Identification of Novel Adenosine A2A Receptor Antagonists by Virtual Screening
- Christopher J. Langmead
- ,
- Stephen P. Andrews
- ,
- Miles Congreve
- ,
- James C. Errey
- ,
- Edward Hurrell
- ,
- Fiona H. Marshall
- ,
- Jonathan S. Mason
- ,
- Christine M. Richardson
- ,
- Nathan Robertson
- ,
- Andrei Zhukov
- , and
- Malcolm Weir
Abstract

Virtual screening was performed against experimentally enabled homology models of the adenosine A2A receptor, identifying a diverse range of ligand efficient antagonists (hit rate 9%). By use of ligand docking and Biophysical Mapping (BPM), hits 1 and 5 were optimized to potent and selective lead molecules (11–13 from 5, pKI = 7.5–8.5, 13- to >100-fold selective versus adenosine A1; 14–16 from 1, pKI = 7.9–9.0, 19- to 59-fold selective).
Introduction
Results and Discussion
Virtual Screening
hit | pKI | LE | LLE | clogP | PSA | MW |
---|---|---|---|---|---|---|
1 | 8.46 | 0.52 | 5.4 | 3.1 | 84.9 | 310.4 |
2 | 5.15 | 0.47 | 4.5 | 0.7 | 72.2 | 222.3 |
3 | 5.75 | 0.44 | 3.9 | 1.9 | 61.7 | 264.3 |
4 | 6.15 | 0.36 | 3.2 | 3.0 | 66.6 | 327.4 |
5 | 5.65 | 0.33 | 3.7 | 1.9 | 85.7 | 331.3 |
6 | 5.62 | 0.31 | 2.6 | 3.0 | 76.7 | 367.9 |
7 | 5.91 | 0.30 | 3.2 | 2.7 | 78.4 | 367.4 |
8 | 5.33 | 0.29 | 3.4 | 1.9 | 79.8 | 340.4 |
9 | 5.70 | 0.29 | 3.9 | 1.8 | 80.1 | 363.4 |
10 | 5.53 | 0.27 | 2.1 | 3.4 | 95.9 | 382.4 |
Figure 1

Figure 1. Structures of virtual screening hits.
Hits to Leads
Scheme 1

Figure 2

Figure 2. Docking of the chromone 12, showing the BPM fingerprint color coded onto the binding site residues and in graphical form as change in pKD. Nonbinding is shown in red (N253A, H250A). Next largest effect is in dark orange (L85A), second largest in amber (N181A, Y271A, I66A), an increase in binding in green (S277A). H-bonding between the nitrogen of the thiazole and the aromatic C–H of the chromone is predicted to Asn2536.55. Selected BPM data are tabulated showing the change in pKD of each binding site mutation.
Scheme 2

Figure 3

Figure 3. Docking of the triazine 15, showing the BPM fingerprint color coded onto the binding site residues and in graphical form as change in pKD. Nonbinding is shown in red (N253A, H250A). Next largest effect is in dark orange (L85A, S277A). H-bonding between the nitrogen of the triazine and the phenol is predicted to Asn2536.55. The polar piperazine substituent is proposed to reach into the region of the binding site occupied by ribose in the natural agonist ligand adenosine and may be the driver of selectivity versus the A1 receptor, as this region of the binding site contains some amino acid differences comparing the two receptors. (14) Selected BPM data are tabulated showing the change in pKD of each binding site mutation.
Conclusion
Experimental Protocols
Virtual Screening Compounds
Computational Chemistry
Adenosine Receptor Assays
Chemical Synthesis
Scheme 3

Scheme aReagents and conditions: (a) THF, iPr2EtN, NH3. (b) For 16: (i) 3-(4-methoxypiperidin-1-yl)phenylboronic acid, Na2CO3, 1,4-dioxane/H2O, Pd(PPh3)4, 90 °C, then (ii) 2-hydroxylphenylboronic acid, Na2CO3, 1,4-dioxane/H2O, Pd(PPh3)4, 90 °C. (c) For 17: (i) 2-benzyloxyphenylboronic acid, Na2CO3, 1,4-dioxane/H2O, Pd(PPh3)4, 70 °C, then (ii) 3-(4-methylpiperazine-1-carbonyl)phenylboronic acid hydrochloride, Na2CO3, 1,4-dioxane/H2O, Pd(PPh3)4, 90 °C; (d) 17, EtOAc, Pd(OH)2/C, 1,4-cyclohexadiene, 140 °C (microwave).
Supporting Information
Chemical synthesis protocols, QC data and binding curves for the top 10 hits, more detailed computational methods including virtual screening workflows, and a table of calculated blood–brain barrier prediction and the closest published adenosine A2A antagonist to each of the hits. This material is available free of charge via the Internet at http://pubs.acs.org.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgment
The authors thank Bissan Al-Lazikani for help with constructing the first generation of homology models, Benjamin Tehan for assistance with computational chemistry, and Nat Monck for assisting with the triaging of screening hits.
BPM | Biophysical Mapping |
PDB | Protein Data Bank |
LE | ligand efficiency |
LLE | ligand lipophilicity efficiency |
SDM | site directed mutagenesis |
References
This article references 25 other publications.
- 1Pinna, A. Novel investigational adenosine A2A receptor antagonists for Parkinson’s disease Expert Opin. Invest. Drugs 2009, 18, 1619– 1631Google ScholarThere is no corresponding record for this reference.
- 2Blagg, J. Structural Alerts for Toxicity. In Burger’s Medicinal Chemistry, Drug Discovery and Development, 7th ed.; Abraham, D. J.; Rotella, D. P., Eds.; Wiley: Hoboken, NJ, 2010; pp 301– 344.Google ScholarThere is no corresponding record for this reference.
- 3Warne, A.; Serrano-Vega, M. J.; Baker, J. G.; Moukhametzianov, R.; Edwards, P. C.; Henderson, R.; Leslie, A. G. W.; Tate, C. G.; Schertler, G. F. X. Structure of the beta1-adrenergic G protein-coupled receptor Nature 2008, 454, 486– 491
(PDB code 2VT4)
Google ScholarThere is no corresponding record for this reference. - 4Zhukov, A.; Andrews, S. P.; Errey, J. C.; Robertson, N.; Tehan, B.; Mason, J. S.; Marshall, F. H.; Weir, M.; Congreve, M. Biophysical Mapping of the adenosine A2A receptor J. Med. Chem. 2011, 54, 4312– 4323Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXnt12nsb0%253D&md5=d7f5c6bf24f9093d823bb41db9867c09Biophysical Mapping of the Adenosine A2A ReceptorZhukov, Andrei; Andrews, Stephen P.; Errey, James C.; Robertson, Nathan; Tehan, Benjamin; Mason, Jonathan S.; Marshall, Fiona H.; Weir, Malcolm; Congreve, MilesJournal of Medicinal Chemistry (2011), 54 (13), 4312-4323CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A new approach to generating information on ligand receptor interactions within the binding pocket of G protein-coupled receptors has been developed, called Biophys. Mapping (BPM). Starting from a stabilized receptor (StaR), minimally engineered for thermostability, addnl. single mutations are then added at positions that could be involved in small mol. interactions. The StaR and a panel of binding site mutants are captured onto Biacore chips to enable characterization of the binding of small mol. ligands using surface plasmon resonance (SPR) measurement. A matrix of binding data for a set of ligands vs. each active site mutation is then generated, providing specific affinity and kinetic information (KD, kon, and koff) of receptor-ligand interactions. This data set, in combination with mol. modeling and docking, is used to map the small mol. binding site for each class of compds. Taken together, the many constraints provided by these data identify key protein-ligand interactions and allow the shape of the site to be refined to produce a high quality three-dimensional picture of ligand binding, thereby facilitating structure based drug design. Results of biophys. mapping of the adenosine A2A receptor are presented.
- 5Halgren, T. A.; Murphy, R. B.; Friesner, R. A.; Beard, H. S.; Frye, L. L.; Pollard, W. T.; Banks, J. L. Glide: a new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screening J. Med. Chem. 2004, 47, 1750– 1759Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhsFyit78%253D&md5=33d68dd968e65626b449df61e44e37beGlide: A new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screeningHalgren, Thomas A.; Murphy, Robert B.; Friesner, Richard A.; Beard, Hege S.; Frye, Leah L.; Pollard, W. Thomas; Banks, Jay L.Journal of Medicinal Chemistry (2004), 47 (7), 1750-1759CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Glide's ability to identify active compds. in a database screen is characterized by applying Glide to a diverse set of nine protein receptors. In many cases, two, or even three, protein sites are employed to probe the sensitivity of the results to the site geometry. To make the database screens as realistic as possible, the screens use sets of "druglike" decoy ligands that have been selected to be representative of what we believe is likely to be found in the compd. collection of a pharmaceutical or biotechnol. company. Results are presented for releases 1.8, 2.0, and 2.5 of Glide. The comparisons show that av. measures for both "early" and "global" enrichment for Glide 2.5 are 3 times higher than for Glide 1.8 and more than 2 times higher than for Glide 2.0 because of better results for the least well-handled screens. This improvement in enrichment stems largely from the better balance of the more widely parametrized GlideScore 2.5 function and the inclusion of terms that penalize ligand-protein interactions that violate established principles of phys. chem., particularly as it concerns the exposure to solvent of charged protein and ligand groups. Comparisons to results for the thymidine kinase and estrogen receptors published by Rognan and co-workers (J. Med. Chem. 2000, 43, 4759-4767) show that Glide 2.5 performs better than GOLD 1.1, FlexX 1.8, or DOCK 4.01.
- 6Congreve, M.; Andrews, S. P.; Dore, A. S.; Hollenstein, K.; Hurrell, E.; Langmead, C. J.; Mason, J. S.; Ng, I. W.; Tehan, B.; Zhukov, A.; Weir, M.; Marshall, F. H. Discovery of 1,2,4-triazine derivatives as adenosine A2A antagonists using structure based drug design. J. Med. Chem. [Online early access]. DOI: DOI: 10.1021/jm201376w . Published Online: Jan 5, 2012.Google ScholarThere is no corresponding record for this reference.
- 7Hopkins, A. L.; Groom, C. R.; Alex, A. Ligand efficiency: a useful metric for lead selection Drug Discovery Today 2004, 9, 430– 431Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2c3ht1ygtw%253D%253D&md5=eb19a1bca53247d0bd9adcb99d5817e6Ligand efficiency: a useful metric for lead selectionHopkins Andrew L; Groom Colin R; Alex AlexanderDrug discovery today (2004), 9 (10), 430-1 ISSN:1359-6446.There is no expanded citation for this reference.
