What is the term for a medication that blocks a receptor in the body?

Consumers and health professionals are advised that serotonin syndrome is a newly identified issue associated with products containing the serotonin-blocking medicines classed as 5-HT3 receptor antagonists.

These medicines are used after surgery and in patients undergoing cancer treatment to prevent nausea and vomiting. They work by blocking serotonin from entering certain cells in the nervous system and brain.

Serotonin syndrome has been seen in patients using 5-HT3 receptor antagonists at the same time as other serotonergic medicines. The 5-HT3 receptor antagonists available in Australia are: granisetron (Sancuso and Kytril), dolasetron (Anzemet), tropisetron (Navoban), ondansetron (Zofran) and palonosetron (Aloxi).

Serotonin syndrome occurs when serotonin accumulates to high levels in the body, as can happen when medicines block the chemical from entering cells. The syndrome is characterised by:

  • altered mental state, e.g. confusion, agitation, restlessness and excitement
  • autonomic dysfunction, e.g. tachycardia, sweating, shivering, hypertension and hyperthermia
  • neuromuscular excitation, e.g. hyperreflexia, tremor.

In some cases serotonin syndrome can lead to loss of consciousness, coma and death.

Health Canada published safety information about this issue after completing a review of 5-HT3 receptor antagonists.

The TGA has previously published an article regarding serotonin syndrome, including information about diagnosis and treatment of this potentially life-threatening condition.

Information for consumers

If you, or someone you care for, is being treated for nausea and vomiting with a serotonin-blocking medicine and experiences the symptoms listed above, contact a health practitioner immediately.

If you have any questions or concerns about this issue, please contact your treating doctor.

Information for health professionals

Health professionals are advised to be alert to this issue.

The TGA is working with the sponsors of the different 5-HT3 receptor antagonists to update their Product Information (PI) regarding the risk of serotonin syndrome. Some sponsors already include this information in their PI.

The updated PI contains a new precaution and information on drug interactions advising that serotonin syndrome has been described following the use of 5-HT3 receptor antagonists when used concomitantly with other serotonergic drugs, including selective serotonin reuptake inhibitors (SSRIs) and serotonin noradrenaline reuptake inhibitors (SNRIs).

If concomitant treatment with a 5-HT3 receptor antagonist and other serotonergic drugs is clinically warranted, it is advised that the patient and caregivers are advised of this issue and that appropriate observation is undertaken.

Reporting problems

Consumers and health professionals are encouraged to report problems with medicines or vaccines. Your report will contribute to the TGA's monitoring of these products.

The TGA cannot give advice about an individual's medical condition. You are strongly encouraged to talk with a health professional if you are concerned about a possible adverse event associated with a medicine or vaccine.

1. Fredholm BB, et al. International Union of Pharmacology. XXV. Nomenclature and classification of adenosine receptors. Pharmacol. Rev. 2001;53:527–552. [PMC free article] [PubMed] [Google Scholar] This is an overview of the current knowledge of adenosine receptor biology, from gene structure to expression, distribution, biochemical properties, pharmacological profiles, signalling and behavioural responses, focusing on receptor nomenclature and classification

2. Fredholm BB, et al. International Union of Basic and Clinical Pharmacology. LXXXI. Nomenclature and classification of adenosine receptors — an update. Pharmacol. Rev. 2011;63:1–34. [PMC free article] [PubMed] [Google Scholar] This is an update of reference 1, based on 10 more years of research.

3. Eltzschig HK, Sitkovsky MV, Robson SC. Purinergic signaling during inflammation. N. Engl. J. Med. 2012;367:2322–2333. [PMC free article] [PubMed] [Google Scholar] This is a comprehensive review summarizing the biomedical implications of extracellular ATP, ADP and adenosine signalling in the context of a broad range of inflammatory diseases.

4. Eltzschig HK. Adenosine: an old drug newly discovered. Anesthesiology. 2009;111:904–915. [PMC free article] [PubMed] [Google Scholar]

5. Johansson SM, Yang JN, Lindgren E, Fredholm BB. Eliminating the antilipolytic adenosine A1 receptor does not lead to compensatory changes in the antilipolytic actions of PGE2 and nicotinic acid. Acta Physiol. 2007;190:87–96. [PubMed] [Google Scholar]

6. Grenz A, et al. Equilibrative nucleoside transporter 1 (ENT1) regulates postischemic blood flow during acute kidney injury in mice. J. Clin. Invest. 2012;122:693–710. [PMC free article] [PubMed] [Google Scholar] Retracted This report shows that a crosstalk pathway — between epithelium-expressed adenosine transporters (such as ENT1) and endothelium-expressed A2B receptors — dampens renal ischaemia and reperfusion injury by preventing a no-reflow phenomenon.

7. Sun D, et al. Mediation of tubuloglomerular feedback by adenosine: evidence from mice lacking adenosine 1 receptors. Proc. Natl Acad. Sci. USA. 2001;98:9983–9988. [PMC free article] [PubMed] [Google Scholar]

8. Rosenberger P, et al. Hypoxia-inducible factor-dependent induction of netrin-1 dampens inflammation caused by hypoxia. Nature Immunol. 2009;10:195–202. [PubMed] [Google Scholar] This study demonstrates that the neuronal guidance molecule netrin 1 has important functions outside brain development, showing that hypoxia-dependent induction of netrin 1 dampens mucosal inflammation by enhancing a purinergic signalling event, particularly through the A2B receptor.

9. Huang ZL, et al. Adenosine A2A, but not A1, receptors mediate the arousal effect of caffeine. Nature Neurosci. 2005;8:858–859. [PubMed] [Google Scholar]

10. Lazarus M, et al. Arousal effect of caffeine depends on adenosine A2A receptors in the shell of the nucleus accumbens. J. Neurosci. 2011;31:10067–10075. [PMC free article] [PubMed] [Google Scholar]

11. Liu XL, et al. Genetic inactivation of the adenosine A2A receptor attenuates pathologic but not developmental angiogenesis in the mouse retina. Invest. Ophthalmol. Vis. Sci. 2010;51:6625–6632. [PMC free article] [PubMed] [Google Scholar]

12. Hasko G, Linden J, Cronstein B, Pacher P. Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nature Rev. Drug Discov. 2008;7:759–770. [PMC free article] [PubMed] [Google Scholar] This is an expert and focused review on the molecular and cellular biology of adenosine receptors and their pharmacological actions, with emphasis on immune and inflammatory responses.

13. Eltzschig HK, Carmeliet P. Hypoxia and inflammation. N. Engl. J. Med. 2011;364:656–665. [PMC free article] [PubMed] [Google Scholar] This review describes the relationship between hypoxia and inflammation, and demonstrates the functional roles of their interdepencence during intestinal inflammation, acute lung injury, ischaemia-reperfusion injury and cancer.

14. Eltzschig HK, Eckle T. Ischemia and reperfusion — from mechanism to translation. Nature Med. 2011;17:1391–1401. [PMC free article] [PubMed] [Google Scholar]

15. Fredholm BB. Adenosine, an endogenous distress signal, modulates tissue damage and repair. Cell Death Differ. 2007;14:1315–1323. [PubMed] [Google Scholar]

16. Jacobson KA, Gao ZG. Adenosine receptors as therapeutic targets. Nature Rev. Drug Discov. 2006;5:247–264. [PMC free article] [PubMed] [Google Scholar] This is an expert and comprehensive review on the medicinal chemistry of the first and second generations of adenosine receptor agonists and antagonists as well as their possible clinical uses.

17. Delacretaz E. Clinical practice. Supraventricular tachycardia. N. Engl. J. Med. 2006;354:1039–1051. [PubMed] [Google Scholar]

18. Muller CE, Jacobson KA. Recent developments in adenosine receptor ligands and their potential as novel drugs. Biochim. Biophys. Acta. 2011;1808:1290–1308. [PMC free article] [PubMed] [Google Scholar]

19. Ghimire G, Hage FG, Heo J, Iskandrian AE. Regadenoson: a focused update. J. Nucl. Cardiol. 2012 Dec 11; doi:10.1007/s12350-012-9661-3. [PubMed] [Google Scholar]

20. Fredholm B, Chen J-F, Masino SA, Vaugeois J-M. Actions of adeosine at its receptors in the CNS: insights from knckouts and drugs. Annu. Rev. Pharmacol. Toxicol. 2005;45:385–412. [PubMed] [Google Scholar] This is a comprehensive review on the development and characterization of genetic knockout models for adenosine receptors (from 1997 to 2005), focusing on new insights into brain functions under physiological and pathological conditions.

21. Massie BM, et al. Rolofylline, an adenosine A1-receptor antagonist, in acute heart failure. N. Engl. J. Med. 2010;363:1419–1428. [PubMed] [Google Scholar] This is a report on the clinical Phase III trial for the A1 receptor antagonist rolofylline in 2,033 patients with acute heart failure; the drug had a disappointing clinical outcome.

22. Teerlink JR, et al. The safety of an adenosine A1-receptor antagonist, rolofylline, in patients with acute heart failure and renal impairment: findings from PROTECT. Drug Saf. 2012;35:233–244. [PubMed] [Google Scholar]

23. Fredholm BB, et al. Structure and function of adenosine receptors and their genes. Naunyn Schmiedebergs Arch. Pharmacol. 2000;362:364–374. [PubMed] [Google Scholar]

24. Daly JW, Padgett WL. Agonist activity of 2- and 5′-substituted adenosine analogs and their N6-cycloalkyl derivatives at A1- and A2-adenosine receptors coupled to adenylate cyclase. Biochem. Pharmacol. 1992;43:1089–1093. [PubMed] [Google Scholar]

25. Koeppen M, Eckle T, Eltzschig HK. Selective deletion of the A1 adenosine receptor abolishes heart-rate slowing effects of intravascular adenosine in vivo. PLoS ONE. 2009;4:e6784. [PMC free article] [PubMed] [Google Scholar]

26. Corvol JC, Studler JM, Schonn JS, Girault JA, Herve D. Gαolf is necessary for coupling D1 and A2a receptors to adenylyl cyclase in the striatum. J. Neurochem. 2001;76:1585–1588. [PubMed] [Google Scholar]

27. Kull B, Svenningsson P, Fredholm BB. Adenosine A2A receptors are colocalized with and activate Golf in rat striatum. Mol. Pharmacol. 2000;58:771–777. [PubMed] [Google Scholar]

28. Schulte G, Fredholm BB. Signalling from adenosine receptors to mitogen-activated protein kinases. Cell. Signal. 2003;15:813–827. [PubMed] [Google Scholar]

29. Eckle T, et al. A2B adenosine receptor dampens hypoxia-induced vascular leak. Blood. 2008;111:2024–2035. [PMC free article] [PubMed] [Google Scholar]

30. Eckle T, et al. Adora2b-elicited Per2 stabilization promotes a HIF-dependent metabolic switch crucial for myocardial adaptation to ischemia. Nature Med. 2012;18:774–782. [PMC free article] [PubMed] [Google Scholar] This study provides a mechanism of how A2B receptor signalling can provide cardioprotection against ischaemia–reperfusion injury. The findings indicate that myocardial A2B signalling leads to the stabilization of the circadian rhythm protein PER2 (period circadian clock 2), which mediates a metabolic switch that enhances myocardial glycolytic capacity, thereby providing enhanced ischaemia tolerance.

