ACS Publications. Most Trusted. Most Cited. Most Read
My Activity
CONTENT TYPES

Discovery of 1,2,4-Triazine Derivatives as Adenosine A2A Antagonists using Structure Based Drug Design

View Author Information
Heptares Therapeutics Limited, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, U.K.
*Phone +44 (0)1707 358638. E-mail: [email protected]
Cite this: J. Med. Chem. 2012, 55, 5, 1898–1903
Publication Date (Web):January 5, 2012
https://doi.org/10.1021/jm201376w
Copyright © 2012 American Chemical Society
  • Open Access

Article Views

9471

Altmetric

-

Citations

LEARN ABOUT THESE METRICS
PDF (2 MB)
Supporting Info (1)»

Abstract

Potent, ligand efficient, selective, and orally efficacious 1,2,4-triazine derivatives have been identified using structure based drug design approaches as antagonists of the adenosine A2A receptor. The X-ray crystal structures of compounds 4e and 4g bound to the GPCR illustrate that the molecules bind deeply inside the orthosteric binding cavity. In vivo pharmacokinetic and efficacy data for compound 4k are presented, demonstrating the potential of this series of compounds for the treatment of Parkinson’s disease.

 Accession Codes

The PDB codes for 4g and 4e are 3UZA and 3UZC, respectively.

Introduction

ARTICLE SECTIONS
Jump To

The adenosine A2A receptor is expressed in the basal ganglia where it functionally opposes the actions of the dopamine D2 receptor, i.e., inhibition of the A2A receptor leads to enhancement of D2 receptor function. Given that the primary pathology in Parkinson’s disease is a loss of nigrostriatal dopamine and hence reduced dopamine D2 receptor activation, adenosine A2A receptor antagonism has emerged as a potential nondopaminergic therapy for this disorder. Preclinically, adenosine A2A receptor antagonists are effective in animal models of Parkinson’s disease, ranging from the reversal of haloperidol-induced catalepsy through to efficacy in more disease-relevant models such as 6-hydroxydopamine lesioned rats and MPTP-lesioned primates. (1) Furthermore, a number of these compounds have progressed into clinical development, the most advanced of which is currently preladenant. This compound was shown to be effective in a phase IIa trial of patients with moderate-to-severe Parkinson’s disease when administered in conjunction with levodopa, increasing on-time with no concomitant increase in dyskinesias. (2)
In the preceding publication, hit molecules derived from a virtual screening strategy were described. (3) One series of 1,3,5-triazine derivatives was identified and optimized to give potent and selective adenosine A2A receptor antagonists. As part of the optimization of this chemotype and by considering the proposed binding mode to the receptor, we hypothesized that the alternative 1,2,4-triazine isomers might also bind to the receptor but could sit more deeply in the receptor pocket accessing the region normally occupied by the ribose group of the natural ligand adenosine, in addition to mimicking the adenine ring itself. Upon testing of the commercially available parent 5,6-diphenyl-1,2,4-triazine-3-amine 4a, we discovered that the molecule was indeed an antagonist of the receptor (Table 1, compound 1; pKi = 6.93). In this paper, we outline our studies in this isomeric chemical series.
Table 1. SAR and in Vitro ADME Data for Compounds 4a to 4la
        SPR data
IDformulaA2ApKiA1pKiLE (17)RLM (min)PPB (%)kinetic solubility (μM)kakdKDpKD
4aX═C;R1–6═H6.936.560.5023ND>100>5 × 107>1 × 1009.03 × 10–65.0
4bX═C;R1═Cl;R2–6═H7.297.250.502997.9133.79 × 1051.68 × 10–14.42 × 10–76.4
4cX═C;R1═R3═Cl;R2,4,5,6 ═H8.407.360.5510899.0385.32 × 1052.43 × 10–24.57 × 10–87.3
4dX═C;R1═R3═Me;R2,4,5,6═H7.676.710.50998.020NDNDNDND
4eX═C;R1═Cl;R2═OH;R3,4,5,6 ═H8.859.790.576998.0454.07 × 1061.01 × 10–32.48 × 10–109.6
4fX═C;R1═R3═Me;R2═OH;R4,5,6═H8.397.780.527593.3438.57 × 1061.36 × 10–31.59 × 10–109.8
4gX═N;R1═R3═Me;R4,5,6═H8.117.070.5310082.1409.92 × 1061.15 × 10–21.16 × 10–98.9
4hX═N;R1═R3═Me;R5═F;R4,6═H7.816.400.4810069.0431.13 × 1071.15 × 10–11.02 × 10–88.0
4iX═N;R1═R3═Me;R4,6═F;R5═H7.566.770.45100ND459.44 × 1068.84 × 10–29.37 × 10–98.0
4jX═N;R1═R3═Me;R4═F; R5,6═H7.986.960.497887.0481.41 × 1074.27 × 10–23.03 × 10–98.5
4kX═N;R1═Me;R3═CF3;R4,5,6═H8.467.500.488692.0351.08 × 1063.73 × 10–33.45 × 10–98.5
4lX═N;R1═Me;R3═CF3;R5═F;R4,6═H8.346.930.459793.0341.55 × 1064.09 × 10–22.63 × 10–87.6
a

RLM rat liver microsome half-life in mins; PPB rat plasma protein binding; SPR kinetics using A2A–StaR (see main text).

Results and Discussion

ARTICLE SECTIONS
Jump To

Design and Synthesis

The binding mode of 1,2,4-triazine derivatives was initially derived from modeling of representative compounds in an “experimentally enhanced” homology model of the adenosine A2A receptor (described in the preceding paper), refined using site directed mutagenesis data both from the literature and our own Biophysical Mapping (BPM) approach. (3-6) Parts A and B of Figure 1 illustrate the proposed binding mode of two analogues 4g and 4e (Table 1) in the orthosteric binding site of the receptor and also show the BPM binding fingerprint around these example ligands, used to refine and improve the model. The residues that, when mutated to alanine, reduce binding of each ligand are colored in red for nonbinding, dark-orange for the largest effect (tier 1), orange for the next largest effect (tier 2), yellow for the smallest effect (tier 3), and in green if the mutation caused an increase in binding of the ligand. The BPM studies have been reported elsewhere and analogues 4e and 4g here equate to examples 3b and 3d in the earlier publication. (4) As well as rationalizing the role of the aminoheterocyclic scaffold binding to Asn2536.55 (superscripts refer to Ballesteros–Weinstein numbering), (7) in particular the BPM fingerprints and modeling suggested that presence of a hydrogen bond acceptor at the para position of ring A of the ligands to His2787.43 with addition of one or more flanking lipophilic substituents on the same ring to interact with Ile662.64 and Ser2777.42 should be one focus of the SAR program (R1, R2, R3 positions in Scheme 1 and Table 1). In addition, careful 3D analysis of the GRID maps, calculated for both A2A and an A1 homology model, enabled the pharmacophoric preferences (hydrophobic, hydrogen-bond donor and acceptor) and shape constrictions/differences to be identified to allow an enhanced evaluation of each ligand docked into the binding site. (8, 9) Very subtle binding site differences, such as A2A–Ser7.42 to A1–Thr7.42 and A2A–Ala2.57 to A1–Val2.57 and also ligand preferences between the two receptor models were exploited to overall enable the design of small, polar, selective, and ligand-efficient compounds.

Figure 1

Figure 1. (A,B) BPM fingerprint of 1,2,4-triazine adenosine A2A antagonists. Compounds 4g (A) and 4e (B) are illustrated bound to the orthosteric pocket of the receptor and the residues lining the pocket that interact with the ligands are labeled. The tier 1, 2, and 3 designation is described in the main text. The key hydrogen bonding to Asn2536.55 of the scaffold is highlighted by green dotted lines. (C,D) Illustration of the A2A–StaR2 ligand binding site in complex with compound 4g (C) and 4e (D). TM helices and visible extracellular regions are depicted in the rainbow format. Ligands are represented as stick models, carbon and chlorine atoms are green, oxygen atoms red, and nitrogen atoms blue. Residues involved in ligand binding are labeled and represented as gray sticks, oxygen atoms are red, and nitrogen atoms are blue. Extracellular loop 2, the key binding site residues and TM’s 1, 2, 5, and 6 are labeled for reference. Potential H-bonds between the ligand and receptor are represented as dashed blue lines. TM3 and TM4 have been omitted for clarity. (E) WaterMap calculation on the binding site of compound 4e (ligand removed for the calculation). Waters calculated are color coded to show the most “unhappy” vs bulk solvent as red (>3.5 kcal/mol), then yellow (2.2–3.5 kcal/mol), with gray intermediate (−1 to 2.2 kcal) and blue “happy” (<−1 kcal/mol). The CPK surface of the ligand 4e is shown as a red dot surface, clearly illustrating that the cluster of red and yellow “unhappy” waters deep in the binding site have been displaced. GRID maps are also shown that highlight the shape (Csp3 (C3) at 1 kcal/mol in light-gray), the lipophilic hotspots (aromatic CH probe (C1═) in yellow at −2.5 kcal/mol), and the water probe hotspots (in green wire mesh at −6.6 kcal/mol). (F) Alignment of the A2A homology model with 4e docked (cyan carbons) onto the crystal structure of A2A4e complex (green carbons). The alignment was generated by the align algorithm in Pymol utilizing only helices where hydrogen bonds are formed with the ligand, helices 6 and 7. Helices 2, 3, and 4 are removed for clarity.

Scheme 1

Scheme 1. Synthesis of 5,6-Biaryl-1,2,4-triazine-3-amine Derivatives (4) and 4-Pyridylboronic Acid Derivatives (6)a

Scheme aReagents and conditions: (a) NBS, DMF, RT; (b) 3, Pd(PPh3)4, K2CO3, 1,4-dioxane/H2O, 150 °C; (c) [Ir(COD)OMe]2, DTBPY, [B(pin)]2, hexane, 50 °C.