- 8Leeson, P. D.; Springthorpe, B. The influence of drug-like concepts on decision-making in medicinal chemistry Nat. Rev. Drug Discovery 2007, 6, 881– 890Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXht1els7rL&md5=5665a9e2dc266a1ee096e20625757ef7The influence of drug-like concepts on decision-making in medicinal chemistryLeeson, Paul D.; Springthorpe, BrianNature Reviews Drug Discovery (2007), 6 (11), 881-890CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Despite the wide acceptance of drug-like principles such as the 'rule of five', this anal. of mols. currently being synthesized in leading pharmaceutical companies reveals that their phys. properties differ significantly from those of recently discovered oral drugs. The marked increase in lipophilicity in particular could increase the likelihood of attrition in drug development. The application of guidelines linked to the concept of drug-likeness, such as the 'rule of five', has gained wide acceptance as an approach to reduce attrition in drug discovery and development. However, despite this acceptance, anal. of recent trends reveals that the phys. properties of mols. that are currently being synthesized in leading drug discovery companies differ significantly from those of recently discovered oral drugs and compds. in clin. development. The consequences of the marked increase in lipophilicity - the most important drug-like phys. property - include a greater likelihood of lack of selectivity and attrition in drug development. Tackling the threat of compd.-related toxicol. attrition needs to move to the mainstream of medicinal chem. decision-making.
- 9Katritch, V.; Jaakola, V.-P.; Lane, J. R.; Lin, J.; Izerman, A. P.; Yaeger, M.; Kufarena, I.; Stevens, R. C.; Abagyan, R. Structure-based discovery of novel chemotypes for adenosine A2a receptor antagonists J. Med. Chem. 2010, 53, 1799– 1809Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXptlKqsw%253D%253D&md5=5a54df43f6edd20d83e7e5942e2f9811Structure-Based Discovery of Novel Chemotypes for Adenosine A2A Receptor AntagonistsKatritch, Vsevolod; Jaakola, Veli-Pekka; Lane, J. Robert; Lin, Judy; IJzerman, Adriaan P.; Yeager, Mark; Kufareva, Irina; Stevens, Raymond C.; Abagyan, RubenJournal of Medicinal Chemistry (2010), 53 (4), 1799-1809CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The recent progress in crystallog. of G-protein coupled receptors opens an unprecedented venue for structure-based GPCR drug discovery. To test efficiency of the structure-based approach, we performed mol. docking and virtual ligand screening (VLS) of more than 4 million com. available "drug-like" and "lead-like" compds. against the A2AAR 2.6 Å resoln. crystal structure. Out of 56 high ranking compds. tested in A2AAR binding assays, 23 showed affinities under 10 μM, 11 of those had sub-μM affinities and two compds. had affinities under 60 nM. The identified hits represent at least 9 different chem. scaffolds and are characterized by very high ligand efficiency (0.3-0.5 kcal/mol per heavy atom). Significant A2AAR antagonist activities were confirmed for 10 out of 13 ligands tested in functional assays. High success rate, novelty, and diversity of the chem. scaffolds and strong ligand efficiency of the A2AAR antagonists identified in this study suggest practical applicability of receptor-based VLS in GPCR drug discovery.
- 10Carlsson, J.; Yoo, L.; Gao, Z. G.; Irwin, J. J.; Shoichet, B. K.; Jacobson, K. A. Structure-based discovery of A2A adenosine receptor ligands J. Med. Chem. 2010, 53, 3748– 3755Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXkvFaqsL8%253D&md5=c36c941d52d2cec06387d79c4c423d46Structure-Based Discovery of A2A Adenosine Receptor LigandsCarlsson, Jens; Yoo, Lena; Gao, Zhan-Guo; Irwin, John J.; Shoichet, Brian K.; Jacobson, Kenneth A.Journal of Medicinal Chemistry (2010), 53 (9), 3748-3755CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The recent detn. of X-ray structures of pharmacol. relevant GPCRs has made these targets accessible to structure-based ligand discovery. Here we explore whether novel chemotypes may be discovered for the A2A adenosine receptor, based on complementarity to its recently detd. structure. The A2A adenosine receptor signals in the periphery and the CNS, with agonists explored as anti-inflammatory drugs and antagonists explored for neurodegenerative diseases. We used mol. docking to screen a 1.4 million compd. database against the X-ray structure computationally and tested 20 high-ranking, previously unknown mols. exptl. Of these 35% showed substantial activity with affinities between 200 nM and 9 μM. For the most potent of these new inhibitors, over 50-fold specificity was obsd. for the A2A vs. the related A1 and A3 subtypes. These high hit rates and affinities at least partly reflect the bias of com. libraries toward GPCR-like chemotypes, an issue that we attempt to investigate quant. Despite this bias, many of the most potent new ligands were novel, dissimilar from known ligands, providing new lead structures for modulation of this medically important target.
- 11Goodford, P. J. A computational procedure for determining energetically favorable binding sites on biologically important macromolecules J. Med. Chem. 1985, 28 (7) 849– 857Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2MXktVeit7c%253D&md5=03e9d22787f16c760ef0dc29ca02aa88A computational procedure for determining energetically favorable binding sites on biologically important macromoleculesGoodford, P. J.Journal of Medicinal Chemistry (1985), 28 (7), 849-57CODEN: JMCMAR; ISSN:0022-2623.The interaction of a probe group with a protein of known structure is computed at sample positions throughout and around the macromol., giving an array of energy values. The probe include H2O, the Me group, amine-N, carboxy-O, and OH. Contour surfaces at appropriate energy levels are calcd. for each probe and displayed by computer graphics together with the protein structure. Contours at neg. energy levels delineate regions of attraction between probe and protein and are found at known ligand binding clefts in particular. The contours also enable other regions of attraction to be identified and facilitate the interpretation of protein-ligand energetics. They may, therefore, be of value for drug design.
- 12Higgs, C.; Beuming, T.; Sherman, W. Hydration site thermodynamics explain SARs for triazolylpurines analogues binding to the A2A receptor ACS Med. Chem. Lett. 2010, 1, 160– 164Google ScholarThere is no corresponding record for this reference.
- 13Congreve, M.; Langmead, C. J.; Mason, J. S.; Marshall, F. H. Progress in structure based drug design for G protein-coupled receptors J. Med. Chem. 2011, 54, 4283– 4311Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXns1emtr0%253D&md5=2610591be5473dad558145b6dea84c2bProgress in Structure Based Drug Design for G Protein-Coupled ReceptorsCongreve, Miles; Langmead, Christopher J.; Mason, Jonathan S.; Marshall, Fiona H.Journal of Medicinal Chemistry (2011), 54 (13), 4283-4311CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. The field of GPCR drug discovery is set to have a new lease of life, invigorated by a quantum leap of progress in structural biol. over the past 3 years. The new era of SBDD for GPCRS should now allow even some of the most challenging targets to yield to the efforts of medicinal chemists. This very well established drug family can expect to see many more first in class agents and better mols. with improved physicochem. properties and selectivity profiles, derived from the careful, rational optimization of hits using structure based and fragment based methods. In particular, a disciplined emphasis on maintaining high ligand efficiency and ligand lipophilicity efficiency should allow the next generation of research on this target family to profit from improved clin. success rates and lead to safe and efficacious medicines over the next decade.
- 14Lebon, G.; Warne, T.; Edwards, P. C.; Bennett, K.; Langmead, C. J.; Leslie, A. G. W.; Tate, C. G. Agonist-bound adenosine A2A receptor structures reveal common features of GPCR activation Nature 2011, 474, 521– 523Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXmsFyqsrg%253D&md5=1b8161216b878db48ad33d7e8e007dc7Agonist-bound adenosine A2A receptor structures reveal common features of GPCR activationLebon, Guillaume; Warne, Tony; Edwards, Patricia C.; Bennett, Kirstie; Langmead, Christopher J.; Leslie, Andrew G. W.; Tate, Christopher G.Nature (London, United Kingdom) (2011), 474 (7352), 521-525CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Adenosine receptors and β-adrenoceptors are G-protein-coupled receptors (GPCRs) that activate intracellular G proteins on binding the agonists adenosine or noradrenaline, resp. GPCRs have similar structures consisting of seven transmembrane helixes that contain well-conserved sequence motifs, indicating that they are probably activated by a common mechanism. Recent structures of β-adrenoceptors highlight residues in transmembrane region 5 that initially bind specifically to agonists rather than to antagonists, indicating that these residues have an important role in agonist-induced activation of receptors. Here we present two crystal structures of the thermostabilized human adenosine A2A receptor (A2AR-GL31) bound to its endogenous agonist adenosine and the synthetic agonist NECA. The structures represent an intermediate conformation between the inactive and active states, because they share all the features of GPCRs that are thought to be in a fully activated state, except that the cytoplasmic end of transmembrane helix 6 partially occludes the G-protein-binding site. The adenine substituent of the agonists binds in a similar fashion to the chem. related region of the inverse agonist ZM241385 (ref. 8). Both agonists contain a ribose group, not found in ZM241385, which extends deep into the ligand-binding pocket where it makes polar interactions with conserved residues in H7 (Ser 2777.42 and His 2787.43; superscripts refer to Ballesteros-Weinstein numbering) and non-polar interactions with residues in H3. In contrast, the inverse agonist ZM241385 does not interact with any of these residues and comparison with the agonist-bound structures indicates that ZM241385 sterically prevents the conformational change in H5 and therefore it acts as an inverse agonist. Comparison of the agonist-bound structures of A2AR with the agonist-bound structures of β-adrenoceptors indicates that the contraction of the ligand-binding pocket caused by the inward motion of helixes 3, 5 and 7 may be a common feature in the activation of all GPCRs.
- 15“Chemicals Available for Purchase” database. Available from http://www.accelrys.com.Google ScholarThere is no corresponding record for this reference.
- 16Berman, H. M; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T. N.; Weissig, H.; Shindyalov, I. N.; Bourne, P. E. The Protein Data Bank Nucleic Acids Res. 2000, 28, 235– 242Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXhvVKjt7w%253D&md5=227fb393f754be2be375ab727bfd05dcThe Protein Data BankBerman, Helen M.; Westbrook, John; Feng, Zukang; Gilliland, Gary; Bhat, T. N.; Weissig, Helge; Shindyalov, Ilya N.; Bourne, Philip E.Nucleic Acids Research (2000), 28 (1), 235-242CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The Protein Data Bank (PDB; http://www.rcsb.org/pdb/)is the single worldwide archive of structural data of biol. macromols. This paper describes the goals of the PDB, the systems in place for data deposition and access, how to obtain further information, and near-term plans for the future development of the resource.
- 17Sali, A.; Blundell, T. L. Comparative protein modeling by satisfaction of spatial restraints J. Mol. Biol. 1993, 234, 779– 815Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXnt1ylug%253D%253D&md5=d4a3c39b2205e36221dc187a3d1a478bComparative protein modeling by satisfaction of spatial restraintsSali, Andrej; Blundell, Tom L.Journal of Molecular Biology (1993), 234 (3), 779-815CODEN: JMOBAK; ISSN:0022-2836.The authors describe a comparative protein modeling method designed to find the most probable structure for a sequence given its alignment with related structures. The three-dimensional (3D) model is obtained by optimally satisfying spatial restraints derived from the alignment and expressed as probability d. functions (pdfs) for the features restrained. For example, the probabilities for main-chain conformations of a modelled residue may be restrained by its residue type, main-chain conformation of an equiv. residue in a related protein, and the local similarity between the two sequences. Several such pdfs are obtained from the correlations between structural features in 17 families of homologous proteins which have been aligned on the basis of their 3D structures. The pdfs restrain Cα-Cα distances, main-chain N-O distances, main-chain and side-chain dihedral angles. A smoothing procedure is used in the derivation of these relationships to minimize the problem of a sparse database. The 3D model of a protein is obtained by optimization of the mol. pdf such that the model violates the input restraints as little as possible. The mol. pdf is derived as a combination of pdfs restraining individual spatial features of the whole mol. The optimization procedure is a variable target function method that applies the conjugate gradients algorithm to positions of all non-hydrogen atoms. The method is automated and is illustrated by the modeling of trypsin from two other serine proteinases.
