Skip to main content
Log in

Orexin/Hypocretin Based Pharmacotherapies for the Treatment of Addiction: DORA or SORA?

  • Review Article
  • Published:
CNS Drugs Aims and scope Submit manuscript

Abstract

Addiction is a chronic relapsing disorder which presents a significant global health burden and unmet medical need. The orexin/hypocretin system is an attractive potential therapeutic target as demonstrated by the successful clinical trials of antagonist medications like Suvorexant for insomnia. It is composed of two neuropeptides, orexin-A and orexin-B and two excitatory and promiscuous G-protein coupled receptors, OX1 and OX2. Orexins are known to have a variety of functions, most notably in regulating arousal, appetite and reward. The orexins have been shown to have a role in mediating the effects of several drugs of abuse, such as cocaine, morphine and alcohol via projections to key brain regions such as the ventral tegmental area, nucleus accumbens and prefrontal cortex. However, it has not yet been demonstrated whether the dual orexin receptor antagonists (DORAs) under development for insomnia are ideal drugs for the treatment of addiction. The question of whether to use a DORA or single orexin receptor antagonist (SORA) for the treatment of addiction is a key question that will need to be answered in order to maximize the clinical utility of orexin receptor antagonists. This review will examine the role of the orexin/hypocretin system in addiction, orexin-based pharmacotherapies under development and factors affecting the selection of one or both orexin receptors as drug targets for the treatment of addiction.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

Notes

  1. The number needed to treat (NNT) is calculated as 1/risk difference. An increase in abstinence rate from 25 to 38 % gives a risk difference of 13 % and an NNT of 8.

References

  1. World Health Organization. International statistical classification of diseases and related health problems (10th revision). Geneva: World Health Organization; 1992.

    Google Scholar 

  2. American Psychiatric Association. Diagnostic and statistical manual of mental disorders: DSM-5. vol (DSM-5™). Arlington: American Psychiatric Association; 2013.

  3. Degenhardt L, Whiteford HA, Ferrari AJ, Baxter AJ, Charlson FJ, Hall WD, et al. Global burden of disease attributable to illicit drug use and dependence: findings from the Global Burden of Disease Study 2010. Lancet. 2013;382(9904):1564–74. doi:10.1016/S0140-6736(13)61530-5.

    PubMed  Google Scholar 

  4. Whiteford HA, Degenhardt L, Rehm J, Baxter AJ, Ferrari AJ, Erskine HE, et al. Global burden of disease attributable to mental and substance use disorders: findings from the Global Burden of Disease Study 2010. Lancet. 2013;382(9904):1575–86. doi:10.1016/S0140-6736(13)61611-6.

    PubMed  Google Scholar 

  5. Pierce RC, O’Brien CP, Kenny PJ, Vanderschuren LJMJ. Rational development of addiction pharmacotherapies: successes, failures, and prospects. Cold Spring Harb Perspect Med 2012;2(6). doi:10.1101/cshperspect.a012880.

  6. Johnson BA, Ait-Daoud N, Wang X, et al. Topiramate for the treatment of cocaine addiction: a randomized clinical trial. JAMA Psychiatry. 2013;70(12):1338–46. doi:10.1001/jamapsychiatry.2013.2295.

    CAS  PubMed  Google Scholar 

  7. Mason BJ, Crean R, Goodell V, Light JM, Quello S, Shadan F, et al. A proof-of-concept randomized controlled study of gabapentin: effects on cannabis use, withdrawal and executive function deficits in cannabis-dependent adults. Neuropsychopharmacology. 2012;37(7):1689–98. doi:10.1038/npp.2012.14.

    CAS  PubMed Central  PubMed  Google Scholar 

  8. Nutt DJ, King LA, Phillips LD. Drug harms in the UK: a multicriteria decision analysis. Lancet. 2010;376(9752):1558–65. doi:10.1016/s0140-6736(10)61462-6.

    PubMed  Google Scholar 

  9. Spanagel R, Vengeliene V, Jandeleit B, Fischer W-N, Grindstaff K, Zhang X, et al. Acamprosate produces its anti-relapse effects via calcium. Neuropsychopharmacology. 2014;39(4):783–91. doi:10.1038/npp.2013.264.

    CAS  PubMed  Google Scholar 

  10. Dupouy J, Fournier J-P, Jouanjus É, Palmaro A, Poutrain J-C, Oustric S, et al. Baclofen for alcohol dependence in France: incidence of treated patients and prescription patterns—a cohort study. Eur Neuropsychopharmacol. 2014;24(2):192–9. doi:10.1016/j.euroneuro.2013.09.008.