During the SAR optimization process, a number of members of the series were successfully cocrystallized in the receptor using the published thermally stabilized adenosine A2A construct A2A–StaR2 (stabilized receptor or StaR) developed in our laboratories. (10) This allowed, for the first time to our knowledge, X-ray structure-directed optimization of a hit series to derive potent and selective leads for a G protein-coupled receptor. This reinforced our efforts focused on the optimization of the substitution patterns around the pendent aryl rings A and B in an atom efficient manner (Scheme 1 and Table 1) to engineer high potency and reduce affinity for the adenosine A1 receptor without significantly increasing molecular weight or lipophilicity. The X-ray structures also validated the BPM approach, which had successfully predicted the binding mode of the compounds but add a further level of understanding particularly with respect to teasing out selectivity for A2A over the A1 receptor.
Synthesis of the target compounds began with a set of purchased 3-amino-5-aryl-1,2,4-triazine derivatives 1, Scheme 1. These building blocks were further elaborated following treatment with NBS at room temperature to afford the corresponding 6-bromotriazines 2. A set of conditions was optimized to enable Suzuki cross-coupling of a diverse range of bromotriazines with commercially available boronic acid derivatives. Thus 2 and 3 were coupled at 150 °C in a sealed vessel in the presence of catalytic quantities of Pd(PPh3)4 to afford the biaryl triazines of interest (4). Access to bespoke 4-pyridylboronic acid derivatives was required to synthesize molecules suggested by the proposed binding mode in the receptor. Using a method described by Hartwig et al., commercial 2,6-disubstituted pyridines 5 were borylated in high yield, under iridium catalysis, with bis(pinacolato)diboron. (11-13)

X-Ray Crystallography

The overall structure of the A2A–StaR2 in complex with compounds 4e and 4g is in close agreement to the previously solved structures in our laboratory and methods for crystallization and a description of the general receptor architecture are described elsewhere. (10) Statistics for data collection and refinement are given in Supporting Information Table S1. The cocrystal structures of the A2A–StaR2 in complex with compounds 4g and 4e (Figure 1C,D) show clear positive omit density at 3.0σ (data not shown) for the presence and position of the ligands in the receptor binding pocket. The structure of A2A–StaR2–4g shows the amino-triazine core makes two critical donor and acceptor H-bonding interactions with the side chain of Asn2536.55 with bonding distances of 2.85 and 2.76 Å, respectively. In addition, the helical portion of extracellular loop (ECL) 2 is positioned for Phe168 to form a perpendicular π–π stack on one side of the core, while the side chain of Met2707.35 makes a hydrophobic interaction on the opposite side, completing the receptor interactions around this region of the ligand. The phenyl substituent from the C5 position of the triazine core occupies a hydrophobic pocket deeper inside the receptor flanked by Leu843.32, Leu853.33, Met1775.38, Asn1815.42, Trp2466.48, Leu2496.51, and His2506.52. The second substituent, dimethyl-pyridine, from the C6 carbon of the triazine core occupies the ribose binding pocket (of the natural agonist adenosine) defined by His2787.43 and Ser2777.42, with one methyl substituent pointing toward a hydrophobic region defined by Ala632.61 and Ile662.64 and the other pointing toward the stabilizing mutation Ser2777.42Ala. (14) Additionally, the side chain of His2787.43 is positioned 4.03 Å away from N4 of the dimethyl-pyridine substituent, precluding a direct H-bond between the ligand and this receptor residue. Compound 4e occupies a similar position overall to that of 4g in the A2A receptor ligand binding site but with specific differences. The amino-triazine core makes a similar set of interactions to the receptor as 4g, however, the bond lengths to Asn2536.55 increase to 3.12 and 3.1 Å in comparison. The driving force for this is an additional hydrogen bond, 2.87 Å in length, formed between the phenolic hydroxyl on the chloro-phenol substituent to Nε of His2787.43. Additionally, Glu131.37 has switched rotamer and is now poised to H-bond to Nδ1 of His2787.43. The additional conjugation of the ligand through the receptor pulls compound 4e ∼1.2 Å deeper into the ribose binding pocket in comparison to 4g (pivoting on the common phenyl group) and perhaps provides a basis for the slow off-rate receptor kinetics of these phenolic compounds (see Table 1). It is noted that two potential conformations for the phenyl and dimethyl-pyridine substituents (compound 4g), and the phenyl and chloro-phenol substituents (compound 4e), involving concomitant ∼50° rotations of each around the bond to the amino-triazine core, can exist in nature. In the structures presented here the conformations with the lower b-factors were submitted to the Research Collaboratory for Structural Bioinformatics Protein Data Bank (RCSB).
An analysis of the A2A binding site (with ligand 4e removed) using the WaterMap software (Schrödinger) (15) shows that these highly ligand efficient ligands (LE = 0.57 for 4e) occupy exactly the region where there is a cluster of “unhappy” waters (shown as red and yellow balls in Figure 1E) and not other less favorable regions (as for example less ligand efficient compounds such as ZM241385 bind, reported elsewhere). (16) The WaterMap software uses a molecular dynamics simulation on a full explicit water network to calculate the enthalpic and entropic energies of waters compared to bulk solvent. Finally, in Figure 1, as the initial SAR work was based on our BPM-optimized homology model, we include a comparison of a docked structure of 4e into the homology model and the protein–ligand X-ray structure, showing that the overall orientation and key hydrogen bond interactions were correctly predicted (Figure 1f). The biophysical mapping fingerprints of compounds 4e and 4g (published previously by Zhukov et al, as compounds 3b and 3d in Table 1), respectively, are in excellent agreement with the crystal structures, highlighting the significant interactions with Ile662.64, Leu853.33, Asn1815.42, and Asn2536.55 and confirming the initial deep placement of the ligand in the ribose sugar pocket of the A2A receptor. (4)

Structure–Activity Relationship

The parent compound 4a was identified as a ligand efficient adenosine A2A receptor antagonist (Table 1). (17) Having examined the putative binding mode in silico, SAR focused on simple substitution of rings A and B (Table 1). During the optimization process, the binding mode predictions were first improved using our BPM approach and then by determination of the crystal structures of compounds 4e and 4g (discussed above). The models suggested only small groups would be tolerated, particularly in ring B, and this was quickly evident because addition of chlorine or methyl in ring A and fluorine in ring B was tolerated or increased affinity (compounds 4ad and 4hj), but larger groups tended to be detrimental to potency (data not shown). The models also suggested the potential to form hydrogen bonding interactions from ring A at the para position R2, and introduction of either a phenolic hydroxyl (compounds 4e and 4f) or a 4-pyridyl nitrogen (compound 4g) were found to increase potency (compare 4f and 4g with 4d). A challenging aspect of the optimization was that affinity for the adenosine A1 receptor was generally also observed, and it was thought desirable to reduce affinity against this target to avoid any potential side effects caused by cross reactivity with this receptor. It was quickly noted, consistent with small differences in the binding sites predicted from a GRID analysis of models of the two receptors (discussed above), that 3,5-disubstitution in ring A introduced a modest selectivity for A2A over A1 (compare 4b with 4c and 4e with 4f). Also, although a very subtle effect, introduction of fluorine at R5 on ring B reduced activity against both receptors but slightly improved the selectivity window for A2A over A1 (compare 4g with 4h). Introduction of fluorine in other substitution patterns tended to reduce affinity further or had no additional selectivity benefit (compounds 4i and 4j). Overall, the derivatives were found to be highly selective over the adenosine A3 receptor, e.g., compounds 4c, 4g, and 4h had antagonist affinity of >10 μM. The series was generally 10-fold selective over the A2B subtype, and inhibition of this target was not thought to be detrimental therapeutically; 4c, 4g, and 4h had pKi values of 7.4, 7.3, and 6.8, respectively (A3 and A2B data were generated by Ricerca Biosciences, Taipei). Fine tuning of affinity by various combinations of small lipophilic substituents quickly led us to pyridyl analogues 4k and 4l, incorporating a CF3 group at the 3-position of ring A with methyl at the 5-position and with or without fluorine at the para position of ring B. These two compounds have the best balance of potency and selectivity, derived by introduction of simple substituents on the scaffold, of the examples shown in the Table.
Table 1 also details the binding kinetics of compounds from the lead series as measured using the adenosine A2A–StaR by surface plasmon resonance on a Biacore instrument (see Supporting Information for full details). The pKD values were generally in good agreement with the radioligand binding data, and the method also allowed comparison of on rates (ka) and off rates (kd) to the receptor between related compounds. Of most note in the data presented here is that the phenolic analogues 4e and 4f had very slow off rates, consistent with the change in binding mode observed in the crystal structure of 4e discussed above. Consideration of relative receptor kinetics was part of the decision making process used for selection of compounds for in vivo efficacy experiments.

Pharmacokinetics and in Vivo Efficacy

The examples in Table 1 generally exhibited good physicochemical and in vitro ADME properties, having moderate to high aqueous solubility, good stability in rat liver microsomes (RLM) and, where a polar substituent had been introduced (such as a pyridyl nitrogen atom), relatively low plasma protein binding (PPB). Another general trend was a lack of inhibition of cytochrome P450 enzymes and the hERG channel, with only the occasional outlier (data not shown). A number of compounds also demonstrated good pharmacokinetic properties in rat dosed either orally or intravenously. Data is given here for example 4k in a rat pharmacokinetic (PK) experiment; PK parameters are shown in Table 2. Compound 4k displayed moderate clearance (42 mL/min/kg), although due to a relatively high steady-state volume of 4.6 L/kg, the compound had an acceptable half-life of 1.2 h. Compound 4k was rapidly absorbed after oral dosing (Tmax = 0.4 h) and displayed good exposure with AUC = 846 ng·h/mL, resulting in an estimated Fpo of 100%. The derivative also displayed excellent brain penetration, as measured by samples at 0.5 h post-IV dose (brain/plasma = 3.2). Furthermore, measured levels in the CSF at the same time point suggested that the unbound fraction of 4k in the brain was 3.6%, reasonably consistent with the measured plasma protein binding in vitro of 92% (unbound plasma fraction = 8%).
Table 2. PK Parameters of Compound 4k in Rat
4k, 1 mg/kg(IV)4k, 2 mg/kg(PO)
plasma clearance42 mL/min/kgTmax0.4 h
Vd(ss)4.6 L/kgCmax244 ng/mL
terminal t1/21.2 hterminal t1/21.1 h
AUCinf397 ng·h/mLAUCinf846 ng·h/mL
brain:plasma (0.5 h)3.2Fpo100%
CSF:brain (0.5 h)0.036  
Given the good overall in vitro ADME and in vivo PK profile of 4k, especially with respect to brain penetration and oral bioavailability, the compound was tested for its ability to reverse haloperidol-induced catalepsy in rats, a simple and well validated in vivo pharmacodynamic model mimicking the loss of striatal dopamine receptor function observed in Parkinson’s disease. (1) Compound 4k was found to very potently reverse catalepsy induced by haloperidol, with ED50 values of 0.2 mg/kg at both 1 and 2 h post dose time points (Figure 2).