- 18Eswar, N.; Marti-Renom, M. A.; Webb, B.; Madhusudhan, M. S.; Eramian, D.; Shen, M.; Pieper, U.; Sali, A. Comparative Protein Structure Modeling with MODELLER. Current Protocols in Bioinformatics; Wiley: New York, 2006; Suppl. 15, pp 5.6.1– 5.6.30.Google ScholarThere is no corresponding record for this reference.
- 19Molecular Operating Environment (MOE), version 2008.10; Chemical Computing Group, Inc.: Montreal, Quebec, Canada, 2008; www.chemcomp.com.Google ScholarThere is no corresponding record for this reference.
- 20Thompson, J. D.; Higgins, D. G.; Gibson, T. J. CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice Nucleic Acids Res. 1994, 22, 4673– 4680Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXitlSgu74%253D&md5=b219f8f0a8ea118b20a3e23461b99ccfCLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choiceThompson, Julie D.; Higgins, Desmond G.; Gibson, Toby J.Nucleic Acids Research (1994), 22 (22), 4673-80CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The sensitivity of the commonly used progressive multiple sequence alignment method has been greatly improved for the alignment of divergent protein sequences. Firstly, individual wts. are assigned to each sequence in a partial alignment in order to down-wt. near-duplicate sequences and up-wt. the most divergent ones. Secondly, amino acid substitution matrixes are varied at different alignment stages according to the divergence of the sequences to be aligned. Thirdly, residue-specific gap penalties and locally reduced gap penalties in hydrophilic regions encourage new gaps in potential loop regions rather than regular secondary structure. Fourthly, positions in early alignments where gaps have been opened receive locally reduced gap penalties to encourage the opening of new gaps at these positions. These modifications are incorporated into a new program, CLUSTAL W which is freely available.
- 21Mirzadegan, T.; Benko, G. Sequence analyses of G-protein coupled receptor: similarities to rhodopsin Biochemistry 2003, 42 (10) 2759– 2767Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXptlCjtw%253D%253D&md5=6a33e7f8bf10b11627ad8aaf36290305Sequence Analyses of G-Protein-Coupled Receptors: Similarities to RhodopsinMirzadegan, Tara; Benko, Gil; Filipek, Slawomir; Palczewski, KrzysztofBiochemistry (2003), 42 (10), 2759-2767CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)The authors performed a comprehensive anal. of the primary sequences of G-protein-coupled receptors (GPCRs) from family A. They demonstrate that the extracellular domain is the least conserved, while GPCRs display considerable conservation toward the cytoplasmic side. Interestingly, the lengths of connecting loops also suggest an importance of these elements in retaining a similar general structure of these receptors. These structural similarities may suggest a common mechanism by which GPCRs activate G-proteins and how these receptors may self-organize within native membranes.
- 22Kim, J.; Wess, J.; van Rhee, M.; Schoneberg, T.; Jacobson, K. A. Site-directed mutagenesis identifies residues involved in ligand recognition in the human A2A adenosine receptor J. Biol. Chem. 1995, 270 (23) 13987– 13997Google ScholarThere is no corresponding record for this reference.
- 23Available from Schrödinger, LLC, New York (http://www.schrodinger.com).Google ScholarThere is no corresponding record for this reference.
- 24Pajouhesh, H.; Lenz, G. R. Medicinal chemical properties of successful central nervous system drugs NeuroRx 2005, 2 (4) 541– 553Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD28%252FpvVKqsg%253D%253D&md5=0815084e2d107b9243b785d0782dfc3aMedicinal chemical properties of successful central nervous system drugsPajouhesh Hassan; Lenz George RNeuroRx : the journal of the American Society for Experimental NeuroTherapeutics (2005), 2 (4), 541-53 ISSN:1545-5343.Fundamental physiochemical features of CNS drugs are related to their ability to penetrate the blood-brain barrier affinity and exhibit CNS activity. Factors relevant to the success of CNS drugs are reviewed. CNS drugs show values of molecular weight, lipophilicity, and hydrogen bond donor and acceptor that in general have a smaller range than general therapeutics. Pharmacokinetic properties can be manipulated by the medicinal chemist to a significant extent. The solubility, permeability, metabolic stability, protein binding, and human ether-ago-go-related gene inhibition of CNS compounds need to be optimized simultaneously with potency, selectivity, and other biological parameters. The balance between optimizing the physiochemical and pharmacokinetic properties to make the best compromises in properties is critical for designing new drugs likely to penetrate the blood brain barrier and affect relevant biological systems. This review is intended as a guide to designing CNS therapeutic agents with better drug-like properties.
- 25Dal Ben, D.; Lambertucci, C.; Marucci, G.; Volpini, R.; Cristalli, G. Adenosine Receptor modeling: What does the A2A crystal structure tell us? Curr. Top. Med. Chem. 2010, 93, 993– 1018Google ScholarThere is no corresponding record for this reference.
Cited By
This article is cited by 117 publications.
- Margarita Stampelou, Anna Suchankova, Efpraxia Tzortzini, Lakshiv Dhingra, Kerry Barkan, Nikolaos Lougiakis, Panagiotis Marakos, Nicole Pouli, Graham Ladds, Antonios Kolocouris. Dual A1/A3 Adenosine Receptor Antagonists: Binding Kinetics and Structure−Activity Relationship Studies Using Mutagenesis and Alchemical Binding Free Energy Calculations. Journal of Medicinal Chemistry 2022, 65
(19)
, 13305-13327. https://doi.org/10.1021/acs.jmedchem.2c01123
- Fazhi Yu, Chenyu Zhu, Qiong Xie, Yonghui Wang. Adenosine A2A Receptor Antagonists for Cancer Immunotherapy. Journal of Medicinal Chemistry 2020, 63
(21)
, 12196-12212. https://doi.org/10.1021/acs.jmedchem.0c00237
- Eleonora Comeo, Nicholas D. Kindon, Mark Soave, Leigh A. Stoddart, Laura E. Kilpatrick, Peter J. Scammells, Stephen J. Hill, Barrie Kellam. Subtype-Selective Fluorescent Ligands as Pharmacological Research Tools for the Human Adenosine A2A Receptor. Journal of Medicinal Chemistry 2020, 63
(5)
, 2656-2672. https://doi.org/10.1021/acs.jmedchem.9b01856
- Mairi M. Littleson, Andrew D. Campbell, Adam Clarke, Mark Dow, Gareth Ensor, Matthew C. Evans, Adam Herring, Bethany A. Jackson, Lucinda V. Jackson, Staffan Karlsson, David J. Klauber, Danny H. Legg, Kevin W. Leslie, Štefan Moravčík, Chris D. Parsons, Thomas O. Ronson, Rebecca E. Meadows. Synthetic Route Design of AZD4635, an A2AR Antagonist. Organic Process Research & Development 2019, 23
(7)
, 1407-1419. https://doi.org/10.1021/acs.oprd.9b00171
- Panagiotis Lagarias, Eleni Vrontaki, George Lambrinidis, Dimitrios Stamatis, Marino Convertino, Gabriella Ortore, Thomas Mavromoustakos, Karl-Norbert Klotz, Antonios Kolocouris. Discovery of Novel Adenosine Receptor Antagonists through a Combined Structure- and Ligand-Based Approach Followed by Molecular Dynamics Investigation of Ligand Binding Mode. Journal of Chemical Information and Modeling 2018, 58
(4)
, 794-815. https://doi.org/10.1021/acs.jcim.7b00455
- Yoonji Lee, Shaherin Basith, and Sun Choi . Recent Advances in Structure-Based Drug Design Targeting Class A G Protein-Coupled Receptors Utilizing Crystal Structures and Computational Simulations. Journal of Medicinal Chemistry 2018, 61
(1)
, 1-46. https://doi.org/10.1021/acs.jmedchem.6b01453
- Francesca Spyrakis, Mostafa H. Ahmed, Alexander S. Bayden, Pietro Cozzini, Andrea Mozzarelli, and Glen E. Kellogg . The Roles of Water in the Protein Matrix: A Largely Untapped Resource for Drug Discovery. Journal of Medicinal Chemistry 2017, 60
(16)
, 6781-6827. https://doi.org/10.1021/acs.jmedchem.7b00057
- Sheng Tian, Xu Wang, Linlang Li, Xiaohu Zhang, Youyong Li, Feng Zhu, Tingjun Hou, and Xuechu Zhen . Discovery of Novel and Selective Adenosine A2A Receptor Antagonists for Treating Parkinson’s Disease through Comparative Structure-Based Virtual Screening. Journal of Chemical Information and Modeling 2017, 57
(6)
, 1474-1487. https://doi.org/10.1021/acs.jcim.7b00188
- Marta Arimont, Shan-Liang Sun, Rob Leurs, Martine Smit, Iwan J. P. de Esch, and Chris de Graaf . Structural Analysis of Chemokine Receptor–Ligand Interactions. Journal of Medicinal Chemistry 2017, 60
(12)
, 4735-4779. https://doi.org/10.1021/acs.jmedchem.6b01309
- Zhong Zheng, Xi-Ping Huang, Thomas J. Mangano, Rodger Zou, Xin Chen, Saheem A. Zaidi, Bryan L. Roth, Raymond C. Stevens, and Vsevolod Katritch . Structure-Based Discovery of New Antagonist and Biased Agonist Chemotypes for the Kappa Opioid Receptor. Journal of Medicinal Chemistry 2017, 60
(7)
, 3070-3081. https://doi.org/10.1021/acs.jmedchem.7b00109
- Ali Jazayeri, Stephen P. Andrews, and Fiona H. Marshall . Structurally Enabled Discovery of Adenosine A2A Receptor Antagonists. Chemical Reviews 2017, 117
(1)
, 21-37. https://doi.org/10.1021/acs.chemrev.6b00119
- Woong-Hee Shin, Charles W. Christoffer, Jibo Wang, and Daisuke Kihara . PL-PatchSurfer2: Improved Local Surface Matching-Based Virtual Screening Method That Is Tolerant to Target and Ligand Structure Variation. Journal of Chemical Information and Modeling 2016, 56