31. Hart ML, et al. Hypoxia-inducible factor-1α-dependent protection from intestinal ischemia/reperfusion injury involves ecto-5′-nucleotidase (CD73) and the A2B adenosine receptor. J. Immunol. 2011;186:4367–4374. [PMC free article] [PubMed] [Google Scholar] Retracted

32. Schingnitz U, et al. Signaling through the A2B adenosine receptor dampens endotoxin-induced acute lung injury. J. Immunol. 2010;184:5271–5279. [PMC free article] [PubMed] [Google Scholar]

33. Frick JS, et al. Contribution of adenosine A2B receptors to inflammatory parameters of experimental colitis. J. Immunol. 2009;182:4957–4964. [PMC free article] [PubMed] [Google Scholar]

34. Zhong H, et al. Activation of murine lung mast cells by the adenosine A3 receptor. J. Immunol. 2003;171:338–345. [PubMed] [Google Scholar]

35. Ochaion A, et al. The anti-inflammatory target A3 adenosine receptor is over-expressed in rheumatoid arthritis, psoriasis and Crohn’s disease. Cell. Immunol. 2009;258:115–122. [PubMed] [Google Scholar]

36. Gessi S, et al. Elevated expression of A3 adenosine receptors in human colorectal cancer is reflected in peripheral blood cells. Clin. Cancer Res. 2004;10:5895–5901. [PubMed] [Google Scholar]

37. Fishman P, Bar-Yehuda S, Liang BT, Jacobson KA. Pharmacological and therapeutic effects of A3 adenosine receptor agonists. Drug Discov. Today. 2012;17:359–366. [PMC free article] [PubMed] [Google Scholar]

38. King AE, Ackley MA, Cass CE, Young JD, Baldwin SA. Nucleoside transporters: from scavengers to novel therapeutic targets. Trends Pharmacol. Sci. 2006;27:416–425. [PubMed] [Google Scholar]

39. MacDonald PE, Braun M, Galvanovskis J, Rorsman P. Release of small transmitters through kiss-and-run fusion pores in rat pancreatic β cells. Cell Metab. 2006;4:283–290. [PubMed] [Google Scholar]

40. Zhang Z, et al. Regulated ATP release from astrocytes through lysosome exocytosis. Nature Cell Biol. 2007;9:945–953. [PubMed] [Google Scholar]

41. Chekeni FB, et al. Pannexin 1 channels mediate ‘find-me’ signal release and membrane permeability during apoptosis. Nature. 2010;467:863–867. [PMC free article] [PubMed] [Google Scholar]

42. Elliott MR, et al. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature. 2009;461:282–286. [PMC free article] [PubMed] [Google Scholar]

43. Anselmi F, et al. ATP release through connexin hemichannels and gap junction transfer of second messengers propagate Ca2+ signals across the inner ear. Proc. Natl. Acad. Sci. USA. 2008;105:18770–18775. [PMC free article] [PubMed] [Google Scholar]

44. Kanneganti TD, et al. Pannexin-1-mediated recognition of bacterial molecules activates the cryopyrin inflammasome independent of Toll-like receptor signaling. Immunity. 2007;26:433–443. [PubMed] [Google Scholar]

45. Faigle M, Seessle J, Zug S, El Kasmi KC, Eltzschig HK. ATP release from vascular endothelia occurs across Cx43 hemichannels and is attenuated during hypoxia. PLoS ONE. 2008;3:e2801. [PMC free article] [PubMed] [Google Scholar]

46. Ballarin M, Fredholm BB, Ambrosio S, Mahy N. Extracellular levels of adenosine and its metabolites in the striatum of awake rats: inhibition of uptake and metabolism. Acta Physiol. Scand. 1991;142:97–103. [PubMed] [Google Scholar]

47. Wei CJ, Li W, Chen JF. Normal and abnormal functions of adenosine receptors in the central nervous system revealed by genetic knockout studies. Biochim. Biophys. Acta. 2011;1808:1358–1379. [PubMed] [Google Scholar]

48. Fedele DE, Li T, Lan JQ, Fredholm BB, Boison D. Adenosine A1 receptors are crucial in keeping an epileptic focus localized. Exp. Neurol. 2006;200:184–190. [PubMed] [Google Scholar]

49. Gimenez-Llort L, et al. Mice lacking the adenosine A1 receptor are anxious and aggressive, but are normal learners with reduced muscle strength and survival rate. Eur. J. Neurosci. 2002;16:547–550. [PubMed] [Google Scholar]

50. Schwarzschild MA, Agnati L, Fuxe K, Chen JF, Morelli M. Targeting adenosine A2A receptors in Parkinson’s disease. Trends Neurosci. 2006;29:647–654. [PubMed] [Google Scholar] This is an expert and focused review on the rationale as well as preclinical and clinical evidence for the efficacy of an A2A receptor antagonist in Parkinson’s disease.

51. Ferre S, Fredholm BB, Morelli M, Popoli P, Fuxe K. Adenosine-dopamine receptor-receptor interactions as an integrative mechanism in the basal ganglia. Trends Neurosci. 1997;20:482–487. [PubMed] [Google Scholar]

52. Richardson PJ, Kase H, Jenner PG. Adenosine A2A receptor antagonists as new agents for the treatment of Parkinson’s disease. Trends Pharmacol. Sci. 1997;18:338–344. [PubMed] [Google Scholar]

53. Wei CJ, et al. Selective inactivation of adenosine A2A receptors in striatal neurons enhances working memory and reversal learning. Learn. Mem. 2011;18:459–474. [PMC free article] [PubMed] [Google Scholar]

54. Zhou SJ, et al. Preferential enhancement of working memory in mice lacking adenosine A2A receptors. Brain Res. 2009;1303:74–83. [PubMed] [Google Scholar]

55. Yu C, Gupta J, Chen JF, Yin HH. Genetic deletion of A2A adenosine receptors in the striatum selectively impairs habit formation. J. Neurosci. 2009;29:15100–15103. [PMC free article] [PubMed] [Google Scholar]

56. Ledent C, et al. Aggressiveness, hypoalgesia and high blood pressure in mice lacking the adenosine A2a receptor. Nature. 1997;388:674–678. [PubMed] [Google Scholar] This milestone study describes the generation and initial characterization of the first genetic knockout mice for adenosine receptors — namely the Adora2a-knockout mice.

57. Chen JF, et al. A2A adenosine receptor deficiency attenuates brain injury induced by transient focal ischemia in mice. J. Neurosci. 1999;19:9192–9200. [PMC free article] [PubMed] [Google Scholar] This study describes the generation and characterization of the second line of the Adora2a-knockout mice and provides compelling evidence that A2A receptor inactivation confers neuroprotection against ischaemia.

58. Xiao D, et al. Forebrain adenosine A2A receptors contribute to l -3,4-dihydroxyphenylalanine-induced dyskinesia in hemiparkinsonian mice. J. Neurosci. 2006;26:13548–13555. [PMC free article] [PubMed] [Google Scholar]

59. Ohta A, Sitkovsky M. Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature. 2001;414:916–920. [PubMed] [Google Scholar] This landmark study demonstrates that endogenous adenosine via the activation of A2A receptors functions to limit excessive inflammation and collateral tissue damage.

60. Day YJ, et al. Renal protection from ischemia mediated by A2A adenosine receptors on bone marrow-derived cells. J. Clin. Invest. 2003;112:883–891. [PMC free article] [PubMed] [Google Scholar]

61. Chen JF, et al. Adenosine A2A receptors and brain injury: broad spectrum of neuroprotection, multifaceted actions and "fine tuning" modulation. Prog. Neurobiol. 2007;83:310–331. [PubMed] [Google Scholar]

62. Yu L, et al. Selective inactivation or reconstitution of adenosine A2A receptors in bone marrow cells reveals their significant contribution to the development of ischemic brain injury. Nature Med. 2004;10:1081–1087. [PubMed] [Google Scholar] This study demonstrates a novel role of the A2A receptor in bone marrow-derived cells: it protects against ischaemic brain injury.

63. Huang ZL, Urade Y, Hayaishi O. The role of adenosine in the regulation of sleep. Curr. Top. Med. Chem. 2011;11:1047–1057. [PubMed] [Google Scholar]

64. Yang D, et al. The A2B adenosine receptor protects against inflammation and excessive vascular adhesion. J. Clin. Invest. 2006;116:1913–1923. [PMC free article] [PubMed] [Google Scholar] This is the first description of Adora2b-knockout mice, which were found to be prone to vascular inflammation — a finding that was later confirmed in many other models of vascular inflammation.

65. Eckle T, et al. Cardioprotection by ecto-5′-nucleotidase (CD73) and A2B adenosine receptors. Circulation. 2007;115:1581–1590. [PubMed] [Google Scholar]

66. Schulte G, Fredholm BB. Human adenosine A1, A2A, A2B, and A3 receptors expressed in Chinese hamster ovary cells all mediate the phosphorylation of extracellular-regulated kinase 1/2. Mol. Pharmacol. 2000;58:477–482. [PubMed] [Google Scholar]

67. Hart ML, Jacobi B, Schittenhelm J, Henn M, Eltzschig HK. Cutting Edge: A2B adenosine receptor signaling provides potent protection during intestinal ischemia/reperfusion injury. J. Immunol. 2009;182:3965–3968. [PubMed] [Google Scholar]

68. Chouker A, et al. In vivo hypoxic preconditioning protects from warm liver ischemia-reperfusion injury through the adenosine A2B receptor. Transplantation. 2012;94:894–902. [PMC free article] [PubMed] [Google Scholar]

69. Grenz A, et al. The reno-vascular A2B adenosine receptor protects the kidney from ischemia. PLoS Med. 2008;5:e137. [PMC free article] [PubMed] [Google Scholar]

70. Eckle T, Grenz A, Laucher S, Eltzschig HK. A2B adenosine receptor signaling attenuates acute lung injury by enhancing alveolar fluid clearance in mice. J. Clin. Invest. 2008;118:3301–3315. [PMC free article] [PubMed] [Google Scholar] This head-to-head comparison of all four Adora-knockout mice revealed that Adora2b-knockout mice are particularly prone to ventilator-induced lung injury. The A2B receptor agonist BAY 60-6583 enhanced alveolar fluid transport, thereby attenuating pulmonary oedema and concomittant lung inflammation during acute lung injury.

71. Takedachi M, et al. CD73-generated adenosine promotes osteoblast differentiation. J. Cell. Physiol. 2012;227:2622–2631. [PMC free article] [PubMed] [Google Scholar]

72. Carroll SH, et al. A2B adenosine receptor promotes mesenchymal stem cell differentiation to osteoblasts and bone formation in vivo. J. Biol. Chem. 2012;287:15718–15727. [PMC free article] [PubMed] [Google Scholar]

73. Johnston-Cox H, et al. The A2b adenosine receptor modulates glucose homeostasis and obesity. PLoS ONE. 2012;7:e40584. [PMC free article] [PubMed] [Google Scholar]

74. Koupenova M, et al. A2b adenosine receptor regulates hyperlipidemia and atherosclerosis. Circulation. 2012;125:354–363. [PMC free article] [PubMed] [Google Scholar]

75. Cekic C, et al. Adenosine A2B receptor blockade slows growth of bladder and breast tumors. J. Immunol. 2012;188:198–205. [PMC free article] [PubMed] [Google Scholar]

76. Dai Y, et al. A2B adenosine receptor-mediated induction of IL-6 promotes CKD. J. Am. Soc. Nephrol. 2011;22:890–901. [PMC free article] [PubMed] [Google Scholar]

77. Wei W, et al. Blocking A2B adenosine receptor alleviates pathogenesis of experimental autoimmune encephalomyelitis via inhibition of IL-6 production and Th17 differentiation. J. Immunol. 2013;190:138–146. [PMC free article] [PubMed] [Google Scholar]

78. Bjorklund O, et al. Decreased behavioral activation following caffeine, amphetamine and darkness in A3 adenosine receptor knock-out mice. Physiol. Behav. 2008;95:668–676. [PubMed] [Google Scholar]

79. Yang JN, Wang Y, Garcia-Roves PM, Bjornholm M, Fredholm BB. Adenosine A3 receptors regulate heart rate, motor activity and body temperature. Acta Physiol. 2010;199:221–230. [PMC free article] [PubMed] [Google Scholar]

80. Eltzschig HK, et al. Coordinated adenine nucleotide phosphohydrolysis and nucleoside signaling in posthypoxic endothelium: role of ectonucleotidases and adenosine A2B receptors. J. Exp. Med. 2003;198:783–796. [PMC free article] [PubMed] [Google Scholar]

81. Thompson LF, et al. Crucial role for ecto-5′-nucleotidase (CD73) in vascular leakage during hypoxia. J. Exp. Med. 2004;200:1395–1405. [PMC free article] [PubMed] [Google Scholar]

82. Sitkovsky MV, et al. Physiological control of immune response and inflammatory tissue damage by hypoxia-inducible factors and adenosine A2A receptors. Annu. Rev. Immunol. 2004;22:657–682. [PubMed] [Google Scholar] This important review discusses the relationship between hypoxia and adenosine signalling through the A2A receptor, and implicates A2A signalling as an endogenous feedback signal to dampen collateral tissue injury.

83. Gomes CV, Kaster MP, Tome AR, Agostinho PM, Cunha R. A. Adenosine receptors and brain diseases: neuroprotection and neurodegeneration. Biochim. Biophys. Acta. 2011;1808:1380–1399. [PubMed] [Google Scholar]

84. Boison D. Adenosine dysfunction in epilepsy. Glia. 2012;60:1234–1243. [PMC free article] [PubMed] [Google Scholar]

85. Linden J. Regulation of leukocyte function by adenosine receptors. Adv. Pharmacol. 2011;61:95–114. [PMC free article] [PubMed] [Google Scholar]

86. Synnestvedt K, et al. Ecto-5′-nucleotidase (CD73) regulation by hypoxia-inducible factor-1 mediates permeability changes in intestinal epithelia. J. Clin. Invest. 2002;110:993–1002. [PMC free article] [PubMed] [Google Scholar]

87. Zetterstrom T, et al. Purine levels in the intact rat brain. Studies with an implanted perfused hollow fibre. Neurosci. Lett. 1982;29:111–115. [PubMed] [Google Scholar]

88. Pedata F, Corsi C, Melani A, Bordoni F, Latini S. Adenosine extracellular brain concentrations and role of A2A receptors in ischemia. Ann. NY Acad. Sci. 2001;939:74–84. [PubMed] [Google Scholar]

89. Andine P, Rudolphi KA, Fredholm BB, Hagberg H. Effect of propentofylline (HWA 285) on extracellular purines and excitatory amino acids in CA1 of rat hippocampus during transient ischaemia. Br. J. Pharmacol. 1990;100:814–818. [PMC free article] [PubMed] [Google Scholar]

90. Dux E, Fastbom J, Ungerstedt U, Rudolphi K, Fredholm BB. Protective effect of adenosine and a novel xanthine derivative propentofylline on the cell damage after bilateral carotid occlusion in the gerbil hippocampus. Brain Res. 1990;516:248–256. [PubMed] [Google Scholar]

91. Johansson B, et al. Hyperalgesia, anxiety, and decreased hypoxic neuroprotection in mice lacking the adenosine A1 receptor. Proc. Natl Acad. Sci. USA. 2001;98:9407–9412. [PMC free article] [PubMed] [Google Scholar] This is the first report of the generation and charaterization of an Adora1-knockout mouse with a focus on its effects in the central nervous system.

92. Eckle T, Koeppen M, Eltzschig HK. Role of extracellular adenosine in acute lung injury. Physiology. 2009;24:298–306. [PubMed] [Google Scholar]

93. Ahmad A, et al. Adenosine A2A receptor is a unique angiogenic target of HIF-2α in pulmonary endothelial cells. Proc. Natl Acad. Sci. USA. 2009;106:10684–10689. [PMC free article] [PubMed] [Google Scholar]

94. Eckle T, Kohler D, Lehmann R, El Kasmi KC, Eltzschig HK. Hypoxia-inducible factor-1 is central to cardioprotection: a new paradigm for ischemic preconditioning. Circulation. 2008;118:166–175. [PubMed] [Google Scholar] These findings reveal a functional role of HIF in mediating cardioprotection during ischaemic preconditioning by enhancing extracelluluar adenosine production and A2B receptor signalling.

95. Kong T, Westerman KA, Faigle M, Eltzschig HK, Colgan SP. HIF-dependent induction of adenosine A2B receptor in hypoxia. FASEB J. 2006;20:2242–2250. [PubMed] [Google Scholar]

96. Eltzschig HK, et al. Central role of Sp1-regulated CD39 in hypoxia/ischemia protection. Blood. 2009;113:224–232. [PMC free article] [PubMed] [Google Scholar] In contrast to NT5E induction by hypoxia, which is mediated by HIF, this study identifies that ENTPD1 induction during conditions of limited oxygen availability is mediated by the transcription factor SP1.

97. Hart ML, Gorzolla IC, Schittenhelm J, Robson SC, Eltzschig HK. SP1-dependent induction of CD39 facilitates hepatic ischemic preconditioning. J. Immunol. 2010;184:4017–4024. [PMC free article] [PubMed] [Google Scholar]

98. Kohler D, et al. CD39/ectonucleoside triphosphate diphosphohydrolase 1 provides myocardial protection during cardiac ischemia/reperfusion injury. Circulation. 2007;116:1784–1794. [PubMed] [Google Scholar]

99. Reutershan J, et al. Adenosine and inflammation: CD39 and CD73 are critical mediators in LPS-induced PMN trafficking into the lungs. FASEB J. 2009;23:473–482. [PubMed] [Google Scholar]

100. Hart ML, et al. Extracellular adenosine production by ecto-5′-nucleotidase protects during murine hepatic ischemic preconditioning. 2008;135:1739–1750.e3. [PubMed] [Google Scholar]

101. Csoka B, et al. A2B adenosine receptors protect against sepsis-induced mortality by dampening excessive inflammation. J. Immunol. 2010;185:542–550. [PMC free article] [PubMed] [Google Scholar]

102. Lappas CM, Day YJ, Marshall MA, Engelhard VH, Linden J. Adenosine A2A receptor activation reduces hepatic ischemia reperfusion injury by inhibiting CD1d-dependent NKT cell activation. J. Exp. Med. 2006;203:2639–2648. [PMC free article] [PubMed] [Google Scholar]

103. Day YJ, et al. A2A adenosine receptors on bone marrow-derived cells protect liver from ischemia-reperfusion injury. J. Immunol. 2005;174:5040–5046. [PubMed] [Google Scholar]

104. Enjyoji K, et al. Targeted disruption of CD39/ATP diphosphohydrolase results in disordered hemostasis and thromboregulation. Nature Med. 1999;5:1010–1017. [PubMed] [Google Scholar]

105. Marcus AJ, et al. Metabolic control of excessive extracellular nucleotide accumulation by CD39/ ecto-nucleotidase-1: implications for ischemic vascular diseases. J. Pharmacol. Exp. Ther. 2003;305:9–16. [PubMed] [Google Scholar]

106. Eckle T, et al. Identification of ectonucleotidases CD39 and CD73 in innate protection during acute lung injury. J. Immunol. 2007;178:8127–8137. [PubMed] [Google Scholar]

107. Grenz A, et al. Contribution of E-NTPDase1 (CD39) to renal protection from ischemia-reperfusion injury. FASEB J. 2007;21:2863–2873. [PubMed] [Google Scholar]

108. Grenz A, et al. Protective role of ecto-5′-nucleotidase (CD73) in renal ischemia. J. Am. Soc. Nephrol. 2007;18:833–845. [PubMed] [Google Scholar]

109. Hasko G, et al. Ecto-5′-nucleotidase (CD73) decreases mortality and organ injury in sepsis. J. Immunol. 2011;187:4256–4267. [PMC free article] [PubMed] [Google Scholar]

110. Castillo CA, Leon D, Ruiz MA, Albasanz JL, Martin M. Modulation of adenosine A1 and A2A receptors in C6 glioma cells during hypoxia: involvement of endogenous adenosine. J. Neurochem. 2008;105:2315–2329. [PubMed] [Google Scholar]

111. Angelatou F, et al. Upregulation of A1 adenosine receptors in human temporal lobe epilepsy: a quantitative autoradiographic study. Neurosci. Lett. 1993;163:11–14. [PubMed] [Google Scholar]

112. Biber K, et al. Interleukin-6 upregulates neuronal adenosine A1 receptors: implications for neuromodulation and neuroprotection. Neuropsychopharmacology. 2008;33:2237–2250. [PubMed] [Google Scholar]

113. Jin D, et al. CD73 on tumor cells impairs antitumor T-cell responses: a novel mechanism of tumor-induced immune suppression. Cancer Res. 2010;70:2245–2255. [PMC free article] [PubMed] [Google Scholar]

114. Sun X, et al. CD39/ENTPD1 expression by CD4+Foxp3+ regulatory T cells promotes hepatic metastatic tumor growth in mice. Gastroenterology. 2010;139:1030–1040. [PMC free article] [PubMed] [Google Scholar]

115. Ohta A, et al. A2A adenosine receptor protects tumors from antitumor T cells. Proc. Natl Acad. Sci. USA. 2006;103:13132–13137. [PMC free article] [PubMed] [Google Scholar]

116. Popoli P, et al. Blockade of striatal adenosine A2A receptor reduces, through a presynaptic mechanism, quinolinic acid-induced excitotoxicity: possible relevance to neuroprotective interventions in neurodegenerative diseases of the striatum. J. Neurosci. 2002;22:1967–1975. [PMC free article] [PubMed] [Google Scholar]

117. Dai SS, et al. Local glutamate level dictates adenosine A2A receptor regulation of neuroinflammation and traumatic brain injury. J. Neurosci. 2010;30:5802–5810. [PMC free article] [PubMed] [Google Scholar] This study demonstrates that a local increase in glutamate levels switches the anti-inflammatory effect mediated by A2A receptor activation to a pro-inflammatory effect and exacerbates traumatic brain injury, providing a partial explanation for the opposing effects on tissue damage caused by A2A receptor activation in the brain versus in peripheral tissues

118. Li W, et al. Genetic inactivation of adenosine A2A receptors attenuates acute traumatic brain injury in the mouse cortical impact model. Exp. Neurol. 2009;215:69–76. [PubMed] [Google Scholar]

119. Chen JF, et al. Neuroprotection by caffeine and A2A adenosine receptor inactivation in a model of Parkinson’s disease. J. Neurosci. 2001;21:RC143. [PMC free article] [PubMed] [Google Scholar]

120. Canas PM, et al. Adenosine A2A receptor blockade prevents synaptotoxicity and memory dysfunction caused by β-amyloid peptides via p38 mitogen-activated protein kinase pathway. J. Neurosci. 2009;29:14741–14751. [PMC free article] [PubMed] [Google Scholar]

121. Blum D, et al. A dual role of adenosine A2A receptors in 3-nitropropionic acid-induced striatal lesions: implications for the neuroprotective potential of A2A antagonists. J. Neurosci. 2003;23:5361–5369. [PMC free article] [PubMed] [Google Scholar]

122. Chou SY, et al. CGS21680 attenuates symptoms of Huntington’s disease in a transgenic mouse model. J. Neurochem. 2005;93:310–320. [PubMed] [Google Scholar]

123. Li Y, et al. Mouse spinal cord compression injury is reduced by either activation of the adenosine A2A receptor on bone marrow-derived cells or deletion of the A2A receptor on non-bone marrow-derived cells. Neuroscience. 2006;141:2029–2039. [PubMed] [Google Scholar]

124. Bar-Yehuda S, et al. The A3 adenosine receptor agonist CF102 induces apoptosis of hepatocellular carcinoma via de-regulation of the Wnt and NF-κB signal transduction pathways. Int. J. Oncol. 2008;33:287–295. [PubMed] [Google Scholar]

125. Deaglio S, et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J. Exp. Med. 2007;204:1257–1265. [PMC free article] [PubMed] [Google Scholar]

126. Eltzschig HK, et al. HIF-1-dependent repression of equilibrative nucleoside transporter (ENT) in hypoxia. J. Exp. Med. 2005;202:1493–1505. [PMC free article] [PubMed] [Google Scholar]

127. Loffler M, Morote-Garcia JC, Eltzschig SA, Coe IR, Eltzschig HK. Physiological roles of vascular nucleoside transporters. Arterioscler. Thromb. Vasc. Biol. 2007;27:1004–1013. [PubMed] [Google Scholar]

128. Morote-Garcia JC, Rosenberger P, Nivillac NM, Coe IR, Eltzschig HK. Hypoxia-inducible factor-dependent repression of equilibrative nucleoside transporter 2 attenuates mucosal inflammation during intestinal hypoxia. Gastroenterology. 2009;136:607–618. [PubMed] [Google Scholar]

129. Sitkovsky MV. Damage control by hypoxia-inhibited AK. Blood. 2008;111:5424–5425. [PubMed] [Google Scholar]

130. Morote-Garcia JC, Rosenberger P, Kuhlicke J, Eltzschig HK. HIF-1-dependent repression of adenosine kinase attenuates hypoxia-induced vascular leak. Blood. 2008;111:5571–5580. [PubMed] [Google Scholar]

131. Khoa ND, et al. Inflammatory cytokines regulate function and expression of adenosine A2A receptors in human monocytic THP-1 cells. J. Immunol. 2001;167:4026–4032. [PubMed] [Google Scholar]

132. Haschemi A, et al. Cross-regulation of carbon monoxide and the adenosine A2a receptor in macrophages. J. Immunol. 2007;178:5921–5929. [PubMed] [Google Scholar]

133. Zhang Y, Palmblad J, Fredholm BB. Biphasic effect of ATP on neutrophil functions mediated by P2U and adenosine A2A receptors. Biochem. Pharmacol. 1996;51:957–965. [PubMed] [Google Scholar]

134. Colgan SP, Eltzschig HK. Adenosine and hypoxia-inducible factor signaling in intestinal injury and recovery. Annu. Rev. Physiol. 2011;74:153–175. [PMC free article] [PubMed] [Google Scholar]

135. Fredholm BB. Adenosine receptors as drug targets. Exp. Cell Res. 2010;316:1284–1288. [PMC free article] [PubMed] [Google Scholar]

136. Dhalla AK, Shryock JC, Shreeniwas R, Belardinelli L. Pharmacology and therapeutic applications of A1 adenosine receptor ligands. Curr. Top. Med. Chem. 2003;3:369–385. [PubMed] [Google Scholar]

137. Chen JF. The adenosine A2A receptor as an attractive target for Parkinson’s disease treatment. Drug News Perspect. 2003;16:597–604. [PubMed] [Google Scholar]

138. Jenner P, et al. Adenosine, adenosine A2A antagonists, and Parkinson’s disease. Parkinsonism Relat. Disord. 2009;15:406–413. [PubMed] [Google Scholar]

139. Hauser RA, et al. Preladenant in patients with Parkinson’s disease and motor fluctuations: a phase 2, double-blind, randomised trial. Lancet Neurol. 2011;10:221–229. [PubMed] [Google Scholar]

140. Guttman M. Efficacy of istradefylline in Parkinson’s diease patients treated with levodopa with motor response complications: results of the KW-6002 US-018 study. Mov. Disord. 2006;21(Suppl. 15):585. [Google Scholar]

141. Meissner WG, et al. Priorities in Parkinson’s disease research. Nature Rev. Drug Discov. 2011;10:377–393. [PubMed] [Google Scholar]

142. Vallon V, Muhlbauer B, Osswald H. Adenosine and kidney function. Physiol. Rev. 2006;86:901–940. [PubMed] [Google Scholar]

143. Cotter G, et al. The PROTECT pilot study: a randomized, placebo-controlled, dose-finding study of the adenosine A1 receptor antagonist rolofylline in patients with acute heart failure and renal impairment. J. Card. Fail. 2008;14:631–640. [PubMed] [Google Scholar]

144. Voors AA, et al. Effects of the adenosine A1 receptor antagonist rolofylline on renal function in patients with acute heart failure and renal dysfunction: results from PROTECT (placebo-controlled randomized study of the selective adenosine A1 receptor antagonist rolofylline for patients hospitalized with acute decompensated heart failure and volume overload to assess treatment effect on congestion and renal function) J. Am. Coll. Cardiol. 2011;57:1899–1907. [PubMed] [Google Scholar] This paper describes the design and clinical outcome of the largest clinical Phase III trial (PROTECT) of the A1 receptor antagonist rolofylline in acute heart failure.

145. Gessi S, et al. Adenosine receptor targeting in health and disease. Expert Opin. Investig. Drugs. 2011;20:1591–1609. [PubMed] [Google Scholar]

146. Silverman MH, et al. Clinical evidence for utilization of the A3 adenosine receptor as a target to treat rheumatoid arthritis: data from a phase II clinical trial. J. Rheumatol. 2008;35:41–48. [PubMed] [Google Scholar]

147. Avni I, et al. Treatment of dry eye syndrome with orally administered CF101: data from a phase 2 clinical trial. Ophthalmology. 2010;117:1287–1293. [PMC free article] [PubMed] [Google Scholar]

148. Montinaro A, et al. Adoptive immunotherapy with Cl-IB-MECA-treated CD8+ T cells reduces melanoma growth in mice. PLoS ONE. 2012;7:e45401. [PMC free article] [PubMed] [Google Scholar]

149. Chen Z, et al. Controlling murine and rat chronic pain through A3 adenosine receptor activation. FASEB J. 2012;26:1855–1865. [PMC free article] [PubMed] [Google Scholar]

150. Gessi S, et al. A3 adenosine receptors in human neutrophils and promyelocytic HL60 cells: a pharmacological and biochemical study. Mol. Pharmacol. 2002;61:415–424. [PubMed] [Google Scholar]

151. Baraldi PG, Preti D, Borea PA, Varani K. Medicinal chemistry of A3 adenosine receptor modulators: pharmacological activities and therapeutic implications. J. Med. Chem. 2012;55:5676–5703. [PubMed] [Google Scholar]

152. Wallace KL, Linden J. Adenosine A2A receptors induced on iNKT and NK cells reduce pulmonary inflammation and injury in mice with sickle cell disease. Blood. 2010;116:5010–5020. [PMC free article] [PubMed] [Google Scholar] This paper shows that adenosine signalling, through the activation of A2A receptors expressed on natural killer T cells, dampens ischaemia-reperfusion injury in models of sickle cell disease.

153. Zhang Y, et al. Detrimental effects of adenosine signaling in sickle cell disease. Nature Med. 2011;17:79–86. [PMC free article] [PubMed] [Google Scholar]

154. Field JJ, Nathan DG, Linden J. Targeting iNKT cells for the treatment of sickle cell disease. Clin. Immunol. 2011;140:177–183. [PMC free article] [PubMed] [Google Scholar]

155. Gladwin MT. Adenosine receptor crossroads in sickle cell disease. Nature Med. 2011;17:38–40. [PubMed] [Google Scholar]

156. Schmitt LI, Sims RE, Dale N, Haydon PG. Wakefulness affects synaptic and network activity by increasing extracellular astrocyte-derived adenosine. J. Neurosci. 2012;32:4417–4425. [PMC free article] [PubMed] [Google Scholar]

157. Lovatt D, et al. Neuronal adenosine release, and not astrocytic ATP release, mediates feedback inhibition of excitatory activity. Proc. Natl Acad. Sci. USA. 2012;109:5913–5914. [PMC free article] [PubMed] [Google Scholar]

158. Ramlackhansingh AF, et al. Adenosine 2A receptor availability in dyskinetic and nondyskinetic patients with Parkinson disease. Neurology. 2011;76:1811–1816. [PMC free article] [PubMed] [Google Scholar]

159. Mishina M, et al. Adenosine A2A receptors measured with CTMSX PET in the striata of Parkinson’s disease patients. PLoS ONE. 2011;6:e17338. [PMC free article] [PubMed] [Google Scholar]

160. Cha JH, et al. Altered brain neurotransmitter receptors in transgenic mice expressing a portion of an abnormal human huntington disease gene. Proc. Natl Acad. Sci. USA. 1998;95:6480–6485. [PMC free article] [PubMed] [Google Scholar]

161. Popoli P, et al. Functions, dysfunctions and possible therapeutic relevance of adenosine A2A receptors in Huntington’s disease. Prog. Neurobiol. 2007;81:331–348. [PubMed] [Google Scholar]

162. Thiel M, et al. Oxygenation inhibits the physiological tissue-protecting mechanism and thereby exacerbates acute inflammatory lung injury. PLoS Biol. 2005;3:e174. [PMC free article] [PubMed] [Google Scholar] This is a landmark paper showing that the therapeutic use of high inspired oxygen concentrations (mechanical ventilation with 100% oxygen) — as is frequently necessary for the treatment of patients suffering from acute lung injury to maintain adequate oxygen tissue levels — has detrimental side effects.

163. Eltzschig HK, Rivera-Nieves J, Colgan SP. Targeting the A2B adenosine receptor during gastrointestinal ischemia and inflammation. Expert Opin. Ther. Targets. 2009;13:1267–1277. [PMC free article] [PubMed] [Google Scholar]

164. Day YJ, et al. Protection from ischemic liver injury by activation of A2A adenosine receptors during reperfusion: inhibition of chemokine induction. Am. J. Physiol. Gastrointest. Liver Physiol. 2004;286:G285–G293. [PubMed] [Google Scholar]

165. Zhou Y, Schneider DJ, Blackburn MR. Adenosine signaling and the regulation of chronic lung disease. Pharmacol. Ther. 2009;123:105–116. [PMC free article] [PubMed] [Google Scholar]

166. Chunn JL, et al. Adenosine-dependent pulmonary fibrosis in adenosine deaminase-deficient mice. J. Immunol. 2005;175:1937–1946. [PubMed] [Google Scholar]

167. Sun CX, et al. Role of A2B receptor signaling in adenosine-dependent pulmonary inflammation and injury. J. Clin. Invest. 2006;116:1–10. [PMC free article] [PubMed] [Google Scholar]

168. Peng Z, et al. Adenosine signaling contributes to ethanol-induced fatty liver in mice. J. Clin. Invest. 2009;119:582–594. [PMC free article] [PubMed] [Google Scholar]

169. Chan ES, et al. Adenosine A2A receptors in diffuse dermal fibrosis: pathogenic role in human dermal fibroblasts and in a murine model of scleroderma. Arthritis Rheum. 2006;54:2632–2642. [PubMed] [Google Scholar]

170. Wen J, et al. Increased adenosine contributes to penile fibrosis, a dangerous feature of priapism, via A2B adenosine receptor signaling. FASEB J. 2010;24:740–749. [PMC free article] [PubMed] [Google Scholar]

171. Mi T, et al. Excess adenosine in murine penile erectile tissues contributes to priapism via A2B adenosine receptor signaling. J. Clin. Invest. 2008;118:1491–1501. [PMC free article] [PubMed] [Google Scholar]

172. Volmer JB, Thompson LF, Blackburn MR. Ecto-5′-nucleotidase (CD73)-mediated adenosine production is tissue protective in a model of bleomycin-induced lung injury. J. Immunol. 2006;176:4449–4458. [PubMed] [Google Scholar]

173. Zhou Y, et al. Enhanced airway inflammation and remodeling in adenosine deaminase-deficient mice lacking the A2B adenosine receptor. J. Immunol. 2009;182:8037–8046. [PMC free article] [PubMed] [Google Scholar]

174. Sun CX, et al. Role of A2B adenosine receptor signaling in adenosine-dependent pulmonary inflammation and injury. J. Clin. Invest. 2006;116:2173–2182. [PMC free article] [PubMed] [Google Scholar]

175. Zhou Y, et al. Distinct roles for the A2B adenosine receptor in acute and chronic stages of bleomycin-induced lung injury. J. Immunol. 2011;186:1097–1106. [PMC free article] [PubMed] [Google Scholar]

176. Sullivan GW, Fang G, Linden J, Scheld WM. A2A adenosine receptor activation improves survival in mouse models of endotoxemia and sepsis. J. Infect. Dis. 2004;189:1897–1904. [PubMed] [Google Scholar]

177. Nemeth ZH, et al. Adenosine A2A receptor inactivation increases survival in polymicrobial sepsis. J. Immunol. 2006;176:5616–5626. [PMC free article] [PubMed] [Google Scholar]

178. Csoka B, et al. A2A adenosine receptors and C/EBPβ are crucially required for IL-10 production by macrophages exposed to Escherichia coli. Blood. 2007;110:2685–2695. [PMC free article] [PubMed] [Google Scholar]

179. Chan ES, et al. Adenosine A2A receptors play a role in the pathogenesis of hepatic cirrhosis. Br. J. Pharmacol. 2006;148:1144–1155. [PMC free article] [PubMed] [Google Scholar]

180. Martire A, et al. Opposite effects of the A2A receptor agonist CGS21680 in the striatum of Huntington’s disease versus wild-type mice. Neurosci. Lett. 2007;417:78–83. [PubMed] [Google Scholar]

181. Obrenovitch TP, Urenjak J. Altered glutamatergic transmission in neurological disorders: from high extracellular glutamate to excessive synaptic efficacy. Prog. Neurobiol. 1997;51:39–87. [PubMed] [Google Scholar]

182. Haydon PG, Carmignoto G. Astrocyte control of synaptic transmission and neurovascular coupling. Physiol. Rev. 2006;86:1009–1031. [PubMed] [Google Scholar]

183. Jacobson KA, von Lubitz DK, Daly JW, Fredholm BB. Adenosine receptor ligands: differences with acute versus chronic treatment. Trends Pharmacol. Sci. 1996;17:108–113. [PMC free article] [PubMed] [Google Scholar]

184. Fredholm BB, Battig K, Holmen J, Nehlig A, Zvartau EE. Actions of caffeine in the brain with special reference to factors that contribute to its widespread use. Pharmacol. Rev. 1999;51:83–133. [PubMed] [Google Scholar] This is an expert and comprehensive review on the complex pharmacology of the actions of caffeine, with an emphasis on the psychostimulatory effects of caffeine in the brain.

185. de Mendonca A, Sebastiao AM, Ribeiro JA. Adenosine: does it have a neuroprotective role after all? Brain Res. Brain Res. Rev. 2000;33:258–274. [PubMed] [Google Scholar]

186. Von Lubitz DK, et al. Chronic administration of selective adenosine A1 receptor agonist or antagonist in cerebral ischemia. Eur. J. Pharmacol. 1994;256:161–167. [PMC free article] [PubMed] [Google Scholar]

187. Von Lubitz DK, et al. Reduction of postischemic brain damage and memory deficits following treatment with the selective adenosine A1 receptor agonist. Eur. J. Pharmacol. 1996;302:43–48. [PMC free article] [PubMed] [Google Scholar]

188. Burgueno J, et al. The adenosine A2A receptor interacts with the actin-binding protein α-actinin. J. Biol. Chem. 2003;278:37545–37552. [PubMed] [Google Scholar]

189. Chern Y, Lai HL, Fong JC, Liang Y. Multiple mechanisms for desensitization of A2a adenosine receptor-mediated cAMP elevation in rat pheochromocytoma PC12 cells. Mol. Pharmacol. 1993;44:950–958. [PubMed] [Google Scholar]

190. Halldner L, Lozza G, Lindstrom K, Fredholm BB. Lack of tolerance to motor stimulant effects of a selective adenosine A2A receptor antagonist. Eur. J. Pharmacol. 2000;406:345–354. [PubMed] [Google Scholar]

191. Pinna A, Fenu S, Morelli M. Motor stimulant effects of the adenosine A2A receptor antagonist SCH 58261 do not develop tolerance after repeated treatments in 6-hydroxydopamine-lesioned rats. Synapse. 2001;39:233–238. [PubMed] [Google Scholar]

192. Bastia E, et al. A crucial role for forebrain adenosine A2A receptors in amphetamine sensitization. Neuropsychopharmacology. 2005;30:891–900. [PubMed] [Google Scholar]

193. Yu L, et al. Adenosine A2A receptor antagonists exert motor and neuroprotective effects by distinct cellular mechanisms. Ann. Neurol. 2008;63:338–346. [PubMed] [Google Scholar]

194. Shen HY, et al. A critical role of the adenosine A2A receptor in extrastriatal neurons in modulating psychomotor activity as revealed by opposite phenotypes of striatum and forebrain A2A receptor knock-outs. J. Neurosci. 2008;28:2970–2975. [PMC free article] [PubMed] [Google Scholar]

195. Scammell TE, et al. Focal deletion of the adenosine A1 receptor in adult mice using an adeno-associated viral vector. J. Neurosci. 2003;23:5762–5770. [PMC free article] [PubMed] [Google Scholar]

196. Adamantidis AR, Zhang F, Aravanis AM, Deisseroth K, de Lecea L. Neural substrates of awakening probed with optogenetic control of hypocretin neurons. Nature. 2007;450:420–424. [PMC free article] [PubMed] [Google Scholar]

197. Deisseroth K, et al. Next-generation optical technologies for illuminating genetically targeted brain circuits. J. Neurosci. 2006;26:10380–10386. [PMC free article] [PubMed] [Google Scholar]

198. Lerchner W, et al. Reversible silencing of neuronal excitability in behaving mice by a genetically targeted, ivermectin-gated Cl-channel. Neuron. 2007;54:35–49. [PubMed] [Google Scholar]

199. Alexander GM, et al. Remote control of neuronal activity in transgenic mice expressing evolved G protein-coupled receptors. Neuron. 2009;63:27–39. [PMC free article] [PubMed] [Google Scholar]

200. Nawaratne V, et al. New insights into the function of M4 muscarinic acetylcholine receptors gained using a novel allosteric modulator and a DREADD (designer receptor exclusively activated by a designer drug) Mol. Pharmacol. 2008;74:1119–1131. [PubMed] [Google Scholar]

201. Aden U, et al. Aggravated brain damage after hypoxic ischemia in immature adenosine A2A knockout mice. Stroke. 2003;34:739–744. [PubMed] [Google Scholar]

202. Burnstock G, Fredholm BB, Verkhratsky A. Adenosine and ATP receptors in the brain. Curr. Top. Med. Chem. 2011;11:973–1011. [PubMed] [Google Scholar]

203. Boison D, Chen JF, Fredholm BB. Adenosine signaling and function in glial cells. Cell Death Differ. 2010;17:1071–1082. [PMC free article] [PubMed] [Google Scholar]

204. Turner CP, et al. A1 adenosine receptors mediate hypoxia-induced ventriculomegaly. Proc. Natl Acad. Sci. USA. 2003;100:11718–11722. [PMC free article] [PubMed] [Google Scholar]

205. Chabre M, le Maire M. Monomeric G-protein-coupled receptor as a functional unit. Biochemistry. 2005;44:9395–9403. [PubMed] [Google Scholar]

206. Whorton MR, et al. A monomeric G protein-coupled receptor isolated in a high-density lipoprotein particle efficiently activates its G protein. Proc. Natl Acad. Sci. USA. 2007;104:7682–7687. [PMC free article] [PubMed] [Google Scholar]

207. Ferre S, et al. Building a new conceptual framework for receptor heteromers. Nature Chem. Biol. 2009;5:131–134. [PMC free article] [PubMed] [Google Scholar]

208. Orru M, Quiroz C, Guitart X, Ferre S. Pharmacological evidence for different populations of postsynaptic adenosine A2A receptors in the rat striatum. Neuropharmacology. 2011;61:967–974. [PMC free article] [PubMed] [Google Scholar]

209. Armentero MT, et al. Past, present and future of A2A adenosine receptor antagonists in the therapy of Parkinson’s disease. Pharmacol. Ther. 2011;132:280–299. [PMC free article] [PubMed] [Google Scholar]

210. Moriyama K, Sitkovsky MV. Adenosine A2A receptor is involved in cell surface expression of A2B receptor. J. Biol. Chem. 2010;285:39271–39288. [PMC free article] [PubMed] [Google Scholar]

211. Canals M, et al. Adenosine A2A-dopamine D2 receptor-receptor heteromerization: qualitative and quantitative assessment by fluorescence and bioluminescence energy transfer. J. Biol. Chem. 2003;278:46741–46749. [PubMed] [Google Scholar]

212. Ciruela F, et al. Presynaptic control of striatal glutamatergic neurotransmission by adenosine A1-A2A receptor heteromers. J. Neurosci. 2006;26:2080–2087. [PMC free article] [PubMed] [Google Scholar]

213. Ferre S, et al. Synergistic interaction between adenosine A2A and glutamate mGlu5 receptors: implications for striatal neuronal function. Proc. Natl Acad. Sci. USA. 2002;99:11940–11945. [PMC free article] [PubMed] [Google Scholar]

214. Pin JP, et al. International Union of Basic and Clinical Pharmacology. LXVII. Recommendations for the recognition and nomenclature of G protein-coupled receptor heteromultimers. Pharmacol. Rev. 2007;59:5–13. [PubMed] [Google Scholar]

215. Yang JN, et al. Mice heterozygous for both A1 and A2A adenosine receptor genes show similarities to mice given long-term caffeine. J. Appl. Physiol. 2009;106:631–639. [PMC free article] [PubMed] [Google Scholar]

216. Ascherio A, et al. Prospective study of caffeine consumption and risk of Parkinson’s disease in men and women. Ann. Neurol. 2001;50:56–63. [PubMed] [Google Scholar]

217. Ross GW, et al. Association of coffee and caffeine intake with the risk of Parkinson disease. JAMA. 2000;283:2674–2679. [PubMed] [Google Scholar] References 216 and 217 provide strong epidemiological evidence — from the large, longitudinal study by the Honolulu Heart Program and the Health Professional Study — that caffeine consumption is inversely correlated with the risk of developing Parkinson’s disease.

218. Lindsay J, et al. Risk factors for Alzheimer’s disease: a prospective analysis from the Canadian Study of Health and Aging. Am. J. Epidemiol. 2002;156:445–453. [PubMed] [Google Scholar]

219. van Boxtel MP, Schmitt JA, Bosma H, Jolles J. The effects of habitual caffeine use on cognitive change: a longitudinal perspective. Pharmacol. Biochem. Behav. 2003;75:921–927. [PubMed] [Google Scholar]

220. Ritchie K, et al. The neuro-protective effects of caffeine: a prospective population study (the Three City Study) Neurology. 2007;69:536–545. [PubMed] [Google Scholar]

221. Higdon JV, Frei B. Coffee and health: a review of recent human research. Crit. Rev. Food Sci. Nutr. 2006;46:101–123. [PubMed] [Google Scholar]

222. Salazar-Martinez E, et al. Coffee consumption and risk for type 2 diabetes mellitus. Ann. Intern. Med. 2004;140:1–8. [PubMed] [Google Scholar]

223. Schmidt B, et al. Caffeine therapy for apnea of prematurity. N. Engl. J. Med. 2006;354:2112–2121. [PubMed] [Google Scholar]

224. Schmidt B, et al. Long-term effects of caffeine therapy for apnea of prematurity. N. Engl. J. Med. 2007;357:1893–1902. [PubMed] [Google Scholar]

225. Saaksjarvi K, et al. Prospective study of coffee consumption and risk of Parkinson’s disease. Eur. J. Clin. Nutr. 2008;62:908–915. [PubMed] [Google Scholar]

226. Powers KM, et al. Combined effects of smoking, coffee, and NSAIDs on Parkinson’s disease risk. Mov. Disord. 2008;23:88–95. [PubMed] [Google Scholar]

227. Fernandez HH, et al. Istradefylline as monotherapy for Parkinson disease: results of the 6002-US-051 trial. Parkinsonism Relat. Disord. 2010;16:16–20. [PubMed] [Google Scholar]

228. Chen JF, et al. What knock-out animals tell us about the effects of caffeine. J. Alzheimers Dis. 2010;20(Suppl. 1):17–24. [PubMed] [Google Scholar]

229. Tejani FH, Thompson RC, Iskandrian AE, McNutt BE, Franks B. Effect of caffeine on SPECT myocardial perfusion imaging during regadenoson pharmacologic stress: rationale and design of a prospective, randomized, multicenter study. J. Nucl. Cardiol. 2011;18:73–81. [PubMed] [Google Scholar]

230. Cabalag MS, et al. Recent caffeine ingestion reduces adenosine efficacy in the treatment of paroxysmal supraventricular tachycardia. Acad. Emerg. Med. 2010;17:44–49. [PubMed] [Google Scholar]

231. Pourcher E, et al. Istradefylline for Parkinson’s disease patients experiencing motor fluctuations: results of the KW-6002-US-018 study. Parkinsonism Relat. Disord. 2012;18:178–184. [PubMed] [Google Scholar]

232. Factor S, et al. A long-term study of istradefylline in subjects with fluctuating Parkinson’s disease. Parkinsonism Relat. Disord. 2010;16:423–426. [PubMed] [Google Scholar]

233. Hauser RA, et al. Study of istradefylline in patients with Parkinson’s disease on levodopa with motor fluctuations. Mov. Disord. 2008;23:2177–2185. [PubMed] [Google Scholar]

234. Rao N, et al. A study of the pharmacokinetic interaction of istradefylline, a novel therapeutic for Parkinson’s disease, and atorvastatin. J. Clin. Pharmacol. 2008;48:1092–1098. [PubMed] [Google Scholar]

235. Stacy M, et al. A 12-week, placebo-controlled study (6002-US-006) of istradefylline in Parkinson disease. Neurology. 2008;70:2233–2240. [PubMed] [Google Scholar]

236. LeWitt PA, et al. Adenosine A2A receptor antagonist istradefylline (KW-6002) reduces "off" time in Parkinson’s disease: a double-blind, randomized, multicenter clinical trial (6002-US-005) Ann. Neurol. 2008;63:295–302. [PubMed] [Google Scholar] This paper describes the design and clinical outcome of one of six of the randomized, placebo-controlled, Phase III trials of the A2A receptor antagonist istradefylline in patients with advanced Parkinson’s disease.

237. Robak T, Robak P. Purine nucleoside analogs in the treatment of rarer chronic lymphoid leukemias. Curr. Pharm. Des. 2012;18:3373–3388. [PubMed] [Google Scholar]

238. Bruns RF, Fergus JH. Allosteric enhancement of adenosine A1 receptor binding and function by 2-amino-3-benzoylthiophenes. Mol. Pharmacol. 1990;38:939–949. [PubMed] [Google Scholar]

239. Katritch V, Cherezov V, Stevens RC. Diversity and modularity of G protein-coupled receptor structures. Trends Pharmacol. Sci. 2012;33:17–27. [PMC free article] [PubMed] [Google Scholar]

240. Chen JF, et al. 8-(3-chlorostyryl)caffeine may attenuate MPTP neurotoxicity through dual actions of monoamine oxidase inhibition and A2A receptor antagonism. J. Biol. Chem. 2002;277:36040–36044. [PubMed] [Google Scholar]

241. Huang NK, et al. A new drug design targeting the adenosinergic system for Huntington’s disease. PLoS ONE. 2011;6:e20934. [PMC free article] [PubMed] [Google Scholar]

242. El-Tayeb A, et al. Nucleoside-5′-monophosphates as prodrugs of adenosine A2A receptor agonists activated by ecto-5′-nucleotidase. J. Med. Chem. 2009;52:7669–7677. [PubMed] [Google Scholar]

243. Flogel U, et al. Selective activation of adenosine A2A receptors on immune cells by a CD73-dependent prodrug suppresses joint inflammation in experimental rheumatoid arthritis. Sci. Transl. Med. 2012;4:146ra108. [PubMed] [Google Scholar]

244. Fredholm BB, Chern Y, Franco R, Sitkovsky M. Aspects of the general biology of adenosine A2A signaling. Prog. Neurobiol. 2007;83:263–276. [PubMed] [Google Scholar]

245. Allen JA, et al. Discovery of β-arrestin-biased dopamine D2 ligands for probing signal transduction pathways essential for antipsychotic efficacy. Proc. Natl Acad. Sci. USA. 2011;108:18488–18493. [PMC free article] [PubMed] [Google Scholar]

246. Langemeijer EV, Verzijl D, Dekker SJ, Ijzerman AP. Functional selectivity of adenosine A1 receptor ligands? Purinergic Signal. 2012;7:171–192. [Google Scholar]

247. Lebon G, et al. Agonist-bound adenosine A2A receptor structures reveal common features of GPCR activation. Nature. 2011;474:521–525. [PMC free article] [PubMed] [Google Scholar]

248. Xu F, et al. Structure of an agonist-bound human A2A adenosine receptor. Science. 2011;332:322–327. [PMC free article] [PubMed] [Google Scholar]

249. Jaakola VP, et al. The 2.6 angstrom crystal structure of a human A2A adenosine receptor bound to an antagonist. Science. 2008;322:1211–1217. [PMC free article] [PubMed] [Google Scholar]

250. Liu W, et al. Structural basis for allosteric regulation of GPCRs by sodium ions. Science. 2012;337:232–236. [PMC free article] [PubMed] [Google Scholar]

251. Kobilka BK. Structural insights into adrenergic receptor function and pharmacology. Trends Pharmacol. Sci. 2011;32:213–218. [PMC free article] [PubMed] [Google Scholar]

252. Hocher B. Adenosine A1 receptor antagonists in clinical research and development. Kidney Int. 2010;78:438–445. [PubMed] [Google Scholar]

253. Bennet DW, Drury AN. Further observations relating to the physiological activity of adenine compounds. J. Physiol. 1931;72:288–320. [PMC free article] [PubMed] [Google Scholar]

254. Drury AN, Szent-Gyorgyi A. The physiological activity of adenine compounds with especial reference to their action upon the mammalian heart. J. Physiol. 1929;68:213–237. [PMC free article] [PubMed] [Google Scholar]

255. Stoner HB, Green HN. Experimental limb ischaemia in man with especial reference to the role of adenosine triphosphate. Clin. Sci. 1945;5:159–175. [PubMed] [Google Scholar]

256. Massion WH. Value of high energy compounds in the treatment of shock. Am. J. Surg. 1965;110:342–347. [PubMed] [Google Scholar]

257. Yang D, et al. A new role for the A2b adenosine receptor in regulating platelet function. J. Thromb. Haemost. 2010;8:817–827. [PubMed] [Google Scholar]

258. Berne RM. Cardiac nucleotides in hypoxia: possible role in regulation of coronary blood flow. Am. J. Physiol. 1963;204:317–322. [PubMed] [Google Scholar]

259. Gerlach E, Deuticke B, Dreisbach RH. Der Nucleotid-Abbau im Herzmuskel bei Sauerstoffmangel und seine mögliche Bedeutung für die Coronardurchblutung. Naturwissenschaften. 1963;50:228–229. in German. [Google Scholar]

260. Mustafa SJ, Berne RM, Rubio R. Adenosine metabolism in cultured chick-embryo heart cells. Am. J. Physiol. 1975;228:1474–1478. [PubMed] [Google Scholar]

261. Duncker DJ, Merkus D. Exercise hyperaemia in the heart: the search for the dilator mechanism. J. Physiol. 2007;583:847–854. [PMC free article] [PubMed] [Google Scholar]

262. Lankford AR, et al. Effect of modulating cardiac A1 adenosine receptor expression on protection with ischemic preconditioning. Am. J. Physiol. Heart Circ. Physiol. 2006;290:H1469–H1473. [PubMed] [Google Scholar]

263. Yang A, Palmer AA, de Wit H. Genetics of caffeine consumption and responses to caffeine. Psychopharmacology. 2010;211:245–257. [PMC free article] [PubMed] [Google Scholar]

264. Retey JV, et al. A functional genetic variation of adenosine deaminase affects the duration and intensity of deep sleep in humans. Proc. Natl Acad. Sci. USA. 2005;102:15676–15681. [PMC free article] [PubMed] [Google Scholar]

265. Landolt HP. Sleep homeostasis: a role for adenosine in humans? Biochem. Pharmacol. 2008;75:2070–2079. [PubMed] [Google Scholar]

266. Retey JV, et al. A genetic variation in the adenosine A2A receptor gene (ADORA2A) contributes to individual sensitivity to caffeine effects on sleep. Clin. Pharmacol. Ther. 2007;81:692–698. [PubMed] [Google Scholar]

267. Childs E, et al. Association between ADORA2A and DRD2 polymorphisms and caffeine-induced anxiety. Neuropsychopharmacology. 2008;33:2791–2800. [PMC free article] [PubMed] [Google Scholar]

268. Domschke K, et al. ADORA2A gene variation, caffeine, and emotional processing: a multi-level interaction on startle reflex. Neuropsychopharmacology. 2012;37:759–769. [PMC free article] [PubMed] [Google Scholar]

269. Alsene K, Deckert J, Sand P, de Wit H. Association between A2a receptor gene polymorphisms and caffeine-induced anxiety. Neuropsychopharmacology. 2003;28:1694–1702. [PubMed] [Google Scholar]

270. Taherzadeh-Fard E, Saft C, Wieczorek S, Epplen JT, Arning L. Age at onset in Huntington’s disease: replication study on the associations of ADORA2A, HAP1 and OGG1. Neurogenetics. 2010;11:435–439. [PubMed] [Google Scholar]

271. Popat RA, et al. ADORA2A, and CYP1A2: the caffeine connection in Parkinson’s disease. Eur. J. Neurol. 2011;18:756–765. [PMC free article] [PubMed] [Google Scholar]

272. Amin N, et al. Genome-wide association analysis of coffee drinking suggests association with CYP1A1/CYP1A2 and NRCAM. Mol. Psychiatry. 2011;17:1116–1129. [PMC free article] [PubMed] [Google Scholar]

273. Hamza TH, et al. Genome-wide gene-environment study identifies glutamate receptor gene GRIN2A as a Parkinson’s disease modifier gene via interaction with coffee. PLoS Genet. 2011;7:e1002237. [PMC free article] [PubMed] [Google Scholar]

274. Tantry US, et al. First analysis of the relation between CYP2C19 genotype and pharmacodynamics in patients treated with ticagrelor versus clopidogrel: the ONSET/OFFSET and RESPOND genotype studies. Circ. Cardiovasc. Genet. 2010;3:556–566. [PubMed] [Google Scholar]

275. Elzein E, Zablocki J. A1 adenosine receptor agonists and their potential therapeutic applications. Expert Opin. Investig. Drugs. 2008;17:1901–1910. [PubMed] [Google Scholar]

276. Pinna A. Novel investigational adenosine A2A receptor antagonists for Parkinson’s disease. Expert Opin. Investig. Drugs. 2009;18:1619–1631. [PubMed] [Google Scholar]

277. Brown R, et al. Abolished tubuloglomerular feedback and increased plasma renin in adenosine A1 receptor-deficient mice. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2001;281:R1362–R1367. [PubMed] [Google Scholar]

278. Tawfik HE, Schnermann J, Oldenburg PJ, Mustafa SJ. Role of A1 adenosine receptors in regulation of vascular tone. Am. J. Physiol. Heart Circ. Physiol. 2005;288:H1411–H1416. [PubMed] [Google Scholar]

279. Wang Y, Yang JN, Arner A, Boels PJ, Fredholm BB. Adenosine A1 receptors and vascular reactivity. Acta Physiol. 2010;199:211–220. [PubMed] [Google Scholar]

280. Hua X, et al. Involvement of A1 adenosine receptors and neural pathways in adenosine-induced bronchoconstriction in mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 2007;293:L25–L32. [PubMed] [Google Scholar]

281. Johansson SM, et al. A1 receptor deficiency causes increased insulin and glucagon secretion in mice. Biochem. Pharmacol. 2007;74:1628–1635. [PubMed] [Google Scholar]

282. Yang JN, et al. Sex differences in mouse heart rate and body temperature and in their regulation by adenosine A1 receptors. Acta Physiol. 2007;190:63–75. [PubMed] [Google Scholar]

283. Kara FM, et al. Adenosine A1 receptors regulate bone resorption in mice: adenosine A1 receptor blockade or deletion increases bone density and prevents ovariectomy-induced bone loss in adenosine A1 receptor-knockout mice. Arthritis Rheum. 2010;62:534–541. [PMC free article] [PubMed] [Google Scholar]

284. Stenberg D, et al. Sleep and its homeostatic regulation in mice lacking the adenosine A1 receptor. J. Sleep Res. 2003;12:283–290. [PubMed] [Google Scholar]

285. Oishi Y, Huang ZL, Fredholm BB, Urade Y, Hayaishi O. Adenosine in the tuberomammillary nucleus inhibits the histaminergic system via A1 receptors and promotes non-rapid eye movement sleep. Proc. Natl Acad. Sci. USA. 2008;105:19992–19997. [PMC free article] [PubMed] [Google Scholar]

286. Wu WP, et al. Increased nociceptive response in mice lacking the adenosine A1 receptor. Pain. 2005;113:395–404. [PubMed] [Google Scholar]

287. Schulte G, et al. Adenosine A receptors are necessary for protection of the murine heart by remote, delayed adaptation to ischaemia. Acta Physiol. Scand. 2004;182:133–143. [PubMed] [Google Scholar]

288. Lappas CM, Rieger JM, Linden J. A2A adenosine receptor induction inhibits IFN-γ production in murine CD4+ T cells. J. Immunol. 2005;174:1073–1080. [PubMed] [Google Scholar]

289. Carman AJ, Mills JH, Krenz A, Kim DG, Bynoe MS. Adenosine receptor signaling modulates permeability of the blood-brain barrier. J. Neurosci. 2011;31:13272–13280. [PMC free article] [PubMed] [Google Scholar]

290. Chen X, Lan X, Roche I, Liu R, Geiger JD. Caffeine protects against MPTP-induced blood-brain barrier dysfunction in mouse striatum. J. Neurochem. 2008;107:1147–1157. [PMC free article] [PubMed] [Google Scholar]

291. Chen X, Gawryluk JW, Wagener JF, Ghribi O, Geiger JD. Caffeine blocks disruption of blood brain barrier in a rabbit model of Alzheimer’s disease. J. Neuroinflamm. 2008;5:12. [PMC free article] [PubMed] [Google Scholar]

292. Morrison RR, Talukder MA, Ledent C, Mustafa SJ. Cardiac effects of adenosine in A2A receptor knockout hearts: uncovering A2B receptors. Am. J. Physiol. Heart Circ. Physiol. 2002;282:H437–H444. [PubMed] [Google Scholar]

293. Montesinos MC, et al. Adenosine promotes wound healing and mediates angiogenesis in response to tissue injury via occupancy of A2A receptors. Am. J. Pathol. 2002;160:2009–2018. [PMC free article] [PubMed] [Google Scholar]

294. Tilley SL, Wagoner VA, Salvatore CA, Jacobson MA, Koller BH. Adenosine and inosine increase cutaneous vasopermeability by activating A3 receptors on mast cells. J. Clin. Invest. 2000;105:361–367. [PMC free article] [PubMed] [Google Scholar]

295. Hua X, et al. Adenosine induces airway hyperresponsiveness through activation of A3 receptors on mast cells. J. Allergy Clin. Immunol. 2008;122:107–113.e7. [PMC free article] [PubMed] [Google Scholar]

296. Li L, et al. Dendritic cells tolerized with adenosine A2AR agonist attenuate acute kidney injury. J. Clin. Invest. 2012;122:3931–3942. [PMC free article] [PubMed] [Google Scholar]

297. Field JJ, et al. Sickle cell vaso-occlusion causes activation of iNKT cells that is decreased by the adenosine A2A receptor agonist regadenoson. Blood. 2013 Feb 1; doi:10.1182/blood-2012-11-465963. [PMC free article] [PubMed] [Google Scholar]


Page 2

Examples of ongoing or recently completed Phase lib–III clinical trials targeting adenosine receptors

Type of compoundPharmacology*
Ki
Purpose or name of studyStatusClinicalTrials.
gov identifer
Refs

What is the term for a medication that blocks a receptor in the body?

A1: 77 nM
A2A: 0.5 nM
A222B: notdetermined

A3: 45 nM

Protection from liver ischaemia
following liver surgery
Ongoing{"type":"clinical-trial","attrs":{"text":"NCT00760708","term_id":"NCT00760708"}}NCT007607082,18
Postconditioning after STEMIOngoing{"type":"clinical-trial","attrs":{"text":"NCT00284323","term_id":"NCT00284323"}}NCT00284323
Pretreatment before stentingOngoing{"type":"clinical-trial","attrs":{"text":"NCT00612521","term_id":"NCT00612521"}}NCT00612521
Does intradermal adenosine
release VEGF and cytokines?
Suspendedfor

re-evaluation

{"type":"clinical-trial","attrs":{"text":"NCT00580905","term_id":"NCT00580905"}}NCT00580905

What is the term for a medication that blocks a receptor in the body?

A1: >10,000 nM
A2A: 290 nM
A2B: >10,000 nM
A3: >10,000 nM
Is regadenoson superior toadenosine for myocardial

perfusion imaging?

Completed{"type":"clinical-trial","attrs":{"text":"NCT00208312","term_id":"NCT00208312"}}NCT0020831218,146,
  191
Myocardial perfusion magneticresonance imaging using

regadenoson

Completed{"type":"clinical-trial","attrs":{"text":"NCT00881218","term_id":"NCT00881218"}}NCT00881218
ASPECT study: effectiveness ofapadenoson in SPECT imaging

compared to adenosine

Completed{"type":"clinical-trial","attrs":{"text":"NCT01085201","term_id":"NCT01085201"}}NCT01085201
Regadenoson approved for
treatment of sickle cell anaemia
Recruiting{"type":"clinical-trial","attrs":{"text":"NCT01566890","term_id":"NCT01566890"}}NCT01566890
Regadenoson blood flow in
type 1 diabetes (RABIT1D)
Completed{"type":"clinical-trial","attrs":{"text":"NCT01019486","term_id":"NCT01019486"}}NCT01019486

What is the term for a medication that blocks a receptor in the body?

No data availableAnalgesic effect of {"type":"entrez-nucleotide","attrs":{"text":"GW493838","term_id":"289072615","term_text":"GW493838"}}GW493838in postherpetic neuralgia or

peripheral nerve injury

Discontinued{"type":"clinical-trial","attrs":{"text":"NCT00376454","term_id":"NCT00376454"}}NCT003764542,18,275
Tolerability and safety ofINO 8875 in glaucoma and

ocular hypertension

Discontinued{"type":"clinical-trial","attrs":{"text":"NCT01123785","term_id":"NCT01123785"}}NCT01123785

What is the term for a medication that blocks a receptor in the body?

CF101:
A1: 51 nM
A2A: 2,900 nM
A2B: 11,000 nM
A3:1.8 nMC3F102:

A1: 220 nM


A2A: 5,360 nM
A2B: >10,000 nM
A3: 1.4 nM
Safety and efficacy of CF101
in psoriasis
Completed{"type":"clinical-trial","attrs":{"text":"NCT00428974","term_id":"NCT00428974"}}NCT004289742,18,37,
146,147
Safety and efficacy of CF101
in rheumatoid arthritis
Completed{"type":"clinical-trial","attrs":{"text":"NCT00556894","term_id":"NCT00556894"}}NCT00556894
Safety and efficacy of CF102
in liver cancer
Ongoing{"type":"clinical-trial","attrs":{"text":"NCT00790218","term_id":"NCT00790218"}}NCT00790218

What is the term for a medication that blocks a receptor in the body?

A1: 10,700 nM
A2A: 23,400 nM
A2B: 33,800 nM
A3:13,300 nM
Treatment of apnoea of
prematurity and dose study
Currentlyrecruiting

participants

{"type":"clinical-trial","attrs":{"text":"NCT01408173","term_id":"NCT01408173"}}NCT01408173  2,18,
223,224
Treatment of apnoea of
prematurity and dose study
Currentlyrecruiting

participants

{"type":"clinical-trial","attrs":{"text":"NCT01349205","term_id":"NCT01349205"}}NCT01349205
Cognitive long-term effects of
caffeine in premature infants
Currentlyrecruiting

participants

{"type":"clinical-trial","attrs":{"text":"NCT00809055","term_id":"NCT00809055"}}NCT00809055
Caffeine for motor manifestations
of Parkinson’s disease
Completed{"type":"clinical-trial","attrs":{"text":"NCT01190735","term_id":"NCT01190735"}}NCT01190735
Caffeine for excessive daytime
somnolence in Parkinson’s disease
Completed{"type":"clinical-trial","attrs":{"text":"NCT00459420","term_id":"NCT00459420"}}NCT00459420

What is the term for a medication that blocks a receptor in the body?

Tonapofylline:
A1: 7.4 nM
A2A: 6,410 nM
A2B: 90 nM
A3: >10,000 nMRolofylline:

A1: 0.72 nM


A2A: 108 nM
A2B: 296 nM
A3:4390 nM
Safety and tolerability ofintravenously administeredtonapofylline in individuals withacute decompensated heartfailure and renal insufficiency

(TRIDENT-1)

Discontinued{"type":"clinical-trial","attrs":{"text":"NCT00709865","term_id":"NCT00709865"}}NCT007098652,18,
21,22
Effect of rolofylline on heart andrenal function in acute heart

failure

Discontinued{"type":"clinical-trial","attrs":{"text":"NCT00328692","term_id":"NCT00328692"}}NCT00328692
Effect of rolofylline on heart andrenal function in acute heart

failure

Discontinued{"type":"clinical-trial","attrs":{"text":"NCT00354458","term_id":"NCT00354458"}}NCT00354458

What is the term for a medication that blocks a receptor in the body?

Istradefylline:
A1: 841 nM
A2A: 12 nM
A2B: >10,000 nM
A3:4470 nMPreladenant:

A1: >1,000 nM


A2A: 0.9 nM
A2B: >1,000 nM
A3: >1,000 nMSYN115:A1: 228.4 nMAm: 0.38 nMA2B: not available

A3: not available

Efficacy of istradefylline inincreasing sleep in patients with

advanced Parkinson’s disease

Ongoing{"type":"clinical-trial","attrs":{"text":"NCT00955526","term_id":"NCT00955526"}}NCT009555262,18,138,
142,234,
238–243,276
Effect of preladenant in early
Parkinson’s disease
Ongoing{"type":"clinical-trial","attrs":{"text":"NCT01155479","term_id":"NCT01155479"}}NCT01155479
Effect of preladenant on ‘off-time’in moderate to severe Parkinson’s

disease

Ongoing{"type":"clinical-trial","attrs":{"text":"NCT01155466","term_id":"NCT01155466"}}NCT01155466
fMRI-aided study of SYN115 onbehaviour and brain activity in

cocaine addicts

Ongoing{"type":"clinical-trial","attrs":{"text":"NCT00783276","term_id":"NCT00783276"}}NCT00783276
Safety and efficacy studyof SYN115 in patients withParkinson’s disease using l-DOPA

to treat end of dose wearing off

Completed{"type":"clinical-trial","attrs":{"text":"NCT01283594","term_id":"NCT01283594"}}NCT01283594
Long-term safety study ofistradefylline in patients

with Parkinson’s disease‡

Completed{"type":"clinical-trial","attrs":{"text":"NCT00957203","term_id":"NCT00957203"}}NCT00957203
Active-controlled extension
study to {"type":"entrez-protein","attrs":{"text":"P04938","term_id":"978128202","term_text":"P04938"}}P04938 and {"type":"entrez-protein","attrs":{"text":"P07037","term_id":"129422","term_text":"P07037"}}P07037
({"type":"entrez-protein","attrs":{"text":"P06153","term_id":"133375","term_text":"P06153"}}P06153 AM3)
Recruiting
participants
{"type":"clinical-trial","attrs":{"text":"NCT01215227","term_id":"NCT01215227"}}NCT01215227
Placebo-controlled study ofpreladenant in participantswith moderate to severe

Parkinsons disease ({"type":"entrez-protein","attrs":{"text":"P07037","term_id":"129422","term_text":"P07037"}}P07037 AM3)

Recruiting
participants
{"type":"clinical-trial","attrs":{"text":"NCT01227265","term_id":"NCT01227265"}}NCT01227265

What is the term for a medication that blocks a receptor in the body?

Protection after cardiac
bypass surgery
Currentlyrecruiting

participants

{"type":"clinical-trial","attrs":{"text":"NCT01295567","term_id":"NCT01295567"}}NCT0129556718,66
Effects on circulating adenosine
levels in relation to genetics
Currentlyrecruiting

participants

{"type":"clinical-trial","attrs":{"text":"NCT00760708","term_id":"NCT00760708"}}NCT00760708
Supplementation withprednisolone in rheumatoid

arthritis

Currentlyrecruiting

participants

{"type":"clinical-trial","attrs":{"text":"NCT01369745","term_id":"NCT01369745"}}NCT01369745