Figure 2

Figure 2. In vivo efficacy of 4k. Dose-dependent effect of 4k (0.1–1 mg/kg, po; 1 and 2 h pretreatment time) to reverse haloperidol-induced catalepsy in rats in comparison with the positive control, istradefylline (1 mg/kg, po).

Conclusions

ARTICLE SECTIONS
Jump To

The studies presented here have shown for the first time that biophysical mapping and cocrystal X-ray structures of ligands to a G protein-coupled receptor can be used to direct optimization of novel, low molecular weight hit molecules into highly potent and selective lead compounds. Compound 4k, described above, has desirable physicochemical and drug-like properties, including high oral bioavailability and very potent in vivo efficacy. Further optimization of this 1,2,4-triazine series of antagonists of the adenosine A2A receptor has subsequently allowed identification of a preclinical candidate for the potential treatment of Parkinson’s disease, and details of the development of this molecule will be the topic of future publications.

Experimental Section

ARTICLE SECTIONS
Jump To

Synthetic Methods

The purity of the final compounds was determined by HPLC or LC/MS analysis to be >95%. Full experimental details of all compounds in Table 1 are described in the Supporting Information. Synthesis of compounds 4g and 4k are described below.

6-Bromo-5-phenyl-1,2,4-triazin-3-amine

A solution of 5-phenyl-1,2,4-triazin-3-amine (1.50 g, 8.70 mmol) in DMF (15 mL) was cooled to −25 °C and treated with a solution of N-bromosuccinimide (4.50 g, 26.6 mmol) in DMF (10 mL) by dropwise addition. The reaction was warmed gradually to room temperature and stirred overnight with TLC monitoring. After completion of the reaction, the mixture was poured into saturated bicarbonate solution (50 mL) and extracted with diethyl ether (25 × 3 mL). The organic phases were combined, dried over Na2SO4, and concentrated in vacuo. The crude compound was purified by gradient flash chromatography, eluting with mixtures of ethyl acetate in hexane to afford 6-bromo-5-phenyl-1,2,4-triazin-3-amine (1.40 g, 64%). HPLC: 99%, 8.31 min (244 nm). Mass spectroscopy: m/z 250.9 [M + H]+. 1H NMR: (400 MHz, DMSO) δ: 7.49–7.57 (m, 5H), 7.72 (m, 2H).

2-(Trifluoromethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-6-methylpyridine

Methoxy(cyclooctadiene)iridium(I) dimer (30 mg, 0.062 mmol), 4,4′-di-tert-butyl-2,2′-bipyridine (33 mg, 0.124 mmol), and bis(pinacolato)diboron (4.09 g, 16.1 mmol) were added to a flask which had been thoroughly purged with nitrogen. The flask was once more purged before adding hexane via syringe (30 mL). The resulting mixture was heated at 50 °C for 10 min until the appearance of a dark-red solution was observed. 2-Trifluoromethyl-6-methyl pyridine (4.0 g, 24.8 mmol) was then added by syringe, and heating continued for a further 6 h. After cooling to room temperature, the crude reaction mixture was concentrated under reduced pressure. The resulting residue was purified by column chromatography, eluting with ethyl acetate/hexane mixtures to afford the target compound 2-(trifluoromethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-6-methylpyridine (5.9 g, 83%). HPLC: 96%, 7.57 min (210 nm). Mass spectroscopy: m/z 287.8 [M + H]+. 1H NMR: (400 MHz, DMSO) δ: 1.31 (s, 12 H), 2.51 (s, 3H), 7.70 (s, 1H), 7.76 (s, 1H).

6-(2,6-Dimethylpyridin-4-yl)-5-phenyl-1,2,4-triazin-3-amine 4g: A Typical Procedure for the Synthesis of 5,6-Biaryl-1,2,4-triazine-3-amine derivatives

A solution of 6-bromo-5-phenyl-1,2,4-triazin-3-amine, (90 mg, 0.358 mmol) in dioxane (2.0 mL) was treated with 2,6-dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (96 mg, 0.412 mmol) and K2CO3 (148 mg, 1.07 mmol). The resulting mixture was diluted with water (1.0 mL), degassed, treated with tetrakis triphenylphosphinepalladium(0) (21 mg, 0.018 mmol), and stirred for 2 h at 150 °C in a sealed vessel. Upon completion of the reaction, the mixture was diluted with water (20 mL) and extracted with ethyl acetate (3 × 20 mL); the combined organic extracts were then dried over Na2SO4 and concentrated under reduced pressure. The crude compound was purified by gradient flash chromatography, eluting with mixtures of ethyl acetate and hexanes to afford 6-(2,6-dimethylpyridin-4-yl)-5-phenyl-1,2,4-triazin-3-amine 4g (44 mg, 43%). HPLC: 98%, 6.09 min (281 nm). Mass spectroscopy: m/z 278.1 [M + H]+. 1H NMR: (400 MHz, DMSO) δ: 2.33 (s, 6H), 6.97 (s, 2H), 7.37–7.43 (m, 4H), 7.48 (m, 1H), 7.58 (bs, 2H).

6-[2-Methyl-6-(trifluoromethyl)pyridin-4-yl]-5-phenyl-1,2,4-triazin-3-amine 4k

6-[2-Methyl-6-(trifluoromethyl)pyridin-4-yl]-5-phenyl-1,2,4-triazin-3-amine 4k (0.32 g, 35%) was prepared from 6-bromo-5-phenyl-1,2,4-triazin-3-amine (0.70 g, 2.78 mmol) and 2-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-6-trifluoromethyl-pyridine (1.2 g, 4.1 mmol) according to the typical procedure described above. HPLC purity: 99%, 10.29 min (269 nm). Mass spectroscopy: m/z 332.0 [M + H]+. 1H NMR: (400 MHz, DMSO) δ: 2.48 (s, 3H), 7.38 (m, 5H), 7.47 (s, 1H), 7.58 (s, 1H), 7.73 (bs, 2H).

Biology Methods

Methods for determination of antagonist potency against human adenosine A2A and A1 receptors, binding constants, and receptor kinetics of compounds binding to the A2A–StaR by surface plasmon resonance and the procedure for determination of in vivo efficacy in rodents by reversal of haloperidol induced catalepsy are detailed in the Supporting Information.

Diffraction Data Collection of A2A–StaR2 in Complex with 4e and 4g

Diffraction data from crystals of A2A–StaR2 in complex with compounds 4e and 4g were collected at I24, Diamond Light Source, Oxford, UK. Statistics for data collection and refinement are given in Supporting Information Table S1. Atomic coordinates and structure factors have been deposited in the RCSB under accession codes 3UZC and 3UZA, respectively.

Computational Chemistry

Homology models were constructed from the avian β1 adrenergic GPCR crystal structure bound to cyanopindolol (PDB: 2VT4) using several computational approaches as detailed in the preceding paper and refined/validated using site-directed mutagenesis and BPM data and known ligands (see Supporting Information for full details). (3, 4) Docking was done using Glide SP and XP (Schrödinger), and GRID analyses of the binding sites was used to evaluate potential docking poses (using the Csp3 (C3) for shape, aromatic CH probe (C1═) for lipophilic hotspots, carbonyl group (O) for hydrogen-bond acceptor hotspots, and amide NH (N1) for hydrogen-bond donor hotspots) and driving the designs. (8, 9)

Supporting Information

ARTICLE SECTIONS
Jump To

Crystallographic table of statistics. Synthesis protocols, 1H NMR, purification details, yields, purities by HPLC and MS or LCMS. Biological protocols for in vitro and in vivo experiments. Computational chemistry methods. SPR binding and kinetic data. This material is available free of charge via the Internet at http://pubs.acs.org.

Terms & Conditions

Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Author Information

ARTICLE SECTIONS
Jump To

  • Corresponding Author
    • Miles Congreve - Heptares Therapeutics Limited, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, U.K. Email: [email protected]
  • Authors
    • Stephen P. Andrews - Heptares Therapeutics Limited, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, U.K.
    • Andrew S. Doré - Heptares Therapeutics Limited, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, U.K.
    • Kaspar Hollenstein - Heptares Therapeutics Limited, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, U.K.
    • Edward Hurrell - Heptares Therapeutics Limited, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, U.K.
    • Christopher J. Langmead - Heptares Therapeutics Limited, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, U.K.
    • Jonathan S. Mason - Heptares Therapeutics Limited, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, U.K.
    • Irene W. Ng - Heptares Therapeutics Limited, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, U.K.
    • Benjamin Tehan - Heptares Therapeutics Limited, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, U.K.
    • Andrei Zhukov - Heptares Therapeutics Limited, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, U.K.
    • Malcolm Weir - Heptares Therapeutics Limited, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, U.K.
    • Fiona H. Marshall - Heptares Therapeutics Limited, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, U.K.

Acknowledgment

ARTICLE SECTIONS
Jump To

We thank Chris Richardson and Bissan Al-Lazikani for help with constructing the first generation of homology models and ligand dockings, David Myszka for helping to establish conditions for use of A2A StaRs on the Biacore format as a precursor to the work in this manuscript, and Nathan Robertson, Asma Baig, Jason Brown, Alistair O’Brien, and Giles Brown at Heptares.

Abbreviations Used

ARTICLE SECTIONS
Jump To

StaR

stabilized receptor

BPM

biophysical mapping

GPCR

G protein-coupled receptor

SPR

surface plasmon resonance

ECL

extracellular loop

TM

transmembrane helix

SAR

structure–activity relationship

PDB

Protein Data Bank

LE

ligand efficiency

RLM

rat liver microsomal turnover

PPB

rat plasma protein binding

RCSB

Research Collaboratory for Structural Bioinformatics Protein Data Bank

References

ARTICLE SECTIONS
Jump To

This article references 17 other publications.

  1. 1
    Shah, U.; Hodgson, R. Recent progress in the discovery of adenosine A2A receptor antagonists for the treatment of Parkinson’s disease Curr. Opin. Drug Discovery Dev. 2010, 13, 466 480
  2. 2
    Salamone, J. D. Preladenant, a novel adenosine A2A receptor antagonist for the potential treatment of parkinsonism and other disorders IDrugs 2010, 13, 723 731
  3. 3
    Langmead, C. J.; Andrews, S.; Congreve, M.; Errey, J.; Hurrell, E.; Marshall, F. H.; Mason, J. S.; Richardson, C.; Robertson, N.; Zhukov, A.; Weir, M. Identification of Novel Adenosine A2A Receptor Antagonists by Virtual Screening.