(9)
, 1676-1691. https://doi.org/10.1021/acs.jcim.6b00163
- Dahlia R. Weiss, Andrea Bortolato, Benjamin Tehan, and Jonathan S. Mason . GPCR-Bench: A Benchmarking Set and Practitioners’ Guide for G Protein-Coupled Receptor Docking. Journal of Chemical Information and Modeling 2016, 56
(4)
, 642-651. https://doi.org/10.1021/acs.jcim.5b00660
- Samuel R. Perry, Weijun Xu, Anna Wirija, Junxian Lim, Mei-Kwan Yau, Martin J. Stoermer, Andrew J. Lucke, and David P. Fairlie . Three Homology Models of PAR2 Derived from Different Templates: Application to Antagonist Discovery. Journal of Chemical Information and Modeling 2015, 55
(6)
, 1181-1191. https://doi.org/10.1021/acs.jcim.5b00087
- Manuel de Lera Ruiz, Yeon-Hee Lim, and Junying Zheng . Adenosine A2A Receptor as a Drug Discovery Target. Journal of Medicinal Chemistry 2014, 57
(9)
, 3623-3650. https://doi.org/10.1021/jm4011669
- Trayder Thomas, Kimberley C. McLean, Fiona M. McRobb, David T. Manallack, David K. Chalmers, and Elizabeth Yuriev . Homology Modeling of Human Muscarinic Acetylcholine Receptors. Journal of Chemical Information and Modeling 2014, 54
(1)
, 243-253. https://doi.org/10.1021/ci400502u
- Dan Chen, Anirudh Ranganathan, Adriaan P. IJzerman, Gregg Siegal, and Jens Carlsson . Complementarity between in Silico and Biophysical Screening Approaches in Fragment-Based Lead Discovery against the A2A Adenosine Receptor. Journal of Chemical Information and Modeling 2013, 53
(10)
, 2701-2714. https://doi.org/10.1021/ci4003156
- Andrea Bortolato, Ben G. Tehan, Michael S. Bodnarchuk, Jonathan W. Essex, and Jonathan S. Mason . Water Network Perturbation in Ligand Binding: Adenosine A2A Antagonists as a Case Study. Journal of Chemical Information and Modeling 2013, 53
(7)
, 1700-1713. https://doi.org/10.1021/ci4001458
- Jianing Li, Amanda L. Jonsson, Thijs Beuming, John C. Shelley, and Gregory A. Voth . Ligand-Dependent Activation and Deactivation of the Human Adenosine A2A Receptor. Journal of the American Chemical Society 2013, 135
(23)
, 8749-8759. https://doi.org/10.1021/ja404391q
- Kenneth A. Jacobson . Structure-Based Approaches to Ligands for G-Protein-Coupled Adenosine and P2Y Receptors, from Small Molecules to Nanoconjugates. Journal of Medicinal Chemistry 2013, 56
(10)
, 3749-3767. https://doi.org/10.1021/jm400422s
- Gerard J. P. van Westen, Olaf O. van den Hoven, Rianne van der Pijl, Thea Mulder-Krieger, Henk de Vries, Jörg K. Wegner, Adriaan P. IJzerman, Herman W. T. van Vlijmen, and Andreas Bender . Identifying Novel Adenosine Receptor Ligands by Simultaneous Proteochemometric Modeling of Rat and Human Bioactivity Data. Journal of Medicinal Chemistry 2012, 55
(16)
, 7010-7020. https://doi.org/10.1021/jm3003069
- Miles Congreve, Stephen P. Andrews, Andrew S. Doré, Kaspar Hollenstein, Edward Hurrell, Christopher J. Langmead, Jonathan S. Mason, Irene W. Ng, Benjamin Tehan, Andrei Zhukov, Malcolm Weir, and Fiona H. Marshall . Discovery of 1,2,4-Triazine Derivatives as Adenosine A2A Antagonists using Structure Based Drug Design. Journal of Medicinal Chemistry 2012, 55
(5)
, 1898-1903. https://doi.org/10.1021/jm201376w
- Miru Tang, Chang Wen, Jie Lin, Hongming Chen, Ting Ran. Discovery of novel A2AR antagonists through deep learning-based virtual screening. Artificial Intelligence in the Life Sciences 2023, 3 , 100058. https://doi.org/10.1016/j.ailsci.2023.100058
- Pierre Matricon, Anh TN. Nguyen, Duc Duy Vo, Jo-Anne Baltos, Mariama Jaiteh, Andreas Luttens, Stefanie Kampen, Arthur Christopoulos, Jan Kihlberg, Lauren Therese May, Jens Carlsson. Structure-based virtual screening discovers potent and selective adenosine A1 receptor antagonists. European Journal of Medicinal Chemistry 2023, 257 , 115419. https://doi.org/10.1016/j.ejmech.2023.115419
- Prakashsingh M. Chauhan, Mayur I. Morja, Bhavik S. Makwana, Kishor H. Chikhalia. Facile synthesis of chromeno fused imidazolidinone derivatives via copper‐catalyzed tandem O‐arylation/oxidative acylation protocol. ChemistrySelect 2023, 8
(31)
https://doi.org/10.1002/slct.202301470
- Jonas Goßen, Rui Pedro Ribeiro, Dirk Bier, Bernd Neumaier, Paolo Carloni, Alejandro Giorgetti, Giulia Rossetti. AI-based identification of therapeutic agents targeting GPCRs: introducing ligand type classifiers and systems biology. Chemical Science 2023, 14
(32)
, 8651-8661. https://doi.org/10.1039/D3SC02352D
- Andrea Spinaci, Michela Buccioni, Cui Chang, Diego Dal Ben, Beatrice Francucci, Catia Lambertucci, Rosaria Volpini, Gabriella Marucci. Adenosine A2A Receptor Antagonists: Chemistry, SARs, and Therapeutic Potential. 2023, 101-141. https://doi.org/10.1007/7355_2023_162
- Wallace Chan, Jiansheng Wu, Eric Bell, Yang Zhang. Virtual Screening and Bioactivity Modeling for G Protein‐Coupled Receptors. 2022, 388-423. https://doi.org/10.1002/9781119564782.ch12
- Theresa Noonan, Katrin Denzinger, Valerij Talagayev, Yu Chen, Kristina Puls, Clemens Alexander Wolf, Sijie Liu, Trung Ngoc Nguyen, Gerhard Wolber. Mind the Gap—Deciphering GPCR Pharmacology Using 3D Pharmacophores and Artificial Intelligence. Pharmaceuticals 2022, 15
(11)
, 1304. https://doi.org/10.3390/ph15111304
- Elissa A. Fink, Jun Xu, Harald Hübner, Joao M. Braz, Philipp Seemann, Charlotte Avet, Veronica Craik, Dorothee Weikert, Maximilian F. Schmidt, Chase M. Webb, Nataliya A. Tolmachova, Yurii S. Moroz, Xi-Ping Huang, Chakrapani Kalyanaraman, Stefan Gahbauer, Geng Chen, Zheng Liu, Matthew P. Jacobson, John J. Irwin, Michel Bouvier, Yang Du, Brian K. Shoichet, Allan I. Basbaum, Peter Gmeiner. Structure-based discovery of nonopioid analgesics acting through the α
2A
-adrenergic receptor. Science 2022, 377
(6614)
https://doi.org/10.1126/science.abn7065
- Zhichao Zhong, Xingrui He, Jiamin Ge, Junlong Zhu, Chuansheng Yao, Hong Cai, Xiang-Yang Ye, Tian Xie, Renren Bai. Discovery of small-molecule compounds and natural products against Parkinson's disease: Pathological mechanism and structural modification. European Journal of Medicinal Chemistry 2022, 237 , 114378. https://doi.org/10.1016/j.ejmech.2022.114378
- Kenneth A. Jacobson, Zhan‐Guo Gao, Pierre Matricon, Matthew T. Eddy, Jens Carlsson. Adenosine A
2A
receptor antagonists: from caffeine to selective non‐xanthines. British Journal of Pharmacology 2022, 179
(14)
, 3496-3511. https://doi.org/10.1111/bph.15103
- Sujin Park, Yujin Ahn, Yongchan Kim, Eun Joo Roh, Yoonji Lee, Chaebin Han, Hee Min Yoo, Jinha Yu. Design, Synthesis and Biological Evaluation of 1,3,5-Triazine Derivatives Targeting hA1 and hA3 Adenosine Receptor. Molecules 2022, 27
(13)
, 4016. https://doi.org/10.3390/molecules27134016
- Chrisna Matthee, Gisella Terre’Blanche, Lesetja J. Legoabe, Helena D. Janse van Rensburg. Exploration of chalcones and related heterocycle compounds as ligands of adenosine receptors: therapeutics development. Molecular Diversity 2022, 26
(3)
, 1779-1821. https://doi.org/10.1007/s11030-021-10257-9
- Tobias Claff, Tim A. Klapschinski, Udaya K. Tiruttani Subhramanyam, Victoria J. Vaaßen, Jonathan G. Schlegel, Christin Vielmuth, Jan H. Voß, Jörg Labahn, Christa E. Müller. Eine einzige stabilisierende Punktmutation ermöglicht hochaufgelöste Co‐Kristallstrukturen des Adenosin‐A
2A
‐Rezeptors mit Preladenant‐Konjugaten. Angewandte Chemie 2022, 134
(22)
https://doi.org/10.1002/ange.202115545
- Tobias Claff, Tim A. Klapschinski, Udaya K. Tiruttani Subhramanyam, Victoria J. Vaaßen, Jonathan G. Schlegel, Christin Vielmuth, Jan H. Voß, Jörg Labahn, Christa E. Müller. Single Stabilizing Point Mutation Enables High‐Resolution Co‐Crystal Structures of the Adenosine A
2A
Receptor with Preladenant Conjugates. Angewandte Chemie International Edition 2022, 61
(22)
https://doi.org/10.1002/anie.202115545
- Miru Tang, Chang Wen, Lin Jie, Hongming Chen, Ting Ran. Discovery of Novel A 2AR Antagonists Through Deep Learning-Based Virtual Screening. SSRN Electronic Journal 2022, 31 https://doi.org/10.2139/ssrn.4188435
- Brian J. Bender, Stefan Gahbauer, Andreas Luttens, Jiankun Lyu, Chase M. Webb, Reed M. Stein, Elissa A. Fink, Trent E. Balius, Jens Carlsson, John J. Irwin, Brian K. Shoichet. A practical guide to large-scale docking. Nature Protocols 2021, 16
(10)
, 4799-4832. https://doi.org/10.1038/s41596-021-00597-z
- Flavio Ballante, Albert J Kooistra, Stefanie Kampen, Chris de Graaf, Jens Carlsson, . Structure-Based Virtual Screening for Ligands of G Protein–Coupled Receptors: What Can Molecular Docking Do for You?. Pharmacological Reviews 2021, 73
(4)
, 1698-1736. https://doi.org/10.1124/pharmrev.120.000246
- João Paulo L Velloso, David B Ascher, Douglas E V Pires, . pdCSM-GPCR: predicting potent GPCR ligands with graph-based signatures. Bioinformatics Advances 2021, 1
(1)
https://doi.org/10.1093/bioadv/vbab031
- Miles Congreve, John A. Christopher, Chris de Graaf. Structure‐Based Drug Design for G Protein‐Coupled Receptors. 2021, 1-59. https://doi.org/10.1002/0471266949.bmc269
- Helena D. Janse van Rensburg, Lesetja J. Legoabe, Gisella Terre’Blanche. C3 amino-substituted chalcone derivative with selective adenosine rA1 receptor affinity in the micromolar range. Chemical Papers 2021, 75