    CAS  PubMed  Google Scholar 

  11. Liu J, Wang L-N. Baclofen for alcohol withdrawal. Cochrane Database Syst Rev. 2013;2013(2). doi:10.1002/14651858.CD008502.pub3.

  12. de Lecea L, Kilduff TS, Peyron C, Gao X-B, Foye PE, Danielson PE, et al. The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity. Proc Natl Acad Sci. 1998;95(1):322–7. doi:10.1073/pnas.95.1.322.

    PubMed Central  PubMed  Google Scholar 

  13. Sakurai T, Amemiya A, Ishii M, Matsuzaki I, Chemelli RM, Tanaka H, et al. Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell. 1998;92(4):573–85. doi:10.1016/s0092-8674(00)80949-6.

    CAS  PubMed  Google Scholar 

  14. Gotter AL, Webber AL, Coleman PJ, Renger JJ, Winrow CJ. International Union of Basic and Clinical Pharmacology. LXXXVI. Orexin receptor function, nomenclature and pharmacology. Pharmacol Rev. 2012;64(3):389–420. doi:10.1124/pr.111.005546.

    CAS  PubMed  Google Scholar 

  15. Sakurai T, Moriguchi T, Furuya K, Kajiwara N, Nakamura T, Yanagisawa M, et al. Structure and function of human prepro-orexin gene. J Biol Chem. 1999;274(25):17771–6. doi:10.1074/jbc.274.25.17771.

    CAS  PubMed  Google Scholar 

  16. Heifetz A, Barker O, Morris GB, Law RJ, Slack M, Biggin PC. Toward an understanding of agonist binding to human Orexin-1 and Orexin-2 receptors with G-protein-coupled receptor modeling and site-directed mutagenesis. Biochemistry. 2013;52(46):8246–60. doi:10.1021/bi401119m.

    CAS  PubMed Central  PubMed  Google Scholar 

  17. Xu TR, Ward RJ, Pediani JD, Milligan G. The orexin OX1 receptor exists predominantly as a homodimer in the basal state: potential regulation of receptor organization by both agonist and antagonist ligands. Biochem J. 2011;439(1):171–83. doi:10.1042/bj20110230.

    CAS  PubMed  Google Scholar 

  18. Jäntti MH, Mandrika I, Kukkonen JP. Human orexin/hypocretin receptors form constitutive homo- and heteromeric complexes with each other and with human CB1 cannabinoid receptors. Biochem Biophys Res Commun. 2014;445(2):486–90. doi:10.1016/j.bbrc.2014.02.026.

    PubMed  Google Scholar 

  19. Ward RJ, Pediani JD, Milligan G. Heteromultimerization of cannabinoid CB1 receptor and orexin OX1 receptor generates a unique complex in which both protomers are regulated by orexin A. J Biol Chem. 2011;286(43):37414–28. doi:10.1074/jbc.M111.287649.

    CAS  PubMed Central  PubMed  Google Scholar 

  20. Chou TC, Lee CE, Lu J, Elmquist JK, Hara J, Willie JT, et al. Orexin (hypocretin) neurons contain dynorphin. J Neurosci. 2001;21(19):RC168.

    CAS  PubMed  Google Scholar 

  21. Furutani N, Hondo M, Kageyama H, Tsujino N, Mieda M, Yanagisawa M, et al. Neurotensin co-expressed in orexin-producing neurons in the lateral hypothalamus plays an important role in regulation of sleep/wakefulness states. PLoS One. 2013;8(4):e62391. doi:10.1371/journal.pone.0062391.

    CAS  PubMed Central  PubMed  Google Scholar 

  22. Rosin DL, Weston MC, Sevigny CP, Stornetta RL, Guyenet PG. Hypothalamic orexin (hypocretin) neurons express vesicular glutamate transporters VGLUT1 or VGLUT2. J Comp Neurol. 2003;465(4):593–603. doi:10.1002/cne.10860.

    CAS  PubMed  Google Scholar 

  23. Henny P, Brischoux F, Mainville L, Stroh T, Jones BE. Immunohistochemical evidence for synaptic release of glutamate from orexin terminals in the locus coeruleus. Neuroscience. 2010;169(3):1150–7. doi:10.1016/j.neuroscience.2010.06.003.

    CAS  PubMed Central  PubMed  Google Scholar 

  24. Harthoorn LF, Sañé A, Nethe M, Heerikhuize JJ. Multi-transcriptional profiling of melanin-concentrating hormone and orexin-containing neurons. Cell Mol Neurobiol. 2005;25(8):1209–23. doi:10.1007/s10571-005-8184-8.

