Discovery of a High Affinity Adenosine A1/A3 Receptor Antagonist with a Novel 7-Amino-pyrazolo[3,4-d]pyridazine Scaffold
- Anna SuchankovaAnna SuchankovaDepartment of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K.More by Anna Suchankova
- ,
- Margarita StampelouMargarita StampelouLaboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, GreeceMore by Margarita Stampelou
- ,
- Klontiana KoutsoukiKlontiana KoutsoukiLaboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, GreeceDepartment of Nutrition & Dietetics, School of Health Sciences and Education, Harokopio University, 17671 Athens, GreeceMore by Klontiana Koutsouki
- ,
- Athanasios PousiasAthanasios PousiasLaboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, GreeceMore by Athanasios Pousias
- ,
- Lakshiv DhingraLakshiv DhingraDepartment of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K.More by Lakshiv Dhingra
- ,
- Kerry BarkanKerry BarkanDepartment of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K.More by Kerry Barkan
- ,
- Nicole PouliNicole PouliLaboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, GreeceMore by Nicole Pouli
- ,
- Panagiotis MarakosPanagiotis MarakosLaboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, GreeceMore by Panagiotis Marakos
- ,
- Roxane TentaRoxane TentaDepartment of Nutrition & Dietetics, School of Health Sciences and Education, Harokopio University, 17671 Athens, GreeceMore by Roxane Tenta
- ,
- Antonios Kolocouris*Antonios Kolocouris*Email: [email protected]Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, GreeceMore by Antonios Kolocouris
- ,
- Nikolaos Lougiakis*Nikolaos Lougiakis*Email: [email protected]Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, GreeceMore by Nikolaos Lougiakis
- , and
- Graham Ladds*Graham Ladds*Email: [email protected]Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K.More by Graham Ladds
Abstract

Here we describe the design and synthesis of pyrazolo[3,4-d]pyridazines as adenosine receptor (AR) ligands. We demonstrate that the introduction of a 3-phenyl group, together with a 7-benzylamino and 1-methyl group at the pyrazolopyridazine scaffold, generated the antagonist compound 10b, which displayed 21 nM affinity and a residence time of ∼60 min, for the human A1R, 55 nM affinity and a residence time of ∼73 min, for the human A3R and 1.7 μΜ affinity for the human A2BR while not being toxic. Strikingly, the 2-methyl analog of 10b, 15b, had no significant affinity. Docking calculations and molecular dynamics simulations of the ligands inside the orthosteric binding area suggested that the 2-methyl group in 15b hinders the formation of hydrogen bonding interactions with N6.55 which are considered critical for the stabilization inside the orthosteric binding cavity. We, therefore, demonstrate that 10a is a novel scaffold for the development of high affinity AR ligands. From the mutagenesis experiments the biggest effect was observed for the Y2717.46A mutation which caused an ∼10-fold reduction in the binding affinity of 10b.
This publication is licensed under
License Summary*
You are free to share (copy and redistribute) this article in any medium or format and to adapt (remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format and to adapt (remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format and to adapt (remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Similarity Calculations
Chemistry
Scheme 1

aReagents and conditions: (a) diethyl oxalate, NaH 60%, toluene dry, 50°C, 2 h; (b) NH2NH2 80%, EtOH, reflux, 90 min; (c) (i) NaH 60%, DMF dry, 0 °C, 15 min, (ii) CH3I, rt, 1 h.
Scheme 2

aReagents and conditions: (a) paraformaldehyde, 33% HBr in AcOH, 90 °C, 3.5 h; (b) Ν-methylmorpholine-Ν-oxide, MeCN dry, rt, 24 h; (c) NH2NH2 (80%), HCl 36%, EtOH, 90°C, 1 h; (d) POCl3, 110 °C, 2.5–8 h; (e) HNR1R2, EtOH, reflux, 2 h.
Scheme 3

aReagents and conditions: (a) paraformaldehyde, 33% HBr in AcOH, 90 °C, 3.5 h; (b) Ν-methylmorpholine-Ν-oxide, MeCN dry, rt, 24 h; (c) NH2NH2 (80%), HCl 36%, EtOH, 90 °C, 1 h; (d) POCl3, 110 °C, 2.5–8 h; (e) HNR1R2, EtOH, reflux, 2 h.
Assessing Biological Activity of Pyrazolo[3,4-d]pyridazine Derivatives
cAMP Assays Assessing Activity at Adenosine Receptors
Figure 1

Figure 1. Characterization of 7-amino-pyrazolo[3,4-d]pyridazines at human A1R and A3R. (A and B) Cells expressing either human A1R (A) or A3R (B) were exposed to 10 μM forskolin and stimulated with increasing concentrations of NECA for 30 min in the presence of a 1 μM concentration of the test compound, and the cAMP accumulation was quantified. (C) cAMP accumulation was measured as detailed in part A using multiple concentrations of 10b. Using pEC50 values, Schild regression analysis was conducted to calculate pA2/pKb values. All values are mean ± SEM expressed as percentage forskolin inhibition, relative to NECA. n ≥ 3 independent experimental repeats were performed in duplicate.

Mean ± SEM; functional activities (pEC50 values of NECA in the presence of either 1 μM ligands or vehicle) as mean ± standard error of the mean (SEM) of at least three independent repeats, conducted in duplicate─values obtained from Figure 1.
Mean ± SEM; equilibrium binding affinities of the ligands measured with NanoBRET against Nluc-A3R or Nluc-A1R; NECA was used as positive control. (3)
Due to the high affinity of MRS1220, 10 nM was used to enable measurement of the full dose–response curve of NECA to determine pEC50.
Statistical significance compared to NECA was determined, at p < 0.05, through one-way ANOVA with Dunnett’s post-test (*, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001).
Quantifying Binding Parameters Using a NanoBRET-Based Saturation Binding Assay
Figure 2

Figure 2. Inhibition of BRET between CA200645 at NLuc-A1R and Nluc-A3R by 10b and 10a. HEK293 cells expressing Nluc-A1R (A) or Nluc-A3R (B) were treated with 5 nM or 20 nM CA200645, respectively, enabling concentration-dependent decreases in the BRET ratio at 10 min to be determined with the response normalized to DMSO. Binding curves were fitted with the Cheng Prusoff equation built into GraphPad Prism 9.3 to enable estimates of the pKi. (19) Comparison of pKi values for A1R (C) and A3R (D) as determined via BRET binding. Each data point represents the mean ± SEM of at least three experiments performed in duplicate. The statistical significance compared to NECA was determined, at p < 0.05, through one-way ANOVA with Dunnett’s post-test (*, p < 0.05; ***, p < 0.001). #Compounds did not fully displace CA200645, so pKi values are estimates preventing statistical analysis.
Determining Kinetic Parameters of 10b Binding at A3R and A1R Using NanoBRET
Simulations
Investigation of the Binding of the 7-Amino-pyrazolo[3,4-d]pyridazines to A1R and A3R
Figure 3


Figure 3. (A–C) 100 ns MD simulations of 10a–c inside the orthosteric binding area of A1R. (D) 100 ns MD simulations of 15b inside the orthosteric binding area of A1R. Starting structures are shown (docking pose), and representative frames from MD simulations, receptor–ligand interaction frequency histograms, and RMSD plots of proteins (RMSDprotein; blue plots) and ligand heavy atoms (RMSDligand; red plots) inside the orthosteric binding area of WT A1R or A3R. Bars are plotted only for residues with interaction frequencies ≥0.2. Color scheme: ligand = brown sticks, receptor = white cartoon and sticks, hydrogen bonding interactions = yellow (dashes or bars), π–π interactions = green (dashes or bars), hydrophobic interactions = gray, water bridges = blue. For the protein models of A1R in complex with 10a–c or 15b, the experimental structure of the inactive form of A1R in complex with an antagonist (PDB ID 5UEN (4)) was used.
Mutagenesis Experiments to Study 10b Binding to A1R
A1R | pKi | Effect on affinity |
---|---|---|
WT | 7.68 ± 0.11 | baseline |
T913.36A | 7.68 ± 0.07 | no change |
E1725.30 A | 7.34 ± 0.06 | no significant change |
L2506.51 A | 7.57 ± 0.04 | no significant change |
H2516.52A | 7.62 ± 0.06 | no significant change |
S2677.42A | 7.86 ± 0.03 | no significant change |
Y2717.46A | 6.99 ± 0.05 | ∼10-fold reduction |
Preliminary Toxicological Analysis of Pyrazolo[3,4-d]pyridazine Derivatives
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsmedchemlett.2c00052.
Scheme S1. Comparison of amino acid residue sequences of the binding area. Table S1. Chemical structures and antagonistic potencies of 7-amino-pyrazolo[3,4-d]pyridazines 10a–c and 15a–c against A2AR and A2BR. Table S2. Mean RMSD values for all compounds against A1R, A2AR, and A2BR and ΔGeff only for 10–10c against A1R. Figure S1. 1H and 13C NMR spectra of the target compounds. Figure S2. Results from the MD simulations of 10b and 15b against A3R and A2BR. Figure S3. Docking poses of 4-(2-phenethyl)amino-1-phenylethyl pyrazolo[3,4-b]pyridine and N9-methyl,N6-benzyl adenine to A1R. Figure S4. Representative frames from 100 ns MD simulations of 10b inside the orthosteric binding area of WT A1R and 10b inside mutant Y271A A1R and the receptor–ligand interaction frequency histogram and RMSD graphs of protein Ca and ligand heavy atoms. Information for the methods and synthetic protocols. (PDF)
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.
Acknowledgments
This research represents part of the master’s thesis of K.K. and part of the Ph.D. theses of A.S. and M.S. We gratefully acknowledge the support of Chiesi Hellas (A.K.), the Cambridge Trust (A.S.), the Leverhulme Trust (G.L.), and the BBSRC (G.L.). This work was supported by computational time granted from the Greek Research & Technology Network (GRNET) in the National HPC facility - ARIS - under project ID pr001007.
ARs | adenosine receptors |
BRET | bioluminescence resonance energy transfer |
GPCRs | G protein-coupled receptors |
HEK | human embryonic kidney |
MD | molecular dynamics |
NECA | 5′-N-ethylcarboxamidoadenosine |
PDB | Protein Data Bank |
RMSD | root-mean-square deviation |
Tc | TanimotoCombo |
tm | mixing time |
TM | transmembrane |
References
This article references 37 other publications.
- 1Fredholm, B. B.; IJzerman, A. P.; Jacobson, K. A.; Linden, J.; Muller, C. E.; Müller, C. E. International Union of Basic and Clinical Pharmacology. LXXXI. Nomenclature and Classification of Adenosine Receptors ─ An Update. Pharmacol. Rev. 2011, 63, 1– 34, DOI: 10.1124/pr.110.003285[Crossref], [PubMed], [CAS], Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXjslGisrY%253D&md5=e184c127e06b800bbcf15402ed27f02aInternational union of basic and clinical pharmacology. LXXXI. Nomenclature and classification of adenosine receptors - an updateFredholm, Bertil B.; IJzerman, Adriaan P.; Jacobson, Kenneth A.; Linden, Joel; Mueller, Christa E.Pharmacological Reviews (2011), 63 (1), 1-34CODEN: PAREAQ; ISSN:0031-6997. (American Society for Pharmacology and Experimental Therapeutics)A review. In the 10 years since our previous International Union of Basic and Clin. Pharmacol. report on the nomenclature and classification of adenosine receptors, no developments have led to major changes in the recommendations. However, there have been so many other developments that an update is needed. The fact that the structure of one of the adenosine receptors has recently been solved has already led to new ways of in silico screening of ligands. The evidence that adenosine receptors can form homo- and hetero-multimers has accumulated, but the functional significance of such complexes remains unclear. The availability of mice with genetic modification of all the adenosine receptors has led to a clarification of the functional roles of adenosine, and to excellent means to study the specificity of drugs. There are also interesting assocns. between disease and structural variants in one or more of the adenosine receptors. Several new selective agonists and antagonists have become available. They provide improved possibilities for receptor classification. There are also developments hinting at the usefulness of allosteric modulators. Many drugs targeting adenosine receptors are in clin. trials, but the established therapeutic use is still very limited.
- 2Schenone, S.; Brullo, C.; Musumeci, F.; Bruno, O.; Botta, M. A1 Receptors Ligands: Past, Present and Future Trends. Curr. Top. Med. Chem. 2010, 10, 878– 901, DOI: 10.2174/156802610791268729[Crossref], [PubMed], [CAS], Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXptVegsL0%253D&md5=19dd32bfed13ee6685e7b31f952ae754A1 receptors ligands: past, present and future trendsSchenone, S.; Brullo, C.; Musumeci, F.; Bruno, O.; Botta, M.Current Topics in Medicinal Chemistry (Sharjah, United Arab Emirates) (2010), 10 (9), 878-901CODEN: CTMCCL; ISSN:1568-0266. (Bentham Science Publishers Ltd.)A review. Adenosine is a neuromodulator that interacting with four receptors, A1, A2A, A2B, and A3, is involved in the regulation of several biol. functions in different organs and tissues, including the central nervous system, the cardiovascular system and the airways; many pathophysiol. states are assocd. with changes of adenosine levels. For these reasons pharmaceutical companies and academicians performed intense efforts to obtain agonists, antagonists and allosteric enhancers selective for each adenosine receptor subtypes as potential clin. candidates. In fact, therapeutic modulation of the adenosine system could offer the possibility of a "soft" treatment of different diseases, but, due to the ubiquitous distribution of adenosine and of its receptors, the challenge in therapy development depends from specificity for the different receptor subtypes. Some A1 agonists and antagonists, very potent and selective, reached clin. trials for the treatment of different diseases. A1 agonists are clin. candidates for atrial arrhythmias, angina, type 2 diabetes and in pain management, while A1 antagonists are in study as potassium-sparing diuretics with kidney-protecting properties and in chronic heart diseases. Several reviews, recently published and herein cited, reported in detail the biol. and clin. aspects of such mols. This review focuses on the A1 adenosine receptor (A1AR) ligands, both agonists and antagonists, appeared in the literature in the last few years, together with their potential therapeutic application, pointing the attention on their chem. structures and SAR (Structure Activity Relationship) and also reporting new findings on preclin. or clin. trials of some important A1AR ligands synthesized in the past.
- 3Squarcialupi, L.; Catarzi, D.; Varano, F.; Betti, M.; Falsini, M.; Vincenzi, F.; Ravani, A.; Ciancetta, A.; Varani, K.; Moro, S.; Colotta, V. Structural Refinement of Pyrazolo[4,3-d]Pyrimidine Derivatives to Obtain Highly Potent and Selective Antagonists for the Human A3 Adenosine Receptor. Eur. J. Med. Chem. 2016, 108, 117– 133, DOI: 10.1016/j.ejmech.2015.11.015[Crossref], [PubMed], [CAS], Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvFantLjL&md5=33cb30d67894345d0d957436482018caStructural refinement of pyrazolo[4,3-d]pyrimidine derivatives to obtain highly potent and selective antagonists for the human A3 adenosine receptorSquarcialupi, Lucia; Catarzi, Daniela; Varano, Flavia; Betti, Marco; Falsini, Matteo; Vincenzi, Fabrizio; Ravani, Annalisa; Ciancetta, Antonella; Varani, Katia; Moro, Stefano; Colotta, VittoriaEuropean Journal of Medicinal Chemistry (2016), 108 (), 117-133CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)In previous research, the authors identified some 7-oxo- and 7-acylamino-substituted pyrazolo[4,3-d]pyrimidine derivs. as potent and selective human (h) A3 adenosine receptor (AR) antagonists. Herein the authors report on the structural refinement of this class of antagonists aimed at achieving improved receptor-ligand recognition. Hence, substituents with different steric bulk, flexibility and lipophilicity (Me, Ar, heteroaryl, CH2Ph) were introduced at the 5- and 2-positions of the bicyclic scaffold of both the 7-oxo and 7-amino derivs., and acyl residues were appended on the 7-amino group of the latter. All the 2-phenylpyrazolo[4,3-d]pyrimidin-7-amines and 7-acylamines bearing a 4-methoxyphenyl- or a 2-thienyl group at the 5-position showed high hA3 affinity and selectivity. In particular, the 2-phenyl-5-(2-thienyl)-pyrazolo[4,3-d]pyrimidin-7-(4-methoxybenzoyl)amine 25 (Ki = 0.027 nM) is one of the most potent and selective hA3 antagonists reported so far. By using an in silico receptor-driven approach the obtained binding data were rationalized and the mol. bases of the obsd. hA3 AR affinities were critically described.
- 4Sun, B.; Bachhawat, P.; Chu, M. L.-H.; Wood, M.; Ceska, T.; Sands, Z. A.; Mercier, J.; Lebon, F.; Kobilka, T. S.; Kobilka, B. K. Crystal Structure of the Adenosine A2A Receptor Bound to an Antagonist Reveals a Potential Allosteric Pocket. Proc. Natl. Acad. Sci. U. S. A. 2017, 114, 2066– 2071, DOI: 10.1073/pnas.1621423114[Crossref], [PubMed], [CAS], Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXitFCksb8%253D&md5=58ac85e930066ef7a6788c4d7f3a72b3Crystal structure of the adenosine A2A receptor bound to an antagonist reveals a potential allosteric pocketSun, Bingfa; Bachhawat, Priti; Chu, Matthew Ling-Hon; Wood, Martyn; Ceska, Tom; Sands, Zara A.; Mercier, Joel; Lebon, Florence; Kobilka, Tong Sun; Kobilka, Brian K.Proceedings of the National Academy of Sciences of the United States of America (2017), 114 (8), 2066-2071CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The adenosine A2A receptor (A2AR) has long been implicated in cardiovascular disorders. As more selective A2AR ligands are being identified, its roles in other disorders, such as Parkinson's disease, are starting to emerge, and A2AR antagonists are important drug candidates for non-dopaminergic anti-Parkinson treatment. Here, the authors report the crystal structure of A2A receptor bound to compd. 1, a novel A2AR/N-methyl-D-aspartate (NMDA) receptor subtype 2B (NR2B; NMDA receptor 2B) dual antagonist and potential anti-Parkinson candidate compd., at 3.5 Å resoln. The A2A receptor with a cytochrome b562-RIL (BRIL) fusion (A2AR-BRIL) in intracellular loop 3 (ICL3) was crystd. in detergent micelles using vapor-phase diffusion. Whereas A2AR-BRIL bound to antagonist ZM241385 was previously been crystd. in the lipidic cubic phase (LCP), structural differences in the compd. 1-bound A2AR-BRIL prevented formation of the lattice obsd. with the ZM241385-bound receptor. The crystals grew with a type II crystal lattice in contrast to the typical type I packing seen from membrane protein structures crystd. in LCP. Compd. 1 bound in a position that overlapped with the native ligand, adenosine, but its methoxyphenyl group extended to an exosite not previously obsd. in other A2AR structures. Structural anal. revealed that compd. 1 binding resulted in the unique conformations of 2 Tyr residues (Tyr91.35 and Tyr2717.36) which are crit. for the formation of the exosite. The structure revealed insights into antagonist binding that were not obsd. in other A2AR structures, highlighting flexibility in the binding pocket that may facilitate the development of A2AR-selective compds. for the treatment of Parkinson's disease.