    Just accepted.

     DOI: DOI: 10.1021/jm201455y . Published online: January17, 2012
  4. 4
    Zhukov, A.; Andrews, S. P.; Errey, J. C.; Robertson, N.; Tehan, B.; Mason, J. S.; Marshall, F. H.; Weir, M.; Congreve, M. Biophysical Mapping of the Adenosine A2A Receptor J. Med. Chem. 2011, 54, 4312 4323
  5. 5
    Dal Ben, D.; Lambertucci, C.; Marucci, G.; Volpini, R.; Cristalli, G. Adenosine Receptor Modeling: What Does the A2A Crystal Structure Tell Us? Curr. Top. Med. Chem. 2010, 93, 993 1018
  6. 6
    Kim, S. K.; Gao, Z.; Van Rompaey, P.; Gross, A. S.; Chen, A.; Van Calenbergh, S.; Jacobson, K. A. Modeling the adenosine receptors: comparison of the binding domains of A2A agonists and antagonists J. Med. Chem. 2003, 46, 4847 4859
  7. 7
    Ballesteros, J. A.; Weinstein, H.; Stuart, C. S. Integrated methods for the construction of three dimensional models and computational probing of structure–function relations in G protein coupled receptors. In Methods in Neurosciences; Academic Press: New York, 1995; Vol. 25, pp 366 428.
  8. 8
    Goodford, P. J. A computational procedure for determining energetically favorable binding sites on biologically important macromolecules J. Med. Chem. 1985, 28, 849 857
  9. 9
    Sciabola, S.; Stanton, R. V.; Mills, J. E.; Flocco, M. M.; Baroni, M.; Cruciani, G.; Perruccio, F.; Mason, J. S. High-throughput virtual screening of proteins using GRID molecular interaction fields J. Chem. Inf. Model. 2010, 50, 155 169
  10. 10
    Doré, A. S.; Robertson, N.; Errey, J. C.; Ng, I.; Hollenstein, K.; Tehan, B.; Hurrell, E.; Bennet, K.; Congreve, M.; Magnani, F.; Tate, C. G.; Weir, M.; Marshall, F. H. Structure of the adenosine A2A receptor in complex with ZM241385 and the xanthines XAC and caffeine Structure 2011, 19, 1283 1293
  11. 11
    J. Takagi, J.; Sato, K.; Hartwig, J. F.; Ishiyama, T.; Miyaura, N. Iridium-catalyzed C–H coupling reaction of heteroaromatic compounds with bis(pinacolato)diboron: regioselective synthesis of heteroarylboronates Tetrahedron Lett. 2002, 43, 5649 5651
  12. 12
    Murphy, J. M.; Liao, X.; Hartwig., J. F. Meta Halogenation of 1,3-Disubstituted Arenes via Iridium-Catalyzed Arene Borylation J. Am. Chem. Soc. 2007, 129, 15434 15435
  13. 13
    Ishiyama, T.; Takagi, J.; Nobuta, Y.; Miyaura., N. Iridium-catalyzed C–H borylation of arenes and heteroarenes: 1-chloro-3-iodo-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzene and 2-(4,4,5,5,-tetramethyl-1,3,2-dioxaborolan-2-yl)indole Org. Synth. 2005, 82, 126 132
  14. 14
    Lebon, G.; Warne, T.; Edwards, P. C.; Bennett, K.; Langmead, C. J.; Leslie, A, G. W.; Tate, C. G. Agonist-bound adenosine A2A receptor structures reveal common features of GPCR activation Nature 2011, 474, 521 523
  15. 15
    Higgs, C.; Beuming, T.; Sherman, W. Hydration site thermodynamics explain SARs for triazolylpurines analogues binding to the A2A receptor ACS Med. Chem. Lett. 2010, 1, 160 164
  16. 16
    Congreve, M.; Langmead, C. J.; Mason, J. S.; Marshall, F. H. Progress in Structure Based Drug Design for G Protein-Coupled Receptors J. Med. Chem. 2011, 54, 4283 4311
  17. 17
    Hopkins, A. L.; Groom, C. R.; Alex, A. Ligand efficiency: a useful metric for lead selection Drug Discovery Today 2004, 9, 430 431

Cited By

ARTICLE SECTIONS
Jump To

This article is cited by 267 publications.