(4)
, 1581-1605. https://doi.org/10.1007/s11696-020-01414-9
- Veronica Salmaso, Kenneth A. Jacobson. Purinergic Signaling: Impact of GPCR Structures on Rational Drug Design. ChemMedChem 2020, 15
(21)
, 1958-1973. https://doi.org/10.1002/cmdc.202000465
- Willem Jespers, Grégory Verdon, Jhonny Azuaje, Maria Majellaro, Henrik Keränen, Xerardo García‐Mera, Miles Congreve, Francesca Deflorian, Chris de Graaf, Andrei Zhukov, Andrew S. Doré, Jonathan S. Mason, Johan Åqvist, Robert M. Cooke, Eddy Sotelo, Hugo Gutiérrez‐de‐Terán. X‐Ray Crystallography and Free Energy Calculations Reveal the Binding Mechanism of A
2A
Adenosine Receptor Antagonists. Angewandte Chemie 2020, 132
(38)
, 16679-16686. https://doi.org/10.1002/ange.202003788
- Willem Jespers, Grégory Verdon, Jhonny Azuaje, Maria Majellaro, Henrik Keränen, Xerardo García‐Mera, Miles Congreve, Francesca Deflorian, Chris de Graaf, Andrei Zhukov, Andrew S. Doré, Jonathan S. Mason, Johan Åqvist, Robert M. Cooke, Eddy Sotelo, Hugo Gutiérrez‐de‐Terán. X‐Ray Crystallography and Free Energy Calculations Reveal the Binding Mechanism of A
2A
Adenosine Receptor Antagonists. Angewandte Chemie International Edition 2020, 59
(38)
, 16536-16543. https://doi.org/10.1002/anie.202003788
- Saleta Vazquez-Rodriguez, Santiago Vilar, Sonja Kachler, Karl-Norbert Klotz, Eugenio Uriarte, Fernanda Borges, Maria João Matos. Adenosine Receptor Ligands: Coumarin–Chalcone Hybrids as Modulating Agents on the Activity of hARs. Molecules 2020, 25
(18)
, 4306. https://doi.org/10.3390/molecules25184306
- Miles Congreve, Chris de Graaf, Nigel A. Swain, Christopher G. Tate. Impact of GPCR Structures on Drug Discovery. Cell 2020, 181
(1)
, 81-91. https://doi.org/10.1016/j.cell.2020.03.003
- Reed M. Stein, Hye Jin Kang, John D. McCorvy, Grant C. Glatfelter, Anthony J. Jones, Tao Che, Samuel Slocum, Xi-Ping Huang, Olena Savych, Yurii S. Moroz, Benjamin Stauch, Linda C. Johansson, Vadim Cherezov, Terry Kenakin, John J. Irwin, Brian K. Shoichet, Bryan L. Roth, Margarita L. Dubocovich. Virtual discovery of melatonin receptor ligands to modulate circadian rhythms. Nature 2020, 579
(7800)
, 609-614. https://doi.org/10.1038/s41586-020-2027-0
- Yu Wei, Mukuo Wang, Yang Li, Zhangyong Hong, Dongmei Li, Jianping Lin. Identification of new potent A1 adenosine receptor antagonists using a multistage virtual screening approach. European Journal of Medicinal Chemistry 2020, 187 , 111936. https://doi.org/10.1016/j.ejmech.2019.111936
- Francesca Deflorian, Jonathan S. Mason, Andrea Bortolato, Benjamin G. Tehan. Impact of Recently Determined Crystallographic Structures of GPCRs on Drug Discovery. 2020, 449-477. https://doi.org/10.1002/9781118681121.ch19
- Jinan Wang, Apurba Bhattarai, Waseem Imtiaz Ahmad, Treyton S. Farnan, Karen Priyadarshini John, Yinglong Miao. Computer-aided GPCR drug discovery. 2020, 283-293. https://doi.org/10.1016/B978-0-12-816228-6.00015-5
- Stacy Gelhaus Wendell, Hao Fan, Cheng Zhang, . G Protein–Coupled Receptors in Asthma Therapy: Pharmacology and Drug Action. Pharmacological Reviews 2020, 72
(1)
, 1-49. https://doi.org/10.1124/pr.118.016899
- Pabitra Narayan Samanta, Supratik Kar, Jerzy Leszczynski. Recent Advances of In-Silico Modeling of Potent Antagonists for the Adenosine Receptors. Current Pharmaceutical Design 2019, 25
(7)
, 750-773. https://doi.org/10.2174/1381612825666190304123545
- Nikhil Agrawal, Balakumar Chandrasekaran, Amal Al-Aboudi. Recent Advances in the In-silico Structure-based and Ligand-based Approaches for the Design and Discovery of Agonists and Antagonists of A2A Adenosine Receptor. Current Pharmaceutical Design 2019, 25
(7)
, 774-782. https://doi.org/10.2174/1381612825666190306162006
- Rita C. Acúrcio, Anna Scomparin, Ronit Satchi‐Fainaro, Helena F. Florindo, Rita C. Guedes. Computer‐aided drug design in new druggable targets for the next generation of immune‐oncology therapies. WIREs Computational Molecular Science 2019, 9
(3)
https://doi.org/10.1002/wcms.1397
- Jiankun Lyu, Sheng Wang, Trent E. Balius, Isha Singh, Anat Levit, Yurii S. Moroz, Matthew J. O’Meara, Tao Che, Enkhjargal Algaa, Kateryna Tolmachova, Andrey A. Tolmachev, Brian K. Shoichet, Bryan L. Roth, John J. Irwin. Ultra-large library docking for discovering new chemotypes. Nature 2019, 566
(7743)
, 224-229. https://doi.org/10.1038/s41586-019-0917-9
- Darinka Gjorgieva Ackova, Jelena Kotur-Stevuljevic, Chandra Bhushan Mishra, Pratibha Mehta Luthra, Luciano Saso. Antioxidant Properties of Synthesized Bicyclic Thiazolopyrimidine Derivatives as Possible Therapeutic Agents. Applied Sciences 2019, 9
(1)
, 113. https://doi.org/10.3390/app9010113
- Miles Congreve, Giles A. Brown, Alexandra Borodovsky, Michelle L. Lamb. Targeting adenosine A
2A
receptor antagonism for treatment of cancer. Expert Opinion on Drug Discovery 2018, 13
(11)
, 997-1003. https://doi.org/10.1080/17460441.2018.1534825
- Victor Jun Yu Lim, Weina Du, Yu Zong Chen, Hao Fan. A benchmarking study on virtual ligand screening against homology models of human GPCRs. Proteins: Structure, Function, and Bioinformatics 2018, 86
(9)
, 978-989. https://doi.org/10.1002/prot.25533
- Petr Popov, Yao Peng, Ling Shen, Raymond C Stevens, Vadim Cherezov, Zhi-Jie Liu, Vsevolod Katritch. Computational design of thermostabilizing point mutations for G protein-coupled receptors. eLife 2018, 7 https://doi.org/10.7554/eLife.34729
- Shaherin Basith, Minghua Cui, Stephani J. Y. Macalino, Jongmi Park, Nina A. B. Clavio, Soosung Kang, Sun Choi. Exploring G Protein-Coupled Receptors (GPCRs) Ligand Space via Cheminformatics Approaches: Impact on Rational Drug Design. Frontiers in Pharmacology 2018, 9 https://doi.org/10.3389/fphar.2018.00128
- Magdalena Korczynska, Mary J. Clark, Celine Valant, Jun Xu, Ee Von Moo, Sabine Albold, Dahlia R. Weiss, Hayarpi Torosyan, Weijiao Huang, Andrew C. Kruse, Brent R. Lyda, Lauren T. May, Jo-Anne Baltos, Patrick M. Sexton, Brian K. Kobilka, Arthur Christopoulos, Brian K. Shoichet, Roger K. Sunahara. Structure-based discovery of selective positive allosteric modulators of antagonists for the M
2
muscarinic acetylcholine receptor. Proceedings of the National Academy of Sciences 2018, 115
(10)
https://doi.org/10.1073/pnas.1718037115
- Agostinho Lemos, Rita Melo, Irina S. Moreira, M. Natália D. S. Cordeiro. Computer-Aided Drug Design Approaches to Study Key Therapeutic Targets in Alzheimer’s Disease. 2018, 61-106. https://doi.org/10.1007/978-1-4939-7404-7_3
- Jason B. Cross. Methods for Virtual Screening of GPCR Targets: Approaches and Challenges. 2018, 233-264. https://doi.org/10.1007/978-1-4939-7465-8_11
- Antonella Ciancetta, Kenneth A. Jacobson. Breakthrough in GPCR Crystallography and Its Impact on Computer-Aided Drug Design. 2018, 45-72. https://doi.org/10.1007/978-1-4939-7465-8_3
- Stefania Baraldi, Pier Giovanni Baraldi, Paola Oliva, Kiran S. Toti, Antonella Ciancetta, Kenneth A. Jacobson. A2A Adenosine Receptor: Structures, Modeling, and Medicinal Chemistry. 2018, 91-136. https://doi.org/10.1007/978-3-319-90808-3_5
- Willem Jespers, Anke C. Schiedel, Laura H. Heitman, Robert M. Cooke, Lisa Kleene, Gerard J.P. van Westen, David E. Gloriam, Christa E. Müller, Eddy Sotelo, Hugo Gutiérrez-de-Terán. Structural Mapping of Adenosine Receptor Mutations: Ligand Binding and Signaling Mechanisms. Trends in Pharmacological Sciences 2018, 39
(1)
, 75-89. https://doi.org/10.1016/j.tips.2017.11.001
- Chen Wang, Jiehui Zhang, Jie Tang, Gang Zou. A Sequential Suzuki Coupling Approach to Unsymmetrical Aryl
s
‐Triazines from Cyanuric Chloride. Advanced Synthesis & Catalysis 2017, 359
(14)
, 2514-2519. https://doi.org/10.1002/adsc.201700260
- Anirudh Ranganathan, David Rodríguez, Jens Carlsson. Structure-Based Discovery of GPCR Ligands from Crystal Structures and Homology Models. 2017, 65-99. https://doi.org/10.1007/7355_2016_25
- M. Congreve, A. Bortolato, G. Brown, R.M. Cooke. Modeling and Design for Membrane Protein Targets. 2017, 145-188. https://doi.org/10.1016/B978-0-12-409547-2.12358-3
- Romain Duroux, Antonella Ciancetta, Philip Mannes, Jinha Yu, Shireesha Boyapati, Elizabeth Gizewski, Said Yous, Francisco Ciruela, John A. Auchampach, Zhan-Guo Gao, Kenneth A. Jacobson. Bitopic fluorescent antagonists of the A
2A
adenosine receptor based on pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidin-5-amine functionalized congeners. MedChemComm 2017, 8
(8)
, 1659-1667. https://doi.org/10.1039/C7MD00247E
- Eelke B. Lenselink, Thijs Beuming, Corine van Veen, Arnault Massink, Woody Sherman, Herman W. T. van Vlijmen, Adriaan P. IJzerman. In search of novel ligands using a structure-based approach: a case study on the adenosine A2A receptor. Journal of Computer-Aided Molecular Design 2016, 30
(10)
, 863-874. https://doi.org/10.1007/s10822-016-9963-7
- Tony Ngo, Irina Kufareva, James LJ Coleman, Robert M Graham, Ruben Abagyan, Nicola J Smith. Identifying ligands at orphan GPCRs: current status using structure‐based approaches. British Journal of Pharmacology 2016, 173
(20)
, 2934-2951. https://doi.org/10.1111/bph.13452