- 5el-Hashim, A.; D’Agostino, B.; Matera, M. G.; Page, C. Characterization of adenosine receptors involved in adenosine-induced bronchoconstriction in allergic rabbits. Br. J. Pharmacol. 1996, 119, 1262– 1268, DOI: 10.1111/j.1476-5381.1996.tb16031.x[Crossref], [PubMed], [CAS], Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28Xntlentbo%253D&md5=58bb082ce7d52a888fd397437f0b015dCharacterization of adenosine receptors involved in adenosine-induced bronchoconstriction in allergic rabbitsEl-Hashim, Ahmed; D'Agostino, Bruno; Matera, Maria Gabriella; Page, CliveBritish Journal of Pharmacology (1996), 119 (6), 1262-1268CODEN: BJPCBM; ISSN:0007-1188. (Stockton)Recent work has suggested that adenosine may be involved in asthma via the activation of A1 receptors. However, the role of the recently cloned A3 receptor in airways is largely unknown. In the present study, the authors have investigated the role of the A3 receptor in adenosine-induced bronchoconstriction in allergic rabbits. Aerosol challenge of antigen (Ag) immunized rabbits with the adenosine precursor, AMP, resulted in a dose-dependent fall in dynamic compliance (Cdyn). The max. fall in Cdyn in these rabbits was significantly greater than that in litter matched, sham immunized animals. However, there was no significant difference in the max. increase in airways resistance (RL) between Ag and sham immunized rabbits. Aerosol challenge of Ag immunized rabbits with cyclopentyl-adenosine (CPA) (A1-receptor agonist) elicited a dose-dependent fall in Cdyn in Ag immunized rabbits and the max. fall in Cdyn in these rabbits was significantly greater than that obsd. in sham immunized rabbits. Similarly, CPA induced dose-dependent increases in RL in Ag immunized rabbits whereas sham immunized rabbits failed to respond to CPA within the same dose range. The max. increase in RL in Ag immunized rabbits was significantly greater than that of sham immunized rabbits. Aerosol challenge of either Ag or sham immunized rabbits with the A3 agonist aminophenylethyladenosine (APNEA) did not elicit dose-dependent changes in either RL or Cdyn. Moreover, there was no significant difference in the max. response, measured by either parameter, between the two animal groups. These data provide further evidence for a role of the A1 receptor in the airways, but do not support a role for the A3 receptor in adenosine-induced bronchoconstriction in the allergic rabbit.
- 6Brown, R. A.; Spina, D.; Page, C. P. Adenosine receptors and asthma. Br. J. Pharmacol. 2008, 153, S446– S456, DOI: 10.1038/bjp.2008.22[Crossref], [PubMed], [CAS], Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXislSjurg%253D&md5=7adce9e6d9f21f823fb63acc5c593e7dAdenosine receptors and asthmaBrown, R. A.; Spina, D.; Page, C. P.British Journal of Pharmacology (2008), 153 (Suppl. 1), S446-S456CODEN: BJPCBM; ISSN:0007-1188. (Nature Publishing Group)A review. The accumulation of evidence implicating a role for adenosine in the pathogenesis of asthma has led to investigations into all adenosine receptor subtypes as potential therapeutic targets for the treatment of asthma. Selective A1 receptor antagonists are currently in preclin. development since adenosine has been shown exptl. to mediate various features of asthma through this receptor such as bronchoconstriction, mucus secretion and inflammation. The A2A receptor is expressed on most inflammatory cells implicated in asthma, and as A2A stimulation activates adenylate cyclase and consequently elevates cAMP, selective A2A receptor agonists have now reached clin. development. However, initial reports concerning their efficacy are inconclusive. A2B receptor antagonists are also under investigation based on the rationale that inhibiting the effects of adenosine on mast cells would be beneficial, in addn. to other reported pro-inflammatory effects mediated by the A2B receptor on cells such as airway smooth muscle, epithelial cells and fibroblasts. While the effects in pre-clin. models are promising, their efficacy in the clin. setting has also yet to be reported. Finally, adenosine A3 receptor stimulation has been demonstrated to mediate inhibitory effects on eosinophils since it also elevates cAMP. However, some exptl. reports suggest that A3 antagonists mediate anti-inflammatory effects, thus the rationale for A3 receptor ligands as therapeutic agents remains to be detd. In conclusion, establishing the precise role of adenosine in the pathogenesis of asthma and developing appropriate subtype selective agonists/antagonists represents an exciting opportunity for the development of novel therapeutics for the treatment of asthma.
- 7Johnson, J. A.; Montgomery, A. P.; Starr, E. R.; Ludwig, J.; Trevitt, J. Dose-dependent effects of adenosine antagonists on tacrine-induced tremulous jaw movements. Eur. J. Pharmacol. 2018, 833, 364– 369, DOI: 10.1016/j.ejphar.2018.06.004[Crossref], [PubMed], [CAS], Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXht1KnurzL&md5=4c6380986c52e264c73789307e5f31acDose-dependent effects of adenosine antagonists on tacrine-induced tremulous jaw movementsJohnson, Joel A.; Montgomery, Aaron P.; Starr, Eric R.; Ludwig, Justin; Trevitt, JenniferEuropean Journal of Pharmacology (2018), 833 (), 364-369CODEN: EJPHAZ; ISSN:0014-2999. (Elsevier B.V.)The present study examines the effect of three adenosine receptor antagonists on tremulous jaw movements (TJMs), an animal model of tremor. Forty-five rats were pre-treated with one adenosine antagonist: caffeine (0.0, 5.0, or 10.0 mg/kg; non-selective adenosine receptor antagonist), 8-cyclopentyltheophylline (CPT; 0.0, 5.0, or 10.0 mg/kg; selective adenosine A1 receptor antagonist), or SCH 58261 (0.0 or 8.0 mg/kg; selective adenosine A2A receptor antagonist) followed by TJM induction with tacrine (0.0, 0.75, or 2.5 mg/kg; acetylcholinesterase inhibitor). CPT and SCH 58261 both significantly reduced TJMs while caffeine did not. Unexpectedly, both SCH 58261 and CPT reduced TJMs even in the absence of tacrine. Also, CPT showed a robust redn. of TJMs, achieved at both (5.0 mg/kg) and (10.0 mg/kg) doses and regardless of tacrine dose. In conclusion, this study shows adenosine receptor antagonism to generally suppress low-dose tacrine-induced TJMs. In concert with two prior studies, these results are suggestive of behavioral evidence for a biphasic effect of adenosine A2A receptor antagonists (caffeine and SCH 58261) that is modulated by tacrine, and a model of this effect is proposed.
- 8Cohen, S.; Fishman, P. Targeting the A3 Adenosine Receptor to Treat Cytokine Release Syndrome in Cancer Immunotherapy. Drug Des. Devel. Ther. 2019, 13, 491– 497, DOI: 10.2147/DDDT.S195294[Crossref], [PubMed], [CAS], Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXit1KitrnP&md5=c145fb65e2c605a4e517de8745c73b4aTargeting the A3 adenosine receptor to treat cytokine release syndrome in cancer immunotherapyCohen, Shira; Fishman, PninaDrug Design, Development and Therapy (2019), 13 (), 491-497CODEN: DDDTAQ; ISSN:1177-8881. (Dove Medical Press Ltd.)Cancer patients undergoing immunotherapy may develop cytokine release syndrome (CRS), an inflmmatory cytokine storm condition, followed by neurotoxic manifestations and may be life-threatening. The current treatments for CRS successfully reduce the inflmmatory response but may limit the anticancer effect of the given immunotherapy and fail to overcome the neurotoxic adverse events. Adenosine, a ubiquitous purine nucleoside, induces a plethora of effects in the body via its binding to four adenosine receptors A1, A2a, A2b, and the A3. Highly selective agonists to the A3 adenosine receptor act as inhibitors of proinflmmatory cytokines, possess robust anti-inflmmatory and anticancer activity, and concomitantly, induce neuroprotective effects. Piclidenoson and namodenoson belong to this group of compds., are effective upon oral administration, show an excellent safety profie in human clin. studies, and therefore, may be considered as drug candidates to treat CRS. In this article, the detailed anti-inflmmatory characteristics of these compds. and the rationale to use them as drugs to combat CRS are described.
- 9Lee, J.; Hwang, I.; Lee, J. H.; Lee, H. W.; Jeong, L.-S.; Ha, H. The Selective A3AR Antagonist LJ-1888 Ameliorates UUO-Induced Tubulointerstitial Fibrosis. Am. J. Pathol. 2013, 183, 1488– 1497, DOI: 10.1016/j.ajpath.2013.07.010[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhs1OqsLfI&md5=25e0faae674396045573507dd5b78e48The Selective A3AR Antagonist LJ-1888 Ameliorates UUO-Induced Tubulointerstitial FibrosisLee, Jiyoun; Hwang, Inah; Lee, Jung H.; Lee, Hyuk W.; Jeong, Lak-Shin; Ha, HunjooAmerican Journal of Pathology (2013), 183 (5), 1488-1497CODEN: AJPAA4; ISSN:0002-9440. (Elsevier B.V.)Adenosine in the normal kidney significantly elevates in response to cellular damage. The renal A3 adenosine receptor (A3AR) is up-regulated under stress, but the therapeutic effects of A3AR antagonists on chronic kidney disease are not fully understood. The present study examd. the effect of LJ-1888 [(2R,3R,4S)-2-[2-chloro-6-(3-iodobenzylamino)-9H-purine-9-yl]-tetrahydrothiophene-3,4-diol], a newly developed potent, selective, species-independent, and orally active A3AR antagonist, on unilateral ureteral obstruction (UUO)-induced renal fibrosis. Pretreatment with LJ-1888 inhibited UUO-induced fibronectin and collagen I up-regulation in a dose-dependent manner. Masson's trichrome staining confirmed that LJ-1888 treatment effectively reduced UUO-induced interstitial collagen accumulation. Furthermore, delayed administration of LJ-1888 showed an equiv. therapeutic effect on tubulointerstitial fibrosis to that of losartan. Small-interfering A3AR transfection effectively inhibited transforming growth factor-β1 (TGF-β1)-induced fibronectin and collagen I up-regulation in proximal tubular cells similar to LJ-1888, confirming that the renoprotective effect of LJ-1888 resulted from A3AR blockade. UUO- or TGF-β1-induced c-Jun N-terminal kinase and extracellular signal-regulated kinase phosphorylation decreased significantly after LJ-1888 administration. A3AR blockade reduced UUO- or TGF-β1-induced up-regulation of lysyl oxidase, which induces crosslinking of extracellular matrix, suggesting that LJ-1888 may also regulate extracellular matrix accumulation via post-translational regulation. In conclusion, the present data demonstrate that the A3AR antagonist, LJ-1888, blocked the development and attenuated the progression of renal fibrosis, and they suggest that LJ-1888 may become a new therapeutic modality for renal interstitial fibrosis.
- 10Wang, Z.; Do, C. W.; Avila, M. Y.; Peterson-Yantorno, K.; Stone, R. A.; Gao, Z.-G.; Joshi, B.; Besada, P.; Jeong, L. S.; Jacobson, K. A.; Civan, M. M. Nucleoside-Derived Antagonists to A3 Adenosine Receptors Lower Mouse Intraocular Pressure and Act across Species. Exp. Eye Res. 2010, 90, 146– 154, DOI: 10.1016/j.exer.2009.10.001[Crossref], [PubMed], [CAS], Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsFantbjM&md5=3d11d9a34194272b31e203a158609fd8Nucleoside-derived antagonists to A3 adenosine receptors lower mouse intraocular pressure and act across speciesWang, Zhao; Do, Chi-Wai; Avila, Marcel Y.; Peterson-Yantorno, Kim; Stone, Richard A.; Gao, Zhan-Guo; Joshi, Bhalchandra; Besada, Pedro; Jeong, Lak-Shin; Jacobson, Kenneth A.; Civan, Mortimer M.Experimental Eye Research (2010), 90 (1), 146-154CODEN: EXERA6; ISSN:0014-4835. (Elsevier B.V.)The purpose of the study was to det. whether novel, selective antagonists of human A3 adenosine receptors (ARs) derived from the A3-selective agonist Cl-IB-MECA lower intraocular pressure (IOP) and act across species. IOP was measured invasively with a micropipette by the Servo-Null Micropipette System (SNMS) and by non-invasive pneumotonometry during topical drug application. Antagonist efficacy was also assayed by measuring inhibition of adenosine-triggered shrinkage of native bovine nonpigmented ciliary epithelial (NPE) cells. Five agonist-based A3AR antagonists lowered mouse IOP measured with SNMS tonometry by 3-5 mm Hg within minutes of topical application. Of the 5 agonist derivs., LJ 1251 was the only antagonist to lower IOP measured by pneumotonometry. No effect was detected pneumotonometrically over 30 min following application of the other 4 compds., consonant with slower, smaller responses previously measured non-invasively following topical application of A3AR agonists and the dihydropyridine A3AR antagonist MRS 1191. Latanoprost similarly lowered SNMS-measured IOP, but not IOP measured non-invasively over 30 min. Like MRS 1191, agonist-based A3AR antagonists applied to native bovine NPE cells inhibited adenosine-triggered shrinkage. In summary, the results indicate that antagonists of human A3ARs derived from the potent, selective A3 agonist Cl-IB-MECA display efficacy in mouse and bovine cells, as well. When intraocular delivery was enhanced by measuring mouse IOP invasively, 5 derivs. of the A3AR agonist Cl-IB-MECA lowered IOP but only one rapidly reduced IOP measured non-invasively after topical application. We conclude that derivs. of the highly-selective A3AR agonist Cl-IB-MECA can reduce IOP upon reaching their intraocular target, and that nucleoside-based derivs. are promising A3 antagonists for study in multiple animal models.
- 11Glukhova, A.; Thal, D. M.; Nguyen, A. T.; Vecchio, E. A.; Jörg, M.; Scammells, P. J.; May, L. T.; Sexton, P. M.; Christopoulos, A. Structure of the Adenosine A1 Receptor Reveals the Basis for Subtype Selectivity. Cell 2017, 168, 867– 877, DOI: 10.1016/j.cell.2017.01.042[Crossref], [PubMed], [CAS], Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjvVWrsrg%253D&md5=a5ff676cfedd6e11a899f65555a1ba62Structure of the Adenosine A1 Receptor Reveals the Basis for Subtype SelectivityGlukhova, Alisa; Thal, David M.; Nguyen, Anh T.; Vecchio, Elizabeth A.; Jorg, Manuela; Scammells, Peter J.; May, Lauren T.; Sexton, Patrick M.; Christopoulos, ArthurCell (Cambridge, MA, United States) (2017), 168 (5), 867-877.e13CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The adenosine A1 receptor (A1-AR) is a G-protein-coupled receptor that plays a vital role in cardiac, renal, and neuronal processes but remains poorly targeted by current drugs. We detd. a 3.2 Å crystal structure of the A1-AR bound to the selective covalent antagonist, DU172, and identified striking differences to the previously solved adenosine A2A receptor (A2A-AR) structure. Mutational and computational anal. of A1-AR revealed a distinct conformation of the second extracellular loop and a wider extracellular cavity with a secondary binding pocket that can accommodate orthosteric and allosteric ligands. We propose that conformational differences in these regions, rather than amino-acid divergence, underlie drug selectivity between these adenosine receptor subtypes. Our findings provide a mol. basis for AR subtype selectivity with implications for understanding the mechanisms governing allosteric modulation of these receptors, allowing the design of more selective agents for the treatment of ischemia-reperfusion injury, renal pathologies, and neuropathic pain.
- 12Cheng, R. K. Y.; Segala, E.; Robertson, N.; Deflorian, F.; Doré, A. S.; Errey, J. C.; Fiez-Vandal, C.; Marshall, F. H.; Cooke, R. M. Structures of Human A1 and A2A Adenosine Receptors with Xanthines Reveal Determinants of Selectivity. Structure 2017, 25, 1275– 1285, DOI: 10.1016/j.str.2017.06.012[Crossref], [PubMed], [CAS], Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFOlsbfJ&md5=e759fd60b25beac2d9429809f3b51b02Structures of Human A1 and A2a Adenosine Receptors with Xanthines Reveal Determinants of SelectivityCheng, Robert K. Y.; Segala, Elena; Robertson, Nathan; Deflorian, Francesca; Dore, Andrew S.; Errey, James C.; Fiez-Vandal, Cedric; Marshall, Fiona H.; Cooke, Robert M.Structure (Oxford, United Kingdom) (2017), 25 (8), 1275-1285.e4CODEN: STRUE6; ISSN:0969-2126. (Elsevier Ltd.)The adenosine A1 and A2a receptors belong to the purinergic family of G protein-coupled receptors, and regulate diverse functions of the cardiovascular, respiratory, renal, inflammation, and CNS. Xanthines such as caffeine and theophylline are weak, non-selective antagonists of adenosine receptors. Here we report the structure of a thermostabilized human A1 receptor at 3.3 Å resoln. with PSB36, an A1-selective xanthine-based antagonist. This is compared with structures of the A2a receptor with PSB36 (2.8 Å resoln.), caffeine (2.1 Å), and theophylline (2.0 Å) to highlight features of ligand recognition which are common across xanthines. The structures of A1R and A2aR were analyzed to identify the differences that are important selectivity determinants for xanthine ligands, and the role of T2707.35 in A1R (M2707.35 in A2aR) in conferring selectivity was confirmed by mutagenesis. The structural differences confirmed to lead to selectivity can be utilized in the design of new subtype-selective A1R or A2aR antagonists.
- 13Draper-Joyce, C. J.; Khoshouei, M.; Thal, D. M.; Liang, Y.-L.; Nguyen, A. T. N.; Furness, S. G. B.; Venugopal, H.; Baltos, J.-A.; Plitzko, J. M.; Danev, R.; Baumeister, W.; May, L. T.; Wootten, D.; Sexton, P. M.; Glukhova, A.; Christopoulos, A. Structure of the Adenosine-Bound Human Adenosine A1 Receptor-Gi Complex. Nature 2018, 558, 559– 563, DOI: 10.1038/s41586-018-0236-6[Crossref], [PubMed], [CAS], Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtFOrsL3P&md5=f31a757364c3ff512bd5548b31e7f1bdStructure of the adenosine-bound human adenosine A1 receptor-Gi complexDraper-Joyce, Christopher J.; Khoshouei, Maryam; Thal, David M.; Liang, Yi-Lynn; Nguyen, Anh T. N.; Furness, Sebastian G. B.; Venugopal, Hariprasad; Baltos, Jo-Anne; Plitzko, Jurgen M.; Danev, Radostin; Baumeister, Wolfgang; May, Lauren T.; Wootten, Denise; Sexton, Patrick M.; Glukhova, Alisa; Christopoulos, ArthurNature (London, United Kingdom) (2018), 558 (7711), 559-563CODEN: NATUAS; ISSN:0028-0836. (Nature Research)The class A adenosine A1 receptor (A1R) is a G-protein-coupled receptor that preferentially couples to inhibitory Gi/o heterotrimeric G proteins, has been implicated in numerous diseases, yet remains poorly targeted. Here we report the 3.6 Å structure of the human A1R in complex with adenosine and heterotrimeric Gi2 protein detd. by Volta phase plate cryo-electron microscopy. Compared to inactive A1R, there is contraction at the extracellular surface in the orthosteric binding site mediated via movement of transmembrane domains 1 and 2. At the intracellular surface, the G protein engages the A1R primarily via amino acids in the C terminus of the Gαi α5-helix, concomitant with a 10.5 Å outward movement of the A1R transmembrane domain 6. Comparison with the agonist-bound β2 adrenergic receptor-Gs-protein complex reveals distinct orientations for each G-protein subtype upon engagement with its receptor. This active A1R structure provides mol. insights into receptor and G-protein selectivity.
- 14Stampelou, M.; Suchankova, A.; Tzortzini, E.; Dhingra, L.; Barkan, K.; Lougiakis, N.; Marakos, P.; Pouli, N.; Ladds, G.; Kolocouris, A. Novel Pyrazolo[3,4-c]Pyridine Antagonists with Nanomolar Affinity for A1/A3 Adenosine Receptors: Binding Kinetics and Exploration of Their Binding Profile Using Mutagenesis Experiments, MD Simulations and TI/MD Calculations. ChemRxiv 2021, DOI: 10.26434/chemrxiv-2021-j18mg-v3 .
