Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
1.
J Med Chem ; 67(12): 10490-10507, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38845345

ABSTRACT

Building on the preceding structural analysis and a structure-activity relationship (SAR) of 8-aryl-2-hexynyl nucleoside hA2AAR antagonist 2a, we strategically inverted C2/C8 substituents and eliminated the ribose moiety. These modifications aimed to mitigate potential steric interactions between ribose and adenosine receptors. The SAR findings indicated that such inversions significantly modulated hA3AR binding affinities depending on the type of ribose, whereas removal of ribose altered the functional efficacy via hA2AAR. Among the synthesized derivatives, 2-aryl-8-hexynyl adenine 4a demonstrated the highest selectivity for hA2AAR (Ki,hA2A = 5.0 ± 0.5 nM, Ki,hA3/Ki,hA2A = 86) and effectively blocked cAMP production and restored IL-2 secretion in PBMCs. Favorable pharmacokinetic properties and a notable enhancement of anticancer effects in combination with an mAb immune checkpoint blockade were observed upon oral administration of 4a. These findings establish 4a as a viable immune-oncology therapeutic candidate.


Subject(s)
Adenine , Adenosine A2 Receptor Antagonists , Nucleosides , Receptor, Adenosine A2A , Ribose , Humans , Structure-Activity Relationship , Animals , Adenine/pharmacology , Adenine/chemistry , Adenine/analogs & derivatives , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/chemical synthesis , Nucleosides/chemistry , Nucleosides/pharmacology , Nucleosides/chemical synthesis , Ribose/chemistry , Ribose/metabolism , Receptor, Adenosine A2A/metabolism , Mice , Molecular Structure , Rats , Female , Cell Line, Tumor
2.
Sci China Life Sci ; 67(5): 986-995, 2024 May.
Article in English | MEDLINE | ID: mdl-38319473

ABSTRACT

The adenosine subfamily G protein-coupled receptors A2AR and A2BR have been identified as promising cancer immunotherapy candidates. One of the A2AR/A2BR dual antagonists, AB928, has progressed to a phase II clinical trial to treat rectal cancer. However, the precise mechanism underlying its dual-antagonistic properties remains elusive. Herein, we report crystal structures of the A2AR complexed with AB928 and a selective A2AR antagonist 2-118. The structures revealed a common binding mode on A2AR, wherein the ligands established extensive interactions with residues from the orthosteric and secondary pockets. In contrast, the cAMP assay and A2AR and A2BR molecular dynamics simulations indicated that the ligands adopted distinct binding modes on A2BR. Detailed analysis of their chemical structures suggested that AB928 readily adapted to the A2BR pocket, while 2-118 did not due to intrinsic differences. This disparity potentially accounted for the difference in inhibitory efficacy between A2BR and A2AR. This study serves as a valuable structural template for the future development of selective or dual inhibitors targeting A2AR/A2BR for cancer therapy.


Subject(s)
Adenosine A2 Receptor Antagonists , Molecular Dynamics Simulation , Receptor, Adenosine A2A , Humans , Adenosine A2 Receptor Antagonists/chemistry , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/metabolism , Binding Sites , Ligands , Crystallography, X-Ray , Protein Binding , Receptor, Adenosine A2B/metabolism , Receptor, Adenosine A2B/chemistry
3.
Int J Mol Sci ; 24(17)2023 Aug 22.
Article in English | MEDLINE | ID: mdl-37685879

ABSTRACT

Numerous basic studies have reported on the neuroprotective properties of several purine derivatives such as caffeine and uric acid (UA). Epidemiological studies have also shown the inverse association of appropriate caffeine intake or serum urate levels with neurodegenerative diseases such as Alzheimer disease (AD) and Parkinson's disease (PD). The well-established neuroprotective mechanisms of caffeine and UA involve adenosine A2A receptor antagonism and antioxidant activity, respectively. Our recent study found that another purine derivative, paraxanthine, has neuroprotective effects similar to those of caffeine and UA. These purine derivatives can promote neuronal cysteine uptake through excitatory amino acid carrier protein 1 (EAAC1) to increase neuronal glutathione (GSH) levels in the brain. This review summarizes the GSH-mediated neuroprotective effects of purine derivatives. Considering the fact that GSH depletion is a manifestation in the brains of AD and PD patients, administration of purine derivatives may be a new therapeutic approach to prevent or delay the onset of these neurodegenerative diseases.


