Adenosine A2A receptor

Last updated
ADORA2A
A2A receptor bilayer.png
Available structures
PDB Ortholog search: PDBe RCSB
Identifiers
Aliases ADORA2A , adenosine A2a receptor, A2aR, ADORA2, RDC8
External IDs OMIM: 102776 MGI: 99402 HomoloGene: 20166 GeneCards: ADORA2A
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_000675
NM_001278497
NM_001278498
NM_001278499
NM_001278500

NM_009630
NM_001331095
NM_001331096

RefSeq (protein)

NP_000666
NP_001265426
NP_001265427
NP_001265428
NP_001265429

NP_001318024
NP_001318025
NP_033760

Location (UCSC) Chr 22: 24.42 – 24.44 Mb Chr 10: 75.15 – 75.17 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

The adenosine A2A receptor, also known as ADORA2A, is an adenosine receptor, and also denotes the human gene encoding it. [5] [6]

Structure

This protein is a member of the G protein-coupled receptor (GPCR) family which possess seven transmembrane alpha helices, as well as an extracellular N-terminus and an intracellular C-terminus. Furthermore, located in the intracellular side close to the membrane is a small alpha helix, often referred to as helix 8 (H8). The crystallographic structure of the adenosine A2A receptor reveals a ligand binding pocket distinct from that of other structurally determined GPCRs (i.e., the beta-2 adrenergic receptor and rhodopsin). [7] Below this primary (orthosteric) binding pocket lies a secondary (allosteric) binding pocket. The crystal-structure of A2A bound to the antagonist ZM241385 (PDB code: 4EIY) showed that a sodium-ion can be found in this location of the protein, thus giving it the name 'sodium-ion binding pocket'. [8]

Heteromers

The actions of the A2A receptor are complicated by the fact that a variety of functional heteromers composed of a mixture of A2A subunits with subunits from other unrelated G-protein coupled receptors have been found in the brain, adding a further degree of complexity to the role of adenosine in modulation of neuronal activity. Heteromers consisting of adenosine A1/A2A, [9] [10] dopamine D2/A2A [11] and D3/A2A, [12] glutamate mGluR5/A2A [13] and cannabinoid CB1/A2A [14] have all been observed, as well as CB1/A2A/D2 heterotrimers, [15] and the functional significance and endogenous role of these hybrid receptors is still only starting to be unravelled. [16] [17] [18]

The receptor's role in immunomodulation in the context of cancer has suggested that it is an important immune checkpoint molecule. [19]

Function

The gene encodes a protein which is one of several receptor subtypes for adenosine. The activity of the encoded protein, a G protein-coupled receptor family member, is mediated by G proteins which activate adenylyl cyclase, which induce synthesis of intracellular cAMP. The A2A receptor binds with the Gs protein at the intracellular site of the receptor. The Gs protein consists of three subunits; Gsα, Gsβ and Gsγ. A crystal structure of the A2A receptor bound with the agonist NECA and a G protein-mimic has been published in 2016 (PDB code: 5g53). [20]

The encoded protein (the A2A receptor) is abundant in basal ganglia, vasculature, T lymphocytes, and platelets and it is a major target of caffeine, which is a competitive antagonist of this protein. [21]

Physiological role

A1 and A2A receptors are believed to regulate myocardial oxygen demand and to increase coronary circulation by vasodilation. In addition, A2A receptor can suppress immune cells, thereby protecting tissue from inflammation. [22]

The A2A receptor is also expressed in the brain, where it has important roles in the regulation of glutamate and dopamine release, making it a potential therapeutic target for the treatment of conditions such as insomnia, pain, depression, and Parkinson's disease. [23] [24] [25] [26] [27] [28] [29]

Ligands

A number of selective A2A ligands have been developed, [30] with several possible therapeutic applications. [31]

Older research on adenosine receptor function, and non-selective adenosine receptor antagonists such as aminophylline, focused mainly on the role of adenosine receptors in the heart, and led to several randomized controlled trials using these receptor antagonists to treat bradyasystolic arrest. [32] [33] [34] [35] [36] [37] [38]

However the development of more highly selective A2A ligands has led towards other applications, with the most significant focus of research currently being the potential therapeutic role for A2A antagonists in the treatment of Parkinson's disease. [39] [40] [41] [42]

Agonists

Antagonists

Interactions

Adenosine A2A receptor has been shown to interact with Dopamine receptor D2. [54] As a result, Adenosine receptor A2A decreases activity in the Dopamine D2 receptors.