  1. Shayma El-Atawneh, Amiram Goldblum. Activity Models of Key GPCR Families in the Central Nervous System: A Tool for Many Purposes. Journal of Chemical Information and Modeling 2023, 63 (11) , 3248-3262. https://doi.org/10.1021/acs.jcim.2c01531
  2. Yuxin Shi, Yi Chen, Liping Deng, Kui Du, Shaoyong Lu, Ting Chen. Structural Understanding of Peptide-Bound G Protein-Coupled Receptors: Peptide–Target Interactions. Journal of Medicinal Chemistry 2023, 66 (2) , 1083-1111. https://doi.org/10.1021/acs.jmedchem.2c01309
  3. Amanda Jane Lyons, Adam Clarke, Heidi Fisk, Bethany Jackson, Peter R. Moore, Samantha Oke, Thomas O. Ronson, Rebecca E. Meadows. Scaling up a C–H Borylation: Addressing the Safety Concerns of an Iridium-Catalyzed Process for Multikilo Scale Manufacture. Organic Process Research & Development 2022, 26 (5) , 1378-1388. https://doi.org/10.1021/acs.oprd.1c00432
  4. Zixi Zhu, Dale L. Boger. N1/N4 1,4-Cycloaddition of 1,2,4,5-Tetrazines with Enamines Promoted by the Lewis Acid ZnCl2. The Journal of Organic Chemistry 2022, 87 (9) , 6288-6301. https://doi.org/10.1021/acs.joc.2c00543
  5. Zhoushun Zhang, Songsong Li, Lei Chen, Zichao Guo. Study on the Safety Design of 3-Amino-6-Bromo-1,2,4-Triazine Drying Process Based on Thermal Analysis Technology. Organic Process Research & Development 2021, 25 (11) , 2516-2524. https://doi.org/10.1021/acs.oprd.1c00293
  6. Jin Cheng, Maozi Chen, Siyi Wang, Tianjian Liang, Hui Chen, Chih-Jung Chen, Zhiwei Feng, Xiang-Qun Xie. Binding Characterization of Agonists and Antagonists by MCCS: A Case Study from Adenosine A2A Receptor. ACS Chemical Neuroscience 2021, 12 (9) , 1606-1620. https://doi.org/10.1021/acschemneuro.1c00082
  7. Nurlybek Amangeldiuly, Dmitry Karlov, Maxim V. Fedorov. Baseline Model for Predicting Protein–Ligand Unbinding Kinetics through Machine Learning. Journal of Chemical Information and Modeling 2020, 60 (12) , 5946-5956. https://doi.org/10.1021/acs.jcim.0c00450
  8. Zixi Zhu, Christopher M. Glinkerman, Dale L. Boger. Selective N1/N4 1,4-Cycloaddition of 1,2,4,5-Tetrazines Enabled by Solvent Hydrogen Bonding. Journal of the American Chemical Society 2020, 142 (49) , 20778-20787. https://doi.org/10.1021/jacs.0c09775
  9. Francesca Deflorian, Laura Perez-Benito, Eelke B Lenselink, Miles Congreve, Herman W. T. van Vlijmen, Jonathan S. Mason, Chris de Graaf, Gary Tresadern. Accurate Prediction of GPCR Ligand Binding Affinity with Free Energy Perturbation. Journal of Chemical Information and Modeling 2020, 60 (11) , 5563-5579. https://doi.org/10.1021/acs.jcim.0c00449
  10. Fazhi Yu, Chenyu Zhu, Qiong Xie, Yonghui Wang. Adenosine A2A Receptor Antagonists for Cancer Immunotherapy. Journal of Medicinal Chemistry 2020, 63 (21) , 12196-12212. https://doi.org/10.1021/acs.jmedchem.0c00237
  11. Maria João Matos, Santiago Vilar, Saleta Vazquez-Rodriguez, Sonja Kachler, Karl-Norbert Klotz, Michela Buccioni, Giovanna Delogu, Lourdes Santana, Eugenio Uriarte, Fernanda Borges. Structure-Based Optimization of Coumarin hA3 Adenosine Receptor Antagonists. Journal of Medicinal Chemistry 2020, 63 (5) , 2577-2587. https://doi.org/10.1021/acs.jmedchem.9b01572
  12. Eleonora Comeo, Nicholas D. Kindon, Mark Soave, Leigh A. Stoddart, Laura E. Kilpatrick, Peter J. Scammells, Stephen J. Hill, Barrie Kellam. Subtype-Selective Fluorescent Ligands as Pharmacological Research Tools for the Human Adenosine A2A Receptor. Journal of Medicinal Chemistry 2020, 63 (5) , 2656-2672. https://doi.org/10.1021/acs.jmedchem.9b01856
  13. Dimitrios Stamatis, Panagiotis Lagarias, Kerry Barkan, Eleni Vrontaki, Graham Ladds, Antonios Kolocouris. Structural Characterization of Agonist Binding to an A3 Adenosine Receptor through Biomolecular Simulations and Mutagenesis Experiments. Journal of Medicinal Chemistry 2019, 62 (19) , 8831-8846. https://doi.org/10.1021/acs.jmedchem.9b01164
  14. Marco Betti, Daniela Catarzi, Flavia Varano, Matteo Falsini, Katia Varani, Fabrizio Vincenzi, Silvia Pasquini, Lorenzo di Cesare Mannelli, Carla Ghelardini, Elena Lucarini, Diego Dal Ben, Andrea Spinaci, Gianluca Bartolucci, Marta Menicatti, Vittoria Colotta. Modifications on the Amino-3,5-dicyanopyridine Core To Obtain Multifaceted Adenosine Receptor Ligands with Antineuropathic Activity. Journal of Medicinal Chemistry 2019, 62 (15) , 6894-6912. https://doi.org/10.1021/acs.jmedchem.9b00106
  15. Mairi M. Littleson, Andrew D. Campbell, Adam Clarke, Mark Dow, Gareth Ensor, Matthew C. Evans, Adam Herring, Bethany A. Jackson, Lucinda V. Jackson, Staffan Karlsson, David J. Klauber, Danny H. Legg, Kevin W. Leslie, Štefan Moravčík, Chris D. Parsons, Thomas O. Ronson, Rebecca E. Meadows. Synthetic Route Design of AZD4635, an A2AR Antagonist. Organic Process Research & Development 2019, 23 (7) , 1407-1419. https://doi.org/10.1021/acs.oprd.9b00171
  16. Andrew Potterton, Fouad S. Husseini, Michelle W. Y. Southey, Mike J. Bodkin, Alexander Heifetz, Peter V. Coveney, Andrea Townsend-Nicholson. Ensemble-Based Steered Molecular Dynamics Predicts Relative Residence Time of A2A Receptor Binders. Journal of Chemical Theory and Computation 2019, 15 (5) , 3316-3330. https://doi.org/10.1021/acs.jctc.8b01270
  17. Jiyue Zheng, Xiaohu Zhang, Xuechu Zhen. Development of Adenosine A2A Receptor Antagonists for the Treatment of Parkinson’s Disease: A Recent Update and Challenge. ACS Chemical Neuroscience 2019, 10 (2) , 783-791. https://doi.org/10.1021/acschemneuro.8b00313
  18. James J. Douglas, Bradley W. V. Adams, Helen Benson, Karl Broberg, Paul M. Gillespie, Oliver Hoult, Ameer K. Ibraheem, Sophie Janbon, Guillaume Janin, Chris D. Parsons, Ralph C. Sigerson, David J. Klauber. Multikilogram-Scale Preparation of AZD4635 via C–H Borylation and Bromination: The Corrosion of Tantalum by a Bromine/Methanol Mixture. Organic Process Research & Development 2019, 23 (1) , 62-68. https://doi.org/10.1021/acs.oprd.8b00342
  19. John A. Christopher, Zoltán Orgován, Miles Congreve, Andrew S. Doré, James C. Errey, Fiona H. Marshall, Jonathan S. Mason, Krzysztof Okrasa, Prakash Rucktooa, Maria J. Serrano-Vega, György G. Ferenczy, György M. Keserű. Structure-Based Optimization Strategies for G Protein-Coupled Receptor (GPCR) Allosteric Modulators: A Case Study from Analyses of New Metabotropic Glutamate Receptor 5 (mGlu5) X-ray Structures. Journal of Medicinal Chemistry 2019, 62 (1) , 207-222. https://doi.org/10.1021/acs.jmedchem.7b01722
  20. Hannah E. Bruce Macdonald, Christopher Cave-Ayland, Gregory A. Ross, Jonathan W. Essex. Ligand Binding Free Energies with Adaptive Water Networks: Two-Dimensional Grand Canonical Alchemical Perturbations. Journal of Chemical Theory and Computation 2018, 14 (12) , 6586-6597. https://doi.org/10.1021/acs.jctc.8b00614
  21. Joshua Pottel, Anat Levit, Magdalena Korczynska, Marcus Fischer, Brian K. Shoichet. The Recognition of Unrelated Ligands by Identical Proteins. ACS Chemical Biology 2018, 13 (9) , 2522-2533. https://doi.org/10.1021/acschembio.8b00443
  22. Panagiotis Lagarias, Eleni Vrontaki, George Lambrinidis, Dimitrios Stamatis, Marino Convertino, Gabriella Ortore, Thomas Mavromoustakos, Karl-Norbert Klotz, Antonios Kolocouris. Discovery of Novel Adenosine Receptor Antagonists through a Combined Structure- and Ligand-Based Approach Followed by Molecular Dynamics Investigation of Ligand Binding Mode. Journal of Chemical Information and Modeling 2018, 58 (4) , 794-815. https://doi.org/10.1021/acs.jcim.7b00455
  23. Matthew S. Dowling, Wenhua Jiao, Jie Hou, Yuchun Jiang, Shangsheng Gong. Modular Synthesis of 3,6-Disubstituted-1,2,4-triazines via the Cyclodehydration of β-Keto-N-acylsulfonamides with Hydrazine Salts. The Journal of Organic Chemistry 2018, 83 (7) , 4229-4238. https://doi.org/10.1021/acs.joc.8b00254
  24. Yoonji Lee, Shaherin Basith, and Sun Choi . Recent Advances in Structure-Based Drug Design Targeting Class A G Protein-Coupled Receptors Utilizing Crystal Structures and Computational Simulations. Journal of Medicinal Chemistry 2018, 61 (1) , 1-46. https://doi.org/10.1021/acs.jmedchem.6b01453
  25. Giuseppina La Sala, Sergio Decherchi, Marco De Vivo, and Walter Rocchia . Allosteric Communication Networks in Proteins Revealed through Pocket Crosstalk Analysis. ACS Central Science 2017, 3 (9) , 949-960. https://doi.org/10.1021/acscentsci.7b00211
  26. Francesca Spyrakis, Mostafa H. Ahmed, Alexander S. Bayden, Pietro Cozzini, Andrea Mozzarelli, and Glen E. Kellogg . The Roles of Water in the Protein Matrix: A Largely Untapped Resource for Drug Discovery. Journal of Medicinal Chemistry 2017, 60 (16) , 6781-6827. https://doi.org/10.1021/acs.jmedchem.7b00057
  27. Sheng Tian, Xu Wang, Linlang Li, Xiaohu Zhang, Youyong Li, Feng Zhu, Tingjun Hou, and Xuechu Zhen . Discovery of Novel and Selective Adenosine A2A Receptor Antagonists for Treating Parkinson’s Disease through Comparative Structure-Based Virtual Screening. Journal of Chemical Information and Modeling 2017, 57 (6) , 1474-1487. https://doi.org/10.1021/acs.jcim.7b00188
  28. Marta Arimont, Shan-Liang Sun, Rob Leurs, Martine Smit, Iwan J. P. de Esch, and Chris de Graaf . Structural Analysis of Chemokine Receptor–Ligand Interactions. Journal of Medicinal Chemistry 2017, 60 (12) , 4735-4779. https://doi.org/10.1021/acs.jmedchem.6b01309
  29. Dong Guo, Laura H. Heitman, and Adriaan P. IJzerman . Kinetic Aspects of the Interaction between Ligand and G Protein-Coupled Receptor: The Case of the Adenosine Receptors. Chemical Reviews 2017, 117 (1) , 38-66. https://doi.org/10.1021/acs.chemrev.6b00025
  30. Ali Jazayeri, Stephen P. Andrews, and Fiona H. Marshall . Structurally Enabled Discovery of Adenosine A2A Receptor Antagonists. Chemical Reviews 2017, 117 (1) , 21-37. https://doi.org/10.1021/acs.chemrev.6b00119
  31. Syeda Rehana Zia, Roberto Gaspari, Sergio Decherchi, and Walter Rocchia . Probing Hydration Patterns in Class-A GPCRs via Biased MD: The A2A Receptor. Journal of Chemical Theory and Computation 2016, 12 (12) , 6049-6061. https://doi.org/10.1021/acs.jctc.6b00475
  32. Terry Z. H. Gani, Efthymios I. Ioannidis, and Heather J. Kulik . Computational Discovery of Hydrogen Bond Design Rules for Electrochemical Ion Separation. Chemistry of Materials 2016, 28 (17) , 6207-6218. https://doi.org/10.1021/acs.chemmater.6b02378