Subject(s)
Alzheimer Disease , Glutathione , Neuroprotection , Neuroprotective Agents , Parkinson Disease , Purines , Humans , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/prevention & control , Brain/metabolism , Cysteine/metabolism , Excitatory Amino Acid Transporter 3/metabolism , Glutathione/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/prevention & control , Purines/chemistry , Purines/pharmacology , Purines/therapeutic use , Receptor, Adenosine A2A , Theophylline/chemistry , Theophylline/pharmacology , Theophylline/therapeutic use , Uric Acid/blood , Caffeine/chemistry , Caffeine/pharmacology , Caffeine/therapeutic use
4.
Biomolecules ; 13(7)2023 07 05.
Article in English | MEDLINE | ID: mdl-37509114

ABSTRACT

Multitarget drugs based on a hybrid dopamine-xanthine core were designed as potential drug candidates for the treatment of neurodegenerative diseases. Monoamine oxidase B (MAO-B) inhibitors with significant ancillary A2A adenosine receptor (A2AAR) antagonistic properties were further developed to exhibit additional phosphodiesterase-4 and -10 (PDE4/10) inhibition and/or dopamine D2 receptor (D2R) agonistic activity. While all of the designed compounds showed MAO-B inhibition in the nanomolar range mostly combined with submicromolar A2AAR affinity, significant enhancement of PDE-inhibitory and D2R-agonistic activity was additionally reached for some compounds through various structural modifications. The final multitarget drugs also showed promising antioxidant properties in vitro. In order to evaluate their potential neuroprotective effect, representative ligands were tested in a cellular model of toxin-induced neurotoxicity. As a result, protective effects against oxidative stress in neuroblastoma cells were observed, confirming the utility of the applied strategy. Further evaluation of the newly developed multitarget ligands in preclinical models of Alzheimer's and Parkinson's diseases is warranted.


Subject(s)
Neurodegenerative Diseases , Humans , Neurodegenerative Diseases/drug therapy , Xanthine/pharmacology , Xanthine/therapeutic use , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/chemistry , Dopamine , Ligands , Structure-Activity Relationship , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/therapeutic use , Monoamine Oxidase/metabolism , Dopamine Agents/pharmacology
5.
Eur J Med Chem ; 257: 115419, 2023 Sep 05.
Article in English | MEDLINE | ID: mdl-37301076

ABSTRACT

Development of subtype-selective leads is essential in drug discovery campaigns targeting G protein-coupled receptors (GPCRs). Herein, a structure-based virtual screening approach to rationally design subtype-selective ligands was applied to the A1 and A2A adenosine receptors (A1R and A2AR). Crystal structures of these closely related subtypes revealed a non-conserved subpocket in the binding sites that could be exploited to identify A1R selective ligands. A library of 4.6 million compounds was screened computationally against both receptors using molecular docking and 20 A1R selective ligands were predicted. Of these, seven antagonized the A1R with micromolar activities and several compounds displayed slight selectivity for this subtype. Twenty-seven analogs of two discovered scaffolds were designed, resulting in antagonists with nanomolar potency and up to 76-fold A1R-selectivity. Our results show the potential of structure-based virtual screening to guide discovery and optimization of subtype-selective ligands, which could facilitate the development of safer drugs.


Subject(s)
Adenosine , Purinergic P1 Receptor Antagonists , Purinergic P1 Receptor Antagonists/pharmacology , Purinergic P1 Receptor Antagonists/chemistry , Molecular Docking Simulation , Ligands , Binding Sites , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A1/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/chemistry
6.
BMC Endocr Disord ; 23(1): 97, 2023 May 04.
Article in English | MEDLINE | ID: mdl-37143025