In cancer immunotherapy

The adenosine A2A receptor has also been shown to play a regulatory role in the adaptive immune system. In this role, it functions similarly to programmed cell death-1 (PD-1) and cytotoxic t-lymphocyte associated protein-4 (CTLA-4) receptors, namely to suppress immunologic response and prevent associated tissue damage. Extracellular adenosine gathers in response to cellular stress and breakdown through interactions with hypoxia induced HIF-1α. [55] Abundant extracellular adenosine can then bind to the A2A receptor resulting in a Gs-protein coupled response, resulting in the accumulation of intracellular cAMP, which functions primarily through protein kinase A to upregulate inhibitory cytokines such as transforming growth factor-beta (TGF-β) and inhibitory receptors (i.e., PD-1). [56] Interactions with FOXP3 stimulates CD4+ T-cells into regulatory Treg cells further inhibiting immune response. [57]

Blockade of A2AR has been attempted to various ends, namely cancer immunotherapy. While several A2A receptor antagonists have progressed to clinical trials for the treatment of Parkinson's disease, A2AR blockade in the context of cancer is less characterized. Mice treated with A2AR antagonists, such as ZM241385 (listed above) or caffeine, show significantly delayed tumor growth due to T-cells resistant to inhibition. [55] This is further highlighted by A2AR knockout mice who show increased tumor rejection. Multiple checkpoint pathway inhibition has been shown to have an additive effect, as shown by an increase in response with blockade to PD-1 and CTLA-4 via monoclonal antibodies as compared to the blockade of a single pathway. The A2AR antogonist CPI-444 has shown this in combination with anti-PD-L1 or anti-CTLA-4 treatment as it eliminated tumors in up to 90% of treated mice, including restoration of immune responses in models that incompletely responded to anti-PD-L1 or anti-CTLA-4 monotherapy. Further, tumor growth was fully inhibited when mice with cleared tumors were later rechallenged, indicating that CPI-444 induced systemic antitumor immune memory. [58] Researchers believe that A2AR blockade could increase the efficacy of such treatments even further. [56] Finally, inhibition of A2AR, either through pharmacologic or genetic targeting, in chimeric antigen receptor (CAR) T-cells reveals promising results. Blockade of A2AR in this setting has shown to increase tumor clearance through CAR T-cell therapy in mice. [59] Targeting of the A2A receptor is an attractive option for the treatment of a variety of cancers, especially with the therapeutic success of the blockade of other checkpoint pathways such as PD-1 and CTLA-4.

Related Research Articles

<span class="mw-page-title-main">Adenosine</span> Chemical compound

Adenosine (symbol A) is an organic compound that occurs widely in nature in the form of diverse derivatives. The molecule consists of an adenine attached to a ribose via a β-N9-glycosidic bond. Adenosine is one of the four nucleoside building blocks of RNA (and its derivative deoxyadenosine is a building block of DNA), which are essential for all life on earth. Its derivatives include the energy carriers adenosine mono-, di-, and triphosphate, also known as AMP/ADP/ATP. Cyclic adenosine monophosphate (cAMP) is pervasive in signal transduction. Adenosine is used as an intravenous medication for some cardiac arrhythmias.

<span class="mw-page-title-main">Dopamine receptor</span> Class of G protein-coupled receptors

Dopamine receptors are a class of G protein-coupled receptors that are prominent in the vertebrate central nervous system (CNS). Dopamine receptors activate different effectors through not only G-protein coupling, but also signaling through different protein interactions. The neurotransmitter dopamine is the primary endogenous ligand for dopamine receptors.

<span class="mw-page-title-main">Adenosine receptor</span> Class of four receptor proteins to the molecule adenosine

The adenosine receptors (or P1 receptors) are a class of purinergic G protein-coupled receptors with adenosine as the endogenous ligand. There are four known types of adenosine receptors in humans: A1, A2A, A2B and A3; each is encoded by a different gene.

Adenosine A<sub>1</sub> receptor Cell surface receptor found in humans

The adenosine A1 receptor (A1AR) is one member of the adenosine receptor group of G protein-coupled receptors with adenosine as endogenous ligand.

<span class="mw-page-title-main">Nociceptin receptor</span> Protein-coding gene in the species Homo sapiens

The nociceptin opioid peptide receptor (NOP), also known as the nociceptin/orphanin FQ (N/OFQ) receptor or kappa-type 3 opioid receptor, is a protein that in humans is encoded by the OPRL1 gene. The nociceptin receptor is a member of the opioid subfamily of G protein-coupled receptors whose natural ligand is the 17 amino acid neuropeptide known as nociceptin (N/OFQ). This receptor is involved in the regulation of numerous brain activities, particularly instinctive and emotional behaviors. Antagonists targeting NOP are under investigation for their role as treatments for depression and Parkinson's disease, whereas NOP agonists have been shown to act as powerful, non-addictive painkillers in non-human primates.

Prostaglandin DP<sub>1</sub> receptor Protein-coding gene in the species Homo sapiens

The prostaglandin D2 receptor 1 (DP1), a G protein-coupled receptor encoded by the PTGDR1 gene (also termed PTGDR), is primarily a receptor for prostaglandin D2 (PGD2). The receptor is a member of the prostaglandin receptors belonging to the subfamily A14 of rhodopsin-like receptors. Activation of DP1 by PGD2 or other cognate receptor ligands is associated with a variety of physiological and pathological responses in animal models.

A heteromer is something that consists of different parts; the antonym of homomeric. Examples are:

Dopamine receptor D<sub>2</sub> Main receptor for most antipsychotic drugs

Dopamine receptor D2, also known as D2R, is a protein that, in humans, is encoded by the DRD2 gene. After work from Paul Greengard's lab had suggested that dopamine receptors were the site of action of antipsychotic drugs, several groups, including those of Solomon Snyder and Philip Seeman used a radiolabeled antipsychotic drug to identify what is now known as the dopamine D2 receptor. The dopamine D2 receptor is the main receptor for most antipsychotic drugs. The structure of DRD2 in complex with the atypical antipsychotic risperidone has been determined.

Dopamine receptor D<sub>1</sub> Protein-coding gene in humans

Dopamine receptor D1, also known as DRD1. It is one of the two types of D1-like receptor family — receptors D1 and D5. It is a protein that in humans is encoded by the DRD1 gene.

Adenosine A<sub>3</sub> receptor Cell surface receptor found in humans

The adenosine A3 receptor, also known as ADORA3, is an adenosine receptor, but also denotes the human gene encoding it.

Adenosine A<sub>2B</sub> receptor Cell surface receptor found in humans

The adenosine A2B receptor, also known as ADORA2B, is a G-protein coupled adenosine receptor, and also denotes the human adenosine A2b receptor gene which encodes it.

Dopamine receptor D<sub>5</sub> Protein-coding gene in humans

Dopamine receptor D5, also known as D1BR, is a protein that in humans is encoded by the DRD5 gene. It belongs to the D1-like receptor family along with the D1 receptor subtype.

<span class="mw-page-title-main">Dipropylcyclopentylxanthine</span> Chemical compound

8-Cyclopentyl-1,3-dipropylxanthine (DPCPX, PD-116,948) is a drug which acts as a potent and selective antagonist for the adenosine A1 receptor. It has high selectivity for A1 over other adenosine receptor subtypes, but as with other xanthine derivatives DPCPX also acts as a phosphodiesterase inhibitor, and is almost as potent as rolipram at inhibiting PDE4. It has been used to study the function of the adenosine A1 receptor in animals, which has been found to be involved in several important functions such as regulation of breathing and activity in various regions of the brain, and DPCPX has also been shown to produce behavioural effects such as increasing the hallucinogen-appropriate responding produced by the 5-HT2A agonist DOI, and the dopamine release induced by MDMA, as well as having interactions with a range of anticonvulsant drugs.

<span class="mw-page-title-main">SCH-58261</span> Chemical compound

SCH-58261 is a drug which acts as a potent and selective antagonist for the adenosine receptor A2A, with more than 50x selectivity for A2A over other adenosine receptors. It has been used to investigate the mechanism of action of caffeine, which is a mixed A1 / A2A antagonist, and has shown that the A2A receptor is primarily responsible for the stimulant and ergogenic effects of caffeine, but blockade of both A1 and A2A receptors is required to accurately replicate caffeine's effects in animals. SCH-58261 has also shown antidepressant, nootropic and neuroprotective effects in a variety of animal models, and has been investigated as a possible treatment for Parkinson's disease.

<span class="mw-page-title-main">Metabotropic glutamate receptor 2</span> Mammalian protein found in humans

Metabotropic glutamate receptor 2 (mGluR2) is a protein that, in humans, is encoded by the GRM2 gene. mGluR2 is a G protein-coupled receptor (GPCR) that couples with the Gi alpha subunit. The receptor functions as an autoreceptor for glutamate, that upon activation, inhibits the emptying of vesicular contents at the presynaptic terminal of glutamatergic neurons.

<span class="mw-page-title-main">GPCR oligomer</span> Class of protein complexes

A GPCR oligomer is a protein complex that consists of a small number of G protein-coupled receptors (GPCRs). It is held together by covalent bonds or by intermolecular forces. The subunits within this complex are called protomers, while unconnected receptors are called monomers. Receptor homomers consist of identical protomers, while heteromers consist of different protomers.

The D1–D2 dopamine receptor heteromer is a receptor heteromer consisting of D1 and D2 protomers.

<span class="mw-page-title-main">Theacrine</span> Chemical compound

Theacrine, also known as 1,3,7,9-tetramethyluric acid, is a purine alkaloid found in Cupuaçu and in a Chinese tea known as kucha. It shows anti-inflammatory and analgesic effects and appears to affect adenosine signalling in a manner similar to caffeine. In kucha leaves, theacrine is synthesized from caffeine in what is thought to be a three-step pathway. Theacrine and caffeine are structurally similar.

<span class="mw-page-title-main">Purinergic signalling</span> Signalling complex involving purine nucleosides and their receptors

Purinergic signalling is a form of extracellular signalling mediated by purine nucleotides and nucleosides such as adenosine and ATP. It involves the activation of purinergic receptors in the cell and/or in nearby cells, thereby regulating cellular functions.

Caffeine-induced anxiety disorder is a subclass of the DSM-5 diagnosis of substance/medication-induced anxiety disorder.

References

  1. 1 2 3 GRCh38: Ensembl release 89: ENSG00000128271 - Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000020178 - Ensembl, May 2017
  3. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. Libert F, Parmentier M, Lefort A, Dinsart C, Van Sande J, Maenhaut C, et al. (May 1989). "Selective amplification and cloning of four new members of the G protein-coupled receptor family". Science. 244 (4904): 569–72. Bibcode:1989Sci...244..569L. doi:10.1126/science.2541503. PMID   2541503.
  6. Libert F, Passage E, Parmentier M, Simons MJ, Vassart G, Mattei MG (September 1991). "Chromosomal mapping of A1 and A2 adenosine receptors, VIP receptor, and a new subtype of serotonin receptor". Genomics. 11 (1): 225–7. doi:10.1016/0888-7543(91)90125-X. PMID   1662665.
  7. PDB: 3EML ; Jaakola VP, Griffith MT, Hanson MA, Cherezov V, Chien EY, Lane JR, et al. (November 2008). "The 2.6 angstrom crystal structure of a human A2A adenosine receptor bound to an antagonist". Science. 322 (5905): 1211–7. Bibcode:2008Sci...322.1211J. doi:10.1126/science.1164772. PMC   2586971 . PMID   18832607.
  8. Liu W, Chun E, Thompson AA, Chubukov P, Xu F, Katritch V, et al. (July 2012). "Structural basis for allosteric regulation of GPCRs by sodium ions". Science. 337 (6091): 232–6. Bibcode:2012Sci...337..232L. doi:10.1126/science.1219218. PMC   3399762 . PMID   22798613.
  9. Ciruela F, Casadó V, Rodrigues RJ, Luján R, Burgueño J, Canals M, et al. (February 2006). "Presynaptic control of striatal glutamatergic neurotransmission by adenosine A1-A2A receptor heteromers". The Journal of Neuroscience. 26 (7): 2080–7. doi:10.1523/JNEUROSCI.3574-05.2006. PMC   6674939 . PMID   16481441.
  10. Ferre S, Ciruela F, Borycz J, Solinas M, Quarta D, Antoniou K, et al. (January 2008). "Adenosine A1-A2A receptor heteromers: new targets for caffeine in the brain". Frontiers in Bioscience. 13 (13): 2391–9. doi: 10.2741/2852 . PMID   17981720.
  11. Fuxe K, Ferré S, Canals M, Torvinen M, Terasmaa A, Marcellino D, et al. (2005). "Adenosine A2A and dopamine D2 heteromeric receptor complexes and their function". Journal of Molecular Neuroscience. 26 (2–3): 209–20. doi:10.1385/JMN:26:2-3:209. PMID   16012194. S2CID   427930.
  12. Torvinen M, Marcellino D, Canals M, Agnati LF, Lluis C, Franco R, Fuxe K (February 2005). "Adenosine A2A receptor and dopamine D3 receptor interactions: evidence of functional A2A/D3 heteromeric complexes". Molecular Pharmacology. 67 (2): 400–7. doi:10.1124/mol.104.003376. PMID   15539641. S2CID   24475855.
  13. Zezula J, Freissmuth M (March 2008). "The A(2A)-adenosine receptor: a GPCR with unique features?". British Journal of Pharmacology. 153 Suppl 1 (S1): S184-90. doi:10.1038/sj.bjp.0707674. PMC   2268059 . PMID   18246094.
  14. Ferré S, Goldberg SR, Lluis C, Franco R (2009). "Looking for the role of cannabinoid receptor heteromers in striatal function". Neuropharmacology. 56 (Suppl 1): 226–34. doi:10.1016/j.neuropharm.2008.06.076. PMC   2635338 . PMID   18691604.
  15. Marcellino D, Carriba P, Filip M, Borgkvist A, Frankowska M, Bellido I, et al. (April 2008). "Antagonistic cannabinoid CB1/dopamine D2 receptor interactions in striatal CB1/D2 heteromers. A combined neurochemical and behavioral analysis". Neuropharmacology. 54 (5): 815–23. doi:10.1016/j.neuropharm.2007.12.011. PMID   18262573. S2CID   195685369.
  16. Ferré S, Ciruela F, Quiroz C, Luján R, Popoli P, Cunha RA, et al. (November 2007). "Adenosine receptor heteromers and their integrative role in striatal function". TheScientificWorldJournal. 7: 74–85. doi: 10.1100/tsw.2007.211 . PMC   5901194 . PMID   17982579.
  17. Wardas J (May 2008). "Potential role of adenosine A2A receptors in the treatment of schizophrenia". Frontiers in Bioscience. 13 (13): 4071–96. doi: 10.2741/2995 . PMID   18508501.
  18. Simola N, Morelli M, Pinna A (2008). "Adenosine A2A receptor antagonists and Parkinson's disease: state of the art and future directions". Current Pharmaceutical Design. 14 (15): 1475–89. doi:10.2174/138161208784480072. PMID   18537671.
  19. Cekic C, Linden J (December 2014). "Adenosine A2A receptors intrinsically regulate CD8+ T cells in the tumor microenvironment". Cancer Research. 74 (24): 7239–49. doi:10.1158/0008-5472.CAN-13-3581. PMC   4459794 . PMID   25341542.
  20. Carpenter B, Nehmé R, Warne T, Leslie AG, Tate CG (August 2016). "Structure of the adenosine A(2A) receptor bound to an engineered G protein". Nature. 536 (7614): 104–7. Bibcode:2016Natur.536..104C. doi:10.1038/nature18966. PMC   4979997 . PMID   27462812.
  21. "Entrez Gene: ADORA2A adenosine A2A receptor".
  22. Ohta A, Sitkovsky M (2001). "Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage". Nature. 414 (6866): 916–20. Bibcode:2001Natur.414..916O. doi:10.1038/414916a. PMID   11780065. S2CID   4386419.
  23. Hack SP, Christie MJ (2003). "Adaptations in adenosine signaling in drug dependence: therapeutic implications". Critical Reviews in Neurobiology. 15 (3–4): 235–74. doi:10.1615/CritRevNeurobiol.v15.i34.30. PMID   15248812.
  24. Morelli M, Di Paolo T, Wardas J, Calon F, Xiao D, Schwarzschild MA (December 2007). "Role of adenosine A2A receptors in parkinsonian motor impairment and l-DOPA-induced motor complications". Progress in Neurobiology. 83 (5): 293–309. doi:10.1016/j.pneurobio.2007.07.001. PMID   17826884. S2CID   27478825.
  25. Schiffmann SN, Fisone G, Moresco R, Cunha RA, Ferré S (December 2007). "Adenosine A2A receptors and basal ganglia physiology". Progress in Neurobiology. 83 (5): 277–92. doi:10.1016/j.pneurobio.2007.05.001. PMC   2148496 . PMID   17646043.
  26. Ferré S, Diamond I, Goldberg SR, Yao L, Hourani SM, Huang ZL, et al. (December 2007). "Adenosine A2A receptors in ventral striatum, hypothalamus and nociceptive circuitry implications for drug addiction, sleep and pain". Progress in Neurobiology. 83 (5): 332–47. doi:10.1016/j.pneurobio.2007.04.002. PMC   2141681 . PMID   17532111.
  27. Brown RM, Short JL (November 2008). "Adenosine A(2A) receptors and their role in drug addiction". The Journal of Pharmacy and Pharmacology. 60 (11): 1409–30. doi:10.1211/jpp/60.11.0001 (inactive 2024-01-24). PMID   18957161.{{cite journal}}: CS1 maint: DOI inactive as of January 2024 (link)
  28. Cunha RA, Ferré S, Vaugeois JM, Chen JF (2008). "Potential therapeutic interest of adenosine A2A receptors in psychiatric disorders". Current Pharmaceutical Design. 14 (15): 1512–24. doi:10.2174/138161208784480090. PMC   2423946 . PMID   18537674.
  29. Mingote S, Font L, Farrar AM, Vontell R, Worden LT, Stopper CM, et al. (September 2008). "Nucleus accumbens adenosine A2A receptors regulate exertion of effort by acting on the ventral striatopallidal pathway". The Journal of Neuroscience. 28 (36): 9037–46. doi:10.1523/JNEUROSCI.1525-08.2008. PMC   2806668 . PMID   18768698.
    • Ongini E, Monopoli A, Cacciari B, Baraldi PG (2001). "Selective adenosine A2A receptor antagonists". Farmaco. 56 (1–2): 87–90. doi:10.1016/S0014-827X(01)01024-2. PMID   11347973.
    • Baraldi PG, Cacciari B, Romagnoli R, Spalluto G, Monopoli A, Ongini E, et al. (January 2002). "7-Substituted 5-amino-2-(2-furyl)pyrazolo[4,3-e]-1,2,4-triazolo[1,5-c]pyrimidines as A2A adenosine receptor antagonists: a study on the importance of modifications at the side chain on the activity and solubility". Journal of Medicinal Chemistry. 45 (1): 115–26. doi:10.1021/jm010924c. PMID   11754583.
    • Baraldi PG, Fruttarolo F, Tabrizi MA, Preti D, Romagnoli R, El-Kashef H, et al. (March 2003). "Design, synthesis, and biological evaluation of C9- and C2-substituted pyrazolo[4,3-e]-1,2,4-triazolo[1,5-c]pyrimidines as new A2A and A3 adenosine receptors antagonists". Journal of Medicinal Chemistry. 46 (7): 1229–41. doi:10.1021/jm021023m. PMID   12646033.
    • Weiss SM, Benwell K, Cliffe IA, Gillespie RJ, Knight AR, Lerpiniere J, et al. (December 2003). "Discovery of nonxanthine adenosine A2A receptor antagonists for the treatment of Parkinson's disease". Neurology. 61 (11 Suppl 6): S101-6. doi:10.1212/01.WNL.0000095581.20961.7D. PMID   14663021. S2CID   12327094.
    • Cristalli G, Lambertucci C, Taffi S, Vittori S, Volpini R (2003). "Medicinal chemistry of adenosine A2A receptor agonists". Current Topics in Medicinal Chemistry. 3 (4): 387–401. doi:10.2174/1568026033392282. PMID   12570757. Archived from the original on 2009-05-04. Retrieved 2018-10-02.
    • Cacciari B, Pastorin G, Spalluto G (2003). "Medicinal chemistry of A2A adenosine receptor antagonists". Current Topics in Medicinal Chemistry. 3 (4): 403–11. doi:10.2174/1568026033392183. PMID   12570758. Archived from the original on 2009-05-04. Retrieved 2018-10-02.
    • Cristalli G, Cacciari B, Dal Ben D, Lambertucci C, Moro S, Spalluto G, Volpini R (March 2007). "Highlights on the development of A(2A) adenosine receptor agonists and antagonists". ChemMedChem. 2 (3): 260–81. doi:10.1002/cmdc.200600193. PMID   17177231. S2CID   6973388.
    • Diniz C, Borges F, Santana L, Uriarte E, Oliveira JM, Gonçalves J, Fresco P (2008). "Ligands and therapeutic perspectives of adenosine A(2A) receptors". Current Pharmaceutical Design. 14 (17): 1698–722. doi:10.2174/138161208784746842. PMID   18673194. Archived from the original on 2009-05-04. Retrieved 2018-10-02.
    • Cristalli G, Lambertucci C, Marucci G, Volpini R, Dal Ben D (2008). "A2A adenosine receptor and its modulators: overview on a druggable GPCR and on structure-activity relationship analysis and binding requirements of agonists and antagonists". Current Pharmaceutical Design. 14 (15): 1525–52. doi:10.2174/138161208784480081. PMID   18537675.
    • Gillespie RJ, Adams DR, Bebbington D, Benwell K, Cliffe IA, Dawson CE, et al. (May 2008). "Antagonists of the human adenosine A2A receptor. Part 1: Discovery and synthesis of thieno[3,2-d]pyrimidine-4-methanone derivatives". Bioorganic & Medicinal Chemistry Letters. 18 (9): 2916–9. doi:10.1016/j.bmcl.2008.03.075. PMID   18406614.
    • Gillespie RJ, Cliffe IA, Dawson CE, Dourish CT, Gaur S, Giles PR, et al. (May 2008). "Antagonists of the human adenosine A2A receptor. Part 2: Design and synthesis of 4-arylthieno[3,2-d]pyrimidine derivatives". Bioorganic & Medicinal Chemistry Letters. 18 (9): 2920–3. doi:10.1016/j.bmcl.2008.03.076. PMID   18407496.
    • Gillespie RJ, Cliffe IA, Dawson CE, Dourish CT, Gaur S, Jordan AM, et al. (May 2008). "Antagonists of the human adenosine A2A receptor. Part 3: Design and synthesis of pyrazolo[3,4-d]pyrimidines, pyrrolo[2,3-d]pyrimidines and 6-arylpurines". Bioorganic & Medicinal Chemistry Letters. 18 (9): 2924–9. doi:10.1016/j.bmcl.2008.03.072. PMID   18411049.
  30. Burton JH, Mass M, Menegazzi JJ, Yealy DM (August 1997). "Aminophylline as an adjunct to standard advanced cardiac life support in prolonged cardiac arrest". Annals of Emergency Medicine. 30 (2): 154–8. doi:10.1016/S0196-0644(97)70134-3. PMID   9250637.
  31. Khoury MY, Moukarbel GV, Obeid MY, Alam SE (May 2001). "Effect of aminophylline on complete atrioventricular block with ventricular asystole following blunt chest trauma". Injury. 32 (4): 335–8. doi:10.1016/S0020-1383(00)00222-9. PMID   11325371.
  32. Mader TJ, Bertolet B, Ornato JP, Gutterman JM (October 2000). "Aminophylline in the treatment of atropine-resistant bradyasystole". Resuscitation. 47 (2): 105–12. doi:10.1016/S0300-9572(00)00234-3. PMID   11008148.
  33. Mader TJ, Smithline HA, Durkin L, Scriver G (March 2003). "A randomized controlled trial of intravenous aminophylline for atropine-resistant out-of-hospital asystolic cardiac arrest". Academic Emergency Medicine. 10 (3): 192–7. doi: 10.1197/aemj.10.3.192 . PMID   12615581.
  34. Mader TJ, Gibson P (August 1997). "Adenosine receptor antagonism in refractory asystolic cardiac arrest: results of a human pilot study". Resuscitation. 35 (1): 3–7. doi:10.1016/S0300-9572(97)01097-6. PMID   9259053.
  35. Perouansky M, Shamir M, Hershkowitz E, Donchin Y (July 1998). "Successful resuscitation using aminophylline in refractory cardiac arrest with asystole". Resuscitation. 38 (1): 39–41. doi:10.1016/S0300-9572(98)00079-3. PMID   9783508.
  36. Viskin S, Belhassen B, Roth A, Reicher M, Averbuch M, Sheps D, et al. (February 1993). "Aminophylline for bradyasystolic cardiac arrest refractory to atropine and epinephrine". Annals of Internal Medicine. 118 (4): 279–81. doi:10.7326/0003-4819-118-4-199302150-00006. PMID   8420445. S2CID   44883687.
  37. Jenner P (December 2003). "A2A antagonists as novel non-dopaminergic therapy for motor dysfunction in PD". Neurology. 61 (11 Suppl 6): S32-8. doi:10.1212/01.WNL.0000095209.59347.79. PMID   14663007. S2CID   28897242.
  38. Mori A, Shindou T (December 2003). "Modulation of GABAergic transmission in the striatopallidal system by adenosine A2A receptors: a potential mechanism for the antiparkinsonian effects of A2A antagonists". Neurology. 61 (11 Suppl 6): S44-8. doi:10.1212/01.WNL.0000095211.71092.A0. PMID   14663009. S2CID   26827799.
  39. Pinna A, Wardas J, Simola N, Morelli M (November 2005). "New therapies for the treatment of Parkinson's disease: adenosine A2A receptor antagonists". Life Sciences. 77 (26): 3259–67. doi:10.1016/j.lfs.2005.04.029. PMID   15979104.
  40. Kelsey JE, Langelier NA, Oriel BS, Reedy C (January 2009). "The effects of systemic, intrastriatal, and intrapallidal injections of caffeine and systemic injections of A2A and A1 antagonists on forepaw stepping in the unilateral 6-OHDA-lesioned rat". Psychopharmacology. 201 (4): 529–39. doi:10.1007/s00213-008-1319-0. PMID   18791705. S2CID   24159282.
  41. 1 2 3 4 5 Jacobson KA, Gao ZG (March 2006). "Adenosine receptors as therapeutic targets". Nature Reviews. Drug Discovery. 5 (3): 247–64. doi:10.1038/nrd1983. PMC   3463109 . PMID   16518376. table 1 lists affinities
  42. Yoneyama F, Yamada H, Satoh K, Taira N (March 1992). "Vasodepressor mechanisms of 2-(1-octynyl)-adenosine (YT-146), a selective adenosine A2 receptor agonist, involve the opening of glibenclamide-sensitive K+ channels". European Journal of Pharmacology. 213 (2): 199–204. doi:10.1016/0014-2999(92)90682-T. PMID   1521559.
  43. https://doi.org/10.1021/acs.jmedchem.0c01856
  44. https://doi.org/10.3390/biomedicines10020515
  45. Burstein S (7 February 2015). "Cannabidiol (CBD) and its analogs: a review of their effects on inflammation". Bioorganic & Medicinal Chemistry. 23 (7): 1377–1385. doi:10.1016/j.bmc.2015.01.059. PMID   25703248.
  46. Doyle SE, Breslin FJ, Rieger JM, Beauglehole A, Lynch WJ (August 2012). "Time and sex-dependent effects of an adenosine A2A/A1 receptor antagonist on motivation to self-administer cocaine in rats". Pharmacology, Biochemistry, and Behavior. 102 (2): 257–63. doi:10.1016/j.pbb.2012.05.001. PMC   3383440 . PMID   22579716.
  47. Kase H, Aoyama S, Ichimura M, Ikeda K, Ishii A, Kanda T, et al. (December 2003). "Progress in pursuit of therapeutic A2A antagonists: the adenosine A2A receptor selective antagonist KW6002: research and development toward a novel nondopaminergic therapy for Parkinson's disease". Neurology. 61 (11 Suppl 6): S97-100. doi:10.1212/01.WNL.0000095219.22086.31. PMID   14663020. S2CID   72084113.
  48. Mott AM, Nunes EJ, Collins LE, Port RG, Sink KS, Hockemeyer J, et al. (May 2009). "The adenosine A2A antagonist MSX-3 reverses the effects of the dopamine antagonist haloperidol on effort-related decision making in a T-maze cost/benefit procedure". Psychopharmacology. 204 (1): 103–12. doi:10.1007/s00213-008-1441-z. PMC   2875244 . PMID   19132351.
  49. Hodgson RA, Bertorelli R, Varty GB, Lachowicz JE, Forlani A, Fredduzzi S, et al. (July 2009). "Characterization of the potent and highly selective A2A receptor antagonists preladenant and SCH 412348 [7-[2-[4-2,4-difluorophenyl]-1-piperazinyl]ethyl]-2-(2-furanyl)-7H-pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidin-5-amine] in rodent models of movement disorders and depression". The Journal of Pharmacology and Experimental Therapeutics. 330 (1): 294–303. doi:10.1124/jpet.108.149617. PMID   19332567. S2CID   22033475.
  50. Pinna A, Fenu S, Morelli M (March 2001). "Motor stimulant effects of the adenosine A2A receptor antagonist SCH 58261 do not develop tolerance after repeated treatments in 6-hydroxydopamine-lesioned rats". Synapse. 39 (3): 233–8. doi:10.1002/1098-2396(20010301)39:3<233::AID-SYN1004>3.0.CO;2-K. PMID   11284438. S2CID   23370571.
  51. Rose S, Jackson MJ, Smith LA, Stockwell K, Johnson L, Carminati P, Jenner P (September 2006). "The novel adenosine A2a receptor antagonist ST1535 potentiates the effects of a threshold dose of L-DOPA in MPTP treated common marmosets". European Journal of Pharmacology. 546 (1–3): 82–7. doi:10.1016/j.ejphar.2006.07.017. PMID   16925991.
  52. Kamiya T, Saitoh O, Yoshioka K, Nakata H (June 2003). "Oligomerization of adenosine A2A and dopamine D2 receptors in living cells". Biochemical and Biophysical Research Communications. 306 (2): 544–9. doi:10.1016/S0006-291X(03)00991-4. PMID   12804599.
  53. 1 2 Sitkovsky MV, Kjaergaard J, Lukashev D, Ohta A (October 2008). "Hypoxia-adenosinergic immunosuppression: tumor protection by T regulatory cells and cancerous tissue hypoxia". Clinical Cancer Research. 14 (19): 5947–52. doi: 10.1158/1078-0432.CCR-08-0229 . PMID   18829471.
  54. 1 2 Leone RD, Lo YC, Powell JD (April 2015). "A2aR antagonists: Next generation checkpoint blockade for cancer immunotherapy". Computational and Structural Biotechnology Journal. 13: 265–72. doi:10.1016/j.csbj.2015.03.008. PMC   4415113 . PMID   25941561.
  55. Pardoll DM (March 2012). "The blockade of immune checkpoints in cancer immunotherapy". Nature Reviews. Cancer. 12 (4): 252–64. doi:10.1038/nrc3239. PMC   4856023 . PMID   22437870.
  56. Willingham SB (October 2018). "A2AR Antagonism with CPI-444 Induces Antitumor Responses and Augments Efficacy to Anti-PD-(L)1 and Anti-CTLA-4 in Preclinical Models". Cancer Immunol Res. 6 (10): 1136–1149. doi:10.1158/2326-6066.CIR-18-0056. PMID   30131376.
  57. Beavis PA, Henderson MA, Giuffrida L, Mills JK, Sek K, Cross RS, et al. (March 2017). "Targeting the adenosine 2A receptor enhances chimeric antigen receptor T cell efficacy". The Journal of Clinical Investigation. 127 (3): 929–941. doi:10.1172/JCI89455. PMC   5330718 . PMID   28165340.

Further reading