    PubMed  Google Scholar 

  25. Ammoun S, Holmqvist T, Shariatmadari R, Oonk HB, Detheux M, Parmentier M, et al. Distinct recognition of OX1 and OX2 receptors by orexin peptides. J Pharmacol Exp Ther. 2003;305(2):507–14. doi:10.1124/jpet.102.048025.

    CAS  PubMed  Google Scholar 

  26. Holmqvist T, Johansson L, Östman M, Ammoun S, Åkerman KEO, Kukkonen JP. OX1 orexin receptors couple to adenylyl cyclase regulation via multiple mechanisms. J Biol Chem. 2005;280(8):6570–9. doi:10.1074/jbc.M407397200.

    CAS  PubMed  Google Scholar 

  27. Zhu Y, Miwa Y, Yamanaka A, Yada T, Shibahara M, Abe Y, et al. Orexin receptor type-1 couples exclusively to pertussis toxin-insensitive G-proteins, while orexin receptor type-2 couples to both pertussis toxin-sensitive and -insensitive G-proteins. J Pharmacol Sci. 2003;92(3):259–66. doi:10.1254/jphs.92.259.

    CAS  PubMed  Google Scholar 

  28. Smart D, Jerman JC, Brough SJ, Rushton SL, Murdock PR, Jewitt F, et al. Characterization of recombinant human orexin receptor pharmacology in a Chinese hamster ovary cell-line using FLIPR. Br J Pharmacol. 1999;128(1):1–3. doi:10.1038/sj.bjp.0702780.

    CAS  PubMed Central  PubMed  Google Scholar 

  29. Magga J, Bart G, Oker-Blom C, Kukkonen JP, Åkerman KEO, Näsman J. Agonist potency differentiates G protein activation and Ca2+ signalling by the orexin receptor type 1. Biochem Pharmacol. 2006;71(6):827–36. doi:10.1016/j.bcp.2005.12.021.

    CAS  PubMed  Google Scholar 

  30. Johansson L, Ekholm ME, Kukkonen JP. Regulation of OX1 orexin/hypocretin receptor-coupling to phospholipase C by Ca2+ influx. Br J Pharmacol. 2007;150(1):97–104. doi:10.1038/sj.bjp.0706959.

    CAS  PubMed Central  PubMed  Google Scholar 

  31. Lund P-E, Shariatmadari R, Uustare A, Detheux M, Parmentier M, Kukkonen JP, et al. The orexin OX1 receptor activates a novel Ca2+ influx pathway necessary for coupling to phospholipase C. J Biol Chem. 2000;275(40):30806–12. doi:10.1074/jbc.M002603200.

    CAS  PubMed  Google Scholar 

  32. Näsman J, Bart G, Larsson K, Louhivuori L, Peltonen H, Åkerman KEO. The orexin OX1 receptor regulates Ca2+ entry via diacylglycerol-activated channels in differentiated neuroblastoma cells. J Neurosci. 2006;26(42):10658–66. doi:10.1523/jneurosci.2609-06.2006.

    PubMed  Google Scholar 

  33. Peltonen HM, Magga JM, Bart G, Turunen PM, Antikainen MSH, Kukkonen JP, et al. Involvement of TRPC3 channels in calcium oscillations mediated by OX1 orexin receptors. Biochem Biophys Res Commun. 2009;385(3):408–12. doi:10.1016/j.bbrc.2009.05.077.

    CAS  PubMed  Google Scholar 

  34. Jäntti MH, Putula J, Somerharju P, Frohman MA, Kukkonen JP. OX1 orexin/hypocretin receptor activation of phospholipase D. Br J Pharmacol. 2012;165(4b):1109–23. doi:10.1111/j.1476-5381.2011.01565.x.

    PubMed Central  PubMed  Google Scholar 

  35. Turunen PM, Ekholm ME, Somerharju P, Kukkonen JP. Arachidonic acid release mediated by OX1 orexin receptors. Br J Pharmacol. 2010;159(1):212–21. doi:10.1111/j.1476-5381.2009.00535.x.

    CAS  PubMed Central  PubMed  Google Scholar 

  36. Turunen PM, Jäntti MH, Kukkonen JP. OX1 orexin/hypocretin receptor signaling through arachidonic acid and endocannabinoid release. Mol Pharmacol. 2012;82(2):156–67. doi:10.1124/mol.112.078063.

    CAS  PubMed  Google Scholar 

  37. Tang J, Chen J, Ramanjaneya M, Punn A, Conner AC, Randeva HS. The signalling profile of recombinant human orexin-2 receptor. Cell Signal. 2008;20(9):1651–61. doi:10.1016/j.cellsig.2008.05.010.