  33. Dilip K. Tosh, Antonella Ciancetta, Eugene Warnick, Robert O’Connor, Zhoumou Chen, Elizabeth Gizewski, Steven Crane, Zhan-Guo Gao, John A. Auchampach, Daniela Salvemini, and Kenneth A. Jacobson . Purine (N)-Methanocarba Nucleoside Derivatives Lacking an Exocyclic Amine as Selective A3 Adenosine Receptor Agonists. Journal of Medicinal Chemistry 2016, 59 (7) , 3249-3263. https://doi.org/10.1021/acs.jmedchem.5b01998
  34. Abdelaziz El Maatougui, Jhonny Azuaje, Manuel González-Gómez, Gabriel Miguez, Abel Crespo, Carlos Carbajales, Luz Escalante, Xerardo García-Mera, Hugo Gutiérrez-de-Terán, and Eddy Sotelo . Discovery of Potent and Highly Selective A2B Adenosine Receptor Antagonist Chemotypes. Journal of Medicinal Chemistry 2016, 59 (5) , 1967-1983. https://doi.org/10.1021/acs.jmedchem.5b01586
  35. Anirudh Ranganathan, Leigh A. Stoddart, Stephen J. Hill, and Jens Carlsson . Fragment-Based Discovery of Subtype-Selective Adenosine Receptor Ligands from Homology Models. Journal of Medicinal Chemistry 2015, 58 (24) , 9578-9590. https://doi.org/10.1021/acs.jmedchem.5b01120
  36. Francesca Spyrakis, Paolo Benedetti, Sergio Decherchi, Walter Rocchia, Andrea Cavalli, Stefano Alcaro, Francesco Ortuso, Massimo Baroni, and Gabriele Cruciani . A Pipeline To Enhance Ligand Virtual Screening: Integrating Molecular Dynamics and Fingerprints for Ligand and Proteins. Journal of Chemical Information and Modeling 2015, 55 (10) , 2256-2274. https://doi.org/10.1021/acs.jcim.5b00169
  37. Andrea Bortolato, Francesca Deflorian, Dahlia R. Weiss, and Jonathan S. Mason . Decoding the Role of Water Dynamics in Ligand–Protein Unbinding: CRF1R as a Test Case. Journal of Chemical Information and Modeling 2015, 55 (9) , 1857-1866. https://doi.org/10.1021/acs.jcim.5b00440
  38. Riccardo Petrelli, Ilaria Torquati, Sonja Kachler, Livio Luongo, Sabatino Maione, Palmarisa Franchetti, Mario Grifantini, Ettore Novellino, Antonio Lavecchia, Karl-Norbert Klotz, and Loredana Cappellacci . 5′-C-Ethyl-tetrazolyl-N6-Substituted Adenosine and 2-Chloro-adenosine Derivatives as Highly Potent Dual Acting A1 Adenosine Receptor Agonists and A3 Adenosine Receptor Antagonists. Journal of Medicinal Chemistry 2015, 58 (5) , 2560-2566. https://doi.org/10.1021/acs.jmedchem.5b00074
  39. Anirudh Ranganathan, Ron O. Dror, and Jens Carlsson . Insights into the Role of Asp792.50 in β2 Adrenergic Receptor Activation from Molecular Dynamics Simulations. Biochemistry 2014, 53 (46) , 7283-7296. https://doi.org/10.1021/bi5008723
  40. Davide Sabbadin, Antonella Ciancetta, and Stefano Moro . Perturbation of Fluid Dynamics Properties of Water Molecules during G Protein-Coupled Receptor–Ligand Recognition: The Human A2A Adenosine Receptor as a Key Study. Journal of Chemical Information and Modeling 2014, 54 (10) , 2846-2855. https://doi.org/10.1021/ci500397y
  41. Antonella Ciancetta, Alberto Cuzzolin, and Stefano Moro . Alternative Quality Assessment Strategy to Compare Performances of GPCR-Ligand Docking Protocols: The Human Adenosine A2A Receptor as a Case Study. Journal of Chemical Information and Modeling 2014, 54 (8) , 2243-2254. https://doi.org/10.1021/ci5002857
  42. Manuel de Lera Ruiz, Yeon-Hee Lim, and Junying Zheng . Adenosine A2A Receptor as a Drug Discovery Target. Journal of Medicinal Chemistry 2014, 57 (9) , 3623-3650. https://doi.org/10.1021/jm4011669
  43. Davide Sabbadin and Stefano Moro . Supervised Molecular Dynamics (SuMD) as a Helpful Tool To Depict GPCR–Ligand Recognition Pathway in a Nanosecond Time Scale. Journal of Chemical Information and Modeling 2014, 54 (2) , 372-376. https://doi.org/10.1021/ci400766b
  44. Davide Sabbadin, Antonella Ciancetta, and Stefano Moro . Bridging Molecular Docking to Membrane Molecular Dynamics To Investigate GPCR–Ligand Recognition: The Human A2A Adenosine Receptor as a Key Study. Journal of Chemical Information and Modeling 2014, 54 (1) , 169-183. https://doi.org/10.1021/ci400532b
  45. Magdalena Bacilieri, Antonella Ciancetta, Silvia Paoletta, Stephanie Federico, Sandro Cosconati, Barbara Cacciari, Sabrina Taliani, Federico Da Settimo, Ettore Novellino, Karl Norbert Klotz, Giampiero Spalluto, and Stefano Moro . Revisiting a Receptor-Based Pharmacophore Hypothesis for Human A2A Adenosine Receptor Antagonists. Journal of Chemical Information and Modeling 2013, 53 (7) , 1620-1637. https://doi.org/10.1021/ci300615u
  46. Andrea Bortolato, Ben G. Tehan, Michael S. Bodnarchuk, Jonathan W. Essex, and Jonathan S. Mason . Water Network Perturbation in Ligand Binding: Adenosine A2A Antagonists as a Case Study. Journal of Chemical Information and Modeling 2013, 53 (7) , 1700-1713. https://doi.org/10.1021/ci4001458
  47. Jianing Li, Amanda L. Jonsson, Thijs Beuming, John C. Shelley, and Gregory A. Voth . Ligand-Dependent Activation and Deactivation of the Human Adenosine A2A Receptor. Journal of the American Chemical Society 2013, 135 (23) , 8749-8759. https://doi.org/10.1021/ja404391q
  48. Hui Wen Ng, Charles A. Laughton, and Stephen W. Doughty . Molecular Dynamics Simulations of the Adenosine A2a Receptor: Structural Stability, Sampling, and Convergence. Journal of Chemical Information and Modeling 2013, 53 (5) , 1168-1178. https://doi.org/10.1021/ci300610w
  49. Dan Chen, James C. Errey, Laura H. Heitman, Fiona H. Marshall, Adriaan P. IJzerman, and Gregg Siegal . Fragment Screening of GPCRs Using Biophysical Methods: Identification of Ligands of the Adenosine A2A Receptor with Novel Biological Activity. ACS Chemical Biology 2012, 7 (12) , 2064-2073. https://doi.org/10.1021/cb300436c
  50. Stephanie Federico, Antonella Ciancetta, Davide Sabbadin, Silvia Paoletta, Giorgia Pastorin, Barbara Cacciari, Karl Norbert Klotz, Stefano Moro, and Giampiero Spalluto . Exploring the Directionality of 5-Substitutions in a New Series of 5-Alkylaminopyrazolo[4,3-e]1,2,4-triazolo[1,5-c]pyrimidine as a Strategy To Design Novel Human A3 Adenosine Receptor Antagonists.. Journal of Medicinal Chemistry 2012, 55 (22) , 9654-9668. https://doi.org/10.1021/jm300899q
  51. Christopher J. Langmead, Stephen P. Andrews, Miles Congreve, James C. Errey, Edward Hurrell, Fiona H. Marshall, Jonathan S. Mason, Christine M. Richardson, Nathan Robertson, Andrei Zhukov, and Malcolm Weir . Identification of Novel Adenosine A2A Receptor Antagonists by Virtual Screening. Journal of Medicinal Chemistry 2012, 55 (5) , 1904-1909. https://doi.org/10.1021/jm201455y
  52. Miru Tang, Chang Wen, Jie Lin, Hongming Chen, Ting Ran. Discovery of novel A2AR antagonists through deep learning-based virtual screening. Artificial Intelligence in the Life Sciences 2023, 3 , 100058. https://doi.org/10.1016/j.ailsci.2023.100058
  53. Hyun Jung Yoon, Sibsankar Kundu, Sangwook Wu. Molecular Dynamics Simulation Study of the Selective Inhibition of Coagulation Factor IXa over Factor Xa. Molecules 2023, 28 (19) , 6909. https://doi.org/10.3390/molecules28196909
  54. Pallab Karjee, Santu Mandal, Bijoy Debnath, Nirali Namdev, Tharmalingam Punniyamurthy. Expedient (3+3)-annulation of in situ generated azaoxyallyl cations with diaziridines. Chemical Communications 2023, 59 (53) , 8270-8273. https://doi.org/10.1039/D3CC02136J
  55. Glen E. Kellogg, Anna Marabotti, Francesca Spyrakis, Andrea Mozzarelli. HINT, a code for understanding the interaction between biomolecules: a tribute to Donald J. Abraham. Frontiers in Molecular Biosciences 2023, 10 https://doi.org/10.3389/fmolb.2023.1194962
  56. Pablo Rayff da Silva, Jéssica Cabral de Andrade, Natália Ferreira de Sousa, Anne Caroline Ribeiro Portela, Hugo Fernandes Oliveira Pires, Maria Caroline Rodrigues Bezerra Remígio, Danielle da Nóbrega Alves, Humberto Hugo Nunes de Andrade, Arthur Lins Dias, Mirian Graciela da Silva Stiebbe Salvadori, Adriana Maria Fernandes de Oliveira Golzio, Ricardo Dias de Castro, Marcus T. Scotti, Cícero Francisco Bezerra Felipe, Reinaldo Nóbrega de Almeida, Luciana Scotti. Computational Studies Applied to Linalool and Citronellal Derivatives Against Alzheimer's and Parkinson's Disorders: A Review with Experimental Approach. Current Neuropharmacology 2023, 21 (4) , 842-866. https://doi.org/10.2174/1570159X21666230221123059
  57. Jean-Louis Banères, Thomas Botzanowski, Jean A. Boutin, Barbara Calamini, Jérôme Castel, Laurent J. Catoire, Sarah Cianférani, Claire Demesmay, Gavin Ferguson, Gilles Ferry, Julie Kniazeff, Isabelle Krimm, Thierry Langer, Guillaume Lebon, Marie Ley, Miklos Nyerges, Magali Schwob, Catherine Venien-Bryan, Renaud Wagner, Gabrielle Zeder-Lutz, Claudia Zilian-Stohrer. Biophysical Dissection of Isolated GPCRs: The Adenosine A2A Receptor under the Bistouries. Receptors 2023, 2 (1) , 47-92. https://doi.org/10.3390/receptors2010004
  58. Andrea Spinaci, Michela Buccioni, Cui Chang, Diego Dal Ben, Beatrice Francucci, Catia Lambertucci, Rosaria Volpini, Gabriella Marucci. Adenosine A2A Receptor Antagonists: Chemistry, SARs, and Therapeutic Potential. 2023, 101-141. https://doi.org/10.1007/7355_2023_162
  59. Ri Han, Hongryul Yoon, Jiho Yoo, Yoonji Lee. Systematic analyses of the sequence conservation and ligand interaction patterns of purinergic P1 and P2Y receptors provide a structural basis for receptor selectivity. Computational and Structural Biotechnology Journal 2023, 21 , 889-898. https://doi.org/10.1016/j.csbj.2023.01.010
  60. Theresa Noonan, Katrin Denzinger, Valerij Talagayev, Yu Chen, Kristina Puls, Clemens Alexander Wolf, Sijie Liu, Trung Ngoc Nguyen, Gerhard Wolber. Mind the Gap—Deciphering GPCR Pharmacology Using 3D Pharmacophores and Artificial Intelligence. Pharmaceuticals 2022, 15 (11) , 1304. https://doi.org/10.3390/ph15111304
  61. Joydip Mondal, Akella Sivaramakrishna. Functionalized Triazines and Tetrazines: Synthesis and Applications. Topics in Current Chemistry 2022, 380 (5) https://doi.org/10.1007/s41061-022-00385-7
  62. Elissa A. Fink, Jun Xu, Harald Hübner, Joao M. Braz, Philipp Seemann, Charlotte Avet, Veronica Craik, Dorothee Weikert, Maximilian F. Schmidt, Chase M. Webb, Nataliya A. Tolmachova, Yurii S. Moroz, Xi-Ping Huang, Chakrapani Kalyanaraman, Stefan Gahbauer, Geng Chen, Zheng Liu, Matthew P. Jacobson, John J. Irwin, Michel Bouvier, Yang Du, Brian K. Shoichet, Allan I. Basbaum, Peter Gmeiner. Structure-based discovery of nonopioid analgesics acting through the α 2A -adrenergic receptor. Science 2022, 377 (6614) https://doi.org/10.1126/science.abn7065