ABSTRACT

BACKGROUND/AIM: Recent research suggests that adenosine receptors (ARs) influence many of the metabolic abnormalities associated with diabetes. A non-xanthine benzylidene indanone derivative 2-(3,4-dihydroxybenzylidene)-4-methoxy-2,3-dihydro-1 H-inden-1-one (2-BI), has shown to exhibit higher affinity at A1/A2A ARs compared to caffeine. Due to its structural similarity to caffeine, and the established antidiabetic effects of caffeine, the current study was initiated to explore the possible antidiabetic effect of 2-BI. METHODS: The study was designed to assess the antidiabetic effects of several A1 and/or A2A AR antagonists, via intestinal glucose absorption and glucose-lowering effects in fructose-streptozotocin (STZ) induced diabetic rats. Six-week-old male Sprague-Dawley rats were induced with diabetes via fructose and streptozotocin. Rats were treated for 4 weeks with AR antagonists, metformin and pioglitazone, respectively. Non-fasting blood glucose (NFBG) was determined weekly and the oral glucose tolerance test (OGTT) was conducted at the end of the intervention period. RESULTS: Dual A1/A2A AR antagonists (caffeine and 2-BI) decreased glucose absorption in the intestinal membrane significantly (p < 0.01), while the selective A2A AR antagonist (Istradefylline), showed the highest significant (p < 0.001) reduction in intestinal glucose absorption. The selective A1 antagonist (DPCPX) had the least significant (p < 0.05) reduction in glucose absorption. Similarly, dual A1/A2A AR antagonists and selective A2A AR antagonists significantly reduced non-fast blood glucose and improved glucose tolerance in diabetic rats from the first week of the treatment. Conversely, the selective A1 AR antagonist did not reduce non-fast blood glucose significantly until the 4th week of treatment. 2-BI, caffeine and istradefylline compared well with standard antidiabetic treatments, metformin and pioglitazone, and in some cases performed even better. CONCLUSION: 2-BI exhibited good antidiabetic activity by reducing intestinal postprandial glucose absorption and improving glucose tolerance in a diabetic animal model. The dual antagonism of A1/A2A ARs presents a positive synergism that could provide a new possibility for the treatment of diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Hyperglycemia , Metformin , Rats , Male , Animals , Purinergic P1 Receptor Antagonists , Caffeine/pharmacology , Streptozocin , Hyperglycemia/chemically induced , Hyperglycemia/drug therapy , Glucose , Pioglitazone , Blood Glucose , Diabetes Mellitus, Experimental/drug therapy , Rats, Sprague-Dawley , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Receptor, Adenosine A1/chemistry , Receptor, Adenosine A1/metabolism , Indans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use
7.
J Chem Inf Model ; 63(10): 3138-3149, 2023 05 22.
Article in English | MEDLINE | ID: mdl-37155356

ABSTRACT

We used a range of computational techniques to assess the effect of selective C-H deuteration on the antagonist istradefylline affinity for the adenosine A2A receptor, which was discussed relative to its structural analogue caffeine, a well-known and likely the most widely used stimulant. The obtained results revealed that smaller caffeine shows high receptor flexibility and exchanges between two distinct poses, which agrees with crystallographic data. In contrast, the additional C8-trans-styryl fragment in istradefylline locks the ligand within a uniform binding pose, while contributing to the affinity through the C-H···π and π···π contacts with surface residues, which, together with its much lower hydration prior to binding, enhances the affinity over caffeine. In addition, the aromatic C8-unit shows a higher deuteration sensitivity over the xanthine part, so when both of its methoxy groups are d6-deuterated, the affinity improvement is -0.4 kcal mol-1, which surpasses the overall affinity gain of -0.3 kcal mol-1 in the perdeuterated d9-caffeine. Yet, the latter predicts around 1.7-fold potency increase, being relevant for its pharmaceutical implementations, and also those within the coffee and energy drink production industries. Still, the full potential of our strategy is achieved in polydeuterated d19-istradefylline, whose A2A affinity improves by -0.6 kcal mol-1, signifying a 2.8-fold potency increase that strongly promotes it as a potential synthetic target. This knowledge supports deuterium application in drug design, and while the literature already reports about over 20 deuterated drugs currently in the clinical development, it is easily foreseen that more examples will hit the market in the years to come. With this in mind, we propose that the devised computational methodology, involving the ONIOM division of the QM region for the ligand and the MM region for its environment, with an implicit quantization of nuclear motions relevant for the H/D exchange, allows fast and efficient estimates of the binding isotope effects in any biological system.