    CAS  PubMed  Google Scholar 

  38. Karteris E, Randeva HS, Grammatopoulos DK, Jaffe RB, Hillhouse EW. Expression and coupling characteristics of the CRH and orexin type 2 receptors in human fetal adrenals. J Clin Endocrinol Metab. 2001;86(9):4512–9. doi:10.1210/jc.86.9.4512.

    CAS  PubMed  Google Scholar 

  39. Urbańska A, Sokołowska P, Woldan-Tambor A, Biegańska K, Brix B, Jöhren O, et al. Orexins/hypocretins acting at Gi protein-coupled OX2 receptors inhibit cyclic AMP synthesis in the primary neuronal cultures. J Mol Neurosci. 2012;46(1):10–7. doi:10.1007/s12031-011-9526-2.

    PubMed Central  PubMed  Google Scholar 

  40. Hoang QV, Bajic D, Yanagisawa M, Nakajima S, Nakajima Y. Effects of orexin (hypocretin) on GIRK channels. J Neurophysiol. 2003;90(2):693–702. doi:10.1152/jn.00001.2003.

    CAS  PubMed  Google Scholar 

  41. Dalrymple MB, Jaeger WC, Eidne KA, Pfleger KDG. Temporal profiling of orexin receptor-arrestin-ubiquitin complexes reveals differences between receptor subtypes. J Biol Chem. 2011;286(19):16726–33. doi:10.1074/jbc.M111.223537.

    CAS  PubMed Central  PubMed  Google Scholar 

  42. Jaeger WC, Seeber RM, Eidne KA, Pfleger KDG. Molecular determinants of orexin receptor-arrestin-ubiquitin complex formation. Br J Pharmacol. 2014;171(2):364–74. doi:10.1111/bph.12481.

    CAS  PubMed  Google Scholar 

  43. Darker JG, Porter RA, Eggleston DS, Smart D, Brough SJ, Sabido-David C, et al. Structure-activity analysis of truncated orexin-A analogues at the orexin-1 receptor. Bioorg Med Chem Lett. 2001;11(5):737–40. doi:10.1016/S0960-894X(01)00043-9.

    CAS  PubMed  Google Scholar 

  44. Lang M, Söll RM, Dürrenberger F, Dautzenberg FM, Beck-Sickinger AG. Structure-activity studies of orexin A and orexin B at the human orexin 1 and orexin 2 receptors led to orexin 2 receptor selective and orexin 1 receptor preferring ligands. J Med Chem. 2004;47(5):1153–60. doi:10.1021/jm030982t.

    CAS  PubMed  Google Scholar 

  45. German NA, Decker AM, Gilmour BP, Thomas BF, Zhang Y. Truncated orexin peptides: structure-activity relationship studies. ACS Med Chem Lett. 2013;4(12):1224–7. doi:10.1021/ml400333a.

    CAS  PubMed Central  PubMed  Google Scholar 

  46. Asahi S, Egashira S-I, Matsuda M, Iwaasa H, Kanatani A, Ohkubo M, et al. Development of an orexin-2 receptor selective agonist, [Ala11, d-Leu15]orexin-B. Bioorg Med Chem Lett. 2003;13(1):111–3. doi:10.1016/S0960-894X(02)00851-X.

    CAS  PubMed  Google Scholar 

  47. Putula J, Turunen PM, Johansson L, Näsman J, Ra R, Korhonen L, et al. Orexin/hypocretin receptor chimaeras reveal structural features important for orexin peptide distinction. FEBS Lett. 2011;585(9):1368–74. doi:10.1016/j.febslet.2011.04.020.

    CAS  PubMed  Google Scholar 

  48. Porter RA, Chan WN, Coulton S, Johns A, Hadley MS, Widdowson K, et al. 1,3-Biarylureas as selective non-peptide antagonists of the orexin-1 receptor. Bioorg Med Chem Lett. 2001;11(14):1907–10. doi:10.1016/S0960-894X(01)00343-2.

    CAS  PubMed  Google Scholar 

  49. Smart D, Sabido-David C, Brough SJ, Jewitt F, Johns A, Porter RA, et al. SB-334867-A: the first selective orexin-1 receptor antagonist. Br J Pharmacol. 2001;132(6):1179–82. doi:10.1038/sj.bjp.0703953.

    CAS  PubMed Central  PubMed  Google Scholar 

  50. Langmead CJ, Jerman JC, Brough SJ, Scott C, Porter RA, Herdon HJ. Characterisation of the binding of [3H]-SB-674042, a novel nonpeptide antagonist, to the human orexin-1 receptor. Br J Pharmacol. 2004;141(2):340–6. doi:10.1038/sj.bjp.0705610.