- Aashish Manglik, Henry Lin, Dipendra K. Aryal, John D. McCorvy, Daniela Dengler, Gregory Corder, Anat Levit, Ralf C. Kling, Viachaslau Bernat, Harald Hübner, Xi-Ping Huang, Maria F. Sassano, Patrick M. Giguère, Stefan Löber, Da Duan, Grégory Scherrer, Brian K. Kobilka, Peter Gmeiner, Bryan L. Roth, Brian K. Shoichet. Structure-based discovery of opioid analgesics with reduced side effects. Nature 2016, 537
(7619)
, 185-190. https://doi.org/10.1038/nature19112
- Celine Lacroix, Inbar Fish, Hayarpi Torosyan, Pranavan Parathaman, John J. Irwin, Brian K. Shoichet, Stephane Angers, . Identification of Novel Smoothened Ligands Using Structure-Based Docking. PLOS ONE 2016, 11
(8)
, e0160365. https://doi.org/10.1371/journal.pone.0160365
- Nagarajan Vaidehi, Reinhard Grisshammer, Christopher G. Tate. How Can Mutations Thermostabilize G-Protein-Coupled Receptors?. Trends in Pharmacological Sciences 2016, 37
(1)
, 37-46. https://doi.org/10.1016/j.tips.2015.09.005
- James Kean, Andrea Bortolato, Kaspar Hollenstein, Fiona H. Marshall, Ali Jazayeri. Conformational thermostabilisation of corticotropin releasing factor receptor 1. Scientific Reports 2015, 5
(1)
https://doi.org/10.1038/srep11954
- Robert M. Cooke, Alastair J.H. Brown, Fiona H. Marshall, Jonathan S. Mason. Structures of G protein-coupled receptors reveal new opportunities for drug discovery. Drug Discovery Today 2015, 20
(11)
, 1355-1364. https://doi.org/10.1016/j.drudis.2015.08.003
- Punita Kumari, Eshan Ghosh, Arun K. Shukla. Emerging Approaches to GPCR Ligand Screening for Drug Discovery. Trends in Molecular Medicine 2015, 21
(11)
, 687-701. https://doi.org/10.1016/j.molmed.2015.09.002
- Ali Jazayeri, Joao M. Dias, Fiona H. Marshall. From G Protein-coupled Receptor Structure Resolution to Rational Drug Design. Journal of Biological Chemistry 2015, 290
(32)
, 19489-19495. https://doi.org/10.1074/jbc.R115.668251
- Delia Preti, Pier Giovanni Baraldi, Allan R. Moorman, Pier Andrea Borea, Katia Varani. History and Perspectives of A
2A
Adenosine Receptor Antagonists as Potential Therapeutic Agents. Medicinal Research Reviews 2015, 35
(4)
, 790-848. https://doi.org/10.1002/med.21344
- Guillaume Lebon, Patricia C. Edwards, Andrew G. W. Leslie, Christopher G. Tate. Molecular Determinants of CGS21680 Binding to the Human Adenosine A
2A
Receptor. Molecular Pharmacology 2015, 87
(6)
, 907-915. https://doi.org/10.1124/mol.114.097360
- Kirstie A Bennett, Andrew S Doré, John A Christopher, Dahlia R Weiss, Fiona H Marshall. Structures of mGluRs shed light on the challenges of drug development of allosteric modulators. Current Opinion in Pharmacology 2015, 20 , 1-7. https://doi.org/10.1016/j.coph.2014.09.022
- Gengyang Yuan, Nicholas G Gedeon, Tanner C Jankins, Graham B Jones. Novel approaches for targeting the adenosine A
2A
receptor. Expert Opinion on Drug Discovery 2015, 10
(1)
, 63-80. https://doi.org/10.1517/17460441.2015.971006
- Thijs Beuming, Bart Lenselink, Daniele Pala, Fiona McRobb, Matt Repasky, Woody Sherman. Docking and Virtual Screening Strategies for GPCR Drug Discovery. 2015, 251-276. https://doi.org/10.1007/978-1-4939-2914-6_17
- Henrik Keränen, Johan Åqvist, Hugo Gutiérrez-de-Terán. Free energy calculations of A
2A
adenosine receptor mutation effects on agonist binding. Chemical Communications 2015, 51
(17)
, 3522-3525. https://doi.org/10.1039/C4CC09517K
- Claudio N. Cavasotto, Damián Palomba. Expanding the horizons of G protein-coupled receptor structure-based ligand discovery and optimization using homology models. Chemical Communications 2015, 51
(71)
, 13576-13594. https://doi.org/10.1039/C5CC05050B
- Henrik Keränen, Hugo Gutiérrez-de-Terán, Johan Åqvist, . Structural and Energetic Effects of A2A Adenosine Receptor Mutations on Agonist and Antagonist Binding. PLoS ONE 2014, 9
(10)
, e108492. https://doi.org/10.1371/journal.pone.0108492
- Maria João Matos, Santiago Vilar, Sonja Kachler, André Fonseca, Lourdes Santana, Eugenio Uriarte, Fernanda Borges, Nicholas P. Tatonetti, Karl‐Norbert Klotz. Insight into the Interactions between Novel Coumarin Derivatives and Human A
3
Adenosine Receptors. ChemMedChem 2014, 9
(10)
, 2245-2253. https://doi.org/10.1002/cmdc.201402205
- Albert J. Kooistra, Chris de Graaf, Henk Timmerman. The Receptor Concept in 3D: From Hypothesis and Metaphor to GPCR–Ligand Structures. Neurochemical Research 2014, 39
(10)
, 1850-1861. https://doi.org/10.1007/s11064-014-1398-8
- David Rodríguez, José Brea, María Isabel Loza, Jens Carlsson. Structure-Based Discovery of Selective Serotonin 5-HT 1B Receptor Ligands. Structure 2014, 22
(8)
, 1140-1151. https://doi.org/10.1016/j.str.2014.05.017
- Rangappa S. Keri, Srinivasa Budagumpi, Ranjith Krishna Pai, R. Geetha Balakrishna. Chromones as a privileged scaffold in drug discovery: A review. European Journal of Medicinal Chemistry 2014, 78 , 340-374. https://doi.org/10.1016/j.ejmech.2014.03.047
- Julia K. Archbold, Jennifer L. Martin, Matthew J. Sweet. Towards selective lysophospholipid GPCR modulators. Trends in Pharmacological Sciences 2014, 35
(5)
, 219-226. https://doi.org/10.1016/j.tips.2014.03.004
- Sophie J Bradley, Sajjad A Riaz, Andrew B Tobin. Employing novel animal models in the design of clinically efficacious GPCR ligands. Current Opinion in Cell Biology 2014, 27 , 117-125. https://doi.org/10.1016/j.ceb.2013.12.002
- Hugo Gutiérrez-de-Terán. The roles of computational chemistry in the ligand design of G protein-coupled receptors: how far have we come and what should we expect?. Future Medicinal Chemistry 2014, 6
(3)
, 251-254. https://doi.org/10.4155/fmc.13.209
- Stephen P. Andrews, Giles A. Brown, John A. Christopher. Structure‐Based and Fragment‐Based GPCR Drug Discovery. ChemMedChem 2014, 9
(2)
, 256-275. https://doi.org/10.1002/cmdc.201300382
- Brian C. Shook. Adenosine A2A Receptor Antagonists. 2014, 1-42. https://doi.org/10.1007/7355_2014_67
- Albert J. Kooistra, Rob Leurs, Iwan J. P. de Esch, Chris de Graaf. From Three-Dimensional GPCR Structure to Rational Ligand Discovery. 2014, 129-157. https://doi.org/10.1007/978-94-007-7423-0_7
- Miles Congreve, João M. Dias, Fiona H. Marshall. Structure-Based Drug Design for G Protein-Coupled Receptors. 2014, 1-63. https://doi.org/10.1016/B978-0-444-63380-4.00001-9
- Lihui Zhang, Tianjun Liu, Xia Wang, Jinan Wang, Guohui Li, Yan Li, Ling Yang, Yonghua Wang. Insight into the binding mode and the structural features of the pyrimidine derivatives as human A2A adenosine receptor antagonists. Biosystems 2014, 115 , 13-22. https://doi.org/10.1016/j.biosystems.2013.04.003
Abstract
Figure 1
Figure 1. Structures of virtual screening hits.
Scheme 1
Scheme 1. Optimization of Chromone Hit 5Figure 2
Figure 2. Docking of the chromone 12, showing the BPM fingerprint color coded onto the binding site residues and in graphical form as change in pKD. Nonbinding is shown in red (N253A, H250A). Next largest effect is in dark orange (L85A), second largest in amber (N181A, Y271A, I66A), an increase in binding in green (S277A). H-bonding between the nitrogen of the thiazole and the aromatic C–H of the chromone is predicted to Asn2536.55. Selected BPM data are tabulated showing the change in pKD of each binding site mutation.
Scheme 2
Scheme 2. Optimization of Triazine Hit 1Figure 3
Figure 3. Docking of the triazine 15, showing the BPM fingerprint color coded onto the binding site residues and in graphical form as change in pKD. Nonbinding is shown in red (N253A, H250A). Next largest effect is in dark orange (L85A, S277A). H-bonding between the nitrogen of the triazine and the phenol is predicted to Asn2536.55. The polar piperazine substituent is proposed to reach into the region of the binding site occupied by ribose in the natural agonist ligand adenosine and may be the driver of selectivity versus the A1 receptor, as this region of the binding site contains some amino acid differences comparing the two receptors. (14) Selected BPM data are tabulated showing the change in pKD of each binding site mutation.
Scheme 3
Scheme 3. Synthesis of 15 and 16aScheme aReagents and conditions: (a) THF, iPr2EtN, NH3. (b) For 16: (i) 3-(4-methoxypiperidin-1-yl)phenylboronic acid, Na2CO3, 1,4-dioxane/H2O, Pd(PPh3)4, 90 °C, then (ii) 2-hydroxylphenylboronic acid, Na2CO3, 1,4-dioxane/H2O, Pd(PPh3)4, 90 °C. (c) For 17: (i) 2-benzyloxyphenylboronic acid, Na2CO3, 1,4-dioxane/H2O, Pd(PPh3)4, 70 °C, then (ii) 3-(4-methylpiperazine-1-carbonyl)phenylboronic acid hydrochloride, Na2CO3, 1,4-dioxane/H2O, Pd(PPh3)4, 90 °C; (d) 17, EtOAc, Pd(OH)2/C, 1,4-cyclohexadiene, 140 °C (microwave).
References
ARTICLE SECTIONSThis article references 25 other publications.
- 1Pinna, A. Novel investigational adenosine A2A receptor antagonists for Parkinson’s disease Expert Opin. Invest. Drugs 2009, 18, 1619– 1631Google ScholarThere is no corresponding record for this reference.
- 2Blagg, J. Structural Alerts for Toxicity. In Burger’s Medicinal Chemistry, Drug Discovery and Development, 7th ed.; Abraham, D. J.; Rotella, D. P., Eds.; Wiley: Hoboken, NJ, 2010; pp 301– 344.Google ScholarThere is no corresponding record for this reference.