- 15Catarzi, D.; Colotta, V.; Varano, F.; Poli, D.; Squarcialupi, L.; Filacchioni, G.; Varani, K.; Vincenzi, F.; Borea, P. A.; Dal Ben, D.; Lambertucci, C.; Cristalli, G. Pyrazolo[1,5-c]Quinazoline Derivatives and Their Simplified Analogues as Adenosine Receptor Antagonists: Synthesis, Structure-Affinity Relationships and Molecular Modeling Studies. Bioorg. Med. Chem. 2013, 21, 283– 294, DOI: 10.1016/j.bmc.2012.10.031[Crossref], [PubMed], [CAS], Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhslSmu7zI&md5=fd01538f48cca49e41bd80f0c5751304Pyrazolo[1,5-c]quinazoline derivatives and their simplified analogues as adenosine receptor antagonists: Synthesis, structure-affinity relationships and molecular modeling studiesCatarzi, Daniela; Colotta, Vittoria; Varano, Flavia; Poli, Daniela; Squarcialupi, Lucia; Filacchioni, Guido; Varani, Katia; Vincenzi, Fabrizio; Borea, Pier Andrea; Dal Ben, Diego; Lambertucci, Catia; Cristalli, GloriaBioorganic & Medicinal Chemistry (2013), 21 (1), 283-294CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)A no. of 5-oxo-pyrazolo[1,5-c]quinazolines, bearing at position-2 the (hetero)aryl moiety but also a carboxylate group, were designed as hA3 AR antagonists. This study produced some interesting compds. endowed with good hA3 receptor affinity and high selectivity, being totally inactive at all the other AR subtypes. In contrast, the corresponding 5-ammino derivs. do not bind or bind with very low affinity at the hA3 AR, the only exception being the 5-N-benzoyl compd. I that shows a hA3Ki value in the high μ-molar range. Evaluation of the synthetic intermediates led to the identification of some 5(3)-(2-aminophenyl)-3(5)-(hetero)arylpyrazoles II [R = Ph, 4-MeOC6H4, 2-furyl, 2-thienyl, CO2Et] with modest affinity but high selectivity toward the hA3 AR subtype. Mol. docking of the herein reported tricyclic and simplified derivs. was carried out to depict their hypothetical binding mode to our model of hA3 receptor.
- 16Manetti, F.; Schenone, S.; Bondavalli, F.; Brullo, C.; Bruno, O.; Ranise, A.; Mosti, L.; Menozzi, G.; Fossa, P.; Trincavelli, M. L.; Martini, C.; Martinelli, A.; Tintori, C.; Botta, M. Synthesis and 3D QSAR of new pyrazolo[3,4-b]pyridines: potent and selective inhibitors of A1 adenosine receptors. J. Med. Chem. 2005, 48, 7172– 7185, DOI: 10.1021/jm050407k[ACS Full Text
], [CAS], Google Scholar
16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhtFWku7vL&md5=58d216826db063b0d522f5e9d6a4474cSynthesis and 3D QSAR of New Pyrazolo[3,4-b]pyridines: Potent and Selective Inhibitors of A1 Adenosine ReceptorsManetti, Fabrizio; Schenone, Silvia; Bondavalli, Francesco; Brullo, Chiara; Bruno, Olga; Ranise, Angelo; Mosti, Luisa; Menozzi, Giulia; Fossa, Paola; Trincavelli, Maria Letizia; Martini, Claudia; Martinelli, Adriano; Tintori, Cristina; Botta, MaurizioJournal of Medicinal Chemistry (2005), 48 (23), 7172-7185CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A no. of 4-(amino)pyrazolo[3,4-b]pyridine-5-carboxylic acid esters were synthesized and evaluated for their binding affinity at the A1, A2A, and A3 adenosine receptors (AR), in bovine cortical membranes, as well as for their affinity toward human A1AR (hA1AR) (A1 purinoceptor antagonists). Some of the new compds. were characterized by a high affinity and selectivity toward the A1 receptor subtype, showing a significant improvement in comparison with other pyrazolo-pyridines previously reported in the literature. In particular two compds., both of them bearing a [(p-methoxyphenyl)ethyl]amino side chain presented Ki values of 6 and 7 nM, resp. To rationalize the relationship between structure and affinity of the novel compds., a 3D QSAR model was also generated starting from compds. belonging to different classes of known A1AR antagonists. - 17Tuccinardi, T.; Zizzari, A. T.; Brullo, C.; Daniele, S.; Musumeci, F.; Schenone, S.; Trincavelli, M. L.; Martini, C.; Martinelli, A.; Giorgi, G.; Botta, M. Substituted pyrazolo[3,4-b]pyridines as human A1 adenosine antagonists: Developments in understanding the receptor stereoselectivity. Org. Biomol. Chem. 2011, 9, 4448– 4455, DOI: 10.1039/c0ob01064b[Crossref], [PubMed], [CAS], Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXntVGgt70%253D&md5=fa192f0519397853d7bef48f22ccb9edSubstituted pyrazolo[3,4-b]pyridines as human A1 adenosine antagonists: Developments in understanding the receptor stereoselectivityTuccinardi, Tiziano; Zizzari, Alessandra Tania; Brullo, Chiara; Daniele, Simona; Musumeci, Francesca; Schenone, Silvia; Trincavelli, Maria Letizia; Martini, Claudia; Martinelli, Adriano; Giorgi, Gianluca; Botta, MaurizioOrganic & Biomolecular Chemistry (2011), 9 (12), 4448-4455CODEN: OBCRAK; ISSN:1477-0520. (Royal Society of Chemistry)A1 adenosine receptor antagonists have been proposed to possess an interesting range of potential therapeutic applications. The authors have already reported the synthesis and the biol. characterization of a family of pyrazolo[3,4-b]pyridine derivs. as A1 adenosine ligands endowed with an antagonistic profile. In the present work, the authors report the LC sepn. of enantiomers of the authors' most active A1 antagonists together with the detn. of their abs. configuration by x-ray crystal structure anal. Biol. assays confirmed a different activity for the two enantiomers, with the R one showing the higher human A1AR affinity. The authors also developed a homol. model of this receptor subtype to suggest a binding disposition of the ligands into the hA1AR. All of the obtained data suggest that the compd.'s chirality plays a key role in A1 affinity.
- 18Cheong, S. L.; Venkatesan, G.; Paira, P.; Jothibasu, R.; Mandel, A. L.; Federico, S.; Spalluto, G.; Pastorin, G. Pyrazolo Derivatives as Potent Adenosine Receptor Antagonists: An Overview on the Structure-Activity Relationships. Int. J. Med. Chem. 2011, 480652, DOI: 10.1155/2011/480652[Crossref], [PubMed], [CAS], Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXltV2gtbw%253D&md5=e31940dc0a60cd1c30079d3edbcdbf56Pyrazolo derivatives as potent adenosine receptor antagonists: an overview on the structure-activity relationshipsCheong, Siew Lee; Venkatesan, Gopalakrishnan; Paira, Priyankar; Jothibasu, Ramasamy; Mandel, Alexander Laurence; Federico, Stephanie; Spalluto, Giampiero; Pastorin, GiorgiaInternational Journal of Medicinal Chemistry (2011), (), 480652, 15 pp.CODEN: IJMCCH; ISSN:2090-2077. (Hindawi Publishing Corp.)A review. In the past few decades, medicinal chem. research towards potent and selective antagonists of human adenosine receptors (namely, A1, A2A, A2B, and A3) has been evolving rapidly. These antagonists are deemed therapeutically beneficial in several pathol. conditions including neurol. and renal disorders, cancer, inflammation, and glaucoma. Up to this point, many classes of compds. have been successfully synthesized and identified as potent human adenosine receptor antagonists. In this paper, an overview of the structure-activity relationship (SAR) profiles of promising nonxanthine pyrazolo derivs. is reported and discussed. We have emphasized the SAR for some representative structures such as pyrazolo-[4,3-e]-1,2,4-triazolo-[1,5-c]pyrimidines; pyrazolo-[3,4-c] or -[4,3-c]quinolines; pyrazolo-[4,3-d]pyrimidinones; pyrazolo-[3,4-d]pyrimidines and pyrazolo-[1,5-a]pyridines. This overview not only clarifies the structural requirements deemed essential for affinity towards individual adenosine receptor subtypes, but it also sheds light on the rational design and optimization of existing structural templates to allow us to conceive new, more potent adenosine receptor antagonists.
- 19Barkan, K.; Lagarias, P.; Stampelou, M.; Stamatis, D.; Hoare, S.; Safitri, D.; Klotz, K.-N.; Vrontaki, E.; Kolocouris, A.; Ladds, G. Pharmacological Characterisation of Novel Adenosine A3 Receptor Antagonists. Sci. Rep. 2020, 10, 20781, DOI: 10.1038/s41598-020-74521-y[Crossref], [PubMed], [CAS], Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisFWltbrL&md5=3710ac8acc7ee7d2b072c0ee95fa3bffPharmacological characterization of novel adenosine A3 receptor antagonistsBarkan, Kerry; Lagarias, Panagiotis; Stampelou, Margarita; Stamatis, Dimitrios; Hoare, Sam; Safitri, Dewi; Klotz, Karl-Norbert; Vrontaki, Eleni; Kolocouris, Antonios; Ladds, GrahamScientific Reports (2020), 10 (1), 20781CODEN: SRCEC3; ISSN:2045-2322. (Nature Research)The adenosine A3 receptor (A3R) belongs to a family of four adenosine receptor (AR) subtypes which all play distinct roles throughout the body. A3R antagonists have been described as potential treatments for numerous diseases including asthma. Given the similarity between (adenosine receptors) orthosteric binding sites, obtaining highly selective antagonists is a challenging but crit. task. Here we screen 39 potential A3R, antagonists using agonist-induced inhibition of cAMP. Pos. hits were assessed for AR subtype selectivity through cAMP accumulation assays. The antagonist affinity was detd. using Schild anal. (pA2 values) and fluorescent ligand binding. Structure-activity relationship investigations revealed that loss of the 3-(dichlorophenyl)-isoxazolyl moiety or the arom. nitrogen heterocycle with nitrogen at α-position to the carbon of carboximidamide group significantly attenuated K18 antagonistic potency. Mutagenic studies supported by mol. dynamic simulations combined with Mol. Mechanics-Poisson Boltzmann Surface Area calcns. identified the residues important for binding in the A3R orthosteric site. We demonstrate that K18, which contains a 3-(dichlorophenyl)-isoxazole group connected through carbonyloxycarboximidamide fragment with a 1,3-thiazole ring, is a specific A3R (< 1 μM) competitive antagonist. Finally, we introduce a model that enables ests. of the equil. binding affinity for rapidly disassocg. compds. from real-time fluorescent ligand-binding studies. These results demonstrate the pharmacol. characterization of a selective competitive A3R antagonist and the description of its orthosteric binding mode. Our findings may provide new insights for drug discovery.
- 20Lagarias, P.; Barkan, K.; Tzortzini, E.; Stampelou, M.; Vrontaki, E.; Ladds, G.; Kolocouris, A. Insights to the Binding of a Selective Adenosine A3 Receptor Antagonist Using Molecular Dynamic Simulations, MM-PBSA and MM-GBSA Free Energy Calculations, and Mutagenesis. J. Chem. Inf. Model. 2019, 59, 5183– 5197, DOI: 10.1021/acs.jcim.9b00751[ACS Full Text
], [CAS], Google Scholar
20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitFGrsL7I&md5=3ad8c6ddc392513238915acb47a00fa0Insights to the Binding of a Selective Adenosine A3 Receptor Antagonist Using Molecular Dynamic Simulations, MM-PBSA and MM-GBSA Free Energy Calculations, and MutagenesisLagarias, Panagiotis; Barkan, Kerry; Tzortzini, Eva; Stampelou, Margarita; Vrontaki, Eleni; Ladds, Graham; Kolocouris, AntoniosJournal of Chemical Information and Modeling (2019), 59 (12), 5183-5197CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Adenosine A3 receptor (A3R) is a promising drug target cancer and for a no. of other conditions like inflammatory diseases, including asthma and rheumatoid arthritis, glaucoma, chronic obstructive pulmonary disease, and ischemic injury. Currently, there is no exptl. detd. structure of A3R. We explored the binding profile of O4-{[3-(2,6-dichlorophenyl)-5-methylisoxazol-4-yl]carbonyl}-2-methyl-1,3-thiazole-4-carbohydroximamide (K18), which is a new specific and competitive antagonist at the orthosteric binding site of A3R. MD simulations and MM-GBSA calcns. of the WT A3R in complex with K18 combined with in vitro mutagenic studies show that the most plausible binding conformation for the dichlorophenyl group of K18 is oriented toward trans-membrane helixes (TM) 5, 6 and reveal important residues for binding. Further, MM-GBSA calcns. distinguish mutations that reduce or maintain or increase antagonistic activity. Our studies show that selectivity of K18 toward A3R is defined not only by direct interactions with residues within the orthosteric binding area but also by remote residues playing a significant role. Although V1695.30 is considered to be a selectivity filter for A3R binders, when it was mutated to glutamic acid, K18 maintained antagonistic potency, in agreement with our previous results obtained for agonists binding profile investigation. Mutation of the direct interacting residue L903.32 in the low region and the remote L2647.35 in the middle/upper region to alanine increases antagonistic potency, suggesting an empty space in the orthosteric area available for increasing antagonist potency. These results approve the computational model for the description of K18 binding at A3R, which we previously performed for agonists binding to A3R, and the design of more effective antagonists based on K18. - 21Papastathopoulos, A.; Lougiakis, N.; Kostakis, I. K.; Marakos, P.; Pouli, N.; Pratsinis, H.; Kletsas, D. New Bioactive 5-Arylcarboximidamidopyrazolo[3,4-c]Pyridines: Synthesis, Cytotoxic Activity, Mechanistic Investigation and Structure-Activity Relationships. Eur. J. Med. Chem. 2021, 218, 113387, DOI: 10.1016/j.ejmech.2021.113387[Crossref], [PubMed], [CAS], Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXns1ehs7s%253D&md5=111d0614bdaa7ddb02eb06e76d1c9eeeNew bioactive 5-arylcarboximidamidopyrazolo[3,4-c]pyridines: Synthesis, cytotoxic activity, mechanistic investigation and structure-activity relationshipsPapastathopoulos, Athanasios; Lougiakis, Nikolaos; Kostakis, Ioannis K.; Marakos, Panagiotis; Pouli, Nicole; Pratsinis, Harris; Kletsas, DimitrisEuropean Journal of Medicinal Chemistry (2021), 218 (), 113387CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)In this study, a series of novel substituted pyrazolo[3,4-c]pyridin-5-ylamidines I [R = Me, (4-methoxyphenyl)methyl; R1 = H, (3,4,5-trimethoxyphenyl)aminyl, cyclohexylaminyl, morpholin-4-yl; R2 = H, Ph; R3 = H, CF3; R4 = H, 4-methylpiperazin-1-yl] and II was synthesized and their cytotoxicity against three cancer cell lines (MDA-MB-231, HT-1080, PC-3), as well as a human normal cell line (AG01523) was evaluated. A no. of derivs. I and II could strongly reduce cancer cells proliferation and exhibit apoptotic induction capability, while reasonable structure-activity relationships could be extd. Certain analogs were endowed with low toxicity against normal cells. Cell cycle anal. revealed that most of the active compds. induced a G0/G1 arrest of HT-1080 cells. Moreover, the potential mechanisms of the cytotoxic activity of the promising compds. I and II were investigated in HT-1080 cells, upon study of their effects on the phosphorylation of Akt, ERK and p38 MAPK. Most of the active derivs. inhibit phosphorylation of Akt and ERK and/or induce p38 MAPK phosphorylation, providing a potential indication on the mode of action of this class.
- 22Bento, A. P.; Gaulton, A.; Hersey, A.; Bellis, L. J.; Chambers, J.; Davies, M.; Krüger, F. A.; Light, Y.; Mak, L.; McGlinchey, S.; Nowotka, M.; Papadatos, G.; Santos, R.; Overington, J. P. The ChEMBL Bioactivity Database: An Update. Nucleic Acids Res. 2014, 42, D1083– D1090, DOI: 10.1093/nar/gkt1031[Crossref], [PubMed], [CAS], Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXoslWl&md5=31b832d03d56ea3065d7aa29618362bcThe ChEMBL bioactivity database: an updateBento, A. Patricia; Gaulton, Anna; Hersey, Anne; Bellis, Louisa J.; Chambers, Jon; Davies, Mark; Krueger, Felix A.; Light, Yvonne; Mak, Lora; McGlinchey, Shaun; Nowotka, Michal; Papadatos, George; Santos, Rita; Overington, John P.Nucleic Acids Research (2014), 42 (D1), D1083-D1090CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)ChEMBL is an open large-scale bioactivity database (https://www.ebi.ac.uk/chembl), previously described in the 2012 Nucleic Acids Research Database Issue. Since then, a variety of new data sources and improvements in functionality have contributed to the growth and utility of the resource. In particular, more comprehensive tracking of compds. from research stages through clin. development to market is provided through the inclusion of data from United States Adopted Name applications; a new richer data model for representing drug targets has been developed; and a no. of methods have been put in place to allow users to more easily identify reliable data. Finally, access to ChEMBL is now available via a new Resource Description Framework format, in addn. to the web-based interface, data downloads and web services.
- 23Hawkins, P. C. D.; Skillman, A. G.; Nicholls, A. Comparison of Shape-Matching and Docking as Virtual Screening Tools. J. Med. Chem. 2007, 50, 74– 82, DOI: 10.1021/jm0603365[ACS Full Text
], [CAS], Google Scholar
23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xhtlansb%252FF&md5=6f97f5c0cc092b4e225f7c2656c1bcf6Comparison of Shape-Matching and Docking as Virtual Screening ToolsHawkins, Paul C. D.; Skillman, A. Geoffrey; Nicholls, AnthonyJournal of Medicinal Chemistry (2007), 50 (1), 74-82CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Ligand docking is a widely used approach in virtual screening. In recent years a large no. 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 no. 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. - 24Pei, Y.; Wickham, B. O. S. Regioselective Syntheses of 3-Aminomethyl-5-Substituted Isoxazoles: A Facile and Chemoselective Reduction of Azide to Amine by Sodium Borohydride Using 1,3-Propanedithiol as a Catalyst. Tetrahedron Lett. 1993, 34, 7509– 7512, DOI: 10.1016/S0040-4039(00)60386-6[Crossref], [CAS], Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXisVCku74%253D&md5=cb8eb0337ff7315bc607e866bacc2e0cRegioselective syntheses of 3-Aminomethyl-5-substituted isoxazoles: a facile and chemoselective reduction of azide to amine by sodium borohydride using 1,3-propanedithiol as a catalystPei, Yazhong; Wickham, Barbara O. S.Tetrahedron Letters (1993), 34 (47), 7509-12CODEN: TELEAY; ISSN:0040-4039.A series of isoxazole azides I (R = Me, Ph, i-Pr; R1 = N3) were prepd. and reduced selectively to isoxazole amines I (R1 = NH2) in quant. yield by sodium borohydride using 1,3-propanedithiol as a catalyst.