    CAS  PubMed Central  PubMed  Google Scholar 

  51. Steiner MA, Gatfield J, Brisbare-Roch C, Dietrich H, Treiber A, Jenck F, et al. Discovery and characterization of ACT-335827, an orally available, brain penetrant orexin receptor type 1 selective antagonist. ChemMedChem. 2013;8(6):898–903. doi:10.1002/cmdc.201300003.

    CAS  PubMed  Google Scholar 

  52. Steiner MA, Sciarretta C, Pasquali A, Jenck F. The selective orexin receptor 1 antagonist ACT-335827 in a rat model of diet-induced obesity associated with metabolic syndrome. Front Pharmacol. 2013;4. doi:10.3389/fphar.2013.00165.

  53. Hirose M, Egashira S-I, Goto Y, Hashihayata T, Ohtake N, Iwaasa H, et al. N-acyl 6,7-dimethoxy-1,2,3,4-tetrahydroisoquinoline: the first orexin-2 receptor selective non-peptidic antagonist. Bioorg Med Chem Lett. 2003;13(24):4497–9. doi:10.1016/j.bmcl.2003.08.038.

    CAS  PubMed  Google Scholar 

  54. McAtee LC, Sutton SW, Rudolph DA, Li X, Aluisio LE, Phuong VK, et al. Novel substituted 4-phenyl-[1,3]dioxanes: potent and selective orexin receptor 2 (OX2R) antagonists. Bioorg Med Chem Lett. 2004;14(16):4225–9. doi:10.1016/j.bmcl.2004.06.032.

    CAS  PubMed  Google Scholar 

  55. Malherbe P, Borroni E, Gobbi L, Knust H, Nettekoven M, Pinard E, et al. Biochemical and behavioural characterization of EMPA, a novel high-affinity, selective antagonist for the OX2 receptor. Br J Pharmacol. 2009;156(8):1326–41. doi:10.1111/j.1476-5381.2009.00127.x.

    CAS  PubMed Central  PubMed  Google Scholar 

  56. Fitch TE, Benvenga MJ, Jesudason CD, Zink C, Vandergriff AB, Menezes M, et al. LSN2424100: a novel, potent orexin-2 receptor antagonist with selectivity over orexin-1 receptors and activity in an animal model predictive of antidepressant-like efficacy. Front Neurosci. 2014;8. doi:10.3389/fnins.2014.00005.

  57. Brisbare-Roch C, Dingemanse J, Koberstein R, Hoever P, Aissaoui H, Flores S, et al. Promotion of sleep by targeting the orexin system in rats, dogs and humans. Nat Med. 2007;13(2):150–5. doi:10.1038/nm1544.

    CAS  PubMed  Google Scholar 

  58. Hoever P, de Haas S, Winkler J, Schoemaker RC, Chiossi E, van Gerven J, et al. Orexin receptor antagonism, a new sleep-promoting paradigm: an ascending single-dose study with almorexant. Clin Pharmacol Ther. 2010;87(5):593–600. doi:10.1038/clpt.2010.19.

    CAS  PubMed  Google Scholar 

  59. Hoever P, de Haas SL, Dorffner G, Chiossi E, van Gerven JM, Dingemanse J. Orexin receptor antagonism: an ascending multiple-dose study with almorexant. J Psychopharmacol. 2012;26(8):1071–80. doi:10.1177/0269881112448946.

    PubMed  Google Scholar 

  60. Hoever P, Dorffner G, Benes H, Penzel T, Danker-Hopfe H, Barbanoj MJ, et al. Orexin receptor antagonism, a new sleep-enabling paradigm: a proof-of-concept clinical trial. Clin Pharmacol Ther. 2012;91(6):975–85. doi:10.1038/clpt.2011.370.

    CAS  PubMed Central  PubMed  Google Scholar 

  61. Heifetz A, Morris GB, Biggin PC, Barker O, Fryatt T, Bentley J, et al. Study of human Orexin-1 and -2 G-protein-coupled receptors with novel and published antagonists by modeling, molecular dynamics simulations, and site-directed mutagenesis. Biochemistry. 2012;51(15):3178–97. doi:10.1021/bi300136h.

    CAS  PubMed  Google Scholar 

  62. Actelion Pharmaceuticals, GlaxoSmithKline. Actelion and GSK discontinue clinical development of almorexant. Allschwil/Basel, Switzerland and London. http://www.actelion.com; 2011. Accessed 21 Dec 2013.

  63. Cruz HG, Hoever P, Chakraborty B, Schoedel K, Sellers EM, Dingemanse J. Assessment of the abuse liability of a dual orexin receptor antagonist: a crossover study of almorexant and zolpidem in recreational drug users. CNS Drugs. 2014;28(4):361–72. doi:10.1007/s40263-014-0150-x.

