Elsevier

Bioorganic Chemistry

Volume 87, June 2019, Pages 380-394
Bioorganic Chemistry

Novel 8-amino-1,2,4-triazolo[4,3-a]pyrazin-3-one derivatives as potent human adenosine A1 and A2A receptor antagonists. Evaluation of their protective effect against β-amyloid-induced neurotoxicity in SH-SY5Y cells

https://doi.org/10.1016/j.bioorg.2019.03.046Get rights and content

Highlights

  • Several derivatives possess nanomolar hA1 and hA2A receptor affinities.

  • Two compounds showed high affinity and a complete selectivity for the hA2A AR.

  • Two compounds were able to prevent β-amyloid peptide (25–35)-induced neurotoxicity.

  • Docking studies were employed to rationalize the affinity data.

Abstract

In this work, an enlarged series of 1,2,4-triazolo[4,3-a]pyrazin-3-ones was designed to target the human (h) A2A adenosine receptor (AR) or both hA1 and hA2A ARs. The novel 8-amino-1,2,4-triazolopyrazin-3-one derivatives 1–25 featured a phenyl or a benzyl pendant at position 2 while different aryl/heteroaryl substituents were placed at position 6. Two compounds (8 and 10) endowed with high affinity (Ki = 7.2 and 10.6 nM) and a complete selectivity for the hA2A AR were identified. Moreover, several derivatives possessed nanomolar affinity for both hA1 and hA2A ARs (both Ki < 20 nM) and different degrees of selectivity versus the hA3 AR. Two selected compounds (10 and 25) demonstrated ability in preventing β-amyloid peptide (25–35)-induced neurotoxicity in SH-SY5Y cells. Results of docking studies at the hA2A and hA1 AR crystal structures helped us to rationalize the observed affinity data and to highlight that the steric hindrance of the substituents at the 2- and 6-position of the bicyclic core affects the binding mode in the receptor cavity.

Introduction

Adenosine is a ubiquitous neuromodulator which controls many physiological and pathological processes, both in the central and peripheral nervous system. Adenosine exerts its effects through activation of G protein-coupled receptors, subdivided into the four subtypes A1, A2A, A2B and A3 [1], [2]. A1 and A3 adenosine receptors (ARs) are negatively coupled to adenylate cyclase while A2A and A2B subtypes activate the enzyme. The hA1 AR subtype is the most abundant in the brain and is traditionally considered a neuroprotective receptor due to its inhibitory effects [3]. For instance, it curtails excitatory neurotransmission thus exerting a protective role in diverse pathological conditions linked to glutamate excitotoxicity such as cerebral ischemia or epilepsy. Nevertheless, there is recent evidence that A1 AR-sustained activation after stroke or ischemia induces AMPA receptor endocytosis and the consequent, persistent synaptic depression may contribute to enhanced neuronal death [4], [5], [6].

The hA2A AR subtype has less widespread localization at central level where it shows higher density in the basal ganglia and lower in the cortex and hippocampus. Blockade of this AR subtype exerts a protective effect in different models of cerebral ischemia and neurodegenerative disorders, such as Parkinson’s or Alzheimer’s diseases [4], [7], [8], [9], [10], [11], [12], [13], [14], [15], [16]. In the last decade, several human studies have highlighted that consumption of caffeine, a non-selective A1 and A2A AR antagonist, negatively correlated with the risk of developing AD and PD [9], [11], [12]. The protective effect of caffeine, investigated in animal models of AD and PD was ascribed to antagonism of the A2A AR subtype, among other pathways [10], [11], [12], [13], [14]. Related to AD models, both caffeine and the potent A2A AR antagonist ZM 241,385 (4-(2-[7-amino-2-(2-furyl)-1,2,4-triazolo[2,3-a]triazin-5-ylamino]ethyl]phenol) prevented cell death after exposure of rat cultured cerebellar granule neurons to the β-amyloid peptide (25–35) [13]. A2A AR antagonists demonstrated an ability to alleviate cognitive deficits caused by administration of the β-amyloid peptides in different in vivo rodent models [14], [15]. However, more recently, also A1 AR antagonism was recognized as affording neuroprotection in a model of combined neurotoxicity. In fact, the protective effect of dual A1 and A2A AR blockade in preventing β-amyloid toxicity in neuroblastoma cells exposed to aluminium chloride was demonstrated [16].