- 25Fadnavis, N. W.; Radhika, K. R. Enantio- and Regiospecific Reduction of Ethyl 4-Phenyl-2,4-Dioxobutyrate with Baker’s Yeast: Preparation of (R)-HPB Ester. Tetrahedron Asymmetry 2004, 15, 3443– 3447, DOI: 10.1016/j.tetasy.2004.09.007[Crossref], [CAS], Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXpt1SqsL0%253D&md5=989deb752b3d4601d170b15688a300b8Enantio- and regiospecific reduction of ethyl 4-phenyl-2,4-dioxobutyrate with baker's yeast: preparation of (R)-HPB esterFadnavis, Nitin W.; Radhika, Kasiraman R.Tetrahedron: Asymmetry (2004), 15 (21), 3443-3447CODEN: TASYE3; ISSN:0957-4166. (Elsevier B.V.)Et 2,4-dioxo-4-phenylbutyrate, obtained by condensation of acetophenone with di-Et oxalate, is reduced enantio- and regiospecifically by baker's yeast in a diisopropyl ether/water two-phase system to give (-)-Et (R)-2-hydroxy-4-oxo-4-phenylbutyrate with an 98% ee in 80% isolated yield. This (hydroxy)keto ester was hydrogenated over Pd-C to obtain (-)-Et (R)-2-hydroxy-4-phenylbutyrate (HPB ester), an important intermediate for the synthesis of ACE inhibitors. Prolonged contact of the redn. product with baker's yeast produced 3-phenyl-3-oxopropanol in 90% yield.
- 26Dang, T. T.; Dang, T. T.; Fischer, C.; Görls, H.; Langer, P. Synthesis of Pyrazole-3-Carboxylates and Pyrazole-1,5-Dicarboxylates by One-Pot Cyclization of Hydrazone Dianions with Diethyl Oxalate. Tetrahedron 2008, 64, 2207– 2215, DOI: 10.1016/j.tet.2007.12.024[Crossref], [CAS], Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsFams7o%253D&md5=9c72ba7ccb12654f2548d94e660e120aSynthesis of pyrazole-3-carboxylates and pyrazole-1,5-dicarboxylates by one-pot cyclization of hydrazone dianions with diethyl oxalateDang, Tung T.; Dang, Tuan T.; Fischer, Christine; Goerls, Helmar; Langer, PeterTetrahedron (2008), 64 (9), 2207-2215CODEN: TETRAB; ISSN:0040-4020. (Elsevier Ltd.)The one-pot cyclization of hydrazone dianions with di-Et oxalate allows a convenient synthesis of pyrazole-3-carboxylates and pyrazole-1,5-dicarboxylates. Under the condition of toluene as solvent and p-TsOH as acid, pyrazole-3-carboxylates are obtained; under the condition of CH2Cl2 as solvent and TFA as acid, pyrazole-1,5-dicarboxylates are obtained.
- 27Yan, Z.; Liu, A.; Huang, M.; Liu, M.; Pei, H.; Huang, L.; Yi, H.; Liu, W.; Hu, A. Design, Synthesis, DFT Study and Antifungal Activity of the Derivatives of Pyrazolecarboxamide Containing Thiazole or Oxazole Ring. Eur. J. Med. Chem. 2018, 149, 170– 181, DOI: 10.1016/j.ejmech.2018.02.036[Crossref], [PubMed], [CAS], Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjvVCgsb0%253D&md5=c7e4e7a0f4bd1b090fdb5fdf41b6a03eDesign, synthesis, DFT study and antifungal activity of the derivatives of pyrazolecarboxamide containing thiazole or oxazole ringYan, Zhongzhong; Liu, Aiping; Huang, Mingzhi; Liu, Minhua; Pei, Hui; Huang, Lu; Yi, Haibo; Liu, Weidong; Hu, AixiEuropean Journal of Medicinal Chemistry (2018), 149 (), 170-181CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)To discover new pyrazolecarboxamide analogs with broad spectrum and high activity, a class of new compds. of pyrazole carboxamide derivs. contg. thiazole or oxazole ring I (R1 = H, 4-Me, 2-Cl, etc.; R2 = H, Cl, Me; R3 = Me, Et, i-Pr, cyclopropyl; R4 = H, Et, Me, Ph; Z = S, O) was designed by scaffold hopping and bioisosterism, and 36 pyrazole carboxamide derivs. with antifungal activity were synthesized. Those compds. I were evaluated against five phytopathogenic fungi, Gibberella zeae, Phytophythora capsici, Sclerotonia sclerotiorum, Erysiphe graminis and Puccinia sorghi. The results indicated that most of the compds. displayed good fungicidal activities, esp. against E. graminis. Theor. calcns. were carried out at the B3LYP/6-31G (d, p) level and the full geometry optimization was carried out using the 6-31G (d, p) basis set, and the frontier orbital energy, at. net charges and, mol. docking were discussed, and the structure-activity relationships were also studied.
- 28Schmidt, A.; Habeck, T.; Kindermann, M. K.; Nieger, M. New Pyrazolium-Carboxylates as Structural Analogues of the Pseudo-Cross-Conjugated Betainic Alkaloid Nigellicine. J. Org. Chem. 2003, 68, 5977– 5982, DOI: 10.1021/jo0344337[ACS Full Text
], [CAS], Google Scholar
28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXkvVOrsbk%253D&md5=34795483a4b72eaabf34c39c093260c9New Pyrazolium-carboxylates as Structural Analogues of the Pseudo-Cross-Conjugated Betainic Alkaloid NigellicineSchmidt, Andreas; Habeck, Tobias; Kindermann, Markus Karl; Nieger, MartinJournal of Organic Chemistry (2003), 68 (15), 5977-5982CODEN: JOCEAH; ISSN:0022-3263. (American Chemical Society)Pyrazolium-3-carboxylates were examd. as relatives of the betainic alkaloid nigellicine and as new examples of the sparsely populated class of heterocyclic pseudo-cross-conjugated mesomeric betaines. The title compds. were prepd. in a 4-step procedure starting from EtO2CCOCH:CROH [R = Me, Ph] which were cyclized with substituted hydrazines. The resulting isomeric pyrazole esters were sepd. and quaternized with di-Me sulfate in the presence of nitrobenzene to pyrazolium esters. Sapon. was best accomplished in dild. sulfuric acid, which resulted in the formation of the pseudo-cross-conjugated mesomeric betaines in one step. Protonation to the corresponding carboxylic acids required the treatment of the betaines with tetrafluoroboric acid in dichloromethane. The effect of neg. solvatochromism proves the charge sepn. in the ground state of the mols. X-ray crystallog. analyses, semiempirical calcns., and ESI mass spectrometric measurements were performed to gain knowledge about the phenomenon of pseudo-cross-conjugation. - 29Knight, A.; Hemmings, J. L.; Winfield, I.; Leuenberger, M.; Frattini, E.; Frenguelli, B. G.; Dowell, S. J.; Lochner, M.; Ladds, G. Discovery of Novel Adenosine Receptor Agonists That Exhibit Subtype Selectivity. J. Med. Chem. 2016, 59, 947– 964, DOI: 10.1021/acs.jmedchem.5b01402[ACS Full Text
], [CAS], Google Scholar
29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xmslemtw%253D%253D&md5=0fd4c845ac9f9b91edac5154e4c8cdccDiscovery of Novel Adenosine Receptor Agonists That Exhibit Subtype SelectivityKnight, Anthony; Hemmings, Jennifer L.; Winfield, Ian; Leuenberger, Michele; Frattini, Eugenia; Frenguelli, Bruno G.; Dowell, Simon J.; Lochner, Martin; Ladds, GrahamJournal of Medicinal Chemistry (2016), 59 (3), 947-964CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A series of N6-bicyclic and N6-(2-hydroxy)cyclopentyl derivs. of adenosine were synthesized as novel A1R agonists and their A1R/A2R selectivity assessed using a simple yeast screening platform. We obsd. that the most selective, high potency ligands were achieved through N6-adamantyl substitution in combination with 5'-N-ethylcarboxamido or 5'-hydroxymethyl groups. In addn., we detd. that 5'-(2-fluoro)thiophenyl derivs. all failed to generate a signaling response despite showing an interaction with the A1R. Some selected compds. were also tested on A1R and A3R in mammalian cells revealing that four of them are entirely A1R-selective agonists. By using in silico homol. modeling and ligand docking, we provide insight into their mechanisms of recognition and activation of the A1R. We believe that given the broad tissue distribution, but contrasting signaling profiles, of adenosine receptor subtypes, these compds. might have therapeutic potential. - 30Stoddart, L. A.; Vernall, A. J.; Denman, J. L.; Briddon, S. J.; Kellam, B.; Hill, S. J. Fragment Screening at Adenosine-A3 Receptors in Living Cells Using a Fluorescence-Based Binding Assay. Chem. Biol. 2012, 19, 1105– 1115, DOI: 10.1016/j.chembiol.2012.07.014[Crossref], [PubMed], [CAS], Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhtl2rs7nI&md5=7f99ec1590a4a1ba1ba97bfbf9b7584cFragment Screening at Adenosine-A3 Receptors in Living Cells Using a Fluorescence-Based Binding AssayStoddart, Leigh A.; Vernall, Andrea J.; Denman, Jessica L.; Briddon, Stephen J.; Kellam, Barrie; Hill, Stephen J.Chemistry & Biology (Oxford, United Kingdom) (2012), 19 (9), 1105-1115CODEN: CBOLE2; ISSN:1074-5521. (Elsevier Ltd.)G protein-coupled receptors (GPCRs) comprise the largest family of transmembrane proteins. For GPCR drug discovery, it is important that ligand affinity is detd. in the correct cellular environment and preferably using an unmodified receptor. The authors developed a live cell high-content screening assay that uses a fluorescent antagonist, CA200645, to det. binding affinity consts. of competing ligands at human adenosine-A1 and -A3 receptors. This method was validated as a tool to screen a library of low mol. wt. fragments, and identified a hit with submicromolar binding affinity (KD). This fragment was structurally unrelated to substructures of known adenosine receptor antagonists and was optimized to show selectivity for the adenosine-A3 receptor. This technol. represents a significant advance that will allow the detn. of ligand and fragment affinities at receptors in their native membrane environment.
- 31Eldridge, M. D.; Murray, C. W.; Auton, T. R.; Paolini, G. V.; Mee, R. P. Empirical Scoring Functions: I. The Development of a Fast Empirical Scoring Function to Estimate the Binding Affinity of Ligands in Receptor Complexes. J. Comput. Aided. Mol. Des. 1997, 11, 425– 445, DOI: 10.1023/A:1007996124545[Crossref], [PubMed], [CAS], Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXnsV2it7o%253D&md5=ece4b881e181f3d5c1287af55efaf6b4Empirical scoring functions: I. The development of a fast empirical scoring function to estimate the binding affinity of ligands in receptor complexesEldridge, Matthew D.; Murray, Christopher W.; Auton, Timothy R.; Paolini, Gaia V.; Mee, Roger P.Journal of Computer-Aided Molecular Design (1997), 11 (5), 425-445CODEN: JCADEQ; ISSN:0920-654X. (Kluwer Academic Publishers)This paper describes the development of a simple empirical scoring function designed to est. the free energy of binding for a protein-ligand complex when the 3D structure of the complex is known or can be approximated. The function uses simple contact terms to est. lipophilic and metal-ligand binding contributions, a simple explicit form for hydrogen bonds and a term which penalizes flexibility. The coeffs. of each term are obtained using a regression based on 82 ligand-receptor complexes for which the binding affinity is known. The function reproduces the binding affinity of the complexes with a cross-validated error of 8.68 kJ/mol. Tests on internal consistency indicate that the coeffs. obtained are stable to changes in the compn. of the training set. The function is also tested on two test sets contg. a further 20 and 10 complexes, resp. The deficiencies of this type of function are discussed and it is compared to approaches by other workers.
- 32Hornak, V.; Abel, R.; Okur, A.; Strockbine, B.; Roitberg, A.; Simmerling, C. Comparison of Multiple Amber Force Fields and Development of Improved Protein Backbone Parameters. Proteins Struct. Funct. Genet. 2006, 65, 712– 725, DOI: 10.1002/prot.21123[Crossref], [PubMed], [CAS], Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtFWqt7fM&md5=de683a26eca9e83ae524726e97ac22faComparison of multiple Amber force fields and development of improved protein backbone parametersHornak, Viktor; Abel, Robert; Okur, Asim; Strockbine, Bentley; Roitberg, Adrian; Simmerling, CarlosProteins: Structure, Function, and Bioinformatics (2006), 65 (3), 712-725CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)The ff94 force field that is commonly assocd. with the Amber simulation package is one of the most widely used parameter sets for biomol. simulation. After a decade of extensive use and testing, limitations in this force field, such as over-stabilization of α-helixes, were reported by the authors and other researchers. This led to a no. of attempts to improve these parameters, resulting in a variety of "Amber" force fields and significant difficulty in detg. which should be used for a particular application. The authors show that several of these continue to suffer from inadequate balance between different secondary structure elements. In addn., the approach used in most of these studies neglected to account for the existence in Amber of two sets of backbone .vphi./ψ dihedral terms. This led to parameter sets that provide unreasonable conformational preferences for glycine. The authors report here an effort to improve the .vphi./ψ dihedral terms in the ff99 energy function. Dihedral term parameters are based on fitting the energies of multiple conformations of glycine and alanine tetrapeptides from high level ab initio quantum mech. calcns. The new parameters for backbone dihedrals replace those in the existing ff99 force field. This parameter set, which the authors denote ff99SB, achieves a better balance of secondary structure elements as judged by improved distribution of backbone dihedrals for glycine and alanine with respect to PDB survey data. It also accomplishes improved agreement with published exptl. data for conformational preferences of short alanine peptides and better accord with exptl. NMR relaxation data of test protein systems.
- 33Massova, I.; Kollman, P. A. Combined molecular mechanical and continuum solvent approach (MM-PBSA/GBSA) to predict ligand binding. Per. Drug Discovery Design 2000, 18, 113– 135, DOI: 10.1023/A:1008763014207[Crossref], [CAS], Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXis12ju7Y%253D&md5=520df05a7c7ed02fdf76d3e1e6f377f9Combined molecular mechanical and continuum solvent approach (MM-PBSA/GBSA) to predict ligand bindingMassova, Irina; Kollman, Peter A.Perspectives in Drug Discovery and Design (2000), 18 (Hydrophobicity and Solvation in Drug Design, Pt. II), 113-135CODEN: PDDDEC; ISSN:0928-2866. (Kluwer Academic Publishers)Significant progress has been achieved in computational methods to treat solvent effects in recent years. Among various techniques, the continuum solvent approach appears to be practically promising because it can be used to calc. reliable interaction and solvation energies in complex systems. A computational scanning mutagenesis method, one of such new approaches, has been recently developed. It combines the mol. mech. and continuum solvent approaches and allows one to identify the "hot spots" in binding interfaces from a single trajectory of a wild type complex. Such techniques can be also used as a tool to optimize the interacting species for the binding, or as a ranking procedure in high throughput screening.
- 34Wang, E.; Sun, H.; Wang, J.; Wang, Z.; Liu, H.; Zhang, J. Z. H.; Hou, T. End-Point Binding Free Energy Calculation with MM/PBSA and MM/GBSA: Strategies and Applications in Drug Design. Chem. Rev. 2019, 119, 9478– 9508, DOI: 10.1021/acs.chemrev.9b00055[ACS Full Text
], [CAS], Google Scholar
34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXht1ajs7%252FF&md5=eacd5dbfe659d27df5d103caec9e1bb5End-point binding free energy calculation with MM/PBSA and MM/GBSA: strategies and applications in drug designWang, Ercheng; Sun, Huiyong; Wang, Junmei; Wang, Zhe; Liu, Hui; Zhang, John Z. H.; Hou, TingjunChemical Reviews (Washington, DC, United States) (2019), 119 (16), 9478-9508CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. Mol. mechanics Poisson-Boltzmann surface area (MM/PBSA) and mol. mechanics generalized Born surface area (MM/GBSA) are arguably very popular methods for binding free energy prediction since they are more accurate than most scoring functions of mol. docking and less computationally demanding than alchem. free energy methods. MM/PBSA and MM/GBSA have been widely used in biomol. studies such as protein folding, protein-ligand binding, protein-protein interaction, etc. In this review, methods to adjust the polar solvation energy and to improve the performance of MM/PBSA and MM/GBSA calcns. are reviewed and discussed. The latest applications of MM/GBSA and MM/PBSA in drug design are also presented. This review intends to provide readers with guidance for practically applying MM/PBSA and MM/GBSA in drug design and related research fields. - 35Kaminski, G.; Friesner, R. A.; Tirado-Rives, J.; Jorgensen, W. L. Evaluation and Reparametrization of the OPLS-AA Force Field for Proteins via Comparison with Accurate Quantum Chemical Calculations on Peptides. Phys. Chem. B 2001, 105 (28), 6474– 6487, DOI: 10.1021/jp003919d[ACS Full Text
], [CAS], Google Scholar
35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXislKhsLk%253D&md5=3ff059626977ee7f6342466f5820f5b7Evaluation and Reparametrization of the OPLS-AA Force Field for Proteins via Comparison with Accurate Quantum Chemical Calculations on PeptidesKaminski, George A.; Friesner, Richard A.; Tirado-Rives, Julian; Jorgensen, William L.Journal of Physical Chemistry B (2001), 105 (28), 6474-6487CODEN: JPCBFK; ISSN:1089-5647. (American Chemical Society)We present results of improving the OPLS-AA force field for peptides by means of refitting the key Fourier torsional coeffs. The fitting technique combines using accurate ab initio data as the target, choosing an efficient fitting subspace of the whole potential-energy surface, and detg. wts. for each of the fitting points based on magnitudes of the potential-energy gradient. The av. energy RMS deviation from the LMP2/cc-pVTZ(-f)//HF/6-31G** data is reduced by ∼40% from 0.81 to 0.47 kcal/mol as a result of the fitting for the electrostatically uncharged dipeptides. Transferability of the parameters is demonstrated by using the same alanine dipeptide-fitted backbone torsional parameters for all of the other dipeptides (with the appropriate side-chain refitting) and the alanine tetrapeptide. Parameters of nonbonded interactions have also been refitted for the sulfur-contg. dipeptides (cysteine and methionine), and the validity of the new Coulombic charges and the van der Waals σ's and ε's is proved through reproducing gas-phase energies of complex formation heats of vaporization and densities of pure model liqs. Moreover, a novel approach to fitting torsional parameters for electrostatically charged mol. systems has been presented and successfully tested on five dipeptides with charged side chains. - 36Shivakumar, D.; Williams, J.; Wu, Y.; Damm, W.; Shelley, J.; Sherman, W. Prediction of Absolute Solvation Free Energies using Molecular Dynamics Free Energy Perturbation and the OPLS Force Field. J. Chem. Theory Comput. 2010, 6, 1509– 19, DOI: 10.1021/ct900587b[ACS Full Text
], [CAS], Google Scholar
36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXkslKhu7g%253D&md5=25d464fb9238e3235e881945c0b50a76Prediction of Absolute Solvation Free Energies using Molecular Dynamics Free Energy Perturbation and the OPLS Force FieldShivakumar, Devleena; Williams, Joshua; Wu, Yujie; Damm, Wolfgang; Shelley, John; Sherman, WoodyJournal of Chemical Theory and Computation (2010), 6 (5), 1509-1519CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The accurate prediction of protein-ligand binding free energies is a primary objective in computer-aided drug design. The solvation free energy of a small mol. provides a surrogate to the desolvation of the ligand in the thermodn. process of protein-ligand binding. Here, we use explicit solvent mol. dynamics free energy perturbation to predict the abs. solvation free energies of a set of 239 small mols., spanning diverse chem. functional groups commonly found in drugs and drug-like mols. We also compare the performance of abs. solvation free energies obtained using the OPLS_2005 force field with two other commonly used small mol. force fields - general AMBER force field (GAFF) with AM1-BCC charges and CHARMm-MSI with CHelpG charges. Using the OPLS_2005 force field, we obtain high correlation with exptl. solvation free energies (R2 = 0.94) and low av. unsigned errors for a majority of the functional groups compared to AM1-BCC/GAFF or CHelpG/CHARMm-MSI. However, OPLS_2005 has errors of over 1.3 kcal/mol for certain classes of polar compds. We show that predictions on these compd. classes can be improved by using a semiempirical charge assignment method with an implicit bond charge correction. - 37Stamatis, D.; Lagarias, P.; Barkan, K.; Vrontaki, E.; Ladds, G.; Kolocouris, A. Structural Characterization of Agonist Binding to an A3 Adenosine Receptor through Biomolecular Simulations and Mutagenesis Experiments. J. Med. Chem. 2019, 62, 8831– 8846, DOI: 10.1021/acs.jmedchem.9b01164[ACS Full Text
], [CAS], Google Scholar
37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslGnsL7O&md5=3c3882463d18e0cb0c999839692b20caStructural Characterization of Agonist Binding to an A3 Adenosine Receptor through Biomolecular Simulations and Mutagenesis ExperimentsStamatis, Dimitrios; Lagarias, Panagiotis; Barkan, Kerry; Vrontaki, Eleni; Ladds, Graham; Kolocouris, AntoniosJournal of Medicinal Chemistry (2019), 62 (19), 8831-8846CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The adenosine A3 receptor (A3R) binds adenosine and is a drug target against cancer cell proliferation. Currently, there is no exptl. structure of A3R. Here, we have generated a mol. model of A3R in complex with two agonists, the nonselective 1-(6-amino-9H-purin-9-yl)-1-deoxy-N-ethyl-β-D-ribofuranuronamide (NECA) and the selective 1-deoxy-1-[6-[[(3-iodophenyl)methyl]amino]-9H-purin-9-yl]-N-methyl-β-D-ribofuranuronamide (IB-MECA). Mol. dynamics simulations of the wild-type A3R in complex with both agonists, combined with in vitro mutagenic studies revealed important residues for binding. Further, mol. mechanics-generalized Born surface area calcns. were able to distinguish mutations that reduce or negate agonistic activity from those that maintained or increased the activity. Our studies reveal that selectivity of IB-MECA toward A3R requires not only direct interactions with residues within the orthosteric binding area but also with remote residues. Although V1695.30 is considered to be a selectivity filter for A3R binders, when it was mutated to glutamic acid or alanine, the activity of IB-MECA increased by making new van der Waals contacts with TM5. This result may have implications in the design of new A3R agonists.