    CAS  PubMed  Google Scholar 

  64. Shakeri-Nejad K, Hoch M, Hoever P, Dingemanse J. Influence of mild and moderate liver impairment on the pharmacokinetics and metabolism of almorexant, a dual orexin receptor antagonist. Eur J Pharm Sci. 2013;49(5):836–44. doi:10.1016/j.ejps.2013.06.002.

    CAS  PubMed  Google Scholar 

  65. Perrey DA, German NA, Gilmour BP, Li J-X, Harris DL, Thomas BF, et al. Substituted tetrahydroisoquinolines as selective antagonists for the orexin 1 receptor. J Med Chem. 2013;56(17):6901–16. doi:10.1021/jm400720h.

    CAS  PubMed  Google Scholar 

  66. Di Fabio R, Pellacani A, Faedo S, Roth A, Piccoli L, Gerrard P, et al. Discovery process and pharmacological characterization of a novel dual orexin 1 and orexin 2 receptor antagonist useful for treatment of sleep disorders. Bioorg Med Chem Lett. 2011;21(18):5562–7. doi:10.1016/j.bmcl.2011.06.086.

    PubMed  Google Scholar 

  67. Bettica P, Nucci G, Pyke C, Squassante L, Zamuner S, Ratti E, et al. Phase I studies on the safety, tolerability, pharmacokinetics and pharmacodynamics of SB-649868, a novel dual orexin receptor antagonist. J Psychopharmacol. 2012;26(8):1058–70. doi:10.1177/0269881111408954.

    PubMed  Google Scholar 

  68. GlaxoSmithKline. A clinical study to evaluate the pharmacokinetic profile of SB-649868 in elderly and female population. clinicaltrials.gov. 2010. clinicaltrials.gov/show/NCT00534872. Accessed 22 Dec 2013.

  69. Roecker AJ, Mercer SP, Schreier JD, Cox CD, Fraley ME, Steen JT, et al. Discovery of 5″-chloro-N-[(5,6-dimethoxypyridin-2-yl)methyl]-2,2′:5′,3″-terpyridine-3′-carboxamide (MK-1064): a selective orexin 2 receptor antagonist (2-SORA) for the treatment of insomnia. ChemMedChem. 2014;9(2):311–22. doi:10.1002/cmdc.201300447.

    CAS  PubMed  Google Scholar 

  70. Coleman PJ, Schreier JD, Cox CD, Breslin MJ, Whitman DB, Bogusky MJ, et al. Discovery of [(2R,5R)-5-{[(5-fluoropyridin-2-yl)oxy]methyl}-2-methylpiperidin-1-yl][5-methyl-2-(pyrimidin-2-yl)phenyl]methanone (MK-6096): a dual orexin receptor antagonist with potent sleep-promoting properties. ChemMedChem. 2012;7(3):415–24. doi:10.1002/cmdc.201200025.

    CAS  PubMed  Google Scholar 

  71. Winrow CJ, Gotter AL, Cox CD, Tannenbaum PL, Garson SL, Doran SM, et al. Pharmacological characterization of MK-6096—a dual orexin receptor antagonist for insomnia. Neuropharmacology. 2012;62(2):978–87. doi:10.1016/j.neuropharm.2011.10.003.

    CAS  PubMed  Google Scholar 

  72. Merck Sharp & Dohme. Polysomnography study of MK6096 in patients with primary insomnia (6096-011). clinicaltrials.gov. 2011. clinicaltrials.gov/show/NCT01021852. Accessed 19 Mar 2014.

  73. Merck Sharp & Dohme. A study of the safety and efficacy of MK-6096 for migraine prophylaxis in participants with episodic migraine (MK-6096-020). clinicaltrials.gov. 2012. clinicaltrials.gov/show/NCT01513291. Accessed 19 Mar 2014.

  74. Merck Sharp & Dohme. Study to evaluate MK-6096 in the treatment of painful diabetic neuropathy (PDN) in adults (MK-6096-021 AM1). clinicaltrials.gov. 2013. clinicaltrials.gov/show/NCT01564459. Accessed 19 Mar 2014.

  75. Merck Sharp & Dohme. Safety and efficacy of MK-6096 as adjunctive therapy in participants with major depressive disorder and partial response to antidepressant monotherapy (MK-6096-022 AM3). clinicaltrials.gov. 2013. clinicaltrials.gov/show/NCT01554176. Accessed 19 Mar 2014.

  76. Merck. Merck receives complete response letter for Suvorexant, Merck’s investigational medicine for insomnia. Whitehouse Station, N.J. http://www.mercknewsroom.com; 2013. Accessed 21 Dec 2013.