  63. Limei M. Gao, Xiaomeng M. Wang, Qinglang L. Wei, Kexin X. Su, Renhong H. Huang, Ju Guo, Yongsheng S. Zheng, Jikai K. Liu. [3+3] Cycloadditions of Azomethine Ylides with Nitrile Imines for the Synthesis of 2,3,4,5‐Tetrahydro‐1,2,4‐Triazine‐5‐Carboxylates. European Journal of Organic Chemistry 2022, 2022 (35) https://doi.org/10.1002/ejoc.202200768
  64. Abid Mahmood, Jamshed Iqbal. Purinergic receptors modulators: An emerging pharmacological tool for disease management. Medicinal Research Reviews 2022, 42 (4) , 1661-1703. https://doi.org/10.1002/med.21888
  65. Zhichao Zhong, Xingrui He, Jiamin Ge, Junlong Zhu, Chuansheng Yao, Hong Cai, Xiang-Yang Ye, Tian Xie, Renren Bai. Discovery of small-molecule compounds and natural products against Parkinson's disease: Pathological mechanism and structural modification. European Journal of Medicinal Chemistry 2022, 237 , 114378. https://doi.org/10.1016/j.ejmech.2022.114378
  66. Kenneth A. Jacobson, Zhan‐Guo Gao, Pierre Matricon, Matthew T. Eddy, Jens Carlsson. Adenosine A 2A receptor antagonists: from caffeine to selective non‐xanthines. British Journal of Pharmacology 2022, 179 (14) , 3496-3511. https://doi.org/10.1111/bph.15103
  67. Atukuri Dorababu. Promising heterocycle-based scaffolds in recent (2019–2021) anti-Alzheimer's drug design and discovery. European Journal of Pharmacology 2022, 920 , 174847. https://doi.org/10.1016/j.ejphar.2022.174847
  68. Adriaan P. IJzerman, Kenneth A. Jacobson, Christa E. Müller, Bruce N. Cronstein, Rodrigo A. Cunha, . International Union of Basic and Clinical Pharmacology. CXII: Adenosine Receptors: A Further Update. Pharmacological Reviews 2022, 74 (2) , 340-372. https://doi.org/10.1124/pharmrev.121.000445
  69. Jinan Wang, Apurba Bhattarai, Hung N. Do, Sana Akhter, Yinglong Miao. Molecular Simulations and Drug Discovery of Adenosine Receptors. Molecules 2022, 27 (7) , 2054. https://doi.org/10.3390/molecules27072054
  70. S.M. Ivanov. 1,2,4-Triazines and Their Benzo Derivatives. 2022, 29-180. https://doi.org/10.1016/B978-0-12-818655-8.00062-7
  71. Anjali Saini, Rajiv Patel, Sobhi Gaba, Gurpreet Singh, G.D. Gupta, Vikramdeep Monga. Adenosine receptor antagonists: Recent advances and therapeutic perspective. European Journal of Medicinal Chemistry 2022, 227 , 113907. https://doi.org/10.1016/j.ejmech.2021.113907
  72. Miru Tang, Chang Wen, Lin Jie, Hongming Chen, Ting Ran. Discovery of Novel A 2AR Antagonists Through Deep Learning-Based Virtual Screening. SSRN Electronic Journal 2022, 31 https://doi.org/10.2139/ssrn.4188435
  73. Milica Karadzic-Banjac, Benjamin Salakovic, Strahinja Kovacevic, Lidija Jevric, Sanja Podunavac-Kuzmanovic. Insight in triazine chromatographic and microbiological analysis: A brief review. Acta Periodica Technologica 2022, (53) , 241-252. https://doi.org/10.2298/APT2253241K
  74. Flavio Ballante, Albert J Kooistra, Stefanie Kampen, Chris de Graaf, Jens Carlsson, . Structure-Based Virtual Screening for Ligands of G Protein–Coupled Receptors: What Can Molecular Docking Do for You?. Pharmacological Reviews 2021, 73 (4) , 1698-1736. https://doi.org/10.1124/pharmrev.120.000246
  75. João Paulo L Velloso, David B Ascher, Douglas E V Pires, . pdCSM-GPCR: predicting potent GPCR ligands with graph-based signatures. Bioinformatics Advances 2021, 1 (1) https://doi.org/10.1093/bioadv/vbab031
  76. Kenneth A. Jacobson, Adriaan P. IJzerman, Christa E. Müller. Medicinal chemistry of P2 and adenosine receptors: Common scaffolds adapted for multiple targets. Biochemical Pharmacology 2021, 187 , 114311. https://doi.org/10.1016/j.bcp.2020.114311
  77. Siyu Zhu, Meixian Wu, Ziwei Huang, Jing An. Trends in application of advancing computational approaches in GPCR ligand discovery. Experimental Biology and Medicine 2021, 246 (9) , 1011-1024. https://doi.org/10.1177/1535370221993422
  78. Miles Congreve, John A. Christopher, Chris de Graaf. Structure‐Based Drug Design for G Protein‐Coupled Receptors. 2021, 1-59. https://doi.org/10.1002/0471266949.bmc269
  79. Andrew W. Buesking, Patrick A. Mayes, Andrew P. Combs. Immuno‐Oncology. 2021, 1-30. https://doi.org/10.1002/0471266949.bmc272
  80. Hung N. Do, Sana Akhter, Yinglong Miao. Pathways and Mechanism of Caffeine Binding to Human Adenosine A2A Receptor. Frontiers in Molecular Biosciences 2021, 8 https://doi.org/10.3389/fmolb.2021.673170
  81. Veronica Salmaso, Shanu Jain, Kenneth A. Jacobson. Purinergic GPCR transmembrane residues involved in ligand recognition and dimerization. 2021, 133-159. https://doi.org/10.1016/bs.mcb.2021.06.001
  82. Hiroaki Hata, Duy Phuoc Tran, Mohamed Marzouk Sobeh, Akio Kitao. Binding free energy of protein/ligand complexes calculated using dissociation Parallel Cascade Selection Molecular Dynamics and Markov state model. Biophysics and Physicobiology 2021, 18 (0) , 305-316. https://doi.org/10.2142/biophysico.bppb-v18.037
  83. Abhinav R. Jain, Claire McGraw, Anne S. Robinson. The Specificity of Downstream Signaling for A1 and A2AR Does Not Depend on the C-Terminus, Despite the Importance of This Domain in Downstream Signaling Strength. Biomedicines 2020, 8 (12) , 603. https://doi.org/10.3390/biomedicines8120603
  84. Doris A. Schuetz, Lars Richter, Riccardo Martini, Gerhard F. Ecker. A structure–kinetic relationship study using matched molecular pair analysis. RSC Medicinal Chemistry 2020, 11 (11) , 1285-1294. https://doi.org/10.1039/D0MD00178C
  85. Veronica Salmaso, Kenneth A. Jacobson. Purinergic Signaling: Impact of GPCR Structures on Rational Drug Design. ChemMedChem 2020, 15 (21) , 1958-1973. https://doi.org/10.1002/cmdc.202000465
  86. Jinjin Chen, Zhaozhao Sun, Fuhong Xiao, Guo-Jun Deng. Base-promoted aerobic oxidative synthesis of fused 1,3,5-triazines under metal-free conditions. Green Chemistry 2020, 22 (20) , 6778-6782. https://doi.org/10.1039/D0GC02691C
  87. Priyanka De, Joyita Roy, Dhananjay Bhattacharyya, Kunal Roy. Chemometric modeling of PET imaging agents for diagnosis of Parkinson’s disease: a QSAR approach. Structural Chemistry 2020, 31 (5) , 1969-1981. https://doi.org/10.1007/s11224-020-01560-6
  88. Willem Jespers, Grégory Verdon, Jhonny Azuaje, Maria Majellaro, Henrik Keränen, Xerardo García‐Mera, Miles Congreve, Francesca Deflorian, Chris de Graaf, Andrei Zhukov, Andrew S. Doré, Jonathan S. Mason, Johan Åqvist, Robert M. Cooke, Eddy Sotelo, Hugo Gutiérrez‐de‐Terán. X‐Ray Crystallography and Free Energy Calculations Reveal the Binding Mechanism of A 2A Adenosine Receptor Antagonists. Angewandte Chemie 2020, 132 (38) , 16679-16686. https://doi.org/10.1002/ange.202003788
  89. Willem Jespers, Grégory Verdon, Jhonny Azuaje, Maria Majellaro, Henrik Keränen, Xerardo García‐Mera, Miles Congreve, Francesca Deflorian, Chris de Graaf, Andrei Zhukov, Andrew S. Doré, Jonathan S. Mason, Johan Åqvist, Robert M. Cooke, Eddy Sotelo, Hugo Gutiérrez‐de‐Terán. X‐Ray Crystallography and Free Energy Calculations Reveal the Binding Mechanism of A 2A Adenosine Receptor Antagonists. Angewandte Chemie International Edition 2020, 59 (38) , 16536-16543. https://doi.org/10.1002/anie.202003788
  90. Saleta Vazquez-Rodriguez, Santiago Vilar, Sonja Kachler, Karl-Norbert Klotz, Eugenio Uriarte, Fernanda Borges, Maria João Matos. Adenosine Receptor Ligands: Coumarin–Chalcone Hybrids as Modulating Agents on the Activity of hARs. Molecules 2020, 25 (18) , 4306. https://doi.org/10.3390/molecules25184306
  91. Jinfeng Zhang, Wenzhong Yan, Wenwen Duan, Kurt Wüthrich, Jianjun Cheng. Tumor Immunotherapy Using A2A Adenosine Receptor Antagonists. Pharmaceuticals 2020, 13 (9) , 237. https://doi.org/10.3390/ph13090237
  92. Guillaume Ferré, Matthew T. Eddy. Structural biology of human GPCR drugs and endogenous ligands - insights from NMR spectroscopy. Methods 2020, 180 , 79-88. https://doi.org/10.1016/j.ymeth.2020.08.008
  93. Flavia Varano, Daniela Catarzi, Erica Vigiani, Fabrizio Vincenzi, Silvia Pasquini, Katia Varani, Vittoria Colotta. Piperazine- and Piperidine-Containing Thiazolo[5,4-d]pyrimidine Derivatives as New Potent and Selective Adenosine A2A Receptor Inverse Agonists. Pharmaceuticals 2020, 13 (8) , 161. https://doi.org/10.3390/ph13080161
  94. Darren V. S. Green, Stephen Pickett, Chris Luscombe, Stefan Senger, David Marcus, Jamel Meslamani, David Brett, Adam Powell, Jonathan Masson. BRADSHAW: a system for automated molecular design. Journal of Computer-Aided Molecular Design 2020, 34 (7) , 747-765. https://doi.org/10.1007/s10822-019-00234-8
  95. Fengxu Wu, Linsheng Zhuo, Fan Wang, Wei Huang, Gefei Hao, Guangfu Yang. Auto In Silico Ligand Directing Evolution to Facilitate the Rapid and Efficient Discovery of Drug Lead. iScience 2020, 23 (6) , 101179. https://doi.org/10.1016/j.isci.2020.101179
  96. Veronica Salmaso, Kenneth A. Jacobson. In Silico Drug Design for Purinergic GPCRs: Overview on Molecular Dynamics Applied to Adenosine and P2Y Receptors. Biomolecules 2020, 10 (6) , 812. https://doi.org/10.3390/biom10060812
  97. Agustín Bruzzese, James A. R. Dalton, Jesús Giraldo, . Insights into adenosine A2A receptor activation through cooperative modulation of agonist and allosteric lipid interactions. PLOS Computational Biology 2020, 16 (4) , e1007818. https://doi.org/10.1371/journal.pcbi.1007818
  98. Miles Congreve, Chris de Graaf, Nigel A. Swain, Christopher G. Tate. Impact of GPCR Structures on Drug Discovery. Cell 2020, 181 (1) , 81-91. https://doi.org/10.1016/j.cell.2020.03.003
  99. Yao-Guo Qin, Zhao-Kai Yang, Jia Fan, Xin Jiang, Xin-Ling Yang, Ju-Lian Chen. Synthesis, Crystal Structure and Bioactivities of N-(5-(4-chlorobenzyl)-1,3,5-Triazinan-2-Ylidene)Nitramide. Crystals 2020, 10 (4) , 245. https://doi.org/10.3390/cryst10040245
  100. Reed M. Stein, Hye Jin Kang, John D. McCorvy, Grant C. Glatfelter, Anthony J. Jones, Tao Che, Samuel Slocum, Xi-Ping Huang, Olena Savych, Yurii S. Moroz, Benjamin Stauch, Linda C. Johansson, Vadim Cherezov, Terry Kenakin, John J. Irwin, Brian K. Shoichet, Bryan L. Roth, Margarita L. Dubocovich. Virtual discovery of melatonin receptor ligands to modulate circadian rhythms. Nature 2020, 579 (7800) , 609-614. https://doi.org/10.1038/s41586-020-2027-0
Load more citations
  • Abstract