Cited By
This article is cited by 1 publications.
- Barbara Preti, Anna Suchankova, Giuseppe Deganutti, Michele Leuenberger, Kerry Barkan, Iga Manulak, Xianglin Huang, Sabrina Carvalho, Graham Ladds, Martin Lochner. Discovery and Structure–Activity Relationship Studies of Novel Adenosine A1 Receptor-Selective Agonists. Journal of Medicinal Chemistry 2022, 65 (21) , 14864-14890. https://doi.org/10.1021/acs.jmedchem.2c01414
Abstract
Scheme 1
Scheme 1. Synthesis of 4a,b and 5a,baaReagents and conditions: (a) diethyl oxalate, NaH 60%, toluene dry, 50°C, 2 h; (b) NH2NH2 80%, EtOH, reflux, 90 min; (c) (i) NaH 60%, DMF dry, 0 °C, 15 min, (ii) CH3I, rt, 1 h.
Scheme 2
Scheme 2. Synthesis of 10a–caaReagents and conditions: (a) paraformaldehyde, 33% HBr in AcOH, 90 °C, 3.5 h; (b) Ν-methylmorpholine-Ν-oxide, MeCN dry, rt, 24 h; (c) NH2NH2 (80%), HCl 36%, EtOH, 90°C, 1 h; (d) POCl3, 110 °C, 2.5–8 h; (e) HNR1R2, EtOH, reflux, 2 h.
Scheme 3
Scheme 3. Synthesis of 15a–caaReagents and conditions: (a) paraformaldehyde, 33% HBr in AcOH, 90 °C, 3.5 h; (b) Ν-methylmorpholine-Ν-oxide, MeCN dry, rt, 24 h; (c) NH2NH2 (80%), HCl 36%, EtOH, 90 °C, 1 h; (d) POCl3, 110 °C, 2.5–8 h; (e) HNR1R2, EtOH, reflux, 2 h.
Figure 1
Figure 1. Characterization of 7-amino-pyrazolo[3,4-d]pyridazines at human A1R and A3R. (A and B) Cells expressing either human A1R (A) or A3R (B) were exposed to 10 μM forskolin and stimulated with increasing concentrations of NECA for 30 min in the presence of a 1 μM concentration of the test compound, and the cAMP accumulation was quantified. (C) cAMP accumulation was measured as detailed in part A using multiple concentrations of 10b. Using pEC50 values, Schild regression analysis was conducted to calculate pA2/pKb values. All values are mean ± SEM expressed as percentage forskolin inhibition, relative to NECA. n ≥ 3 independent experimental repeats were performed in duplicate.
Figure 2
Figure 2. Inhibition of BRET between CA200645 at NLuc-A1R and Nluc-A3R by 10b and 10a. HEK293 cells expressing Nluc-A1R (A) or Nluc-A3R (B) were treated with 5 nM or 20 nM CA200645, respectively, enabling concentration-dependent decreases in the BRET ratio at 10 min to be determined with the response normalized to DMSO. Binding curves were fitted with the Cheng Prusoff equation built into GraphPad Prism 9.3 to enable estimates of the pKi. (19) Comparison of pKi values for A1R (C) and A3R (D) as determined via BRET binding. Each data point represents the mean ± SEM of at least three experiments performed in duplicate. The statistical significance compared to NECA was determined, at p < 0.05, through one-way ANOVA with Dunnett’s post-test (*, p < 0.05; ***, p < 0.001). #Compounds did not fully displace CA200645, so pKi values are estimates preventing statistical analysis.
Figure 3
Figure 3. (A–C) 100 ns MD simulations of 10a–c inside the orthosteric binding area of A1R. (D) 100 ns MD simulations of 15b inside the orthosteric binding area of A1R. Starting structures are shown (docking pose), and representative frames from MD simulations, receptor–ligand interaction frequency histograms, and RMSD plots of proteins (RMSDprotein; blue plots) and ligand heavy atoms (RMSDligand; red plots) inside the orthosteric binding area of WT A1R or A3R. Bars are plotted only for residues with interaction frequencies ≥0.2. Color scheme: ligand = brown sticks, receptor = white cartoon and sticks, hydrogen bonding interactions = yellow (dashes or bars), π–π interactions = green (dashes or bars), hydrophobic interactions = gray, water bridges = blue. For the protein models of A1R in complex with 10a–c or 15b, the experimental structure of the inactive form of A1R in complex with an antagonist (PDB ID 5UEN (4)) was used.
References
ARTICLE SECTIONSThis article references 37 other publications.
- 1Fredholm, B. B.; IJzerman, A. P.; Jacobson, K. A.; Linden, J.; Muller, C. E.; Müller, C. E. International Union of Basic and Clinical Pharmacology. LXXXI. Nomenclature and Classification of Adenosine Receptors ─ An Update. Pharmacol. Rev. 2011, 63, 1– 34, DOI: 10.1124/pr.110.003285[Crossref], [PubMed], [CAS], Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXjslGisrY%253D&md5=e184c127e06b800bbcf15402ed27f02aInternational union of basic and clinical pharmacology. LXXXI. Nomenclature and classification of adenosine receptors - an updateFredholm, Bertil B.; IJzerman, Adriaan P.; Jacobson, Kenneth A.; Linden, Joel; Mueller, Christa E.Pharmacological Reviews (2011), 63 (1), 1-34CODEN: PAREAQ; ISSN:0031-6997. (American Society for Pharmacology and Experimental Therapeutics)A review. In the 10 years since our previous International Union of Basic and Clin. Pharmacol. report on the nomenclature and classification of adenosine receptors, no developments have led to major changes in the recommendations. However, there have been so many other developments that an update is needed. The fact that the structure of one of the adenosine receptors has recently been solved has already led to new ways of in silico screening of ligands. The evidence that adenosine receptors can form homo- and hetero-multimers has accumulated, but the functional significance of such complexes remains unclear. The availability of mice with genetic modification of all the adenosine receptors has led to a clarification of the functional roles of adenosine, and to excellent means to study the specificity of drugs. There are also interesting assocns. between disease and structural variants in one or more of the adenosine receptors. Several new selective agonists and antagonists have become available. They provide improved possibilities for receptor classification. There are also developments hinting at the usefulness of allosteric modulators. Many drugs targeting adenosine receptors are in clin. trials, but the established therapeutic use is still very limited.
- 2Schenone, S.; Brullo, C.; Musumeci, F.; Bruno, O.; Botta, M. A1 Receptors Ligands: Past, Present and Future Trends. Curr. Top. Med. Chem. 2010, 10, 878– 901, DOI: 10.2174/156802610791268729[Crossref], [PubMed], [CAS], Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXptVegsL0%253D&md5=19dd32bfed13ee6685e7b31f952ae754A1 receptors ligands: past, present and future trendsSchenone, S.; Brullo, C.; Musumeci, F.; Bruno, O.; Botta, M.Current Topics in Medicinal Chemistry (Sharjah, United Arab Emirates) (2010), 10 (9), 878-901CODEN: CTMCCL; ISSN:1568-0266. (Bentham Science Publishers Ltd.)A review. Adenosine is a neuromodulator that interacting with four receptors, A1, A2A, A2B, and A3, is involved in the regulation of several biol. functions in different organs and tissues, including the central nervous system, the cardiovascular system and the airways; many pathophysiol. states are assocd. with changes of adenosine levels. For these reasons pharmaceutical companies and academicians performed intense efforts to obtain agonists, antagonists and allosteric enhancers selective for each adenosine receptor subtypes as potential clin. candidates. In fact, therapeutic modulation of the adenosine system could offer the possibility of a "soft" treatment of different diseases, but, due to the ubiquitous distribution of adenosine and of its receptors, the challenge in therapy development depends from specificity for the different receptor subtypes. Some A1 agonists and antagonists, very potent and selective, reached clin. trials for the treatment of different diseases. A1 agonists are clin. candidates for atrial arrhythmias, angina, type 2 diabetes and in pain management, while A1 antagonists are in study as potassium-sparing diuretics with kidney-protecting properties and in chronic heart diseases. Several reviews, recently published and herein cited, reported in detail the biol. and clin. aspects of such mols. This review focuses on the A1 adenosine receptor (A1AR) ligands, both agonists and antagonists, appeared in the literature in the last few years, together with their potential therapeutic application, pointing the attention on their chem. structures and SAR (Structure Activity Relationship) and also reporting new findings on preclin. or clin. trials of some important A1AR ligands synthesized in the past.
- 3Squarcialupi, L.; Catarzi, D.; Varano, F.; Betti, M.; Falsini, M.; Vincenzi, F.; Ravani, A.; Ciancetta, A.; Varani, K.; Moro, S.; Colotta, V. Structural Refinement of Pyrazolo[4,3-d]Pyrimidine Derivatives to Obtain Highly Potent and Selective Antagonists for the Human A3 Adenosine Receptor. Eur. J. Med. Chem. 2016, 108, 117– 133, DOI: 10.1016/j.ejmech.2015.11.015[Crossref], [PubMed], [CAS], Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvFantLjL&md5=33cb30d67894345d0d957436482018caStructural refinement of pyrazolo[4,3-d]pyrimidine derivatives to obtain highly potent and selective antagonists for the human A3 adenosine receptorSquarcialupi, Lucia; Catarzi, Daniela; Varano, Flavia; Betti, Marco; Falsini, Matteo; Vincenzi, Fabrizio; Ravani, Annalisa; Ciancetta, Antonella; Varani, Katia; Moro, Stefano; Colotta, VittoriaEuropean Journal of Medicinal Chemistry (2016), 108 (), 117-133CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)In previous research, the authors identified some 7-oxo- and 7-acylamino-substituted pyrazolo[4,3-d]pyrimidine derivs. as potent and selective human (h) A3 adenosine receptor (AR) antagonists. Herein the authors report on the structural refinement of this class of antagonists aimed at achieving improved receptor-ligand recognition. Hence, substituents with different steric bulk, flexibility and lipophilicity (Me, Ar, heteroaryl, CH2Ph) were introduced at the 5- and 2-positions of the bicyclic scaffold of both the 7-oxo and 7-amino derivs., and acyl residues were appended on the 7-amino group of the latter. All the 2-phenylpyrazolo[4,3-d]pyrimidin-7-amines and 7-acylamines bearing a 4-methoxyphenyl- or a 2-thienyl group at the 5-position showed high hA3 affinity and selectivity. In particular, the 2-phenyl-5-(2-thienyl)-pyrazolo[4,3-d]pyrimidin-7-(4-methoxybenzoyl)amine 25 (Ki = 0.027 nM) is one of the most potent and selective hA3 antagonists reported so far. By using an in silico receptor-driven approach the obtained binding data were rationalized and the mol. bases of the obsd. hA3 AR affinities were critically described.
- 4Sun, B.; Bachhawat, P.; Chu, M. L.-H.; Wood, M.; Ceska, T.; Sands, Z. A.; Mercier, J.; Lebon, F.; Kobilka, T. S.; Kobilka, B. K. Crystal Structure of the Adenosine A2A Receptor Bound to an Antagonist Reveals a Potential Allosteric Pocket. Proc. Natl. Acad. Sci. U. S. A. 2017, 114, 2066– 2071, DOI: 10.1073/pnas.1621423114[Crossref], [PubMed], [CAS], Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXitFCksb8%253D&md5=58ac85e930066ef7a6788c4d7f3a72b3Crystal structure of the adenosine A2A receptor bound to an antagonist reveals a potential allosteric pocketSun, Bingfa; Bachhawat, Priti; Chu, Matthew Ling-Hon; Wood, Martyn; Ceska, Tom; Sands, Zara A.; Mercier, Joel; Lebon, Florence; Kobilka, Tong Sun; Kobilka, Brian K.Proceedings of the National Academy of Sciences of the United States of America (2017), 114 (8), 2066-2071CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The adenosine A2A receptor (A2AR) has long been implicated in cardiovascular disorders. As more selective A2AR ligands are being identified, its roles in other disorders, such as Parkinson's disease, are starting to emerge, and A2AR antagonists are important drug candidates for non-dopaminergic anti-Parkinson treatment. Here, the authors report the crystal structure of A2A receptor bound to compd. 1, a novel A2AR/N-methyl-D-aspartate (NMDA) receptor subtype 2B (NR2B; NMDA receptor 2B) dual antagonist and potential anti-Parkinson candidate compd., at 3.5 Å resoln. The A2A receptor with a cytochrome b562-RIL (BRIL) fusion (A2AR-BRIL) in intracellular loop 3 (ICL3) was crystd. in detergent micelles using vapor-phase diffusion. Whereas A2AR-BRIL bound to antagonist ZM241385 was previously been crystd. in the lipidic cubic phase (LCP), structural differences in the compd. 1-bound A2AR-BRIL prevented formation of the lattice obsd. with the ZM241385-bound receptor. The crystals grew with a type II crystal lattice in contrast to the typical type I packing seen from membrane protein structures crystd. in LCP. Compd. 1 bound in a position that overlapped with the native ligand, adenosine, but its methoxyphenyl group extended to an exosite not previously obsd. in other A2AR structures. Structural anal. revealed that compd. 1 binding resulted in the unique conformations of 2 Tyr residues (Tyr91.35 and Tyr2717.36) which are crit. for the formation of the exosite. The structure revealed insights into antagonist binding that were not obsd. in other A2AR structures, highlighting flexibility in the binding pocket that may facilitate the development of A2AR-selective compds. for the treatment of Parkinson's disease.
- 5el-Hashim, A.; D’Agostino, B.; Matera, M. G.; Page, C. Characterization of adenosine receptors involved in adenosine-induced bronchoconstriction in allergic rabbits. Br. J. Pharmacol. 1996, 119, 1262– 1268, DOI: 10.1111/j.1476-5381.1996.tb16031.x[Crossref], [PubMed], [CAS], Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28Xntlentbo%253D&md5=58bb082ce7d52a888fd397437f0b015dCharacterization of adenosine receptors involved in adenosine-induced bronchoconstriction in allergic rabbitsEl-Hashim, Ahmed; D'Agostino, Bruno; Matera, Maria Gabriella; Page, CliveBritish Journal of Pharmacology (1996), 119 (6), 1262-1268CODEN: BJPCBM; ISSN:0007-1188. (Stockton)Recent work has suggested that adenosine may be involved in asthma via the activation of A1 receptors. However, the role of the recently cloned A3 receptor in airways is largely unknown. In the present study, the authors have investigated the role of the A3 receptor in adenosine-induced bronchoconstriction in allergic rabbits. Aerosol challenge of antigen (Ag) immunized rabbits with the adenosine precursor, AMP, resulted in a dose-dependent fall in dynamic compliance (Cdyn). The max. fall in Cdyn in these rabbits was significantly greater than that in litter matched, sham immunized animals. However, there was no significant difference in the max. increase in airways resistance (RL) between Ag and sham immunized rabbits. Aerosol challenge of Ag immunized rabbits with cyclopentyl-adenosine (CPA) (A1-receptor agonist) elicited a dose-dependent fall in Cdyn in Ag immunized rabbits and the max. fall in Cdyn in these rabbits was significantly greater than that obsd. in sham immunized rabbits. Similarly, CPA induced dose-dependent increases in RL in Ag immunized rabbits whereas sham immunized rabbits failed to respond to CPA within the same dose range. The max. increase in RL in Ag immunized rabbits was significantly greater than that of sham immunized rabbits. Aerosol challenge of either Ag or sham immunized rabbits with the A3 agonist aminophenylethyladenosine (APNEA) did not elicit dose-dependent changes in either RL or Cdyn. Moreover, there was no significant difference in the max. response, measured by either parameter, between the two animal groups. These data provide further evidence for a role of the A1 receptor in the airways, but do not support a role for the A3 receptor in adenosine-induced bronchoconstriction in the allergic rabbit.
- 6Brown, R. A.; Spina, D.; Page, C. P. Adenosine receptors and asthma. Br. J. Pharmacol. 2008, 153, S446– S456, DOI: 10.1038/bjp.2008.22[Crossref], [PubMed], [CAS], Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXislSjurg%253D&md5=7adce9e6d9f21f823fb63acc5c593e7dAdenosine receptors and asthmaBrown, R. A.; Spina, D.; Page, C. P.British Journal of Pharmacology (2008), 153 (Suppl. 1), S446-S456CODEN: BJPCBM; ISSN:0007-1188. (Nature Publishing Group)A review. The accumulation of evidence implicating a role for adenosine in the pathogenesis of asthma has led to investigations into all adenosine receptor subtypes as potential therapeutic targets for the treatment of asthma. Selective A1 receptor antagonists are currently in preclin. development since adenosine has been shown exptl. to mediate various features of asthma through this receptor such as bronchoconstriction, mucus secretion and inflammation. The A2A receptor is expressed on most inflammatory cells implicated in asthma, and as A2A stimulation activates adenylate cyclase and consequently elevates cAMP, selective A2A receptor agonists have now reached clin. development. However, initial reports concerning their efficacy are inconclusive. A2B receptor antagonists are also under investigation based on the rationale that inhibiting the effects of adenosine on mast cells would be beneficial, in addn. to other reported pro-inflammatory effects mediated by the A2B receptor on cells such as airway smooth muscle, epithelial cells and fibroblasts. While the effects in pre-clin. models are promising, their efficacy in the clin. setting has also yet to be reported. Finally, adenosine A3 receptor stimulation has been demonstrated to mediate inhibitory effects on eosinophils since it also elevates cAMP. However, some exptl. reports suggest that A3 antagonists mediate anti-inflammatory effects, thus the rationale for A3 receptor ligands as therapeutic agents remains to be detd. In conclusion, establishing the precise role of adenosine in the pathogenesis of asthma and developing appropriate subtype selective agonists/antagonists represents an exciting opportunity for the development of novel therapeutics for the treatment of asthma.