  77. Cox CD, Breslin MJ, Whitman DB, Schreier JD, McGaughey GB, Bogusky MJ, et al. Discovery of the dual orexin receptor antagonist [(7R)-4-(5-chloro-1,3-benzoxazol-2-yl)-7-methyl-1,4-diazepan-1-yl][5-methyl-2-(2H-1,2,3-triazol-2-yl)phenyl]methanone (MK-4305) for the treatment of insomnia. J Med Chem. 2010;53(14):5320–32. doi:10.1021/jm100541c.

    CAS  PubMed  Google Scholar 

  78. Winrow CJ, Gotter AL, Cox CD, Doran SM, Tannenbaum PL, Breslin MJ, et al. Promotion of sleep by suvorexant—a novel dual orexin receptor antagonist. J Neurogenet. 2011;25(1–2):52–61. doi:10.3109/01677063.2011.566953.

    CAS  PubMed  Google Scholar 

  79. Herring WJMDP, Snyder EP, Budd KBS, Hutzelmann JMS, Snavely DMA, Liu KP, et al. Orexin receptor antagonism for treatment of insomnia: a randomized clinical trial of suvorexant. Neurology. 2012;79(23):2265–74. doi:10.1212/WNL.0b013e31827688ee.

    CAS  PubMed  Google Scholar 

  80. Sun H, Kennedy WP, Wilbraham D, Lewis N, Calder N, Li X, et al. Effects of suvorexant, an orexin receptor antagonist, on sleep parameters as measured by polysomnography in healthy men. Sleep. 2013;36(2):259–67. doi:10.5665/sleep.2386.

    PubMed Central  PubMed  Google Scholar 

  81. Allard JS, Tizabi Y, Shaffery JP, Ovid Trouth C, Manaye K. Stereological analysis of the hypothalamic hypocretin/orexin neurons in an animal model of depression. Neuropeptides. 2004;38(5):311–5. doi:10.1016/j.npep.2004.06.004.

    CAS  PubMed  Google Scholar 

  82. Thannickal TC, Moore RY, Nienhuis R, Ramanathan L, Gulyani S, Aldrich M, et al. Reduced number of hypocretin neurons in human narcolepsy. Neuron. 2000;27(3):469–74. doi:10.1016/S0896-6273(00)00058-1.

    CAS  PubMed  Google Scholar 

  83. Thannickal TC, Nienhuis R, Siegel JM. Localized loss of hypocretin (orexin) cells in narcolepsy without cataplexy. Sleep. 2009;32(8):993–8.

    PubMed Central  PubMed  Google Scholar 

  84. Peyron C, Tighe DK, van den Pol AN, de Lecea L, Heller HC, Sutcliffe JG, et al. Neurons containing hypocretin (orexin) project to multiple neuronal systems. J Neurosci. 1998;18(23):9996–10015.

    CAS  PubMed  Google Scholar 

  85. Baldo BA, Daniel RA, Berridge CW, Kelley AE. Overlapping distributions of orexin/hypocretin- and dopamine-β-hydroxylase immunoreactive fibers in rat brain regions mediating arousal, motivation, and stress. J Comp Neurol. 2003;464(2):220–37. doi:10.1002/cne.10783.

    PubMed  Google Scholar 

  86. Nambu T, Sakurai T, Mizukami K, Hosoya Y, Yanagisawa M, Goto K. Distribution of orexin neurons in the adult rat brain. Brain Res. 1999;827(1–2):243–60. doi:10.1016/S0006-8993(99)01336-0.

    CAS  PubMed  Google Scholar 

  87. Schmitt O, Usunoff KG, Lazarov NE, Itzev DE, Eipert P, Rolfs A, et al. Orexinergic innervation of the extended amygdala and basal ganglia in the rat. Brain Struct Funct. 2012;217(2):233–56. doi:10.1007/s00429-011-0343-8.

    CAS  PubMed  Google Scholar 

  88. Fadel J, Deutch AY. Anatomical substrates of orexin-dopamine interactions: lateral hypothalamic projections to the ventral tegmental area. Neuroscience. 2002;111(2):379–87. doi:10.1016/s0306-4522(02)00017-9.

    CAS  PubMed  Google Scholar 

  89. Marcus JN, Aschkenasi CJ, Lee CE, Chemelli RM, Saper CB, Yanagisawa M, et al. Differential expression of orexin receptors 1 and 2 in the rat brain. J Comp Neurol. 2001;435(1):6–25. doi:10.1002/cne.1190.

    CAS  PubMed  Google Scholar 

  90. Trivedi P, Yu H, MacNeil DJ, Van der Ploeg LHT, Guan X-M. Distribution of orexin receptor mRNA in the rat brain. FEBS Lett. 1998;438(1–2):71–5. doi:10.1016/s0014-5793(98)01266-6.