Subject(s)
Caffeine , Receptor, Adenosine A2A , Caffeine/pharmacology , Caffeine/chemistry , Ligands , Receptor, Adenosine A2A/metabolism , Adenosine , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/chemistry
8.
J Integr Bioinform ; 19(4)2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36112816

ABSTRACT

Parkinson's disease is considered the second most frequent neurodegenerative disease. It is described by the loss of dopaminergic neurons in the mid-brain. For many decades, L-DOPA has been considered as the gold standard for treating Parkinson's disease motor symptoms, however, due to the decrease of efficacy, in the long run, there is an urgent need for novel antiparkinsonian drugs. Caffeine derivatives have been reported several times for their neuroprotective properties and dual blockade of monoamine oxidase (MAO) and adenosine A2A receptors (AA2AR). Natural products are currently attracting more focus due to structural diversity and safety in contrast to synthetic drugs. In the present work, computational studies were conducted on natural product-like caffeine derivatives to search for novel potent candidates acting as dual MAO-B inhibitors/AA2AR antagonists for Parkinson's disease. Our findings revealed two natural products among the top hits: CNP0202316 and CNP0365210 fulfill the requirements of drugs acting on the brain. The selected lead compounds were further studied using molecular dynamics simulation to assess their stability with MAO-B. Current findings might shift the interest towards natural-based compounds and could be exploited to further optimize caffeine derivatives into a successful dual-target-directed drug for managing and halting the neuronal damage in Parkinson's disease patients.


Subject(s)
Neurodegenerative Diseases , Parkinson Disease , Humans , Caffeine/pharmacology , Caffeine/therapeutic use , Parkinson Disease/drug therapy , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/therapeutic use , Monoamine Oxidase Inhibitors/chemistry , Neurodegenerative Diseases/drug therapy , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/therapeutic use , Adenosine A2 Receptor Antagonists/chemistry , Monoamine Oxidase/therapeutic use
9.
J Med Chem ; 65(17): 11648-11657, 2022 09 08.
Article in English | MEDLINE | ID: mdl-35977382

ABSTRACT

Modulators of the G protein-coupled A2A adenosine receptor (A2AAR) have been considered promising agents to treat Parkinson's disease, inflammation, cancer, and central nervous system disorders. Herein, we demonstrate that a thiophene modification at the C8 position in the common adenine scaffold converted an A2AAR agonist into an antagonist. We synthesized and characterized a novel A2AAR antagonist, 2 (LJ-4517), with Ki = 18.3 nM. X-ray crystallographic structures of 2 in complex with two thermostabilized A2AAR constructs were solved at 2.05 and 2.80 Å resolutions. In contrast to A2AAR agonists, which simultaneously interact with both Ser2777.42 and His2787.43, 2 only transiently contacts His2787.43, which can be direct or water-mediated. The n-hexynyl group of 2 extends into an A2AAR exosite. Structural analysis revealed that the introduced thiophene modification restricted receptor conformational rearrangements required for subsequent activation. This approach can expand the repertoire of adenosine receptor antagonists that can be designed based on available agonist scaffolds.


Subject(s)
Nucleosides , Receptor, Adenosine A2A , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Crystallography, X-Ray , Molecular Conformation , Receptor, Adenosine A2A/chemistry , Thiophenes
10.
Eur J Med Chem ; 241: 114620, 2022 Nov 05.
Article in English | MEDLINE | ID: mdl-35933788

ABSTRACT

The past fifty years have been marked by the surge of neurodegenerative diseases. Unfortunately, current treatments are only symptomatic. Hence, the search for new and innovative therapeutic targets for curative treatments becomes a major challenge. Among these targets, the adenosine A2A receptor (A2AAR) has been the subject of much research in recent years. In this paper, we report the design, synthesis and pharmacological analysis of quinazoline derivatives as A2AAR antagonists with high ligand efficiency. This class of molecules has been discovered by a virtual screening and bears no structural semblance with reference antagonist ZM-241385. More precisely, we identified a series of 2-aminoquinazoline as promising A2AAR antagonists. Among them, one compound showed a high affinity towards A2AAR (21a, Ki = 20 nM). We crystallized this ligand in complex with A2AAR, confirming one of our predicted docking poses and opening up possibilities for further optimization to derive selective ligands for specific adenosine receptor subtypes.


Subject(s)
Adenosine A2 Receptor Antagonists , Purinergic P1 Receptor Antagonists , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Ligands , Molecular Docking Simulation , Purinergic P1 Receptor Antagonists/pharmacology , Quinazolines/pharmacology , Receptor, Adenosine A2A/chemistry , Structure-Activity Relationship
11.
Molecules ; 27(8)2022 Apr 07.
Article in English | MEDLINE | ID: mdl-35458588

ABSTRACT

The A2A adenosine receptor (A2AAR) is one of the four subtypes activated by nucleoside adenosine, and the molecules able to selectively counteract its action are attractive tools for neurodegenerative disorders. In order to find novel A2AAR ligands, two series of compounds based on purine and triazolotriazine scaffolds were synthesized and tested at ARs. Compound 13 was also tested in an in vitro model of neuroinflammation. Some compounds were found to possess high affinity for A2AAR, and it was observed that compound 13 exerted anti-inflammatory properties in microglial cells. Molecular modeling studies results were in good agreement with the binding affinity data and underlined that triazolotriazine and purine scaffolds are interchangeable only when 5- and 2-positions of the triazolotriazine moiety (corresponding to the purine 2- and 8-positions) are substituted.


Subject(s)
Adenosine A2 Receptor Antagonists , Purinergic P1 Receptor Antagonists , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Purinergic P1 Receptor Antagonists/pharmacology , Purines/chemistry , Receptor, Adenosine A2A/metabolism , Structure-Activity Relationship
12.
J Biomol Struct Dyn ; 40(20): 9592-9601, 2022.
Article in English | MEDLINE | ID: mdl-34180379

ABSTRACT

Sickle cell disease (SCD) is a disease resulting from mutation in the globin portion of hemoglobin caused by the replacement of adenine for thymine in the codon of the ß globin gene. In Brazil, SCD affects about 0.3% of the black and Caucasian population. Until now, there is no specific treatment and the available drugs have several serious adverse effects which makes the search for new drugs an emergently need. The use of computational techniques can accelerate the drug development process by prioritization of molecules with affinity against essential targets. Adenosine A2b receptor (rA2b) has been studied in SCD due to its relationship with red blood cells concentration of 2,3-diphosphoglycerate which reduces the hemoglobin affinity for oxygen (O2), facilitating its availability for the tissues. Then, development of rA2b antagonists could be helpful for the treatment of SCD. However, there is still no 3D structure of rA2b and to overcome this limitation, homology modeling should be applied. In this scenario, this study aims to build a suitable 3D model of rA2b by SWISS MODEL and to evaluate the structural aspects of rA2b with known antagonists that may be useful for the identification of new potential antagonists by molecular dynamics on a lipid bilayer environment using GROMACS 5.1.4. The complexes with antagonists ZINC223070016 and ZINC17974526 interacted with key residues by hydrophobic contacts and hydrogen bonds which stabilized them at the rA2b binding site. This intermolecular profile can contribute to the development of more potent rA2b antagonists. Communicated by Ramaswamy H. Sarma.


Subject(s)
Adenosine A2 Receptor Antagonists , Anemia, Sickle Cell , Humans , Adenosine A2 Receptor Antagonists/chemistry , Receptor, Adenosine A2B/chemistry , Anemia, Sickle Cell/drug therapy , Molecular Dynamics Simulation , Hydrogen Bonding
13.
PLoS Comput Biol ; 17(11): e1009152, 2021 11.
Article in English | MEDLINE | ID: mdl-34818333

ABSTRACT

Transmembranal G Protein-Coupled Receptors (GPCRs) transduce extracellular chemical signals to the cell, via conformational change from a resting (inactive) to an active (canonically bound to a G-protein) conformation. Receptor activation is normally modulated by extracellular ligand binding, but mutations in the receptor can also shift this equilibrium by stabilizing different conformational states. In this work, we built structure-energetic relationships of receptor activation based on original thermodynamic cycles that represent the conformational equilibrium of the prototypical A2A adenosine receptor (AR). These cycles were solved with efficient free energy perturbation (FEP) protocols, allowing to distinguish the pharmacological profile of different series of A2AAR agonists with different efficacies. The modulatory effects of point mutations on the basal activity of the receptor or on ligand efficacies could also be detected. This methodology can guide GPCR ligand design with tailored pharmacological properties, or allow the identification of mutations that modulate receptor activation with potential clinical implications.


Subject(s)
Receptor, Adenosine A2A/chemistry , Adenosine A2 Receptor Agonists/chemistry , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Amino Acid Substitution , Computational Biology , Humans , Ligands , Models, Molecular , Molecular Dynamics Simulation , Point Mutation , Protein Conformation/drug effects , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Thermodynamics
14.
ChemistryOpen ; 10(6): 630-638, 2021 06.
Article in English | MEDLINE | ID: mdl-34102706

ABSTRACT

Two novel alkaloids compounds together with fifteen know metabolites were identified from Aspergillus ochraceus. The stereochemistry features of the new molecules were determined via HRESIMS, NMR, ECD, and XRD analyses. Amongst these, compounds two compounds exhibited potential efficacy as anti-Parkinson's disease with the EC50 values of 2.30 and 2.45 µM, respectively. ADMET prediction showed that these compounds owned favorable drug-like characteristics and safe toxicity scores towards CNS drugs. Virtual screening analyses manifested that the compounds exhibited not only robust and reliable interactions to adenosine receptors A2A , but also higher binding selectivity to A2A receptors than to A1 and A3 receptors. Molecular dynamics simulation demonstrated the reliability of molecular docking results and the stability of the complexes obtained with the novel compounds and A2A receptors in natural environments. It is the first time that anti-PD lead compounds have been identified from Aspergillus ochraceus and targeting adenosine A2A receptors.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Antiparkinson Agents/pharmacology , Aspergillus ochraceus/chemistry , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/metabolism , Adenosine A2 Receptor Antagonists/pharmacokinetics , Animals , Antiparkinson Agents/chemistry , Antiparkinson Agents/metabolism , Antiparkinson Agents/pharmacokinetics , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , Humans , Male , Mice , Molecular Docking Simulation , Molecular Dynamics Simulation , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Rats , Stereoisomerism
15.
ACS Chem Biol ; 16(6): 991-1002, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34048655

ABSTRACT

Allosteric ligands provide new opportunities to modulate G protein-coupled receptor (GPCR) function and present therapeutic benefits over orthosteric molecules. Negative allosteric modulators (NAMs) can inhibit the activation of a receptor and downstream signal transduction. Screening NAMs for a GPCR target is particularly challenging because of the difficulty in distinguishing NAMs from antagonists bound to the orthosteric site as they both show inhibitory effects in receptor signaling assays. Here we report an affinity mass spectrometry (MS)-based approach tailored to screening potential NAMs of a GPCR target especially from fragment libraries. Compared to regular surface plasmon resonance or NMR-based methods for fragment screening, our approach features a reduction of the protein and compound consumption by 2-4 orders of magnitude and an increase in the data acquisition speed by 2-3 orders of magnitude. Our affinity MS-based fragment screening led to the identification of a new NAM of the adenosine A2A receptor (A2AAR) bearing an unprecedented azetidine moiety predicted to occupy the allosteric sodium binding site. Molecular dynamics simulations, ligand structure-activity relationship (SAR) studies, and in-solution NMR analyses further revealed the unique binding mode and antagonistic property of this compound that differs considerably from HMA (5-(N,N-hexamethylene)amiloride), a well-characterized NAM of A2AAR. Taken together, our work would facilitate fragment-based screening of allosteric modulators, as well as guide the design of novel NAMs acting at the sodium ion pocket of class A GPCRs.


Subject(s)
Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Allosteric Regulation/drug effects , Receptor, Adenosine A2A/metabolism , Sodium/metabolism , Adenosine A2 Receptor Agonists/chemistry , Adenosine A2 Receptor Antagonists/chemistry , Allosteric Site/drug effects , Binding Sites/drug effects , Drug Discovery , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Receptor, Adenosine A2A/chemistry
16.
Int J Mol Sci ; 22(5)2021 Feb 25.
Article in English | MEDLINE | ID: mdl-33669003

ABSTRACT

The adenosine A2A receptor (A2AR) represents a potential therapeutic target for neurodegenerative diseases. Aiming at the development of a positron emission tomography (PET) radiotracer to monitor changes of receptor density and/or occupancy during the A2AR-tailored therapy, we designed a library of fluorinated analogs based on a recently published lead compound (PPY). Among those, the highly affine 4-fluorobenzyl derivate (PPY1; Ki(hA2AR) = 5.3 nM) and the 2-fluorobenzyl derivate (PPY2; Ki(hA2AR) = 2.1 nM) were chosen for 18F-labeling via an alcohol-enhanced copper-mediated procedure starting from the corresponding boronic acid pinacol ester precursors. Investigations of the metabolic stability of [18F]PPY1 and [18F]PPY2 in CD-1 mice by radio-HPLC analysis revealed parent fractions of more than 76% of total activity in the brain. Specific binding of [18F]PPY2 on mice brain slices was demonstrated by in vitro autoradiography. In vivo PET/magnetic resonance imaging (MRI) studies in CD-1 mice revealed a reasonable high initial brain uptake for both radiotracers, followed by a fast clearance.


Subject(s)
Brain/diagnostic imaging , Fluorine Radioisotopes/chemistry , Hydrocarbons, Fluorinated/chemistry , Positron-Emission Tomography/methods , Radiopharmaceuticals/chemistry , Receptor, Adenosine A2A/metabolism , Adenosine/metabolism , Adenosine A2 Receptor Antagonists/chemistry , Animals , Autoradiography , Brain/metabolism , Chromatography, High Pressure Liquid , Cricetinae , Hydrocarbons, Fluorinated/chemical synthesis , Magnetic Resonance Imaging , Mice , Molecular Docking Simulation , Structure-Activity Relationship
17.
Eur J Med Chem ; 214: 113214, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33548636

ABSTRACT

With the aim to obtain potent adenosine A2A receptor (A2AR) ligands, a series of eighteen derivatives of 4-hydroxy-N-(4-methoxy-7-morpholin-4-yl-1,3-benzo[d]thiazol-2-yl)-4-methylpiperidine-1-carboxamide (SYN-115, Tozadenant) were designed and synthesized. The target compounds were obtained by a chemical building block principle that involved reaction of the appropriate aminobenzothiazole phenyl carbamates with either commercially available or readily synthesized functionalized piperidines. Their affinity and subtype selectivity with regard to human adenosine A1-and A2A receptors were determined using radioligand binding assays. Ki values for human A2AR ranged from 2.4 to 38 nM, with more than 120-fold selectivity over A1 receptors for all evaluated compounds except 13k which had a Ki of 361 nM and 18-fold selectivity. The most potent fluorine-containing derivatives 13e, 13g and 13l exhibited Ki values of 4.9 nM, 3.6 nM and 2.8 nM for the human A2AR. Interestingly, the corresponding values for rat A2AR were found to be four to five times higher. Their binding to A2AR was further confirmed by radiolabeling with 18F and in vitro autoradiography in rat brain slices, which showed almost exclusive striatal binding and complete displacement by the A2AR antagonist ZM 241385. We conclude that these compounds represent potential candidates for the visualization of the A2A receptor and open pathways to novel therapeutic treatments of neurodegenerative disorders or cancer.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Benzothiazoles/pharmacology , Drug Design , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Antagonists/chemical synthesis , Adenosine A2 Receptor Antagonists/chemistry , Animals , Benzothiazoles/chemical synthesis , Benzothiazoles/chemistry , CHO Cells , Cells, Cultured , Cricetulus , Dose-Response Relationship, Drug , Humans , Ligands , Molecular Structure , Structure-Activity Relationship
18.
Eur J Med Chem ; 210: 113040, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33316692

ABSTRACT

Antagonists of adenosine receptor are under exploration as potential drug candidates for treatment of neurological disorders, depression, certain cancers and potentially used as a cancer immunotherapy. Herein, we describe design and synthesis of novel scaffold benzo[4,5]imidazo [1,2-a]pyrazin-1-amine (6) derivatives. All the compounds were evaluated for A2A AR antagonist activity and displayed encouraging results (IC50 9-300 nM) of A2A AR antagonist binding affinity in biochemical assay. Compound 27 exhibits good activity in A2A AR antagonist cAMP functional assay (IC50 31 nM) and further this compound shows T-cell activation at the IL-2 production assay (EC50 165 nM). Molecular docking studies were carried out to rationalize the observed binding affinity of compound 27.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Drug Design , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Antagonists/chemical synthesis , Adenosine A2 Receptor Antagonists/chemistry , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Molecular Structure , Structure-Activity Relationship
19.
J Biochem Mol Toxicol ; 35(3): e22659, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33156955

ABSTRACT

Parkinson's disease (PD) is a chronic neuro-degenerative ailment characterized by impairment in various motor and nonmotor functions of the body. In the past few years, adenosine A2 A receptor (A2 AR) antagonists have attracted much attention due to significant relief in PD. Therefore, in the current study, we intend to disclose the development of novel 1,3,5-triazines as A2 AR antagonist. The radioligand binding and selectivity of analogs were tested in HEK293 (human embryonic kidney) and the cells were transfected with pcDNA 3.1(+) containing full-length human A2 AR cDNA and pcDNA 3.1(+) containing full-length human A1 R cDNA, where they exhibit selective affinity for A2 AR. Molecular docking analysis was also conducted to rationalize the probable mode of action, binding affinity, and orientation of the most potent molecule (7c) at the active site of A2 AR. It has been shown that compound 7c form numerous nonbonded interactions in the active site of A2 AR by interacting with Ala59, Ala63, Ile80, Val84 Glu169, Phe168, Met270, and Ile274. The study revealed 1,3,5-triazines as a novel class of A2 AR antagonists.


Subject(s)
Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/chemical synthesis , Molecular Docking Simulation , Parkinson Disease/drug therapy , Receptor, Adenosine A2A/chemistry , Triazines/chemistry , Triazines/chemical synthesis , Adenosine A2 Receptor Antagonists/therapeutic use , HEK293 Cells , Humans , Parkinson Disease/metabolism , Receptor, Adenosine A2A/metabolism , Triazines/therapeutic use
20.
Eur J Med Chem ; 201: 112478, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32659606

ABSTRACT

This paper describes identification of the first-in-class multi-target adenosine A2A receptor antagonists-carbonic anhydrase (CA) IX and XII inhibitors, as new potential antitumor agents. To obtain the multi-acting ligands, the 8-amino-2,6-diphenyltriazolo[4,3-a]pyrazin-3-one, a potent adenosine hA2A receptor (AR) antagonist, was taken as lead compound. To address activity against the tumor-associated CA isoforms, it was modified by introduction of different substituents (OH, COOH, CONHOH, SO2NH2) on the 6-phenyl ring or on a phenyl pendant connected to the former through different spacers. Among the new triazolopyrazines 1-23, the most active were those featuring the sulfonamide residue. Derivative 20, featuring a 4-sulfonamidophenyl residue attached through a CONH(CH2)2CONH spacer at the para-position of the 6-phenyl ring, showed the best combination of activity at the three targets. In fact, it inhibited both the tumor-associated hCA IX and XII isozymes at nanomolar concentration (Ki = 5.0 and 27.0 nM), and also displayed a quite good affinity for the hA2A AR (Ki = 108 nM). Compound 14, bearing the 4-sulfonamidophenyl residue linked at the para-position of the 6-phenyl ring by a CONH spacer, was remarkable because both its hA2A AR affinity and hCA XII inhibitory potency were in the low nanomolar range (Ki = 6.4 and 6.2 nM, respectively). Molecular docking studies highlighted the interaction mode of selected triazolopyrazines in the hA2A AR recognition pocket and in the active site of hCA II, IX and XII isoforms.


Subject(s)
Adenosine A2 Receptor Antagonists/chemistry , Antineoplastic Agents/chemistry , Carbonic Anhydrase Inhibitors/chemistry , Pyrazines/chemistry , Triazoles/chemistry , Adenosine A2 Receptor Antagonists/chemical synthesis , Adenosine A2 Receptor Antagonists/metabolism , Animals , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , CHO Cells , Carbonic Anhydrase IX/chemistry , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/metabolism , Carbonic Anhydrases/chemistry , Carbonic Anhydrases/metabolism , Catalytic Domain , Cricetulus , Enzyme Assays , Humans , Molecular Docking Simulation , Molecular Structure , Protein Binding , Pyrazines/chemical synthesis , Pyrazines/metabolism , Receptor, Adenosine A2A/metabolism , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...