In recent years, as part of a research program aimed at finding new adenosine receptor antagonists [17], [18], [19], [20], [21], [22], [23], [24], [25], we disclosed the 8-amino-1,2,4-triazolo[4,3-a]pyrazin-3-one as a new decorable scaffold to obtain potent antagonists able to bind selectively the hA2A AR or both the hA1 and hA2A subtypes [21] (Fig. 1). The SAR study showed that the unsubstituted phenyl ring at position 2 was the best group for obtaining an efficient hA2A receptor interaction, and that the presence of small para alkoxy groups (OMe, OEt, O-isopropyl, O-propargyl) on the 6-phenyl pendant afforded nanomolar affinity and a complete selectivity for this AR subtype.

To continue investigations of the 8-amino-1,2,4-triazolopyrazin-3-one series, we designed and synthesized an enlarged series of derivatives to obtain antagonists for the hA2A AR or both hA1 and hA2A ARs. These types of ligands were of our interest for their ability to induce neuroprotection in different neurodegenerative diseases. Thus, a new set of triazolopyrazines, featuring the unsubstituted phenyl ring at position 2 and heteroaryl or aryl groups at position 6 (Fig. 1 compounds 1–22), were synthesized. Simple substituents, endowed with different electronic, steric and lipophilic properties, were probed on the 6-phenyl ring (X = OMe, NO2, NH2, Br, Cl). The NH2 group was also inserted to construct the piperazine moiety appended on the 6-phenyl moiety (compounds 17–22). Finally, derivatives featuring a benzyl chain at position 2 and a phenyl ring or a 2-furyl/2-(5-methylfuryl) group at position 6 were synthesized (derivatives 23–25) since the benzyl pendant and the furyl moiety were thought to enhance compound solubility.

Section snippets

Chemistry

The new 8-amino-1,2,4-triazolo[4,3-a]pyrazin-3-one derivatives 1–25 were prepared as described in Scheme 1, Scheme 2, Scheme 3. Scheme 1 depicts the synthesis of the 2-phenyl derivatives 1–16 which were obtained starting from ethyl 1-phenyl-5-oxo-1H-1,2,4-triazole-3-carboxylate 26 [21].

N4-Alkylation of 26 with the suitable α-aloketones in DMF/CH3CN, and in the presence of potassium carbonate, afforded the ethyl −5-oxo-4-phenyl-1,2,4-triazole-3-carboxylate derivatives 27–38 whose cyclization

Conclusion

In this work, an enlarged set of 1,2,4-triazolo[4,3-a]pyrazine-3-one derivatives was synthesized to deepen SAR studies and to obtain hA2A AR selective antagonists or dual-targeting hA1 and hA2A AR antagonists which were of our interest for their potential neuroprotective effect. The aim of the work can be considered satisfied. In fact, compounds 8 and 10, featuring a 2-phenyl ring and, respectively, a 4-nitro and 4-bromo substituent on the 6-phenyl moiety, showed high hA2A AR affinity (Ki = 7.2

Chemistry

The microwave-assisted syntheses were performed using an Initiator EXP Microwave Biotage instrument (frequency of irradiation: 2.45 GHz). Silica gel 60 (Merck, 70–230 mesh) was used for analytical TLC, and for column chromatography, respectively. All melting points were determined on a Gallenkamp melting point apparatus and are uncorrected. Elemental analyses were performed with a Flash E1112 Thermofinnigan elemental analyzer for C, H, N and the results were within 0.4% of the theoretical

Acknowledgements

The work was financially supported by the Italian Ministry for University and Research (MIUR, PRIN 2010-2011, 20103W4779_004 project) and the University of Florence (Research Fund 2017).