    Figure 1

    Figure 1. (A,B) BPM fingerprint of 1,2,4-triazine adenosine A2A antagonists. Compounds 4g (A) and 4e (B) are illustrated bound to the orthosteric pocket of the receptor and the residues lining the pocket that interact with the ligands are labeled. The tier 1, 2, and 3 designation is described in the main text. The key hydrogen bonding to Asn2536.55 of the scaffold is highlighted by green dotted lines. (C,D) Illustration of the A2A–StaR2 ligand binding site in complex with compound 4g (C) and 4e (D). TM helices and visible extracellular regions are depicted in the rainbow format. Ligands are represented as stick models, carbon and chlorine atoms are green, oxygen atoms red, and nitrogen atoms blue. Residues involved in ligand binding are labeled and represented as gray sticks, oxygen atoms are red, and nitrogen atoms are blue. Extracellular loop 2, the key binding site residues and TM’s 1, 2, 5, and 6 are labeled for reference. Potential H-bonds between the ligand and receptor are represented as dashed blue lines. TM3 and TM4 have been omitted for clarity. (E) WaterMap calculation on the binding site of compound 4e (ligand removed for the calculation). Waters calculated are color coded to show the most “unhappy” vs bulk solvent as red (>3.5 kcal/mol), then yellow (2.2–3.5 kcal/mol), with gray intermediate (−1 to 2.2 kcal) and blue “happy” (<−1 kcal/mol). The CPK surface of the ligand 4e is shown as a red dot surface, clearly illustrating that the cluster of red and yellow “unhappy” waters deep in the binding site have been displaced. GRID maps are also shown that highlight the shape (Csp3 (C3) at 1 kcal/mol in light-gray), the lipophilic hotspots (aromatic CH probe (C1═) in yellow at −2.5 kcal/mol), and the water probe hotspots (in green wire mesh at −6.6 kcal/mol). (F) Alignment of the A2A homology model with 4e docked (cyan carbons) onto the crystal structure of A2A4e complex (green carbons). The alignment was generated by the align algorithm in Pymol utilizing only helices where hydrogen bonds are formed with the ligand, helices 6 and 7. Helices 2, 3, and 4 are removed for clarity.

    Scheme 1

    Scheme 1. Synthesis of 5,6-Biaryl-1,2,4-triazine-3-amine Derivatives (4) and 4-Pyridylboronic Acid Derivatives (6)a

    Scheme aReagents and conditions: (a) NBS, DMF, RT; (b) 3, Pd(PPh3)4, K2CO3, 1,4-dioxane/H2O, 150 °C; (c) [Ir(COD)OMe]2, DTBPY, [B(pin)]2, hexane, 50 °C.

    Figure 2

    Figure 2. In vivo efficacy of 4k. Dose-dependent effect of 4k (0.1–1 mg/kg, po; 1 and 2 h pretreatment time) to reverse haloperidol-induced catalepsy in rats in comparison with the positive control, istradefylline (1 mg/kg, po).

  • References

    ARTICLE SECTIONS
    Jump To

    This article references 17 other publications.

    1. 1
      Shah, U.; Hodgson, R. Recent progress in the discovery of adenosine A2A receptor antagonists for the treatment of Parkinson’s disease Curr. Opin. Drug Discovery Dev. 2010, 13, 466 480
    2. 2
      Salamone, J. D. Preladenant, a novel adenosine A2A receptor antagonist for the potential treatment of parkinsonism and other disorders IDrugs 2010, 13, 723 731
    3. 3
      Langmead, C. J.; Andrews, S.; Congreve, M.; Errey, J.; Hurrell, E.; Marshall, F. H.; Mason, J. S.; Richardson, C.; Robertson, N.; Zhukov, A.; Weir, M. Identification of Novel Adenosine A2A Receptor Antagonists by Virtual Screening.

      Just accepted.

       DOI: DOI: 10.1021/jm201455y . Published online: January17, 2012
    4. 4
      Zhukov, A.; Andrews, S. P.; Errey, J. C.; Robertson, N.; Tehan, B.; Mason, J. S.; Marshall, F. H.; Weir, M.; Congreve, M. Biophysical Mapping of the Adenosine A2A Receptor J. Med. Chem. 2011, 54, 4312 4323
    5. 5
      Dal Ben, D.; Lambertucci, C.; Marucci, G.; Volpini, R.; Cristalli, G. Adenosine Receptor Modeling: What Does the A2A Crystal Structure Tell Us? Curr. Top. Med. Chem. 2010, 93, 993 1018
    6. 6
      Kim, S. K.; Gao, Z.; Van Rompaey, P.; Gross, A. S.; Chen, A.; Van Calenbergh, S.; Jacobson, K. A. Modeling the adenosine receptors: comparison of the binding domains of A2A agonists and antagonists J. Med. Chem. 2003, 46, 4847 4859
    7. 7
      Ballesteros, J. A.; Weinstein, H.; Stuart, C. S. Integrated methods for the construction of three dimensional models and computational probing of structure–function relations in G protein coupled receptors. In Methods in Neurosciences; Academic Press: New York, 1995; Vol. 25, pp 366 428.
    8. 8
      Goodford, P. J. A computational procedure for determining energetically favorable binding sites on biologically important macromolecules J. Med. Chem. 1985, 28, 849 857
    9. 9
      Sciabola, S.; Stanton, R. V.; Mills, J. E.; Flocco, M. M.; Baroni, M.; Cruciani, G.; Perruccio, F.; Mason, J. S. High-throughput virtual screening of proteins using GRID molecular interaction fields J. Chem. Inf. Model. 2010, 50, 155 169
    10. 10
      Doré, A. S.; Robertson, N.; Errey, J. C.; Ng, I.; Hollenstein, K.; Tehan, B.; Hurrell, E.; Bennet, K.; Congreve, M.; Magnani, F.; Tate, C. G.; Weir, M.; Marshall, F. H. Structure of the adenosine A2A receptor in complex with ZM241385 and the xanthines XAC and caffeine Structure 2011, 19, 1283 1293
    11. 11
      J. Takagi, J.; Sato, K.; Hartwig, J. F.; Ishiyama, T.; Miyaura, N. Iridium-catalyzed C–H coupling reaction of heteroaromatic compounds with bis(pinacolato)diboron: regioselective synthesis of heteroarylboronates Tetrahedron Lett. 2002, 43, 5649 5651
    12. 12
      Murphy, J. M.; Liao, X.; Hartwig., J. F. Meta Halogenation of 1,3-Disubstituted Arenes via Iridium-Catalyzed Arene Borylation J. Am. Chem. Soc. 2007, 129, 15434 15435
    13. 13
      Ishiyama, T.; Takagi, J.; Nobuta, Y.; Miyaura., N. Iridium-catalyzed C–H borylation of arenes and heteroarenes: 1-chloro-3-iodo-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzene and 2-(4,4,5,5,-tetramethyl-1,3,2-dioxaborolan-2-yl)indole Org. Synth. 2005, 82, 126 132
    14. 14
      Lebon, G.; Warne, T.; Edwards, P. C.; Bennett, K.; Langmead, C. J.; Leslie, A, G. W.; Tate, C. G. Agonist-bound adenosine A2A receptor structures reveal common features of GPCR activation Nature 2011, 474, 521 523
    15. 15
      Higgs, C.; Beuming, T.; Sherman, W. Hydration site thermodynamics explain SARs for triazolylpurines analogues binding to the A2A receptor ACS Med. Chem. Lett. 2010, 1, 160 164
    16. 16
      Congreve, M.; Langmead, C. J.; Mason, J. S.; Marshall, F. H. Progress in Structure Based Drug Design for G Protein-Coupled Receptors J. Med. Chem. 2011, 54, 4283 4311
    17. 17
      Hopkins, A. L.; Groom, C. R.; Alex, A. Ligand efficiency: a useful metric for lead selection Drug Discovery Today 2004, 9, 430 431
  • Supporting Information

    Supporting Information

    ARTICLE SECTIONS
    Jump To

    Crystallographic table of statistics. Synthesis protocols, 1H NMR, purification details, yields, purities by HPLC and MS or LCMS. Biological protocols for in vitro and in vivo experiments. Computational chemistry methods. SPR binding and kinetic data. This material is available free of charge via the Internet at http://pubs.acs.org.


    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect