bioinformatics technologies such as Molecular docking, Scaffold hopping, Virtual screening, Pharmacophore modeling, Molecular dynamics (MD) simulation for the development of potentially new drug candidates with a drug-repurpose approach against Candida albicans within a limited time frame and also cost reductive.
Collapse
|
3
|
Qin X, Vila-Sanjurjo C, Singh R, Philipp B, Goycoolea FM. Screening of Bacterial Quorum Sensing Inhibitors in a Vibrio fischeri LuxR-Based Synthetic Fluorescent E. coli Biosensor. Pharmaceuticals (Basel) 2020; 13:E263. [PMID: 32971993 PMCID: PMC7559085 DOI: 10.3390/ph13090263] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
A library of 23 pure compounds of varying structural and chemical characteristics was screened for their quorum sensing (QS) inhibition activity using a synthetic fluorescent Escherichia coli biosensor that incorporates a modified version of lux regulon of Vibrio fischeri. Four such compounds exhibited QS inhibition activity without compromising bacterial growth, namely, phenazine carboxylic acid (PCA), 2-heptyl-3-hydroxy-4-quinolone (PQS), 1H-2-methyl-4-quinolone (MOQ) and genipin. When applied at 50 µM, these compounds reduced the QS response of the biosensor to 33.7% ± 2.6%, 43.1% ± 2.7%, 62.2% ± 6.3% and 43.3% ± 1.2%, respectively. A series of compounds only showed activity when tested at higher concentrations. This was the case of caffeine, which, when applied at 1 mM, reduced the QS to 47% ± 4.2%. In turn, capsaicin, caffeic acid phenethyl ester (CAPE), furanone and polygodial exhibited antibacterial activity when applied at 1mM, and reduced the bacterial growth by 12.8% ± 10.1%, 24.4% ± 7.0%, 91.4% ± 7.4% and 97.5% ± 3.8%, respectively. Similarly, we confirmed that trans-cinnamaldehyde and vanillin, when tested at 1 mM, reduced the QS response to 68.3% ± 4.9% and 27.1% ± 7.4%, respectively, though at the expense of concomitantly reducing cell growth by 18.6% ± 2.5% and 16% ± 2.2%, respectively. Two QS natural compounds of Pseudomonas aeruginosa, namely PQS and PCA, and the related, synthetic compounds MOQ, 1H-3-hydroxyl-4-quinolone (HOQ) and 1H-2-methyl-3-hydroxyl-4-quinolone (MHOQ) were used in molecular docking studies with the binding domain of the QS receptor TraR as a target. We offer here a general interpretation of structure-function relationships in this class of compounds that underpins their potential application as alternatives to antibiotics in controlling bacterial virulence.
Collapse
Affiliation(s)
- Xiaofei Qin
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China;
- Laboratory of Nanobiotechnology, Institute of Plant Biology and Biotechnology, University of Münster, Schlossplatz 8, D-48143 Münster, Germany;
| | - Celina Vila-Sanjurjo
- Laboratory of Nanobiotechnology, Institute of Plant Biology and Biotechnology, University of Münster, Schlossplatz 8, D-48143 Münster, Germany;
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela. Campus Vida, s/n, 15782 Santiago de Compostela, Spain
| | - Ratna Singh
- Laboratory of Molecular Phytopathology and Renewable Resources, Institute of Plant Biology and Biotechnology, University of Münster, Schlossplatz 8, D-48143 Münster, Germany;
| | - Bodo Philipp
- Institute of Molecular Microbiology and Biotechnology, University of Münster, Corrensstraße 3, D-48149 Münster, Germany;
| | - Francisco M. Goycoolea
- Laboratory of Nanobiotechnology, Institute of Plant Biology and Biotechnology, University of Münster, Schlossplatz 8, D-48143 Münster, Germany;
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
4
|
Marconi G, Aiello D, Kindiger B, Storchi L, Marrone A, Reale L, Terzaroli N, Albertini E. The Role of APOSTART in Switching between Sexuality and Apomixis in Poa pratensis. Genes (Basel) 2020; 11:genes11080941. [PMID: 32824095 PMCID: PMC7464379 DOI: 10.3390/genes11080941] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/11/2020] [Accepted: 08/11/2020] [Indexed: 12/20/2022] Open
Abstract
The production of seeds without sex is considered the holy grail of plant biology. The transfer of apomixis to various crop species has the potential to transform plant breeding, since it will allow new varieties to retain valuable traits thorough asexual reproduction. Therefore, a greater molecular understanding of apomixis is fundamental. In a previous work we identified a gene, namely APOSTART, that seemed to be involved in this asexual mode of reproduction, which is very common in Poa pratensis L., and here we present a detailed work aimed at clarifying its role in apomixis. In situ hybridization showed that PpAPOSTART is expressed in reproductive tissues from pre-meiosis to embryo development. Interestingly, it is expressed early in few nucellar cells of apomictic individuals possibly switching from a somatic to a reproductive cell as in aposporic apomixis. Moreover, out of 13 APOSTART members, we identified one, APOSTART_6, as specifically expressed in flower tissue. APOSTART_6 also exhibited delayed expression in apomictic genotypes when compared with sexual types. Most importantly, the SCAR (Sequence Characterized Amplified Region) derived from the APOSTART_6 sequence completely co-segregated with apomixis.
Collapse
Affiliation(s)
- Gianpiero Marconi
- Dipartimento di Scienze Agrarie, Alimentari e Ambientali, Università degli Studi di Perugia, Borgo XX Giugno 74, 06121 Perugia, Italy; (G.M.); (D.A.); (L.R.); (N.T.)
| | - Domenico Aiello
- Dipartimento di Scienze Agrarie, Alimentari e Ambientali, Università degli Studi di Perugia, Borgo XX Giugno 74, 06121 Perugia, Italy; (G.M.); (D.A.); (L.R.); (N.T.)
| | - Bryan Kindiger
- USDA-ARS, Grazinglands Research Laboratory, 7207 West Cheyenne St., El Reno, OK 73036, USA;
| | - Loriano Storchi
- Dipartimento di Farmacia, Università G. d’Annunzio, via dei Vestini 31, 66100 Chieti, Italy; (L.S.); (A.M.)
- Molecular Discovery Limited, Elstree WD6 3FG, UK
| | - Alessandro Marrone
- Dipartimento di Farmacia, Università G. d’Annunzio, via dei Vestini 31, 66100 Chieti, Italy; (L.S.); (A.M.)
| | - Lara Reale
- Dipartimento di Scienze Agrarie, Alimentari e Ambientali, Università degli Studi di Perugia, Borgo XX Giugno 74, 06121 Perugia, Italy; (G.M.); (D.A.); (L.R.); (N.T.)
| | - Niccolò Terzaroli
- Dipartimento di Scienze Agrarie, Alimentari e Ambientali, Università degli Studi di Perugia, Borgo XX Giugno 74, 06121 Perugia, Italy; (G.M.); (D.A.); (L.R.); (N.T.)
| | - Emidio Albertini
- Dipartimento di Scienze Agrarie, Alimentari e Ambientali, Università degli Studi di Perugia, Borgo XX Giugno 74, 06121 Perugia, Italy; (G.M.); (D.A.); (L.R.); (N.T.)
- Correspondence:
| |
Collapse
|
5
|
Paciotti R, Agamennone M, Coletti C, Storchi L. Characterization of PD-L1 binding sites by a combined FMO/GRID-DRY approach. J Comput Aided Mol Des 2020; 34:897-914. [PMID: 32185582 DOI: 10.1007/s10822-020-00306-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/09/2020] [Indexed: 12/17/2022]
Abstract
The programmed cell death protein 1 (PD-1) and its ligand, PD-L1, constitute an important co-inhibitory immune checkpoint leading to downregulation of immune system. Tumor cells developed a strategy to trigger PD-1/PD-L1 pathway reducing the T cell anticancer activity. Anti-PD-L1 small drugs, generally with improved pharmacokinetic and technological profiles than monoclonal antibodies, became an attractive research topic. Nevertheless, still few works have been published on the chemical features of possible binding sites. In this work, we applied a novel computational protocol based on the combination of the ab initio Fragment Molecular Orbital (FMO) method and a newly developed GRID-DRY approach in order to characterize the PD-L1 binding sites, starting from PD-1/PD-L1 and PD-L1/BMS-ligands (Bristol-Mayers Squibb ligands) complexes. The FMO method allows the calculation of the pair-residues as well as the ligand-residues interactions with ab initio accuracy, whereas the GRID-DRY approach is an effective tool to investigate hydrophobic interactions, not easily detectable by ab initio methods. The present GRID-DRY protocol is able to determine the energy contributions of each ligand atoms to each hydrophobic interaction, both qualitatively and quantitatively. We were also able to identify the three specific hot regions involved in PD-1/PD-L1 protein-protein interaction and in PD-L1/BMS-ligand interactions, in agreement with preceding theoretical/experimental results, and to suggest a specific pharmacophore for PD-L1 inhibitors.
Collapse
Affiliation(s)
- Roberto Paciotti
- Department of Pharmacy, Università "G. D'Annunzio" Di Chieti-Pescara, Chieti, Italy.
| | | | - Cecilia Coletti
- Department of Pharmacy, Università "G. D'Annunzio" Di Chieti-Pescara, Chieti, Italy
| | - Loriano Storchi
- Department of Pharmacy, Università "G. D'Annunzio" Di Chieti-Pescara, Chieti, Italy. .,Molecular Discovery Limited, Middlesex, London, UK.
| |
Collapse
|
6
|
Radchenko T, Fontaine F, Morettoni L, Zamora I. WebMetabase: cleavage sites analysis tool for natural and unnatural substrates from diverse data source. Bioinformatics 2019; 35:650-655. [PMID: 30052776 DOI: 10.1093/bioinformatics/bty667] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/29/2018] [Accepted: 07/24/2018] [Indexed: 11/14/2022] Open
Abstract
SUMMARY More than 150 peptide therapeutics are globally in clinical development. Many enzymatic barriers should be crossed by a successful drug to be prosperous in such a process. Therefore, the new peptide drugs must be designed preventing the potential protease cleavage to make the compound less susceptible to protease reaction. We present a new data analysis tool developed in WebMetabase, an approach that stores the information from liquid chromatography mass spectrometry-based experimental data or from external sources such as the MEROPS database. The tool is a chemically aware system where each peptide substrate is presented as a sequence of structural blocks (SBs) connected by amide bonds and not being limited to the natural amino acids. Each SB is characterized by its pharmacophoric and physicochemical properties including a similarity score that describes likelihood between a SB and each one of the other SBs in the database. This methodology can be used to perform a frequency analysis to discover the most frequent cleavage sites for similar amide bonds, defined based on the similarity of the SB that participate in such a bond within the experimentally derived and/or public database. AVAILABILITY AND IMPLEMENTATION http://webmetabase.com:8182/WebMetabaseBioinformatics/. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Tatiana Radchenko
- Pompeu Fabra University, Barcelona, Spain.,Lead Molecular Design, S.L., Sant Cugat del Vallés, Spain
| | | | | | - Ismael Zamora
- Pompeu Fabra University, Barcelona, Spain.,Lead Molecular Design, S.L., Sant Cugat del Vallés, Spain
| |
Collapse
|
7
|
Tran-Nguyen VK, Da Silva F, Bret G, Rognan D. All in One: Cavity Detection, Druggability Estimate, Cavity-Based Pharmacophore Perception, and Virtual Screening. J Chem Inf Model 2019; 59:573-585. [PMID: 30563339 DOI: 10.1021/acs.jcim.8b00684] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Discovering the very first ligands of pharmacologically important targets in a fast and cost-efficient manner is an important issue in drug discovery. In the absence of structural information on either endogenous or synthetic ligands, computational chemists classically identify the very first hits by docking compound libraries to a binding site of interest, with well-known biases arising from the usage of scoring functions. We herewith propose a novel computational method tailored to ligand-free protein structures and consisting in the generation of simple cavity-based pharmacophores to which potential ligands could be aligned by the use of a smooth Gaussian function. The method, embedded in the IChem toolkit, automatically detects ligand-binding cavities, then predicts their structural druggability, and last creates a structure-based pharmacophore for predicted druggable binding sites. A companion tool (Shaper2) was designed to align ligands to cavity-derived pharmacophoric features. The proposed method is as efficient as state-of-the-art virtual screening methods (ROCS, Surflex-Dock) in both posing and virtual screening challenges. Interestingly, IChem-Shaper2 is clearly orthogonal to these latter methods in retrieving unique chemotypes from high-throughput virtual screening data.
Collapse
Affiliation(s)
- Viet-Khoa Tran-Nguyen
- Laboratoire d'Innovation Thérapeutique , UMR 7200 CNRS-Université de Strasbourg , 67400 Illkirch , France
| | - Franck Da Silva
- Laboratoire d'Innovation Thérapeutique , UMR 7200 CNRS-Université de Strasbourg , 67400 Illkirch , France
| | - Guillaume Bret
- Laboratoire d'Innovation Thérapeutique , UMR 7200 CNRS-Université de Strasbourg , 67400 Illkirch , France
| | - Didier Rognan
- Laboratoire d'Innovation Thérapeutique , UMR 7200 CNRS-Université de Strasbourg , 67400 Illkirch , France
| |
Collapse
|
8
|
Design, synthesis and pharmacological evaluation of N4,N6-disubstituted pyrimidine-4,6-diamine derivatives as potent EGFR inhibitors in non-small cell lung cancer. Eur J Med Chem 2018; 157:1300-1325. [PMID: 30195240 DOI: 10.1016/j.ejmech.2018.08.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/11/2018] [Accepted: 08/11/2018] [Indexed: 02/06/2023]
Abstract
A novel series of 4, 6-disubstituted pyrimidines derivatives were designed, synthesized, and evaluated as epidermal growth factor receptor (EGFR) inhibitors for non-small cell lung cancer(NSCLC). 4, 6-disubstituted pyrimidines as core structure was utilized to substitute the lead structure AZD3759 of the quinazoline basic skeleton via an approach involving scaffold hopping. It was found that compound Yfq07 exhibited the best inhibitory effect compared with AZD3759 in vitro and in vivo: Yfq07 exhibited a competitive ATP inhibitory effect, multiple target effects, and further featured a stronger activity against H3255, A431, HCC827, PC-9 and H1975 compared to AZD3759. Moreover, a stronger pro-apoptotic effect, inhibition of cell G2/M phase on A431, H3255, HCC827 and H1975 could also be observed. In this study, the ultimate goal was changing the core structure to improve other epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) properties while retaining the overall potency. Yfq07 was further explored as an effective 4, 6-pyrimidine anticancer agent for the treatment of human NSCLC.
Collapse
|
9
|
Structure-based design of small-molecule protein–protein interaction modulators: the story so far. Future Med Chem 2014; 6:343-57. [DOI: 10.4155/fmc.13.204] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
As the pivotal role of protein–protein interactions in cell growth, transcriptional activity, intracellular trafficking, signal transduction and pathological conditions has been assessed, experimental and in silico strategies have been developed to design protein–protein interaction modulators. State-of-the-art structure-based design methods, mainly pharmacophore modeling and docking, which have succeeded in the identification of enzyme inhibitors, receptor agonists and antagonists, and new tools specifically conceived to target protein–protein interfaces (e.g., hot-spot and druggable pocket prediction methods) have been applied in the search for small-molecule protein–protein interaction modulators. Many successful applications of structure-based design approaches that, despite the challenge of targeting protein–protein interfaces with small molecules, have led to the identification of micromolar and submicromolar hits are reviewed here.
Collapse
|
10
|
Di Pizio A, Laghezza A, Tortorella P, Agamennone M. Probing the S1' site for the identification of non-zinc-binding MMP-2 inhibitors. ChemMedChem 2013; 8:1475-82, 1421. [PMID: 23873724 DOI: 10.1002/cmdc.201300186] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 07/01/2013] [Indexed: 11/05/2022]
Abstract
Matrix metalloproteinases (MMPs) are zinc-dependent enzymes involved in several pathological states. Among them, MMP-2 is a relevant therapeutic target because of its role in cancer development and progression. Many MMP inhibitors (MMPIs) have been discovered over the last 30 years, and the majority of them contain a functional group that binds the zinc ion (zinc-binding group; ZBG). Unfortunately, no MMPIs have reached the market yet, owing to toxic effects due to unselective interactions of the ZBG. The new generation of MMPIs that do not bind the zinc ion could overcome problems of selectivity and toxicity, but have so far been developed only for MMP-8, -12, and -13. In this work, a virtual screening protocol was established by combining ligand- and structure-based methods to identify non-zinc-binding MMP-2 inhibitors using a new-generation MMP-8 inhibitor as a probe to find unexplored interactions in the MMP-2 S1' site. The screening allowed the identification of micromolar MMP-2 inhibitors that putatively avoid binding the zinc ion, as demonstrated by docking calculations. The LIA model, built to correlate predicted and experimental binding energies of the identified non-zinc-binding MMP-2 hits, underpins the reliability of the predicted docking poses.
Collapse
Affiliation(s)
- Antonella Di Pizio
- Dipartimento di Farmacia, Università "G. d'Annunzio" Chieti, Via dei Vestini 31, 66013 Chieti, Italy
| | | | | | | |
Collapse
|
11
|
Peptide Scaffolds: Flexible Molecular Structures With Diverse Therapeutic Potentials. Int J Pept Res Ther 2012. [DOI: 10.1007/s10989-011-9286-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
12
|
Sanders MPA, McGuire R, Roumen L, de Esch IJP, de Vlieg J, Klomp JPG, de Graaf C. From the protein's perspective: the benefits and challenges of protein structure-based pharmacophore modeling. MEDCHEMCOMM 2012. [DOI: 10.1039/c1md00210d] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Protein structure-based pharmacophore (SBP) models derive the molecular features a ligand must contain to be biologically active by conversion of protein properties to reciprocal ligand space. SBPs improve molecular understanding of ligand–protein interactions and can be used as valuable tools for hit and lead optimization, compound library design, and target hopping.
Collapse
Affiliation(s)
- Marijn P. A. Sanders
- Computational Drug Discovery Group
- CMBI
- Radboud University Nijmegen
- Nijmegen
- The Netherlands
| | | | - Luc Roumen
- Division of Medicinal Chemistry
- LACDR
- VU University Amsterdam
- Amsterdam
- The Netherlands
| | - Iwan J. P. de Esch
- Division of Medicinal Chemistry
- LACDR
- VU University Amsterdam
- Amsterdam
- The Netherlands
| | - Jacob de Vlieg
- Computational Drug Discovery Group
- CMBI
- Radboud University Nijmegen
- Nijmegen
- The Netherlands
| | | | - Chris de Graaf
- Division of Medicinal Chemistry
- LACDR
- VU University Amsterdam
- Amsterdam
- The Netherlands
| |
Collapse
|
13
|
Liu X, Jiang H, Li H. SHAFTS: A Hybrid Approach for 3D Molecular Similarity Calculation. 1. Method and Assessment of Virtual Screening. J Chem Inf Model 2011; 51:2372-85. [DOI: 10.1021/ci200060s] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Xiaofeng Liu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
- Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hualiang Jiang
- Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Honglin Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| |
Collapse
|
14
|
Broccatelli F, Carosati E, Neri A, Frosini M, Goracci L, Oprea TI, Cruciani G. A novel approach for predicting P-glycoprotein (ABCB1) inhibition using molecular interaction fields. J Med Chem 2011; 54:1740-51. [PMID: 21341745 DOI: 10.1021/jm101421d] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
P-glycoprotein (Pgp or ABCB1) is an ABC transporter protein involved in intestinal absorption, drug metabolism, and brain penetration, and its inhibition can seriously alter a drug's bioavailability and safety. In addition, inhibitors of Pgp can be used to overcome multidrug resistance. Given this dual purpose, reliable in silico procedures to predict Pgp inhibition are of great interest. A large and accurate literature collection yielded more than 1200 structures; a model was then constructed using various molecular interaction field-based technologies, considering pharmacophoric features and those physicochemical properties related to membrane partitioning. High accuracy was demonstrated internally with two different validation sets and, moreover, using a number of molecules, for which Pgp inhibition was not experimentally available but was evaluated in-house. All of the validations confirmed the robustness of the model and its suitability to help medicinal chemists in drug discovery. The information derived from the model was rationalized as a pharmacophore for competitive Pgp inhibition.
Collapse
Affiliation(s)
- Fabio Broccatelli
- Laboratory of Chemometrics, Department of Chemistry, University of Perugia, Via Elce di Sotto 10, I-06123 Perugia, Italy
| | | | | | | | | | | | | |
Collapse
|
15
|
Sarkisov PD, Butusov OB, Meshalkin VP. Computer-aided tools for molecular systems engineering and wavelet-morphometric analysis of the texture of nanomaterials. THEORETICAL FOUNDATIONS OF CHEMICAL ENGINEERING 2011. [DOI: 10.1134/s004057951101009x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
16
|
Nasonov AF. Computational methods and software in computer-aided combinatorial library design. RUSS J GEN CHEM+ 2011. [DOI: 10.1134/s1070363210120248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
17
|
Cross S, Baroni M, Carosati E, Benedetti P, Clementi S. FLAP: GRID molecular interaction fields in virtual screening. validation using the DUD data set. J Chem Inf Model 2010; 50:1442-50. [PMID: 20690627 DOI: 10.1021/ci100221g] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The performance of FLAP (Fingerprints for Ligands and Proteins) in virtual screening is assessed using a subset of the DUD (Directory of Useful Decoys) benchmarking data set containing 13 targets each with more than 15 different chemotype classes. A variety of ligand and receptor-based virtual screening approaches are examined, using combinations of individual templates 2D structures of known actives, a cocrystallized ligand, a receptor structure, or a cocrystallized ligand-biased receptor structure. We examine several data fusion approaches to combine the results of the individual virtual screens. In doing so, we show that excellent chemotype enrichment is achieved in both single target ligand-based and receptor-based approaches, of approximately 17-fold over random on average at a false positive rate of 1%. We also show that using as much starting knowledge as possible improves chemotype enrichment, and that data fusion using Pareto ranking is an effective method to do this giving up to 50% improvement in enrichment over the single methods. Finally we show that if inactivity or decoy data is incorporated, automatically training the scoring function in FLAP improves recovery still further, with almost 2-fold improvement over the enrichments shown by the single methods. The results clearly demonstrate the utility of FLAP for virtual screening when either a limited or wide range of prior knowledge is available.
Collapse
Affiliation(s)
- Simon Cross
- Molecular Discovery Limited, 215 Marsh Road, Pinner, Middlesex, London HA5 5NE, United Kingdom.
| | | | | | | | | |
Collapse
|
18
|
Brincat JP, Carosati E, Sabatini S, Manfroni G, Fravolini A, Raygada JL, Patel D, Kaatz GW, Cruciani G. Discovery of novel inhibitors of the NorA multidrug transporter of Staphylococcus aureus. J Med Chem 2010; 54:354-65. [PMID: 21141825 DOI: 10.1021/jm1011963] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Four novel inhibitors of the NorA efflux pump of Staphylococcus aureus, discovered through a virtual screening process, are reported. The four compounds belong to different chemical classes and were tested for their in vitro ability to block the efflux of a well-known NorA substrate, as well as for their ability to potentiate the effect of ciprofloxacin (CPX) on several strains of S. aureus, including a NorA overexpressing strain. Additionally, the MIC values of each of the compounds individually are reported. A structure-activity relationship study was also performed on these novel chemotypes, revealing three new compounds that are also potent NorA inhibitors. The virtual screening procedure employed FLAP, a new methodology based on GRID force field descriptors.
Collapse
|
19
|
Abstract
3D ligand-based similarity approaches are widely used in the early phases of drug discovery for tasks such as hit finding by virtual screening or compound design with quantitative structure–activity relationships. Here in we review widely used software for performing such tasks. Some techniques are based on relatively mature technology, shape-based similarity for instance. Typically, these methods remained in the realm of the expert user, the experienced modeler. However, advances in implementation and speed have improved usability and allow these methods to be applied to databases comprising millions of compounds. There are now many reports of such methods impacting drug-discovery projects. As such, the medicinal chemistry community has become the intended market for some of these new tools, yet they may consider the wide array and choice of approaches somewhat disconcerting. Each method has subtle differences and is better suited to certain tasks than others. In this article we review some of the widely used computational methods via application, provide straightforward background on the underlying theory and provide examples for the interested reader to pursue in more detail. In the new era of preclinical drug discovery there will be ever more pressure to move faster and more efficiently, and computational approaches based on 3D ligand similarity will play an increasing role in in this process.
Collapse
|
20
|
Lead generation and optimization based on protein-ligand complementarity. Molecules 2010; 15:4382-400. [PMID: 20657448 PMCID: PMC6264460 DOI: 10.3390/molecules15064382] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 06/07/2010] [Indexed: 01/16/2023] Open
Abstract
This work proposes a computational procedure for structure-based lead generation and optimization, which relies on the complementarity of the protein-ligand interactions. This procedure takes as input the known structure of a protein-ligand complex. Retaining the positions of the ligand heavy atoms in the protein binding site it designs structurally similar compounds considering all possible combinations of atomic species (N, C, O, CH3, NH, etc). Compounds are ranked based on a score which incorporates energetic contributions evaluated using molecular mechanics force fields. This procedure was used to design new inhibitor molecules for three serine/threonine protein kinases (p38 MAP kinase, p42 MAP kinase (ERK2), and c-Jun N-terminal kinase 3 (JNK3)). For each enzyme, the calculations produce a set of potential inhibitors whose scores are in agreement with IC50 data and Ki values. Furthermore, the native ligands for each protein target, scored within the five top-ranking compounds predicted by our method, one of the top-ranking compounds predicted to inhibit JNK3 was synthesized and his inhibitory activity confirmed against ATP hydrolysis. Our computational procedure is therefore deemed to be a useful tool for generating chemically diverse molecules active against known target proteins.
Collapse
|
21
|
A similarity search using molecular topological graphs. J Biomed Biotechnol 2009; 2009:231780. [PMID: 20037730 PMCID: PMC2796334 DOI: 10.1155/2009/231780] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2009] [Revised: 07/26/2009] [Accepted: 09/19/2009] [Indexed: 11/22/2022] Open
Abstract
A molecular similarity measure has been developed using molecular topological graphs and atomic partial charges. Two kinds of topological graphs were used. One is the ordinary adjacency matrix and the other is a matrix which represents the minimum path length between two atoms of the molecule. The ordinary adjacency matrix is suitable to compare the local structures of molecules such as functional groups, and the other matrix is suitable to compare the global structures of molecules. The combination of these two matrices gave a similarity measure. This method was applied to in silico drug screening, and the results showed that it was effective as a similarity measure.
Collapse
|
22
|
Jahn A, Hinselmann G, Fechner N, Zell A. Optimal assignment methods for ligand-based virtual screening. J Cheminform 2009; 1:14. [PMID: 20150995 PMCID: PMC2820492 DOI: 10.1186/1758-2946-1-14] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Accepted: 08/25/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ligand-based virtual screening experiments are an important task in the early drug discovery stage. An ambitious aim in each experiment is to disclose active structures based on new scaffolds. To perform these "scaffold-hoppings" for individual problems and targets, a plethora of different similarity methods based on diverse techniques were published in the last years. The optimal assignment approach on molecular graphs, a successful method in the field of quantitative structure-activity relationships, has not been tested as a ligand-based virtual screening method so far. RESULTS We evaluated two already published and two new optimal assignment methods on various data sets. To emphasize the "scaffold-hopping" ability, we used the information of chemotype clustering analyses in our evaluation metrics. Comparisons with literature results show an improved early recognition performance and comparable results over the complete data set. A new method based on two different assignment steps shows an increased "scaffold-hopping" behavior together with a good early recognition performance. CONCLUSION The presented methods show a good combination of chemotype discovery and enrichment of active structures. Additionally, the optimal assignment on molecular graphs has the advantage to investigate and interpret the mappings, allowing precise modifications of internal parameters of the similarity measure for specific targets. All methods have low computation times which make them applicable to screen large data sets.
Collapse
Affiliation(s)
- Andreas Jahn
- University of Tübingen, Center for Bioinformatics Tübingen (ZBIT), Sand 1, 72076 Tübingen, Germany
| | | | | | | |
Collapse
|
23
|
Fontaine F, Cross S, Plasencia G, Pastor M, Zamora I. SHOP: A Method For Structure-Based Fragment and Scaffold Hopping. ChemMedChem 2009; 4:427-39. [DOI: 10.1002/cmdc.200800355] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
24
|
Cheeseright TJ, Mackey MD, Melville JL, Vinter JG. FieldScreen: Virtual Screening Using Molecular Fields. Application to the DUD Data Set. J Chem Inf Model 2008; 48:2108-17. [DOI: 10.1021/ci800110p] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Timothy J. Cheeseright
- Cresset BioMolecular Discovery Ltd., BioPark Hertfordshire, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, United Kingdom
| | - Mark D. Mackey
- Cresset BioMolecular Discovery Ltd., BioPark Hertfordshire, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, United Kingdom
| | - James L. Melville
- Cresset BioMolecular Discovery Ltd., BioPark Hertfordshire, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, United Kingdom
| | - Jeremy G. Vinter
- Cresset BioMolecular Discovery Ltd., BioPark Hertfordshire, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, United Kingdom
| |
Collapse
|
25
|
Carosati E, Budriesi R, Ioan P, Ugenti MP, Frosini M, Fusi F, Corda G, Cosimelli B, Spinelli D, Chiarini A, Cruciani G. Discovery of Novel and Cardioselective Diltiazem-like Calcium Channel Blockers via Virtual Screening. J Med Chem 2008; 51:5552-65. [DOI: 10.1021/jm800151n] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Emanuele Carosati
- Dipartimento di Chimica, Università degli Studi di Perugia, Via Elce di Sotto 10, 06123 Perugia, Italia, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Bologna, Via Belmeloro 6, 40126 Bologna, Italia, Dipartimento di Scienze Biomediche, Sezione di Farmacologia, Fisiologia e Tossicologia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italia, Dipartimento di Chimica Farmaceutica e Tossicologica, Università degli Studi di Napoli “Federico II”, Via Montesano 49, 80131 Napoli,
| | - Roberta Budriesi
- Dipartimento di Chimica, Università degli Studi di Perugia, Via Elce di Sotto 10, 06123 Perugia, Italia, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Bologna, Via Belmeloro 6, 40126 Bologna, Italia, Dipartimento di Scienze Biomediche, Sezione di Farmacologia, Fisiologia e Tossicologia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italia, Dipartimento di Chimica Farmaceutica e Tossicologica, Università degli Studi di Napoli “Federico II”, Via Montesano 49, 80131 Napoli,
| | - Pierfranco Ioan
- Dipartimento di Chimica, Università degli Studi di Perugia, Via Elce di Sotto 10, 06123 Perugia, Italia, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Bologna, Via Belmeloro 6, 40126 Bologna, Italia, Dipartimento di Scienze Biomediche, Sezione di Farmacologia, Fisiologia e Tossicologia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italia, Dipartimento di Chimica Farmaceutica e Tossicologica, Università degli Studi di Napoli “Federico II”, Via Montesano 49, 80131 Napoli,
| | - Maria P. Ugenti
- Dipartimento di Chimica, Università degli Studi di Perugia, Via Elce di Sotto 10, 06123 Perugia, Italia, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Bologna, Via Belmeloro 6, 40126 Bologna, Italia, Dipartimento di Scienze Biomediche, Sezione di Farmacologia, Fisiologia e Tossicologia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italia, Dipartimento di Chimica Farmaceutica e Tossicologica, Università degli Studi di Napoli “Federico II”, Via Montesano 49, 80131 Napoli,
| | - Maria Frosini
- Dipartimento di Chimica, Università degli Studi di Perugia, Via Elce di Sotto 10, 06123 Perugia, Italia, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Bologna, Via Belmeloro 6, 40126 Bologna, Italia, Dipartimento di Scienze Biomediche, Sezione di Farmacologia, Fisiologia e Tossicologia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italia, Dipartimento di Chimica Farmaceutica e Tossicologica, Università degli Studi di Napoli “Federico II”, Via Montesano 49, 80131 Napoli,
| | - Fabio Fusi
- Dipartimento di Chimica, Università degli Studi di Perugia, Via Elce di Sotto 10, 06123 Perugia, Italia, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Bologna, Via Belmeloro 6, 40126 Bologna, Italia, Dipartimento di Scienze Biomediche, Sezione di Farmacologia, Fisiologia e Tossicologia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italia, Dipartimento di Chimica Farmaceutica e Tossicologica, Università degli Studi di Napoli “Federico II”, Via Montesano 49, 80131 Napoli,
| | - Gaetano Corda
- Dipartimento di Chimica, Università degli Studi di Perugia, Via Elce di Sotto 10, 06123 Perugia, Italia, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Bologna, Via Belmeloro 6, 40126 Bologna, Italia, Dipartimento di Scienze Biomediche, Sezione di Farmacologia, Fisiologia e Tossicologia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italia, Dipartimento di Chimica Farmaceutica e Tossicologica, Università degli Studi di Napoli “Federico II”, Via Montesano 49, 80131 Napoli,
| | - Barbara Cosimelli
- Dipartimento di Chimica, Università degli Studi di Perugia, Via Elce di Sotto 10, 06123 Perugia, Italia, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Bologna, Via Belmeloro 6, 40126 Bologna, Italia, Dipartimento di Scienze Biomediche, Sezione di Farmacologia, Fisiologia e Tossicologia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italia, Dipartimento di Chimica Farmaceutica e Tossicologica, Università degli Studi di Napoli “Federico II”, Via Montesano 49, 80131 Napoli,
| | - Domenico Spinelli
- Dipartimento di Chimica, Università degli Studi di Perugia, Via Elce di Sotto 10, 06123 Perugia, Italia, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Bologna, Via Belmeloro 6, 40126 Bologna, Italia, Dipartimento di Scienze Biomediche, Sezione di Farmacologia, Fisiologia e Tossicologia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italia, Dipartimento di Chimica Farmaceutica e Tossicologica, Università degli Studi di Napoli “Federico II”, Via Montesano 49, 80131 Napoli,
| | - Alberto Chiarini
- Dipartimento di Chimica, Università degli Studi di Perugia, Via Elce di Sotto 10, 06123 Perugia, Italia, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Bologna, Via Belmeloro 6, 40126 Bologna, Italia, Dipartimento di Scienze Biomediche, Sezione di Farmacologia, Fisiologia e Tossicologia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italia, Dipartimento di Chimica Farmaceutica e Tossicologica, Università degli Studi di Napoli “Federico II”, Via Montesano 49, 80131 Napoli,
| | - Gabriele Cruciani
- Dipartimento di Chimica, Università degli Studi di Perugia, Via Elce di Sotto 10, 06123 Perugia, Italia, Dipartimento di Scienze Farmaceutiche, Università degli Studi di Bologna, Via Belmeloro 6, 40126 Bologna, Italia, Dipartimento di Scienze Biomediche, Sezione di Farmacologia, Fisiologia e Tossicologia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italia, Dipartimento di Chimica Farmaceutica e Tossicologica, Università degli Studi di Napoli “Federico II”, Via Montesano 49, 80131 Napoli,
| |
Collapse
|
26
|
Barillari C, Marcou G, Rognan D. Hot-spots-guided receptor-based pharmacophores (HS-Pharm): a knowledge-based approach to identify ligand-anchoring atoms in protein cavities and prioritize structure-based pharmacophores. J Chem Inf Model 2008; 48:1396-410. [PMID: 18570371 DOI: 10.1021/ci800064z] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The design of biologically active compounds from ligand-free protein structures using a structure-based approach is still a major challenge. In this paper, we present a fast knowledge-based approach (HS-Pharm) that allows the prioritization of cavity atoms that should be targeted for ligand binding, by training machine learning algorithms with atom-based fingerprints of known ligand-binding pockets. The knowledge of hot spots for ligand binding is here used for focusing structure-based pharmacophore models. Three targets of pharmacological interest (neuraminidase, beta2 adrenergic receptor, and cyclooxygenase-2) were used to test the evaluated methodology, and the derived structure-based pharmacophores were used in retrospective virtual screening studies. The current study shows that structure-based pharmacophore screening is a powerful technique for the fast identification of potential hits in a chemical library, and that it is a valid alternative to virtual screening by molecular docking.
Collapse
Affiliation(s)
- Caterina Barillari
- Bioinformatics of the Drug, UMR 7175 CNRS-ULP (Universite Louis Pasteur-Strasbourg I), 74 route du Rhin, B.P. 24, F-67400 Illkirch, France
| | | | | |
Collapse
|
27
|
Abstract
The advent of therapeutic strategies aimed at targeting specific macromolecular components of deregulated signaling pathways associated with particular disease states has given rise to the idea that it should be possible to design ligands as drug candidates to these targets from first principles. This concept has been beckoning for a long time but structure-based ligand design only became feasible once it was possible to determine the 3-D structures of molecular targets at atomic resolution. However, structure-based design turned out to be difficult, chiefly because under physiological conditions both receptors and ligands are not static but they behave dynamically. While it is possible to design ligands with high steric and electronic complementarity to a receptor site, it is always uncertain how biologically relevant the assumed conformations of both ligand and receptor actually are. The fact that it remains beyond our current abilities to predict with sufficient accuracy the affinity between hypothetical ligand and receptor poses is in part connected with this problem and continues to confound the reliable prediction of drug-like ligands for therapeutic targets. Nevertheless, significant progress has been made and so-called virtual screening methods that use computational methods to dock candidate ligands into receptor sites and to score the resulting complexes are now used routinely as one of the components in drug discovery screening campaigns. Here an overview is given of the underlying principles, implementations, and applications of structure-guided computational design technologies. Although the emphasis is on receptor-based strategies, mention will also be made of some of the more established ligand-based approaches, such as similarity analyses and quantitative structure-activity relationship methods.
Collapse
Affiliation(s)
- Peter M Fischer
- Centre for Biomolecular Sciences and School of Pharmacy, University of Nottingham, University Park, Nottingham, UK.
| |
Collapse
|
28
|
Venhorst J, Núñez S, Terpstra JW, Kruse CG. Assessment of Scaffold Hopping Efficiency by Use of Molecular Interaction Fingerprints. J Med Chem 2008; 51:3222-9. [PMID: 18447336 DOI: 10.1021/jm8001058] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jennifer Venhorst
- Solvay Pharmaceuticals, Research Laboratories, C. J. van Houtenlaan 36, 1381 CP Weesp, The Netherlands
| | - Sara Núñez
- Solvay Pharmaceuticals, Research Laboratories, C. J. van Houtenlaan 36, 1381 CP Weesp, The Netherlands
| | - Jan Willem Terpstra
- Solvay Pharmaceuticals, Research Laboratories, C. J. van Houtenlaan 36, 1381 CP Weesp, The Netherlands
| | - Chris G. Kruse
- Solvay Pharmaceuticals, Research Laboratories, C. J. van Houtenlaan 36, 1381 CP Weesp, The Netherlands
| |
Collapse
|
29
|
ParaFrag--an approach for surface-based similarity comparison of molecular fragments. J Mol Model 2008; 14:547-58. [PMID: 18449578 DOI: 10.1007/s00894-008-0302-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Accepted: 03/12/2008] [Indexed: 10/22/2022]
Abstract
A frequent task in computer-aided drug design is to identify novel chemotypes similar in activity but structurally different to a given reference structure. Here we report the development of a novel method for atom-independent similarity comparison of molecular fragments (substructures of drug-like molecules). The fragments are characterized by their local surface properties coded in the form of 3D pharmacophores. As surface properties, we used the electrostatic potential (MEP), the local ionization energy (IE(L)), local electron affinity (EA(L)) and local polarizability (POL) calculated on isodensity surfaces. A molecular fragment can then be represented by a minimal set of extremes for each surface property. We defined a tolerance sphere for each of these extremes, thus allowing us to assess the similarity of fragments in an analogous manner to classical pharmacophore comparison. As a first application of this method we focused on comparing rigid fragments suitable for scaffold hopping. A retrospective analysis of successful scaffold hopping reported for Factor Xa inhibitors [Wood MR et al (2006) J Med Chem 49:1231] showed that our method performs well where atom-based similarity metrics fail.
Collapse
|
30
|
Gillet VJ. New directions in library design and analysis. Curr Opin Chem Biol 2008; 12:372-8. [PMID: 18331851 DOI: 10.1016/j.cbpa.2008.02.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Accepted: 02/06/2008] [Indexed: 10/22/2022]
Abstract
The high costs associated with high-throughput screening (HTS) coupled with the limited coverage and bias of current screening collections is such that diversity analysis continues to be an important criterion in lead generation. Whereas early approaches to diversity analysis were based on traditional descriptors such as two-dimensional fingerprints a recent emphasis has been on assessing scaffold coverage to ensure that a variety of different chemotypes are represented. Moreover, whether designing diverse or focused libraries, it is widely recognised that designs should aim to achieve a balance in a number of different properties and multiobjective optimisation provides an effective way of achieving such designs.
Collapse
Affiliation(s)
- Valerie J Gillet
- Department of Information Studies, University of Sheffield, Sheffield, UK.
| |
Collapse
|
31
|
Ahlström MM, Zamora I. Characterization of Type II Ligands in CYP2C9 and CYP3A4. J Med Chem 2008; 51:1755-63. [DOI: 10.1021/jm701121y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Marie M. Ahlström
- Discovery DMPK and Bioanalytical Chemistry, AstraZeneca R&D Mölndal, SE-431 83 Mölndal, Sweden, Department of Chemistry, Medicinal Chemistry, Göteborg University, SE-412 96 Gothenburg, Sweden, Lead Molecular Design, S.L., Vallés 96-102 (27) E-08190, Sant Cugat del Vallés, Spain, and Institut Municipal d’Investigació Medica (IMIM), Universitat Pompeu Fabra, Doctor Aiguader 80, 08003 Barcelona, Spain
| | - Ismael Zamora
- Discovery DMPK and Bioanalytical Chemistry, AstraZeneca R&D Mölndal, SE-431 83 Mölndal, Sweden, Department of Chemistry, Medicinal Chemistry, Göteborg University, SE-412 96 Gothenburg, Sweden, Lead Molecular Design, S.L., Vallés 96-102 (27) E-08190, Sant Cugat del Vallés, Spain, and Institut Municipal d’Investigació Medica (IMIM), Universitat Pompeu Fabra, Doctor Aiguader 80, 08003 Barcelona, Spain
| |
Collapse
|
32
|
Murali S, Hojo S, Tsujishita H, Nakamura H, Fukunishi Y. In-silico drug screening method based on the protein–compound affinity matrix using the factor selection technique. Eur J Med Chem 2007; 42:966-76. [PMID: 17307278 DOI: 10.1016/j.ejmech.2006.12.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Revised: 12/14/2006] [Accepted: 12/21/2006] [Indexed: 11/25/2022]
Abstract
We have developed a new in-silico drug screening method, a modified version of a docking score index (DSI) method, based on a protein-compound docking affinity matrix. By using this method, the docking scores are converted to the docking score indexes by the principal component analysis (PCA) method and each compound is projected into a PCA space. In this study, we propose a method to select a set of suitable principal component axes and evaluate the database enrichment for 12 target proteins. This method selects the new active compounds or hits, which are close to the known active compounds, thereby enhancing the database enrichment.
Collapse
Affiliation(s)
- Sukumaran Murali
- Japan Biological Information Research Center, Japan Biological Informatics Consortium, 2-41-6, Aomi, Koto-ku, Tokyo 135-0064, Japan
| | | | | | | | | |
Collapse
|
33
|
Bergmann R, Linusson A, Zamora I. SHOP: scaffold HOPping by GRID-based similarity searches. J Med Chem 2007; 50:2708-17. [PMID: 17489578 DOI: 10.1021/jm061259g] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A new GRID-based method for scaffold hopping (SHOP) is presented. In a fully automatic manner, scaffolds were identified in a database based on three types of 3D-descriptors. SHOP's ability to recover scaffolds was assessed and validated by searching a database spiked with fragments of known ligands of three different protein targets relevant for drug discovery using a rational approach based on statistical experimental design. Five out of eight and seven out of eight thrombin scaffolds and all seven HIV protease scaffolds were recovered within the top 10 and 31 out of 31 neuraminidase scaffolds were in the 31 top-ranked scaffolds. SHOP also identified new scaffolds with substantially different chemotypes from the queries. Docking analysis indicated that the new scaffolds would have similar binding modes to those of the respective query scaffolds observed in X-ray structures. The databases contained scaffolds from published combinatorial libraries to ensure that identified scaffolds could be feasibly synthesized.
Collapse
Affiliation(s)
- Rikke Bergmann
- Department of Medicinal Chemistry, University of Copenhagen, Faculty of Pharmaceutical Sciences, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark.
| | | | | |
Collapse
|
34
|
Hawkins PCD, Skillman AG, Nicholls A. Comparison of shape-matching and docking as virtual screening tools. J Med Chem 2007; 50:74-82. [PMID: 17201411 DOI: 10.1021/jm0603365] [Citation(s) in RCA: 707] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ligand docking is a widely used approach in virtual screening. In recent years a large number of publications have appeared in which docking tools are compared and evaluated for their effectiveness in virtual screening against a wide variety of protein targets. These studies have shown that the effectiveness of docking in virtual screening is highly variable due to a large number of possible confounding factors. Another class of method that has shown promise in virtual screening is the shape-based, ligand-centric approach. Several direct comparisons of docking with the shape-based tool ROCS have been conducted using data sets from some of these recent docking publications. The results show that a shape-based, ligand-centric approach is more consistent than, and often superior to, the protein-centric approach taken by docking.
Collapse
|
35
|
Bender A, Jenkins JL, Li Q, Adams SE, Cannon EO, Glen RC. Chapter 9 Molecular Similarity: Advances in Methods, Applications and Validations in Virtual Screening and QSAR. ACTA ACUST UNITED AC 2006; 2:141-168. [PMID: 32362803 PMCID: PMC7185533 DOI: 10.1016/s1574-1400(06)02009-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
This chapter discusses recent developments in some of the areas that exploit the molecular similarity principle, novel approaches to capture molecular properties by the use of novel descriptors, focuses on a crucial aspect of computational models-their validity, and discusses additional ways to examine data available, such as those from high-throughput screening (HTS) campaigns and to gain more knowledge from this data. The chapter also presents some of the recent applications of methods discussed focusing on the successes of virtual screening applications, database clustering and comparisons (such as drug- and in-house-likeness), and the recent large-scale validations of docking and scoring programs. While a great number of descriptors and modeling methods has been proposed until today, the recent trend toward proper model validation is very much appreciated. Although some of their limitations are surely because of underlying principles and limitations of fundamental concepts, others will certainly be eliminated in the future.
Collapse
Affiliation(s)
- Andreas Bender
- Unilever Centre for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.,Lead Discovery Center, Novartis Institutes for BioMedical Research Inc., 250 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Jeremy L Jenkins
- Lead Discovery Center, Novartis Institutes for BioMedical Research Inc., 250 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Qingliang Li
- College of Chemistry and Molecular Engineering, Center for Theoretical Biology, Peking University, Beijing 100871, China
| | - Sam E Adams
- Unilever Centre for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Edward O Cannon
- Unilever Centre for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Robert C Glen
- Unilever Centre for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| |
Collapse
|
36
|
Bohl M, Loeprecht B, Wendt B, Heritage T, Richmond NJ, Willett P. Unsupervised 3D Ring Template Searching as an Ideas Generator for Scaffold Hopping: Use of the LAMDA, RigFit, and Field-Based Similarity Search (FBSS) Methods. J Chem Inf Model 2006; 46:1882-90. [PMID: 16995717 DOI: 10.1021/ci049657k] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Crystal structures taken from the Cambridge Structural Database were used to build a ring scaffold database containing 19 050 3D structures, with each such scaffold then being used to generate a centroid connecting path (CCP) representation. The CCP is a novel object that connects ring centroids, ring linker atoms, and other important points on the connection path between ring centroids. Unsupervised searching in the scaffold and CCP data sets was carried out using the atom-based LAMDA and RigFit search methods and the field-based similarity search method. The performance of these methods was tested with three different ring scaffold queries. These searches demonstrated that unsupervised 3D scaffold searching methods can find not only the types of ring systems that might be retrieved in carefully defined pharmacophore searches (supervised approach) but also additional, structurally diverse ring systems that could form the starting point for lead discovery programs or other scaffold-hopping applications. Not only are the methods effective but some are sufficiently rapid to permit scaffold searching in large chemical databases on a routine basis.
Collapse
Affiliation(s)
- Martin Bohl
- Tripos GmbH, Martin-Kollar-Strasse 17, D-81829 Munich, Germany.
| | | | | | | | | | | |
Collapse
|