References (42)

  • L. Di Cesare Mannelli et al.

    Oxaliplatin-induced oxidative stress in nervous system-derived cellular models: could it correlate with in vivo neuropathy?

    Free Radic. Biol. Med.

    (2013)
  • J. Faria et al.

    Comparative study of the neurotoxicological effects of tramadol and tapentadol in SH-SY5Y cells

    Toxicology

    (2016)
  • J.J.V. Branca et al.

    Selenium and zinc: Two key players against cadmium-induced neuronal toxicity

    Toxicol. In Vitro

    (2018)
  • G. Jones et al.

    Development and validation of a genetic algorithm for flexible docking

    J. Mol. Biol.

    (1997)
  • G.A. Bennett et al.

    Studies on antifungal agents 30. Novel substituted 3-(2-furanyl)-3- (1H-imidazol-1-ylmethyl)-2-methyl-5-phenyl(or phenyloxymethy)-isoxazolidines

    Eur. J. Med. Chem.

    (1989)
  • B.B. Fredholm et al.

    International union of Pharmacology XXV. Nomenclature and classification of adenosine receptors

    Pharmacol. Rev.

    (2001)
  • B.B. Fredholm et al.

    International union of Pharmacology LXXXI. Nomenclature and classification of adenosine receptors. An up date

    Pharmacol. Rev.

    (2011)
  • R.A. Cunha

    Neuroprotection by adenosine in the brain: from A1 receptor activation to A2A receptor blockade

    Purinergic Signal.

    (2005)
  • J. Stockwell et al.

    Adenosine A1 and A2A receptors in the brain: current research and their role in neurodegeneration

    Molecules

    (2017)
  • Z. Chen et al.

    Adenosine A1 receptor-mediated endocytosis of AMPA receptors contributes to impairments in long-term potentiation (LTP) in the middle-aged rat hippocampus

    Neurochem. Res.

    (2016)
  • P.A. Borea et al.

    Pharmacology of adenosine receptors: the state of art

    Physiol. Rev.

    (2018)
  • Cited by (14)

    • Discovery of first-in-class multi-target adenosine A<inf>2A</inf> receptor antagonists-carbonic anhydrase IX and XII inhibitors. 8-Amino-6-aryl-2-phenyl-1,2,4-triazolo [4,3-a]pyrazin-3-one derivatives as new potential antitumor agents

      2020, European Journal of Medicinal Chemistry
      Citation Excerpt :

      hA2A AR affinity of 4 and 4a were quite scarce, particularly of the latter. The lower affinity of 4a, with respect to 4, was probably due to the loss of hydrogen-bonding interaction that the 8-amino group typically engages with Asn254 residue [18,22,24] (see also molecular modeling studies section). The triazolopyrazines 5 and 6, featuring, respectively, an ethyl glycine and a succinamide moiety at the para-position of the 6-phenyl ring, showed only some hA2A AR affinity while they were not able to inhibit the hCAs.

    • New 8-amino-1,2,4-triazolo[4,3-a]pyrazin-3-one derivatives. Evaluation of different moieties on the 6-aryl ring to obtain potent and selective human A<inf>2A</inf> adenosine receptor antagonists

      2020, Bioorganic and Medicinal Chemistry Letters
      Citation Excerpt :

      This binding mode was observed also for analogues of these compounds at the same receptor.15,19,22 The substituted 6-aryl group modulates the interaction with the receptor, depending on the physicochemical profile of its functional groups, as evidenced for previously reported compounds of this series.15,19,22 In particular, the interaction is made with residues located at the entrance of the binding cavity, such as Ser61.32, Ser672.65, Gly692.67, Leu167 (EL2), Leu267 (EL3) and Tyr2717.36 (Fig. 3).

    View all citing articles on Scopus
    View full text