- 7Johnson, J. A.; Montgomery, A. P.; Starr, E. R.; Ludwig, J.; Trevitt, J. Dose-dependent effects of adenosine antagonists on tacrine-induced tremulous jaw movements. Eur. J. Pharmacol. 2018, 833, 364– 369, DOI: 10.1016/j.ejphar.2018.06.004[Crossref], [PubMed], [CAS], Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXht1KnurzL&md5=4c6380986c52e264c73789307e5f31acDose-dependent effects of adenosine antagonists on tacrine-induced tremulous jaw movementsJohnson, Joel A.; Montgomery, Aaron P.; Starr, Eric R.; Ludwig, Justin; Trevitt, JenniferEuropean Journal of Pharmacology (2018), 833 (), 364-369CODEN: EJPHAZ; ISSN:0014-2999. (Elsevier B.V.)The present study examines the effect of three adenosine receptor antagonists on tremulous jaw movements (TJMs), an animal model of tremor. Forty-five rats were pre-treated with one adenosine antagonist: caffeine (0.0, 5.0, or 10.0 mg/kg; non-selective adenosine receptor antagonist), 8-cyclopentyltheophylline (CPT; 0.0, 5.0, or 10.0 mg/kg; selective adenosine A1 receptor antagonist), or SCH 58261 (0.0 or 8.0 mg/kg; selective adenosine A2A receptor antagonist) followed by TJM induction with tacrine (0.0, 0.75, or 2.5 mg/kg; acetylcholinesterase inhibitor). CPT and SCH 58261 both significantly reduced TJMs while caffeine did not. Unexpectedly, both SCH 58261 and CPT reduced TJMs even in the absence of tacrine. Also, CPT showed a robust redn. of TJMs, achieved at both (5.0 mg/kg) and (10.0 mg/kg) doses and regardless of tacrine dose. In conclusion, this study shows adenosine receptor antagonism to generally suppress low-dose tacrine-induced TJMs. In concert with two prior studies, these results are suggestive of behavioral evidence for a biphasic effect of adenosine A2A receptor antagonists (caffeine and SCH 58261) that is modulated by tacrine, and a model of this effect is proposed.
- 8Cohen, S.; Fishman, P. Targeting the A3 Adenosine Receptor to Treat Cytokine Release Syndrome in Cancer Immunotherapy. Drug Des. Devel. Ther. 2019, 13, 491– 497, DOI: 10.2147/DDDT.S195294[Crossref], [PubMed], [CAS], Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXit1KitrnP&md5=c145fb65e2c605a4e517de8745c73b4aTargeting the A3 adenosine receptor to treat cytokine release syndrome in cancer immunotherapyCohen, Shira; Fishman, PninaDrug Design, Development and Therapy (2019), 13 (), 491-497CODEN: DDDTAQ; ISSN:1177-8881. (Dove Medical Press Ltd.)Cancer patients undergoing immunotherapy may develop cytokine release syndrome (CRS), an inflmmatory cytokine storm condition, followed by neurotoxic manifestations and may be life-threatening. The current treatments for CRS successfully reduce the inflmmatory response but may limit the anticancer effect of the given immunotherapy and fail to overcome the neurotoxic adverse events. Adenosine, a ubiquitous purine nucleoside, induces a plethora of effects in the body via its binding to four adenosine receptors A1, A2a, A2b, and the A3. Highly selective agonists to the A3 adenosine receptor act as inhibitors of proinflmmatory cytokines, possess robust anti-inflmmatory and anticancer activity, and concomitantly, induce neuroprotective effects. Piclidenoson and namodenoson belong to this group of compds., are effective upon oral administration, show an excellent safety profie in human clin. studies, and therefore, may be considered as drug candidates to treat CRS. In this article, the detailed anti-inflmmatory characteristics of these compds. and the rationale to use them as drugs to combat CRS are described.
- 9Lee, J.; Hwang, I.; Lee, J. H.; Lee, H. W.; Jeong, L.-S.; Ha, H. The Selective A3AR Antagonist LJ-1888 Ameliorates UUO-Induced Tubulointerstitial Fibrosis. Am. J. Pathol. 2013, 183, 1488– 1497, DOI: 10.1016/j.ajpath.2013.07.010[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhs1OqsLfI&md5=25e0faae674396045573507dd5b78e48The Selective A3AR Antagonist LJ-1888 Ameliorates UUO-Induced Tubulointerstitial FibrosisLee, Jiyoun; Hwang, Inah; Lee, Jung H.; Lee, Hyuk W.; Jeong, Lak-Shin; Ha, HunjooAmerican Journal of Pathology (2013), 183 (5), 1488-1497CODEN: AJPAA4; ISSN:0002-9440. (Elsevier B.V.)Adenosine in the normal kidney significantly elevates in response to cellular damage. The renal A3 adenosine receptor (A3AR) is up-regulated under stress, but the therapeutic effects of A3AR antagonists on chronic kidney disease are not fully understood. The present study examd. the effect of LJ-1888 [(2R,3R,4S)-2-[2-chloro-6-(3-iodobenzylamino)-9H-purine-9-yl]-tetrahydrothiophene-3,4-diol], a newly developed potent, selective, species-independent, and orally active A3AR antagonist, on unilateral ureteral obstruction (UUO)-induced renal fibrosis. Pretreatment with LJ-1888 inhibited UUO-induced fibronectin and collagen I up-regulation in a dose-dependent manner. Masson's trichrome staining confirmed that LJ-1888 treatment effectively reduced UUO-induced interstitial collagen accumulation. Furthermore, delayed administration of LJ-1888 showed an equiv. therapeutic effect on tubulointerstitial fibrosis to that of losartan. Small-interfering A3AR transfection effectively inhibited transforming growth factor-β1 (TGF-β1)-induced fibronectin and collagen I up-regulation in proximal tubular cells similar to LJ-1888, confirming that the renoprotective effect of LJ-1888 resulted from A3AR blockade. UUO- or TGF-β1-induced c-Jun N-terminal kinase and extracellular signal-regulated kinase phosphorylation decreased significantly after LJ-1888 administration. A3AR blockade reduced UUO- or TGF-β1-induced up-regulation of lysyl oxidase, which induces crosslinking of extracellular matrix, suggesting that LJ-1888 may also regulate extracellular matrix accumulation via post-translational regulation. In conclusion, the present data demonstrate that the A3AR antagonist, LJ-1888, blocked the development and attenuated the progression of renal fibrosis, and they suggest that LJ-1888 may become a new therapeutic modality for renal interstitial fibrosis.
- 10Wang, Z.; Do, C. W.; Avila, M. Y.; Peterson-Yantorno, K.; Stone, R. A.; Gao, Z.-G.; Joshi, B.; Besada, P.; Jeong, L. S.; Jacobson, K. A.; Civan, M. M. Nucleoside-Derived Antagonists to A3 Adenosine Receptors Lower Mouse Intraocular Pressure and Act across Species. Exp. Eye Res. 2010, 90, 146– 154, DOI: 10.1016/j.exer.2009.10.001[Crossref], [PubMed], [CAS], Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsFantbjM&md5=3d11d9a34194272b31e203a158609fd8Nucleoside-derived antagonists to A3 adenosine receptors lower mouse intraocular pressure and act across speciesWang, Zhao; Do, Chi-Wai; Avila, Marcel Y.; Peterson-Yantorno, Kim; Stone, Richard A.; Gao, Zhan-Guo; Joshi, Bhalchandra; Besada, Pedro; Jeong, Lak-Shin; Jacobson, Kenneth A.; Civan, Mortimer M.Experimental Eye Research (2010), 90 (1), 146-154CODEN: EXERA6; ISSN:0014-4835. (Elsevier B.V.)The purpose of the study was to det. whether novel, selective antagonists of human A3 adenosine receptors (ARs) derived from the A3-selective agonist Cl-IB-MECA lower intraocular pressure (IOP) and act across species. IOP was measured invasively with a micropipette by the Servo-Null Micropipette System (SNMS) and by non-invasive pneumotonometry during topical drug application. Antagonist efficacy was also assayed by measuring inhibition of adenosine-triggered shrinkage of native bovine nonpigmented ciliary epithelial (NPE) cells. Five agonist-based A3AR antagonists lowered mouse IOP measured with SNMS tonometry by 3-5 mm Hg within minutes of topical application. Of the 5 agonist derivs., LJ 1251 was the only antagonist to lower IOP measured by pneumotonometry. No effect was detected pneumotonometrically over 30 min following application of the other 4 compds., consonant with slower, smaller responses previously measured non-invasively following topical application of A3AR agonists and the dihydropyridine A3AR antagonist MRS 1191. Latanoprost similarly lowered SNMS-measured IOP, but not IOP measured non-invasively over 30 min. Like MRS 1191, agonist-based A3AR antagonists applied to native bovine NPE cells inhibited adenosine-triggered shrinkage. In summary, the results indicate that antagonists of human A3ARs derived from the potent, selective A3 agonist Cl-IB-MECA display efficacy in mouse and bovine cells, as well. When intraocular delivery was enhanced by measuring mouse IOP invasively, 5 derivs. of the A3AR agonist Cl-IB-MECA lowered IOP but only one rapidly reduced IOP measured non-invasively after topical application. We conclude that derivs. of the highly-selective A3AR agonist Cl-IB-MECA can reduce IOP upon reaching their intraocular target, and that nucleoside-based derivs. are promising A3 antagonists for study in multiple animal models.
- 11Glukhova, A.; Thal, D. M.; Nguyen, A. T.; Vecchio, E. A.; Jörg, M.; Scammells, P. J.; May, L. T.; Sexton, P. M.; Christopoulos, A. Structure of the Adenosine A1 Receptor Reveals the Basis for Subtype Selectivity. Cell 2017, 168, 867– 877, DOI: 10.1016/j.cell.2017.01.042[Crossref], [PubMed], [CAS], Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjvVWrsrg%253D&md5=a5ff676cfedd6e11a899f65555a1ba62Structure of the Adenosine A1 Receptor Reveals the Basis for Subtype SelectivityGlukhova, Alisa; Thal, David M.; Nguyen, Anh T.; Vecchio, Elizabeth A.; Jorg, Manuela; Scammells, Peter J.; May, Lauren T.; Sexton, Patrick M.; Christopoulos, ArthurCell (Cambridge, MA, United States) (2017), 168 (5), 867-877.e13CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The adenosine A1 receptor (A1-AR) is a G-protein-coupled receptor that plays a vital role in cardiac, renal, and neuronal processes but remains poorly targeted by current drugs. We detd. a 3.2 Å crystal structure of the A1-AR bound to the selective covalent antagonist, DU172, and identified striking differences to the previously solved adenosine A2A receptor (A2A-AR) structure. Mutational and computational anal. of A1-AR revealed a distinct conformation of the second extracellular loop and a wider extracellular cavity with a secondary binding pocket that can accommodate orthosteric and allosteric ligands. We propose that conformational differences in these regions, rather than amino-acid divergence, underlie drug selectivity between these adenosine receptor subtypes. Our findings provide a mol. basis for AR subtype selectivity with implications for understanding the mechanisms governing allosteric modulation of these receptors, allowing the design of more selective agents for the treatment of ischemia-reperfusion injury, renal pathologies, and neuropathic pain.
- 12Cheng, R. K. Y.; Segala, E.; Robertson, N.; Deflorian, F.; Doré, A. S.; Errey, J. C.; Fiez-Vandal, C.; Marshall, F. H.; Cooke, R. M. Structures of Human A1 and A2A Adenosine Receptors with Xanthines Reveal Determinants of Selectivity. Structure 2017, 25, 1275– 1285, DOI: 10.1016/j.str.2017.06.012[Crossref], [PubMed], [CAS], Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFOlsbfJ&md5=e759fd60b25beac2d9429809f3b51b02Structures of Human A1 and A2a Adenosine Receptors with Xanthines Reveal Determinants of SelectivityCheng, Robert K. Y.; Segala, Elena; Robertson, Nathan; Deflorian, Francesca; Dore, Andrew S.; Errey, James C.; Fiez-Vandal, Cedric; Marshall, Fiona H.; Cooke, Robert M.Structure (Oxford, United Kingdom) (2017), 25 (8), 1275-1285.e4CODEN: STRUE6; ISSN:0969-2126. (Elsevier Ltd.)The adenosine A1 and A2a receptors belong to the purinergic family of G protein-coupled receptors, and regulate diverse functions of the cardiovascular, respiratory, renal, inflammation, and CNS. Xanthines such as caffeine and theophylline are weak, non-selective antagonists of adenosine receptors. Here we report the structure of a thermostabilized human A1 receptor at 3.3 Å resoln. with PSB36, an A1-selective xanthine-based antagonist. This is compared with structures of the A2a receptor with PSB36 (2.8 Å resoln.), caffeine (2.1 Å), and theophylline (2.0 Å) to highlight features of ligand recognition which are common across xanthines. The structures of A1R and A2aR were analyzed to identify the differences that are important selectivity determinants for xanthine ligands, and the role of T2707.35 in A1R (M2707.35 in A2aR) in conferring selectivity was confirmed by mutagenesis. The structural differences confirmed to lead to selectivity can be utilized in the design of new subtype-selective A1R or A2aR antagonists.
- 13Draper-Joyce, C. J.; Khoshouei, M.; Thal, D. M.; Liang, Y.-L.; Nguyen, A. T. N.; Furness, S. G. B.; Venugopal, H.; Baltos, J.-A.; Plitzko, J. M.; Danev, R.; Baumeister, W.; May, L. T.; Wootten, D.; Sexton, P. M.; Glukhova, A.; Christopoulos, A. Structure of the Adenosine-Bound Human Adenosine A1 Receptor-Gi Complex. Nature 2018, 558, 559– 563, DOI: 10.1038/s41586-018-0236-6[Crossref], [PubMed], [CAS], Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtFOrsL3P&md5=f31a757364c3ff512bd5548b31e7f1bdStructure of the adenosine-bound human adenosine A1 receptor-Gi complexDraper-Joyce, Christopher J.; Khoshouei, Maryam; Thal, David M.; Liang, Yi-Lynn; Nguyen, Anh T. N.; Furness, Sebastian G. B.; Venugopal, Hariprasad; Baltos, Jo-Anne; Plitzko, Jurgen M.; Danev, Radostin; Baumeister, Wolfgang; May, Lauren T.; Wootten, Denise; Sexton, Patrick M.; Glukhova, Alisa; Christopoulos, ArthurNature (London, United Kingdom) (2018), 558 (7711), 559-563CODEN: NATUAS; ISSN:0028-0836. (Nature Research)The class A adenosine A1 receptor (A1R) is a G-protein-coupled receptor that preferentially couples to inhibitory Gi/o heterotrimeric G proteins, has been implicated in numerous diseases, yet remains poorly targeted. Here we report the 3.6 Å structure of the human A1R in complex with adenosine and heterotrimeric Gi2 protein detd. by Volta phase plate cryo-electron microscopy. Compared to inactive A1R, there is contraction at the extracellular surface in the orthosteric binding site mediated via movement of transmembrane domains 1 and 2. At the intracellular surface, the G protein engages the A1R primarily via amino acids in the C terminus of the Gαi α5-helix, concomitant with a 10.5 Å outward movement of the A1R transmembrane domain 6. Comparison with the agonist-bound β2 adrenergic receptor-Gs-protein complex reveals distinct orientations for each G-protein subtype upon engagement with its receptor. This active A1R structure provides mol. insights into receptor and G-protein selectivity.
- 14Stampelou, M.; Suchankova, A.; Tzortzini, E.; Dhingra, L.; Barkan, K.; Lougiakis, N.; Marakos, P.; Pouli, N.; Ladds, G.; Kolocouris, A. Novel Pyrazolo[3,4-c]Pyridine Antagonists with Nanomolar Affinity for A1/A3 Adenosine Receptors: Binding Kinetics and Exploration of Their Binding Profile Using Mutagenesis Experiments, MD Simulations and TI/MD Calculations. ChemRxiv 2021, DOI: 10.26434/chemrxiv-2021-j18mg-v3 .
- 15Catarzi, D.; Colotta, V.; Varano, F.; Poli, D.; Squarcialupi, L.; Filacchioni, G.; Varani, K.; Vincenzi, F.; Borea, P. A.; Dal Ben, D.; Lambertucci, C.; Cristalli, G. Pyrazolo[1,5-c]Quinazoline Derivatives and Their Simplified Analogues as Adenosine Receptor Antagonists: Synthesis, Structure-Affinity Relationships and Molecular Modeling Studies. Bioorg. Med. Chem. 2013, 21, 283– 294, DOI: 10.1016/j.bmc.2012.10.031[Crossref], [PubMed], [CAS], Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhslSmu7zI&md5=fd01538f48cca49e41bd80f0c5751304Pyrazolo[1,5-c]quinazoline derivatives and their simplified analogues as adenosine receptor antagonists: Synthesis, structure-affinity relationships and molecular modeling studiesCatarzi, Daniela; Colotta, Vittoria; Varano, Flavia; Poli, Daniela; Squarcialupi, Lucia; Filacchioni, Guido; Varani, Katia; Vincenzi, Fabrizio; Borea, Pier Andrea; Dal Ben, Diego; Lambertucci, Catia; Cristalli, GloriaBioorganic & Medicinal Chemistry (2013), 21 (1), 283-294CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)A no. of 5-oxo-pyrazolo[1,5-c]quinazolines, bearing at position-2 the (hetero)aryl moiety but also a carboxylate group, were designed as hA3 AR antagonists. This study produced some interesting compds. endowed with good hA3 receptor affinity and high selectivity, being totally inactive at all the other AR subtypes. In contrast, the corresponding 5-ammino derivs. do not bind or bind with very low affinity at the hA3 AR, the only exception being the 5-N-benzoyl compd. I that shows a hA3Ki value in the high μ-molar range. Evaluation of the synthetic intermediates led to the identification of some 5(3)-(2-aminophenyl)-3(5)-(hetero)arylpyrazoles II [R = Ph, 4-MeOC6H4, 2-furyl, 2-thienyl, CO2Et] with modest affinity but high selectivity toward the hA3 AR subtype. Mol. docking of the herein reported tricyclic and simplified derivs. was carried out to depict their hypothetical binding mode to our model of hA3 receptor.
- 16Manetti, F.; Schenone, S.; Bondavalli, F.; Brullo, C.; Bruno, O.; Ranise, A.; Mosti, L.; Menozzi, G.; Fossa, P.; Trincavelli, M. L.; Martini, C.; Martinelli, A.; Tintori, C.; Botta, M. Synthesis and 3D QSAR of new pyrazolo[3,4-b]pyridines: potent and selective inhibitors of A1 adenosine receptors. J. Med. Chem. 2005, 48, 7172– 7185, DOI: 10.1021/jm050407k[ACS Full Text
], [CAS], Google Scholar
16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhtFWku7vL&md5=58d216826db063b0d522f5e9d6a4474cSynthesis and 3D QSAR of New Pyrazolo[3,4-b]pyridines: Potent and Selective Inhibitors of A1 Adenosine ReceptorsManetti, Fabrizio; Schenone, Silvia; Bondavalli, Francesco; Brullo, Chiara; Bruno, Olga; Ranise, Angelo; Mosti, Luisa; Menozzi, Giulia; Fossa, Paola; Trincavelli, Maria Letizia; Martini, Claudia; Martinelli, Adriano; Tintori, Cristina; Botta, MaurizioJournal of Medicinal Chemistry (2005), 48 (23), 7172-7185CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A no. of 4-(amino)pyrazolo[3,4-b]pyridine-5-carboxylic acid esters were synthesized and evaluated for their binding affinity at the A1, A2A, and A3 adenosine receptors (AR), in bovine cortical membranes, as well as for their affinity toward human A1AR (hA1AR) (A1 purinoceptor antagonists). Some of the new compds. were characterized by a high affinity and selectivity toward the A1 receptor subtype, showing a significant improvement in comparison with other pyrazolo-pyridines previously reported in the literature. In particular two compds., both of them bearing a [(p-methoxyphenyl)ethyl]amino side chain presented Ki values of 6 and 7 nM, resp. To rationalize the relationship between structure and affinity of the novel compds., a 3D QSAR model was also generated starting from compds. belonging to different classes of known A1AR antagonists. - 17Tuccinardi, T.; Zizzari, A. T.; Brullo, C.; Daniele, S.; Musumeci, F.; Schenone, S.; Trincavelli, M. L.; Martini, C.; Martinelli, A.; Giorgi, G.; Botta, M. Substituted pyrazolo[3,4-b]pyridines as human A1 adenosine antagonists: Developments in understanding the receptor stereoselectivity. Org. Biomol. Chem. 2011, 9, 4448– 4455, DOI: 10.1039/c0ob01064b[Crossref], [PubMed], [CAS], Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXntVGgt70%253D&md5=fa192f0519397853d7bef48f22ccb9edSubstituted pyrazolo[3,4-b]pyridines as human A1 adenosine antagonists: Developments in understanding the receptor stereoselectivityTuccinardi, Tiziano; Zizzari, Alessandra Tania; Brullo, Chiara; Daniele, Simona; Musumeci, Francesca; Schenone, Silvia; Trincavelli, Maria Letizia; Martini, Claudia; Martinelli, Adriano; Giorgi, Gianluca; Botta, MaurizioOrganic & Biomolecular Chemistry (2011), 9 (12), 4448-4455CODEN: OBCRAK; ISSN:1477-0520. (Royal Society of Chemistry)A1 adenosine receptor antagonists have been proposed to possess an interesting range of potential therapeutic applications. The authors have already reported the synthesis and the biol. characterization of a family of pyrazolo[3,4-b]pyridine derivs. as A1 adenosine ligands endowed with an antagonistic profile. In the present work, the authors report the LC sepn. of enantiomers of the authors' most active A1 antagonists together with the detn. of their abs. configuration by x-ray crystal structure anal. Biol. assays confirmed a different activity for the two enantiomers, with the R one showing the higher human A1AR affinity. The authors also developed a homol. model of this receptor subtype to suggest a binding disposition of the ligands into the hA1AR. All of the obtained data suggest that the compd.'s chirality plays a key role in A1 affinity.
- 18Cheong, S. L.; Venkatesan, G.; Paira, P.; Jothibasu, R.; Mandel, A. L.; Federico, S.; Spalluto, G.; Pastorin, G. Pyrazolo Derivatives as Potent Adenosine Receptor Antagonists: An Overview on the Structure-Activity Relationships. Int. J. Med. Chem. 2011, 480652, DOI: 10.1155/2011/480652[Crossref], [PubMed], [CAS], Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXltV2gtbw%253D&md5=e31940dc0a60cd1c30079d3edbcdbf56Pyrazolo derivatives as potent adenosine receptor antagonists: an overview on the structure-activity relationshipsCheong, Siew Lee; Venkatesan, Gopalakrishnan; Paira, Priyankar; Jothibasu, Ramasamy; Mandel, Alexander Laurence; Federico, Stephanie; Spalluto, Giampiero; Pastorin, GiorgiaInternational Journal of Medicinal Chemistry (2011), (), 480652, 15 pp.CODEN: IJMCCH; ISSN:2090-2077. (Hindawi Publishing Corp.)A review. In the past few decades, medicinal chem. research towards potent and selective antagonists of human adenosine receptors (namely, A1, A2A, A2B, and A3) has been evolving rapidly. These antagonists are deemed therapeutically beneficial in several pathol. conditions including neurol. and renal disorders, cancer, inflammation, and glaucoma. Up to this point, many classes of compds. have been successfully synthesized and identified as potent human adenosine receptor antagonists. In this paper, an overview of the structure-activity relationship (SAR) profiles of promising nonxanthine pyrazolo derivs. is reported and discussed. We have emphasized the SAR for some representative structures such as pyrazolo-[4,3-e]-1,2,4-triazolo-[1,5-c]pyrimidines; pyrazolo-[3,4-c] or -[4,3-c]quinolines; pyrazolo-[4,3-d]pyrimidinones; pyrazolo-[3,4-d]pyrimidines and pyrazolo-[1,5-a]pyridines. This overview not only clarifies the structural requirements deemed essential for affinity towards individual adenosine receptor subtypes, but it also sheds light on the rational design and optimization of existing structural templates to allow us to conceive new, more potent adenosine receptor antagonists.
- 19Barkan, K.; Lagarias, P.; Stampelou, M.; Stamatis, D.; Hoare, S.; Safitri, D.; Klotz, K.-N.; Vrontaki, E.; Kolocouris, A.; Ladds, G. Pharmacological Characterisation of Novel Adenosine A3 Receptor Antagonists. Sci. Rep. 2020, 10, 20781, DOI: 10.1038/s41598-020-74521-y[Crossref], [PubMed], [CAS], Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisFWltbrL&md5=3710ac8acc7ee7d2b072c0ee95fa3bffPharmacological characterization of novel adenosine A3 receptor antagonistsBarkan, Kerry; Lagarias, Panagiotis; Stampelou, Margarita; Stamatis, Dimitrios; Hoare, Sam; Safitri, Dewi; Klotz, Karl-Norbert; Vrontaki, Eleni; Kolocouris, Antonios; Ladds, GrahamScientific Reports (2020), 10 (1), 20781CODEN: SRCEC3; ISSN:2045-2322. (Nature Research)The adenosine A3 receptor (A3R) belongs to a family of four adenosine receptor (AR) subtypes which all play distinct roles throughout the body. A3R antagonists have been described as potential treatments for numerous diseases including asthma. Given the similarity between (adenosine receptors) orthosteric binding sites, obtaining highly selective antagonists is a challenging but crit. task. Here we screen 39 potential A3R, antagonists using agonist-induced inhibition of cAMP. Pos. hits were assessed for AR subtype selectivity through cAMP accumulation assays. The antagonist affinity was detd. using Schild anal. (pA2 values) and fluorescent ligand binding. Structure-activity relationship investigations revealed that loss of the 3-(dichlorophenyl)-isoxazolyl moiety or the arom. nitrogen heterocycle with nitrogen at α-position to the carbon of carboximidamide group significantly attenuated K18 antagonistic potency. Mutagenic studies supported by mol. dynamic simulations combined with Mol. Mechanics-Poisson Boltzmann Surface Area calcns. identified the residues important for binding in the A3R orthosteric site. We demonstrate that K18, which contains a 3-(dichlorophenyl)-isoxazole group connected through carbonyloxycarboximidamide fragment with a 1,3-thiazole ring, is a specific A3R (< 1 μM) competitive antagonist. Finally, we introduce a model that enables ests. of the equil. binding affinity for rapidly disassocg. compds. from real-time fluorescent ligand-binding studies. These results demonstrate the pharmacol. characterization of a selective competitive A3R antagonist and the description of its orthosteric binding mode. Our findings may provide new insights for drug discovery.
- 20Lagarias, P.; Barkan, K.; Tzortzini, E.; Stampelou, M.; Vrontaki, E.; Ladds, G.; Kolocouris, A. Insights to the Binding of a Selective Adenosine A3 Receptor Antagonist Using Molecular Dynamic Simulations, MM-PBSA and MM-GBSA Free Energy Calculations, and Mutagenesis. J. Chem. Inf. Model. 2019, 59, 5183– 5197, DOI: 10.1021/acs.jcim.9b00751[ACS Full Text
], [CAS], Google Scholar
20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitFGrsL7I&md5=3ad8c6ddc392513238915acb47a00fa0Insights to the Binding of a Selective Adenosine A3 Receptor Antagonist Using Molecular Dynamic Simulations, MM-PBSA and MM-GBSA Free Energy Calculations, and MutagenesisLagarias, Panagiotis; Barkan, Kerry; Tzortzini, Eva; Stampelou, Margarita; Vrontaki, Eleni; Ladds, Graham; Kolocouris, AntoniosJournal of Chemical Information and Modeling (2019), 59 (12), 5183-5197CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Adenosine A3 receptor (A3R) is a promising drug target cancer and for a no. of other conditions like inflammatory diseases, including asthma and rheumatoid arthritis, glaucoma, chronic obstructive pulmonary disease, and ischemic injury. Currently, there is no exptl. detd. structure of A3R. We explored the binding profile of O4-{[3-(2,6-dichlorophenyl)-5-methylisoxazol-4-yl]carbonyl}-2-methyl-1,3-thiazole-4-carbohydroximamide (K18), which is a new specific and competitive antagonist at the orthosteric binding site of A3R. MD simulations and MM-GBSA calcns. of the WT A3R in complex with K18 combined with in vitro mutagenic studies show that the most plausible binding conformation for the dichlorophenyl group of K18 is oriented toward trans-membrane helixes (TM) 5, 6 and reveal important residues for binding. Further, MM-GBSA calcns. distinguish mutations that reduce or maintain or increase antagonistic activity. Our studies show that selectivity of K18 toward A3R is defined not only by direct interactions with residues within the orthosteric binding area but also by remote residues playing a significant role. Although V1695.30 is considered to be a selectivity filter for A3R binders, when it was mutated to glutamic acid, K18 maintained antagonistic potency, in agreement with our previous results obtained for agonists binding profile investigation. Mutation of the direct interacting residue L903.32 in the low region and the remote L2647.35 in the middle/upper region to alanine increases antagonistic potency, suggesting an empty space in the orthosteric area available for increasing antagonist potency. These results approve the computational model for the description of K18 binding at A3R, which we previously performed for agonists binding to A3R, and the design of more effective antagonists based on K18. - 21Papastathopoulos, A.; Lougiakis, N.; Kostakis, I. K.; Marakos, P.; Pouli, N.; Pratsinis, H.; Kletsas, D. New Bioactive 5-Arylcarboximidamidopyrazolo[3,4-c]Pyridines: Synthesis, Cytotoxic Activity, Mechanistic Investigation and Structure-Activity Relationships. Eur. J. Med. Chem. 2021, 218, 113387, DOI: 10.1016/j.ejmech.2021.113387[Crossref], [PubMed], [CAS], Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXns1ehs7s%253D&md5=111d0614bdaa7ddb02eb06e76d1c9eeeNew bioactive 5-arylcarboximidamidopyrazolo[3,4-c]pyridines: Synthesis, cytotoxic activity, mechanistic investigation and structure-activity relationshipsPapastathopoulos, Athanasios; Lougiakis, Nikolaos; Kostakis, Ioannis K.; Marakos, Panagiotis; Pouli, Nicole; Pratsinis, Harris; Kletsas, DimitrisEuropean Journal of Medicinal Chemistry (2021), 218 (), 113387CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)In this study, a series of novel substituted pyrazolo[3,4-c]pyridin-5-ylamidines I [R = Me, (4-methoxyphenyl)methyl; R1 = H, (3,4,5-trimethoxyphenyl)aminyl, cyclohexylaminyl, morpholin-4-yl; R2 = H, Ph; R3 = H, CF3; R4 = H, 4-methylpiperazin-1-yl] and II was synthesized and their cytotoxicity against three cancer cell lines (MDA-MB-231, HT-1080, PC-3), as well as a human normal cell line (AG01523) was evaluated. A no. of derivs. I and II could strongly reduce cancer cells proliferation and exhibit apoptotic induction capability, while reasonable structure-activity relationships could be extd. Certain analogs were endowed with low toxicity against normal cells. Cell cycle anal. revealed that most of the active compds. induced a G0/G1 arrest of HT-1080 cells. Moreover, the potential mechanisms of the cytotoxic activity of the promising compds. I and II were investigated in HT-1080 cells, upon study of their effects on the phosphorylation of Akt, ERK and p38 MAPK. Most of the active derivs. inhibit phosphorylation of Akt and ERK and/or induce p38 MAPK phosphorylation, providing a potential indication on the mode of action of this class.
- 22Bento, A. P.; Gaulton, A.; Hersey, A.; Bellis, L. J.; Chambers, J.; Davies, M.; Krüger, F. A.; Light, Y.; Mak, L.; McGlinchey, S.; Nowotka, M.; Papadatos, G.; Santos, R.; Overington, J. P. The ChEMBL Bioactivity Database: An Update. Nucleic Acids Res. 2014, 42, D1083– D1090, DOI: 10.1093/nar/gkt1031[Crossref], [PubMed], [CAS], Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXoslWl&md5=31b832d03d56ea3065d7aa29618362bcThe ChEMBL bioactivity database: an updateBento, A. Patricia; Gaulton, Anna; Hersey, Anne; Bellis, Louisa J.; Chambers, Jon; Davies, Mark; Krueger, Felix A.; Light, Yvonne; Mak, Lora; McGlinchey, Shaun; Nowotka, Michal; Papadatos, George; Santos, Rita; Overington, John P.Nucleic Acids Research (2014), 42 (D1), D1083-D1090CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)ChEMBL is an open large-scale bioactivity database (https://www.ebi.ac.uk/chembl), previously described in the 2012 Nucleic Acids Research Database Issue. Since then, a variety of new data sources and improvements in functionality have contributed to the growth and utility of the resource. In particular, more comprehensive tracking of compds. from research stages through clin. development to market is provided through the inclusion of data from United States Adopted Name applications; a new richer data model for representing drug targets has been developed; and a no. of methods have been put in place to allow users to more easily identify reliable data. Finally, access to ChEMBL is now available via a new Resource Description Framework format, in addn. to the web-based interface, data downloads and web services.
- 23Hawkins, P. C. D.; Skillman, A. G.; Nicholls, A. Comparison of Shape-Matching and Docking as Virtual Screening Tools. J. Med. Chem. 2007, 50, 74– 82, DOI: 10.1021/jm0603365[ACS Full Text
], [CAS], Google Scholar
23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xhtlansb%252FF&md5=6f97f5c0cc092b4e225f7c2656c1bcf6Comparison of Shape-Matching and Docking as Virtual Screening ToolsHawkins, Paul C. D.; Skillman, A. Geoffrey; Nicholls, AnthonyJournal of Medicinal Chemistry (2007), 50 (1), 74-82CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Ligand docking is a widely used approach in virtual screening. In recent years a large no. 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 no. 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. - 24Pei, Y.; Wickham, B. O. S. Regioselective Syntheses of 3-Aminomethyl-5-Substituted Isoxazoles: A Facile and Chemoselective Reduction of Azide to Amine by Sodium Borohydride Using 1,3-Propanedithiol as a Catalyst. Tetrahedron Lett. 1993, 34, 7509– 7512, DOI: 10.1016/S0040-4039(00)60386-6[Crossref], [CAS], Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXisVCku74%253D&md5=cb8eb0337ff7315bc607e866bacc2e0cRegioselective syntheses of 3-Aminomethyl-5-substituted isoxazoles: a facile and chemoselective reduction of azide to amine by sodium borohydride using 1,3-propanedithiol as a catalystPei, Yazhong; Wickham, Barbara O. S.Tetrahedron Letters (1993), 34 (47), 7509-12CODEN: TELEAY; ISSN:0040-4039.A series of isoxazole azides I (R = Me, Ph, i-Pr; R1 = N3) were prepd. and reduced selectively to isoxazole amines I (R1 = NH2) in quant. yield by sodium borohydride using 1,3-propanedithiol as a catalyst.
- 25Fadnavis, N. W.; Radhika, K. R. Enantio- and Regiospecific Reduction of Ethyl 4-Phenyl-2,4-Dioxobutyrate with Baker’s Yeast: Preparation of (R)-HPB Ester. Tetrahedron Asymmetry 2004, 15, 3443– 3447, DOI: 10.1016/j.tetasy.2004.09.007[Crossref], [CAS], Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXpt1SqsL0%253D&md5=989deb752b3d4601d170b15688a300b8Enantio- and regiospecific reduction of ethyl 4-phenyl-2,4-dioxobutyrate with baker's yeast: preparation of (R)-HPB esterFadnavis, Nitin W.; Radhika, Kasiraman R.Tetrahedron: Asymmetry (2004), 15 (21), 3443-3447CODEN: TASYE3; ISSN:0957-4166. (Elsevier B.V.)Et 2,4-dioxo-4-phenylbutyrate, obtained by condensation of acetophenone with di-Et oxalate, is reduced enantio- and regiospecifically by baker's yeast in a diisopropyl ether/water two-phase system to give (-)-Et (R)-2-hydroxy-4-oxo-4-phenylbutyrate with an 98% ee in 80% isolated yield. This (hydroxy)keto ester was hydrogenated over Pd-C to obtain (-)-Et (R)-2-hydroxy-4-phenylbutyrate (HPB ester), an important intermediate for the synthesis of ACE inhibitors. Prolonged contact of the redn. product with baker's yeast produced 3-phenyl-3-oxopropanol in 90% yield.
- 26Dang, T. T.; Dang, T. T.; Fischer, C.; Görls, H.; Langer, P. Synthesis of Pyrazole-3-Carboxylates and Pyrazole-1,5-Dicarboxylates by One-Pot Cyclization of Hydrazone Dianions with Diethyl Oxalate. Tetrahedron 2008, 64, 2207– 2215, DOI: 10.1016/j.tet.2007.12.024[Crossref], [CAS], Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsFams7o%253D&md5=9c72ba7ccb12654f2548d94e660e120aSynthesis of pyrazole-3-carboxylates and pyrazole-1,5-dicarboxylates by one-pot cyclization of hydrazone dianions with diethyl oxalateDang, Tung T.; Dang, Tuan T.; Fischer, Christine; Goerls, Helmar; Langer, PeterTetrahedron (2008), 64 (9), 2207-2215CODEN: TETRAB; ISSN:0040-4020. (Elsevier Ltd.)The one-pot cyclization of hydrazone dianions with di-Et oxalate allows a convenient synthesis of pyrazole-3-carboxylates and pyrazole-1,5-dicarboxylates. Under the condition of toluene as solvent and p-TsOH as acid, pyrazole-3-carboxylates are obtained; under the condition of CH2Cl2 as solvent and TFA as acid, pyrazole-1,5-dicarboxylates are obtained.
- 27Yan, Z.; Liu, A.; Huang, M.; Liu, M.; Pei, H.; Huang, L.; Yi, H.; Liu, W.; Hu, A. Design, Synthesis, DFT Study and Antifungal Activity of the Derivatives of Pyrazolecarboxamide Containing Thiazole or Oxazole Ring. Eur. J. Med. Chem. 2018, 149, 170– 181, DOI: 10.1016/j.ejmech.2018.02.036[Crossref], [PubMed], [CAS], Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjvVCgsb0%253D&md5=c7e4e7a0f4bd1b090fdb5fdf41b6a03eDesign, synthesis, DFT study and antifungal activity of the derivatives of pyrazolecarboxamide containing thiazole or oxazole ringYan, Zhongzhong; Liu, Aiping; Huang, Mingzhi; Liu, Minhua; Pei, Hui; Huang, Lu; Yi, Haibo; Liu, Weidong; Hu, AixiEuropean Journal of Medicinal Chemistry (2018), 149 (), 170-181CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)To discover new pyrazolecarboxamide analogs with broad spectrum and high activity, a class of new compds. of pyrazole carboxamide derivs. contg. thiazole or oxazole ring I (R1 = H, 4-Me, 2-Cl, etc.; R2 = H, Cl, Me; R3 = Me, Et, i-Pr, cyclopropyl; R4 = H, Et, Me, Ph; Z = S, O) was designed by scaffold hopping and bioisosterism, and 36 pyrazole carboxamide derivs. with antifungal activity were synthesized. Those compds. I were evaluated against five phytopathogenic fungi, Gibberella zeae, Phytophythora capsici, Sclerotonia sclerotiorum, Erysiphe graminis and Puccinia sorghi. The results indicated that most of the compds. displayed good fungicidal activities, esp. against E. graminis. Theor. calcns. were carried out at the B3LYP/6-31G (d, p) level and the full geometry optimization was carried out using the 6-31G (d, p) basis set, and the frontier orbital energy, at. net charges and, mol. docking were discussed, and the structure-activity relationships were also studied.
- 28Schmidt, A.; Habeck, T.; Kindermann, M. K.; Nieger, M. New Pyrazolium-Carboxylates as Structural Analogues of the Pseudo-Cross-Conjugated Betainic Alkaloid Nigellicine. J. Org. Chem. 2003, 68, 5977– 5982, DOI: 10.1021/jo0344337[ACS Full Text
], [CAS], Google Scholar
28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXkvVOrsbk%253D&md5=34795483a4b72eaabf34c39c093260c9New Pyrazolium-carboxylates as Structural Analogues of the Pseudo-Cross-Conjugated Betainic Alkaloid NigellicineSchmidt, Andreas; Habeck, Tobias; Kindermann, Markus Karl; Nieger, MartinJournal of Organic Chemistry (2003), 68 (15), 5977-5982CODEN: JOCEAH; ISSN:0022-3263. (American Chemical Society)Pyrazolium-3-carboxylates were examd. as relatives of the betainic alkaloid nigellicine and as new examples of the sparsely populated class of heterocyclic pseudo-cross-conjugated mesomeric betaines. The title compds. were prepd. in a 4-step procedure starting from EtO2CCOCH:CROH [R = Me, Ph] which were cyclized with substituted hydrazines. The resulting isomeric pyrazole esters were sepd. and quaternized with di-Me sulfate in the presence of nitrobenzene to pyrazolium esters. Sapon. was best accomplished in dild. sulfuric acid, which resulted in the formation of the pseudo-cross-conjugated mesomeric betaines in one step. Protonation to the corresponding carboxylic acids required the treatment of the betaines with tetrafluoroboric acid in dichloromethane. The effect of neg. solvatochromism proves the charge sepn. in the ground state of the mols. X-ray crystallog. analyses, semiempirical calcns., and ESI mass spectrometric measurements were performed to gain knowledge about the phenomenon of pseudo-cross-conjugation. - 29Knight, A.; Hemmings, J. L.; Winfield, I.; Leuenberger, M.; Frattini, E.; Frenguelli, B. G.; Dowell, S. J.; Lochner, M.; Ladds, G. Discovery of Novel Adenosine Receptor Agonists That Exhibit Subtype Selectivity. J. Med. Chem. 2016, 59, 947– 964, DOI: 10.1021/acs.jmedchem.5b01402[ACS Full Text
], [CAS], Google Scholar
29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xmslemtw%253D%253D&md5=0fd4c845ac9f9b91edac5154e4c8cdccDiscovery of Novel Adenosine Receptor Agonists That Exhibit Subtype SelectivityKnight, Anthony; Hemmings, Jennifer L.; Winfield, Ian; Leuenberger, Michele; Frattini, Eugenia; Frenguelli, Bruno G.; Dowell, Simon J.; Lochner, Martin; Ladds, GrahamJournal of Medicinal Chemistry (2016), 59 (3), 947-964CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A series of N6-bicyclic and N6-(2-hydroxy)cyclopentyl derivs. of adenosine were synthesized as novel A1R agonists and their A1R/A2R selectivity assessed using a simple yeast screening platform. We obsd. that the most selective, high potency ligands were achieved through N6-adamantyl substitution in combination with 5'-N-ethylcarboxamido or 5'-hydroxymethyl groups. In addn., we detd. that 5'-(2-fluoro)thiophenyl derivs. all failed to generate a signaling response despite showing an interaction with the A1R. Some selected compds. were also tested on A1R and A3R in mammalian cells revealing that four of them are entirely A1R-selective agonists. By using in silico homol. modeling and ligand docking, we provide insight into their mechanisms of recognition and activation of the A1R. We believe that given the broad tissue distribution, but contrasting signaling profiles, of adenosine receptor subtypes, these compds. might have therapeutic potential. - 30Stoddart, L. A.; Vernall, A. J.; Denman, J. L.; Briddon, S. J.; Kellam, B.; Hill, S. J. Fragment Screening at Adenosine-A3 Receptors in Living Cells Using a Fluorescence-Based Binding Assay. Chem. Biol. 2012, 19, 1105– 1115, DOI: 10.1016/j.chembiol.2012.07.014[Crossref], [PubMed], [CAS], Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhtl2rs7nI&md5=7f99ec1590a4a1ba1ba97bfbf9b7584cFragment Screening at Adenosine-A3 Receptors in Living Cells Using a Fluorescence-Based Binding AssayStoddart, Leigh A.; Vernall, Andrea J.; Denman, Jessica L.; Briddon, Stephen J.; Kellam, Barrie; Hill, Stephen J.Chemistry & Biology (Oxford, United Kingdom) (2012), 19 (9), 1105-1115CODEN: CBOLE2; ISSN:1074-5521. (Elsevier Ltd.)G protein-coupled receptors (GPCRs) comprise the largest family of transmembrane proteins. For GPCR drug discovery, it is important that ligand affinity is detd. in the correct cellular environment and preferably using an unmodified receptor. The authors developed a live cell high-content screening assay that uses a fluorescent antagonist, CA200645, to det. binding affinity consts. of competing ligands at human adenosine-A1 and -A3 receptors. This method was validated as a tool to screen a library of low mol. wt. fragments, and identified a hit with submicromolar binding affinity (KD). This fragment was structurally unrelated to substructures of known adenosine receptor antagonists and was optimized to show selectivity for the adenosine-A3 receptor. This technol. represents a significant advance that will allow the detn. of ligand and fragment affinities at receptors in their native membrane environment.
- 31Eldridge, M. D.; Murray, C. W.; Auton, T. R.; Paolini, G. V.; Mee, R. P. Empirical Scoring Functions: I. The Development of a Fast Empirical Scoring Function to Estimate the Binding Affinity of Ligands in Receptor Complexes. J. Comput. Aided. Mol. Des. 1997, 11, 425– 445, DOI: 10.1023/A:1007996124545[Crossref], [PubMed], [CAS], Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXnsV2it7o%253D&md5=ece4b881e181f3d5c1287af55efaf6b4Empirical scoring functions: I. The development of a fast empirical scoring function to estimate the binding affinity of ligands in receptor complexesEldridge, Matthew D.; Murray, Christopher W.; Auton, Timothy R.; Paolini, Gaia V.; Mee, Roger P.Journal of Computer-Aided Molecular Design (1997), 11 (5), 425-445CODEN: JCADEQ; ISSN:0920-654X. (Kluwer Academic Publishers)This paper describes the development of a simple empirical scoring function designed to est. the free energy of binding for a protein-ligand complex when the 3D structure of the complex is known or can be approximated. The function uses simple contact terms to est. lipophilic and metal-ligand binding contributions, a simple explicit form for hydrogen bonds and a term which penalizes flexibility. The coeffs. of each term are obtained using a regression based on 82 ligand-receptor complexes for which the binding affinity is known. The function reproduces the binding affinity of the complexes with a cross-validated error of 8.68 kJ/mol. Tests on internal consistency indicate that the coeffs. obtained are stable to changes in the compn. of the training set. The function is also tested on two test sets contg. a further 20 and 10 complexes, resp. The deficiencies of this type of function are discussed and it is compared to approaches by other workers.
- 32Hornak, V.; Abel, R.; Okur, A.; Strockbine, B.; Roitberg, A.; Simmerling, C. Comparison of Multiple Amber Force Fields and Development of Improved Protein Backbone Parameters. Proteins Struct. Funct. Genet. 2006, 65, 712– 725, DOI: 10.1002/prot.21123[Crossref], [PubMed], [CAS], Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtFWqt7fM&md5=de683a26eca9e83ae524726e97ac22faComparison of multiple Amber force fields and development of improved protein backbone parametersHornak, Viktor; Abel, Robert; Okur, Asim; Strockbine, Bentley; Roitberg, Adrian; Simmerling, CarlosProteins: Structure, Function, and Bioinformatics (2006), 65 (3), 712-725CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)The ff94 force field that is commonly assocd. with the Amber simulation package is one of the most widely used parameter sets for biomol. simulation. After a decade of extensive use and testing, limitations in this force field, such as over-stabilization of α-helixes, were reported by the authors and other researchers. This led to a no. of attempts to improve these parameters, resulting in a variety of "Amber" force fields and significant difficulty in detg. which should be used for a particular application. The authors show that several of these continue to suffer from inadequate balance between different secondary structure elements. In addn., the approach used in most of these studies neglected to account for the existence in Amber of two sets of backbone .vphi./ψ dihedral terms. This led to parameter sets that provide unreasonable conformational preferences for glycine. The authors report here an effort to improve the .vphi./ψ dihedral terms in the ff99 energy function. Dihedral term parameters are based on fitting the energies of multiple conformations of glycine and alanine tetrapeptides from high level ab initio quantum mech. calcns. The new parameters for backbone dihedrals replace those in the existing ff99 force field. This parameter set, which the authors denote ff99SB, achieves a better balance of secondary structure elements as judged by improved distribution of backbone dihedrals for glycine and alanine with respect to PDB survey data. It also accomplishes improved agreement with published exptl. data for conformational preferences of short alanine peptides and better accord with exptl. NMR relaxation data of test protein systems.
- 33Massova, I.; Kollman, P. A. Combined molecular mechanical and continuum solvent approach (MM-PBSA/GBSA) to predict ligand binding. Per. Drug Discovery Design 2000, 18, 113– 135, DOI: 10.1023/A:1008763014207[Crossref], [CAS], Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXis12ju7Y%253D&md5=520df05a7c7ed02fdf76d3e1e6f377f9Combined molecular mechanical and continuum solvent approach (MM-PBSA/GBSA) to predict ligand bindingMassova, Irina; Kollman, Peter A.Perspectives in Drug Discovery and Design (2000), 18 (Hydrophobicity and Solvation in Drug Design, Pt. II), 113-135CODEN: PDDDEC; ISSN:0928-2866. (Kluwer Academic Publishers)Significant progress has been achieved in computational methods to treat solvent effects in recent years. Among various techniques, the continuum solvent approach appears to be practically promising because it can be used to calc. reliable interaction and solvation energies in complex systems. A computational scanning mutagenesis method, one of such new approaches, has been recently developed. It combines the mol. mech. and continuum solvent approaches and allows one to identify the "hot spots" in binding interfaces from a single trajectory of a wild type complex. Such techniques can be also used as a tool to optimize the interacting species for the binding, or as a ranking procedure in high throughput screening.
- 34Wang, E.; Sun, H.; Wang, J.; Wang, Z.; Liu, H.; Zhang, J. Z. H.; Hou, T. End-Point Binding Free Energy Calculation with MM/PBSA and MM/GBSA: Strategies and Applications in Drug Design. Chem. Rev. 2019, 119, 9478– 9508, DOI: 10.1021/acs.chemrev.9b00055[ACS Full Text
], [CAS], Google Scholar
34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXht1ajs7%252FF&md5=eacd5dbfe659d27df5d103caec9e1bb5End-point binding free energy calculation with MM/PBSA and MM/GBSA: strategies and applications in drug designWang, Ercheng; Sun, Huiyong; Wang, Junmei; Wang, Zhe; Liu, Hui; Zhang, John Z. H.; Hou, TingjunChemical Reviews (Washington, DC, United States) (2019), 119 (16), 9478-9508CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. Mol. mechanics Poisson-Boltzmann surface area (MM/PBSA) and mol. mechanics generalized Born surface area (MM/GBSA) are arguably very popular methods for binding free energy prediction since they are more accurate than most scoring functions of mol. docking and less computationally demanding than alchem. free energy methods. MM/PBSA and MM/GBSA have been widely used in biomol. studies such as protein folding, protein-ligand binding, protein-protein interaction, etc. In this review, methods to adjust the polar solvation energy and to improve the performance of MM/PBSA and MM/GBSA calcns. are reviewed and discussed. The latest applications of MM/GBSA and MM/PBSA in drug design are also presented. This review intends to provide readers with guidance for practically applying MM/PBSA and MM/GBSA in drug design and related research fields. - 35Kaminski, G.; Friesner, R. A.; Tirado-Rives, J.; Jorgensen, W. L. Evaluation and Reparametrization of the OPLS-AA Force Field for Proteins via Comparison with Accurate Quantum Chemical Calculations on Peptides. Phys. Chem. B 2001, 105 (28), 6474– 6487, DOI: 10.1021/jp003919d[ACS Full Text
], [CAS], Google Scholar
35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXislKhsLk%253D&md5=3ff059626977ee7f6342466f5820f5b7Evaluation and Reparametrization of the OPLS-AA Force Field for Proteins via Comparison with Accurate Quantum Chemical Calculations on PeptidesKaminski, George A.; Friesner, Richard A.; Tirado-Rives, Julian; Jorgensen, William L.Journal of Physical Chemistry B (2001), 105 (28), 6474-6487CODEN: JPCBFK; ISSN:1089-5647. (American Chemical Society)We present results of improving the OPLS-AA force field for peptides by means of refitting the key Fourier torsional coeffs. The fitting technique combines using accurate ab initio data as the target, choosing an efficient fitting subspace of the whole potential-energy surface, and detg. wts. for each of the fitting points based on magnitudes of the potential-energy gradient. The av. energy RMS deviation from the LMP2/cc-pVTZ(-f)//HF/6-31G** data is reduced by ∼40% from 0.81 to 0.47 kcal/mol as a result of the fitting for the electrostatically uncharged dipeptides. Transferability of the parameters is demonstrated by using the same alanine dipeptide-fitted backbone torsional parameters for all of the other dipeptides (with the appropriate side-chain refitting) and the alanine tetrapeptide. Parameters of nonbonded interactions have also been refitted for the sulfur-contg. dipeptides (cysteine and methionine), and the validity of the new Coulombic charges and the van der Waals σ's and ε's is proved through reproducing gas-phase energies of complex formation heats of vaporization and densities of pure model liqs. Moreover, a novel approach to fitting torsional parameters for electrostatically charged mol. systems has been presented and successfully tested on five dipeptides with charged side chains. - 36Shivakumar, D.; Williams, J.; Wu, Y.; Damm, W.; Shelley, J.; Sherman, W. Prediction of Absolute Solvation Free Energies using Molecular Dynamics Free Energy Perturbation and the OPLS Force Field. J. Chem. Theory Comput. 2010, 6, 1509– 19, DOI: 10.1021/ct900587b[ACS Full Text
], [CAS], Google Scholar
36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXkslKhu7g%253D&md5=25d464fb9238e3235e881945c0b50a76Prediction of Absolute Solvation Free Energies using Molecular Dynamics Free Energy Perturbation and the OPLS Force FieldShivakumar, Devleena; Williams, Joshua; Wu, Yujie; Damm, Wolfgang; Shelley, John; Sherman, WoodyJournal of Chemical Theory and Computation (2010), 6 (5), 1509-1519CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The accurate prediction of protein-ligand binding free energies is a primary objective in computer-aided drug design. The solvation free energy of a small mol. provides a surrogate to the desolvation of the ligand in the thermodn. process of protein-ligand binding. Here, we use explicit solvent mol. dynamics free energy perturbation to predict the abs. solvation free energies of a set of 239 small mols., spanning diverse chem. functional groups commonly found in drugs and drug-like mols. We also compare the performance of abs. solvation free energies obtained using the OPLS_2005 force field with two other commonly used small mol. force fields - general AMBER force field (GAFF) with AM1-BCC charges and CHARMm-MSI with CHelpG charges. Using the OPLS_2005 force field, we obtain high correlation with exptl. solvation free energies (R2 = 0.94) and low av. unsigned errors for a majority of the functional groups compared to AM1-BCC/GAFF or CHelpG/CHARMm-MSI. However, OPLS_2005 has errors of over 1.3 kcal/mol for certain classes of polar compds. We show that predictions on these compd. classes can be improved by using a semiempirical charge assignment method with an implicit bond charge correction. - 37Stamatis, D.; Lagarias, P.; Barkan, K.; Vrontaki, E.; Ladds, G.; Kolocouris, A. Structural Characterization of Agonist Binding to an A3 Adenosine Receptor through Biomolecular Simulations and Mutagenesis Experiments. J. Med. Chem. 2019, 62, 8831– 8846, DOI: 10.1021/acs.jmedchem.9b01164[ACS Full Text
], [CAS], Google Scholar
37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslGnsL7O&md5=3c3882463d18e0cb0c999839692b20caStructural Characterization of Agonist Binding to an A3 Adenosine Receptor through Biomolecular Simulations and Mutagenesis ExperimentsStamatis, Dimitrios; Lagarias, Panagiotis; Barkan, Kerry; Vrontaki, Eleni; Ladds, Graham; Kolocouris, AntoniosJournal of Medicinal Chemistry (2019), 62 (19), 8831-8846CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The adenosine A3 receptor (A3R) binds adenosine and is a drug target against cancer cell proliferation. Currently, there is no exptl. structure of A3R. Here, we have generated a mol. model of A3R in complex with two agonists, the nonselective 1-(6-amino-9H-purin-9-yl)-1-deoxy-N-ethyl-β-D-ribofuranuronamide (NECA) and the selective 1-deoxy-1-[6-[[(3-iodophenyl)methyl]amino]-9H-purin-9-yl]-N-methyl-β-D-ribofuranuronamide (IB-MECA). Mol. dynamics simulations of the wild-type A3R in complex with both agonists, combined with in vitro mutagenic studies revealed important residues for binding. Further, mol. mechanics-generalized Born surface area calcns. were able to distinguish mutations that reduce or negate agonistic activity from those that maintained or increased the activity. Our studies reveal that selectivity of IB-MECA toward A3R requires not only direct interactions with residues within the orthosteric binding area but also with remote residues. Although V1695.30 is considered to be a selectivity filter for A3R binders, when it was mutated to glutamic acid or alanine, the activity of IB-MECA increased by making new van der Waals contacts with TM5. This result may have implications in the design of new A3R agonists.
Supporting Information
Supporting Information
ARTICLE SECTIONSThe Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsmedchemlett.2c00052.
Scheme S1. Comparison of amino acid residue sequences of the binding area. Table S1. Chemical structures and antagonistic potencies of 7-amino-pyrazolo[3,4-d]pyridazines 10a–c and 15a–c against A2AR and A2BR. Table S2. Mean RMSD values for all compounds against A1R, A2AR, and A2BR and ΔGeff only for 10–10c against A1R. Figure S1. 1H and 13C NMR spectra of the target compounds. Figure S2. Results from the MD simulations of 10b and 15b against A3R and A2BR. Figure S3. Docking poses of 4-(2-phenethyl)amino-1-phenylethyl pyrazolo[3,4-b]pyridine and N9-methyl,N6-benzyl adenine to A1R. Figure S4. Representative frames from 100 ns MD simulations of 10b inside the orthosteric binding area of WT A1R and 10b inside mutant Y271A A1R and the receptor–ligand interaction frequency histogram and RMSD graphs of protein Ca and ligand heavy atoms. Information for the methods and synthetic protocols. (PDF)
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.