- 3Warne, A.; Serrano-Vega, M. J.; Baker, J. G.; Moukhametzianov, R.; Edwards, P. C.; Henderson, R.; Leslie, A. G. W.; Tate, C. G.; Schertler, G. F. X. Structure of the beta1-adrenergic G protein-coupled receptor Nature 2008, 454, 486– 491
(PDB code 2VT4)
Google ScholarThere is no corresponding record for this reference. - 4Zhukov, A.; Andrews, S. P.; Errey, J. C.; Robertson, N.; Tehan, B.; Mason, J. S.; Marshall, F. H.; Weir, M.; Congreve, M. Biophysical Mapping of the adenosine A2A receptor J. Med. Chem. 2011, 54, 4312– 4323Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXnt12nsb0%253D&md5=d7f5c6bf24f9093d823bb41db9867c09Biophysical Mapping of the Adenosine A2A ReceptorZhukov, Andrei; Andrews, Stephen P.; Errey, James C.; Robertson, Nathan; Tehan, Benjamin; Mason, Jonathan S.; Marshall, Fiona H.; Weir, Malcolm; Congreve, MilesJournal of Medicinal Chemistry (2011), 54 (13), 4312-4323CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A new approach to generating information on ligand receptor interactions within the binding pocket of G protein-coupled receptors has been developed, called Biophys. Mapping (BPM). Starting from a stabilized receptor (StaR), minimally engineered for thermostability, addnl. single mutations are then added at positions that could be involved in small mol. interactions. The StaR and a panel of binding site mutants are captured onto Biacore chips to enable characterization of the binding of small mol. ligands using surface plasmon resonance (SPR) measurement. A matrix of binding data for a set of ligands vs. each active site mutation is then generated, providing specific affinity and kinetic information (KD, kon, and koff) of receptor-ligand interactions. This data set, in combination with mol. modeling and docking, is used to map the small mol. binding site for each class of compds. Taken together, the many constraints provided by these data identify key protein-ligand interactions and allow the shape of the site to be refined to produce a high quality three-dimensional picture of ligand binding, thereby facilitating structure based drug design. Results of biophys. mapping of the adenosine A2A receptor are presented.
- 5Halgren, T. A.; Murphy, R. B.; Friesner, R. A.; Beard, H. S.; Frye, L. L.; Pollard, W. T.; Banks, J. L. Glide: a new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screening J. Med. Chem. 2004, 47, 1750– 1759Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhsFyit78%253D&md5=33d68dd968e65626b449df61e44e37beGlide: A new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screeningHalgren, Thomas A.; Murphy, Robert B.; Friesner, Richard A.; Beard, Hege S.; Frye, Leah L.; Pollard, W. Thomas; Banks, Jay L.Journal of Medicinal Chemistry (2004), 47 (7), 1750-1759CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Glide's ability to identify active compds. in a database screen is characterized by applying Glide to a diverse set of nine protein receptors. In many cases, two, or even three, protein sites are employed to probe the sensitivity of the results to the site geometry. To make the database screens as realistic as possible, the screens use sets of "druglike" decoy ligands that have been selected to be representative of what we believe is likely to be found in the compd. collection of a pharmaceutical or biotechnol. company. Results are presented for releases 1.8, 2.0, and 2.5 of Glide. The comparisons show that av. measures for both "early" and "global" enrichment for Glide 2.5 are 3 times higher than for Glide 1.8 and more than 2 times higher than for Glide 2.0 because of better results for the least well-handled screens. This improvement in enrichment stems largely from the better balance of the more widely parametrized GlideScore 2.5 function and the inclusion of terms that penalize ligand-protein interactions that violate established principles of phys. chem., particularly as it concerns the exposure to solvent of charged protein and ligand groups. Comparisons to results for the thymidine kinase and estrogen receptors published by Rognan and co-workers (J. Med. Chem. 2000, 43, 4759-4767) show that Glide 2.5 performs better than GOLD 1.1, FlexX 1.8, or DOCK 4.01.
- 6Congreve, M.; Andrews, S. P.; Dore, A. S.; Hollenstein, K.; Hurrell, E.; Langmead, C. J.; Mason, J. S.; Ng, I. W.; Tehan, B.; Zhukov, A.; Weir, M.; Marshall, F. H. Discovery of 1,2,4-triazine derivatives as adenosine A2A antagonists using structure based drug design. J. Med. Chem. [Online early access]. DOI: DOI: 10.1021/jm201376w . Published Online: Jan 5, 2012.Google ScholarThere is no corresponding record for this reference.
- 7Hopkins, A. L.; Groom, C. R.; Alex, A. Ligand efficiency: a useful metric for lead selection Drug Discovery Today 2004, 9, 430– 431Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2c3ht1ygtw%253D%253D&md5=eb19a1bca53247d0bd9adcb99d5817e6Ligand efficiency: a useful metric for lead selectionHopkins Andrew L; Groom Colin R; Alex AlexanderDrug discovery today (2004), 9 (10), 430-1 ISSN:1359-6446.There is no expanded citation for this reference.
- 8Leeson, P. D.; Springthorpe, B. The influence of drug-like concepts on decision-making in medicinal chemistry Nat. Rev. Drug Discovery 2007, 6, 881– 890Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXht1els7rL&md5=5665a9e2dc266a1ee096e20625757ef7The influence of drug-like concepts on decision-making in medicinal chemistryLeeson, Paul D.; Springthorpe, BrianNature Reviews Drug Discovery (2007), 6 (11), 881-890CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Despite the wide acceptance of drug-like principles such as the 'rule of five', this anal. of mols. currently being synthesized in leading pharmaceutical companies reveals that their phys. properties differ significantly from those of recently discovered oral drugs. The marked increase in lipophilicity in particular could increase the likelihood of attrition in drug development. The application of guidelines linked to the concept of drug-likeness, such as the 'rule of five', has gained wide acceptance as an approach to reduce attrition in drug discovery and development. However, despite this acceptance, anal. of recent trends reveals that the phys. properties of mols. that are currently being synthesized in leading drug discovery companies differ significantly from those of recently discovered oral drugs and compds. in clin. development. The consequences of the marked increase in lipophilicity - the most important drug-like phys. property - include a greater likelihood of lack of selectivity and attrition in drug development. Tackling the threat of compd.-related toxicol. attrition needs to move to the mainstream of medicinal chem. decision-making.
- 9Katritch, V.; Jaakola, V.-P.; Lane, J. R.; Lin, J.; Izerman, A. P.; Yaeger, M.; Kufarena, I.; Stevens, R. C.; Abagyan, R. Structure-based discovery of novel chemotypes for adenosine A2a receptor antagonists J. Med. Chem. 2010, 53, 1799– 1809Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXptlKqsw%253D%253D&md5=5a54df43f6edd20d83e7e5942e2f9811Structure-Based Discovery of Novel Chemotypes for Adenosine A2A Receptor AntagonistsKatritch, Vsevolod; Jaakola, Veli-Pekka; Lane, J. Robert; Lin, Judy; IJzerman, Adriaan P.; Yeager, Mark; Kufareva, Irina; Stevens, Raymond C.; Abagyan, RubenJournal of Medicinal Chemistry (2010), 53 (4), 1799-1809CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The recent progress in crystallog. of G-protein coupled receptors opens an unprecedented venue for structure-based GPCR drug discovery. To test efficiency of the structure-based approach, we performed mol. docking and virtual ligand screening (VLS) of more than 4 million com. available "drug-like" and "lead-like" compds. against the A2AAR 2.6 Å resoln. crystal structure. Out of 56 high ranking compds. tested in A2AAR binding assays, 23 showed affinities under 10 μM, 11 of those had sub-μM affinities and two compds. had affinities under 60 nM. The identified hits represent at least 9 different chem. scaffolds and are characterized by very high ligand efficiency (0.3-0.5 kcal/mol per heavy atom). Significant A2AAR antagonist activities were confirmed for 10 out of 13 ligands tested in functional assays. High success rate, novelty, and diversity of the chem. scaffolds and strong ligand efficiency of the A2AAR antagonists identified in this study suggest practical applicability of receptor-based VLS in GPCR drug discovery.
- 10Carlsson, J.; Yoo, L.; Gao, Z. G.; Irwin, J. J.; Shoichet, B. K.; Jacobson, K. A. Structure-based discovery of A2A adenosine receptor ligands J. Med. Chem. 2010, 53, 3748– 3755Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXkvFaqsL8%253D&md5=c36c941d52d2cec06387d79c4c423d46Structure-Based Discovery of A2A Adenosine Receptor LigandsCarlsson, Jens; Yoo, Lena; Gao, Zhan-Guo; Irwin, John J.; Shoichet, Brian K.; Jacobson, Kenneth A.Journal of Medicinal Chemistry (2010), 53 (9), 3748-3755CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The recent detn. of X-ray structures of pharmacol. relevant GPCRs has made these targets accessible to structure-based ligand discovery. Here we explore whether novel chemotypes may be discovered for the A2A adenosine receptor, based on complementarity to its recently detd. structure. The A2A adenosine receptor signals in the periphery and the CNS, with agonists explored as anti-inflammatory drugs and antagonists explored for neurodegenerative diseases. We used mol. docking to screen a 1.4 million compd. database against the X-ray structure computationally and tested 20 high-ranking, previously unknown mols. exptl. Of these 35% showed substantial activity with affinities between 200 nM and 9 μM. For the most potent of these new inhibitors, over 50-fold specificity was obsd. for the A2A vs. the related A1 and A3 subtypes. These high hit rates and affinities at least partly reflect the bias of com. libraries toward GPCR-like chemotypes, an issue that we attempt to investigate quant. Despite this bias, many of the most potent new ligands were novel, dissimilar from known ligands, providing new lead structures for modulation of this medically important target.
- 11Goodford, P. J. A computational procedure for determining energetically favorable binding sites on biologically important macromolecules J. Med. Chem. 1985, 28 (7) 849– 857Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2MXktVeit7c%253D&md5=03e9d22787f16c760ef0dc29ca02aa88A computational procedure for determining energetically favorable binding sites on biologically important macromoleculesGoodford, P. J.Journal of Medicinal Chemistry (1985), 28 (7), 849-57CODEN: JMCMAR; ISSN:0022-2623.The interaction of a probe group with a protein of known structure is computed at sample positions throughout and around the macromol., giving an array of energy values. The probe include H2O, the Me group, amine-N, carboxy-O, and OH. Contour surfaces at appropriate energy levels are calcd. for each probe and displayed by computer graphics together with the protein structure. Contours at neg. energy levels delineate regions of attraction between probe and protein and are found at known ligand binding clefts in particular. The contours also enable other regions of attraction to be identified and facilitate the interpretation of protein-ligand energetics. They may, therefore, be of value for drug design.
- 12Higgs, C.; Beuming, T.; Sherman, W. Hydration site thermodynamics explain SARs for triazolylpurines analogues binding to the A2A receptor ACS Med. Chem. Lett. 2010, 1, 160– 164Google ScholarThere is no corresponding record for this reference.
- 13Congreve, M.; Langmead, C. J.; Mason, J. S.; Marshall, F. H. Progress in structure based drug design for G protein-coupled receptors J. Med. Chem. 2011, 54, 4283– 4311Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXns1emtr0%253D&md5=2610591be5473dad558145b6dea84c2bProgress in Structure Based Drug Design for G Protein-Coupled ReceptorsCongreve, Miles; Langmead, Christopher J.; Mason, Jonathan S.; Marshall, Fiona H.Journal of Medicinal Chemistry (2011), 54 (13), 4283-4311CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. The field of GPCR drug discovery is set to have a new lease of life, invigorated by a quantum leap of progress in structural biol. over the past 3 years. The new era of SBDD for GPCRS should now allow even some of the most challenging targets to yield to the efforts of medicinal chemists. This very well established drug family can expect to see many more first in class agents and better mols. with improved physicochem. properties and selectivity profiles, derived from the careful, rational optimization of hits using structure based and fragment based methods. In particular, a disciplined emphasis on maintaining high ligand efficiency and ligand lipophilicity efficiency should allow the next generation of research on this target family to profit from improved clin. success rates and lead to safe and efficacious medicines over the next decade.
- 14Lebon, G.; Warne, T.; Edwards, P. C.; Bennett, K.; Langmead, C. J.; Leslie, A. G. W.; Tate, C. G. Agonist-bound adenosine A2A receptor structures reveal common features of GPCR activation Nature 2011, 474, 521– 523Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXmsFyqsrg%253D&md5=1b8161216b878db48ad33d7e8e007dc7Agonist-bound adenosine A2A receptor structures reveal common features of GPCR activationLebon, Guillaume; Warne, Tony; Edwards, Patricia C.; Bennett, Kirstie; Langmead, Christopher J.; Leslie, Andrew G. W.; Tate, Christopher G.Nature (London, United Kingdom) (2011), 474 (7352), 521-525CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Adenosine receptors and β-adrenoceptors are G-protein-coupled receptors (GPCRs) that activate intracellular G proteins on binding the agonists adenosine or noradrenaline, resp. GPCRs have similar structures consisting of seven transmembrane helixes that contain well-conserved sequence motifs, indicating that they are probably activated by a common mechanism. Recent structures of β-adrenoceptors highlight residues in transmembrane region 5 that initially bind specifically to agonists rather than to antagonists, indicating that these residues have an important role in agonist-induced activation of receptors. Here we present two crystal structures of the thermostabilized human adenosine A2A receptor (A2AR-GL31) bound to its endogenous agonist adenosine and the synthetic agonist NECA. The structures represent an intermediate conformation between the inactive and active states, because they share all the features of GPCRs that are thought to be in a fully activated state, except that the cytoplasmic end of transmembrane helix 6 partially occludes the G-protein-binding site. The adenine substituent of the agonists binds in a similar fashion to the chem. related region of the inverse agonist ZM241385 (ref. 8). Both agonists contain a ribose group, not found in ZM241385, which extends deep into the ligand-binding pocket where it makes polar interactions with conserved residues in H7 (Ser 2777.42 and His 2787.43; superscripts refer to Ballesteros-Weinstein numbering) and non-polar interactions with residues in H3. In contrast, the inverse agonist ZM241385 does not interact with any of these residues and comparison with the agonist-bound structures indicates that ZM241385 sterically prevents the conformational change in H5 and therefore it acts as an inverse agonist. Comparison of the agonist-bound structures of A2AR with the agonist-bound structures of β-adrenoceptors indicates that the contraction of the ligand-binding pocket caused by the inward motion of helixes 3, 5 and 7 may be a common feature in the activation of all GPCRs.
- 15“Chemicals Available for Purchase” database. Available from http://www.accelrys.com.Google ScholarThere is no corresponding record for this reference.
- 16Berman, H. M; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T. N.; Weissig, H.; Shindyalov, I. N.; Bourne, P. E. The Protein Data Bank Nucleic Acids Res. 2000, 28, 235– 242Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXhvVKjt7w%253D&md5=227fb393f754be2be375ab727bfd05dcThe Protein Data BankBerman, Helen M.; Westbrook, John; Feng, Zukang; Gilliland, Gary; Bhat, T. N.; Weissig, Helge; Shindyalov, Ilya N.; Bourne, Philip E.Nucleic Acids Research (2000), 28 (1), 235-242CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The Protein Data Bank (PDB; http://www.rcsb.org/pdb/)is the single worldwide archive of structural data of biol. macromols. This paper describes the goals of the PDB, the systems in place for data deposition and access, how to obtain further information, and near-term plans for the future development of the resource.
- 17Sali, A.; Blundell, T. L. Comparative protein modeling by satisfaction of spatial restraints J. Mol. Biol. 1993, 234, 779– 815Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXnt1ylug%253D%253D&md5=d4a3c39b2205e36221dc187a3d1a478bComparative protein modeling by satisfaction of spatial restraintsSali, Andrej; Blundell, Tom L.Journal of Molecular Biology (1993), 234 (3), 779-815CODEN: JMOBAK; ISSN:0022-2836.The authors describe a comparative protein modeling method designed to find the most probable structure for a sequence given its alignment with related structures. The three-dimensional (3D) model is obtained by optimally satisfying spatial restraints derived from the alignment and expressed as probability d. functions (pdfs) for the features restrained. For example, the probabilities for main-chain conformations of a modelled residue may be restrained by its residue type, main-chain conformation of an equiv. residue in a related protein, and the local similarity between the two sequences. Several such pdfs are obtained from the correlations between structural features in 17 families of homologous proteins which have been aligned on the basis of their 3D structures. The pdfs restrain Cα-Cα distances, main-chain N-O distances, main-chain and side-chain dihedral angles. A smoothing procedure is used in the derivation of these relationships to minimize the problem of a sparse database. The 3D model of a protein is obtained by optimization of the mol. pdf such that the model violates the input restraints as little as possible. The mol. pdf is derived as a combination of pdfs restraining individual spatial features of the whole mol. The optimization procedure is a variable target function method that applies the conjugate gradients algorithm to positions of all non-hydrogen atoms. The method is automated and is illustrated by the modeling of trypsin from two other serine proteinases.
- 18Eswar, N.; Marti-Renom, M. A.; Webb, B.; Madhusudhan, M. S.; Eramian, D.; Shen, M.; Pieper, U.; Sali, A. Comparative Protein Structure Modeling with MODELLER. Current Protocols in Bioinformatics; Wiley: New York, 2006; Suppl. 15, pp 5.6.1– 5.6.30.Google ScholarThere is no corresponding record for this reference.
- 19Molecular Operating Environment (MOE), version 2008.10; Chemical Computing Group, Inc.: Montreal, Quebec, Canada, 2008; www.chemcomp.com.Google ScholarThere is no corresponding record for this reference.
- 20Thompson, J. D.; Higgins, D. G.; Gibson, T. J. CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice Nucleic Acids Res. 1994, 22, 4673– 4680Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXitlSgu74%253D&md5=b219f8f0a8ea118b20a3e23461b99ccfCLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choiceThompson, Julie D.; Higgins, Desmond G.; Gibson, Toby J.Nucleic Acids Research (1994), 22 (22), 4673-80CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The sensitivity of the commonly used progressive multiple sequence alignment method has been greatly improved for the alignment of divergent protein sequences. Firstly, individual wts. are assigned to each sequence in a partial alignment in order to down-wt. near-duplicate sequences and up-wt. the most divergent ones. Secondly, amino acid substitution matrixes are varied at different alignment stages according to the divergence of the sequences to be aligned. Thirdly, residue-specific gap penalties and locally reduced gap penalties in hydrophilic regions encourage new gaps in potential loop regions rather than regular secondary structure. Fourthly, positions in early alignments where gaps have been opened receive locally reduced gap penalties to encourage the opening of new gaps at these positions. These modifications are incorporated into a new program, CLUSTAL W which is freely available.
- 21Mirzadegan, T.; Benko, G. Sequence analyses of G-protein coupled receptor: similarities to rhodopsin Biochemistry 2003, 42 (10) 2759– 2767Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXptlCjtw%253D%253D&md5=6a33e7f8bf10b11627ad8aaf36290305Sequence Analyses of G-Protein-Coupled Receptors: Similarities to RhodopsinMirzadegan, Tara; Benko, Gil; Filipek, Slawomir; Palczewski, KrzysztofBiochemistry (2003), 42 (10), 2759-2767CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)The authors performed a comprehensive anal. of the primary sequences of G-protein-coupled receptors (GPCRs) from family A. They demonstrate that the extracellular domain is the least conserved, while GPCRs display considerable conservation toward the cytoplasmic side. Interestingly, the lengths of connecting loops also suggest an importance of these elements in retaining a similar general structure of these receptors. These structural similarities may suggest a common mechanism by which GPCRs activate G-proteins and how these receptors may self-organize within native membranes.
- 22Kim, J.; Wess, J.; van Rhee, M.; Schoneberg, T.; Jacobson, K. A. Site-directed mutagenesis identifies residues involved in ligand recognition in the human A2A adenosine receptor J. Biol. Chem. 1995, 270 (23) 13987– 13997Google ScholarThere is no corresponding record for this reference.
- 23Available from Schrödinger, LLC, New York (http://www.schrodinger.com).Google ScholarThere is no corresponding record for this reference.
- 24Pajouhesh, H.; Lenz, G. R. Medicinal chemical properties of successful central nervous system drugs NeuroRx 2005, 2 (4) 541– 553Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD28%252FpvVKqsg%253D%253D&md5=0815084e2d107b9243b785d0782dfc3aMedicinal chemical properties of successful central nervous system drugsPajouhesh Hassan; Lenz George RNeuroRx : the journal of the American Society for Experimental NeuroTherapeutics (2005), 2 (4), 541-53 ISSN:1545-5343.Fundamental physiochemical features of CNS drugs are related to their ability to penetrate the blood-brain barrier affinity and exhibit CNS activity. Factors relevant to the success of CNS drugs are reviewed. CNS drugs show values of molecular weight, lipophilicity, and hydrogen bond donor and acceptor that in general have a smaller range than general therapeutics. Pharmacokinetic properties can be manipulated by the medicinal chemist to a significant extent. The solubility, permeability, metabolic stability, protein binding, and human ether-ago-go-related gene inhibition of CNS compounds need to be optimized simultaneously with potency, selectivity, and other biological parameters. The balance between optimizing the physiochemical and pharmacokinetic properties to make the best compromises in properties is critical for designing new drugs likely to penetrate the blood brain barrier and affect relevant biological systems. This review is intended as a guide to designing CNS therapeutic agents with better drug-like properties.
- 25Dal Ben, D.; Lambertucci, C.; Marucci, G.; Volpini, R.; Cristalli, G. Adenosine Receptor modeling: What does the A2A crystal structure tell us? Curr. Top. Med. Chem. 2010, 93, 993– 1018Google ScholarThere is no corresponding record for this reference.
Supporting Information
Supporting Information
ARTICLE SECTIONSChemical synthesis protocols, QC data and binding curves for the top 10 hits, more detailed computational methods including virtual screening workflows, and a table of calculated blood–brain barrier prediction and the closest published adenosine A2A antagonist to each of the hits. This material is available free of charge via the Internet at http://pubs.acs.org.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.