    CAS  PubMed  Google Scholar 

  91. Hervieu GJ, Cluderay JE, Harrison DC, Roberts JC, Leslie RA. Gene expression and protein distribution of the orexin-1 receptor in the rat brain and spinal cord. Neuroscience. 2001;103(3):777–97. doi:10.1016/s0306-4522(01)00033-1.

    CAS  PubMed  Google Scholar 

  92. Cluderay JE, Harrison DC, Hervieu GJ. Protein distribution of the orexin-2 receptor in the rat central nervous system. Regul Pept. 2002;104(1–3):131–44. doi:10.1016/s0167-0115(01)00357-3.

    CAS  PubMed  Google Scholar 

  93. Martin G, Fabre V, Siggins GR, de Lecea L. Interaction of the hypocretins with neurotransmitters in the nucleus accumbens. Regul Pept. 2002;104(1–3):111–7. doi:10.1016/s0167-0115(01)00354-8.

    CAS  PubMed  Google Scholar 

  94. Porkka-Heiskanen T, Alanko L, Kalinchuk A, Heiskanen S, Stenberg D. The effect of age on prepro-orexin gene expression and contents of orexin A and B in the rat brain. Neurobiol Aging. 2004;25(2):231–8. doi:10.1016/S0197-4580(03)00043-5.

    CAS  PubMed  Google Scholar 

  95. Sasaki K, Suzuki M, Mieda M, Tsujino N, Roth B, Sakurai T. Pharmacogenetic modulation of orexin neurons alters sleep/wakefulness states in mice. PLoS One. 2011;6(5):e20360. doi:10.1371/journal.pone.0020360.

    CAS  PubMed Central  PubMed  Google Scholar 

  96. Yamanaka A, Beuckmann CT, Willie JT, Hara J, Tsujino N, Mieda M, et al. Hypothalamic orexin neurons regulate arousal according to energy balance in mice. Neuron. 2003;38(5):701–13. doi:10.1016/S0896-6273(03)00331-3.

    CAS  PubMed  Google Scholar 

  97. Mieda M, Williams SC, Sinton CM, Richardson JA, Sakurai T, Yanagisawa M. Orexin neurons function in an efferent pathway of a food-entrainable circadian oscillator in eliciting food-anticipatory activity and wakefulness. J Neurosci. 2004;24(46):10493–501. doi:10.1523/jneurosci.3171-04.2004.

    CAS  PubMed  Google Scholar 

  98. Muschamp JW, Dominguez JM, Sato SM, Shen R-Y, Hull EM. A role for hypocretin (orexin) in male sexual behavior. J Neurosci. 2007;27(11):2837–45. doi:10.1523/jneurosci.4121-06.2007.

    CAS  PubMed  Google Scholar 

  99. Georgescu D, Zachariou V, Barrot M, Mieda M, Willie JT, Eisch AJ, et al. Involvement of the lateral hypothalamic peptide orexin in morphine dependence and withdrawal. J Neurosci. 2003;23(8):3106–11.

    CAS  PubMed  Google Scholar 

  100. Laorden ML, Ferenczi S, Pintér-Kübler B, González-Martín LL, Lasheras MC, Kovács KJ, et al. Hypothalamic orexin—a neurons are involved in the response of the brain stress system to morphine withdrawal. PLoS One. 2012;7(5):e36871. doi:10.1371/journal.pone.0036871.

    CAS  PubMed Central  PubMed  Google Scholar 

  101. Harris GC, Wimmer M, Aston-Jones G. A role for lateral hypothalamic orexin neurons in reward seeking. Nature. 2005;437(7058):556–9. doi:10.1038/nature04071.

    CAS  PubMed  Google Scholar 

  102. Yun IA, Wakabayashi KT, Fields HL, Nicola SM. The ventral tegmental area is required for the behavioral and nucleus accumbens neuronal firing responses to incentive cues. J Neurosci. 2004;24(12):2923–33. doi:10.1523/jneurosci.5282-03.2004.

    CAS  PubMed  Google Scholar 

  103. Balcita-Pedicino JJ, Sesack SR. Orexin axons in the rat ventral tegmental area synapse infrequently onto dopamine and γ-aminobutyric acid neurons. J Comp Neurol. 2007;503(5):668–84. doi:10.1002/cne.21420.

    PubMed  Google Scholar 

  104. Narita M, Nagumo Y, Hashimoto S, Narita M, Khotib J, Miyatake M, et al. Direct involvement of orexinergic systems in the activation of the mesolimbic dopamine pathway and related behaviors induced by morphine. J Neurosci. 2006;26(2):398–405. doi:10.1523/jneurosci.2761-05.2006.

    CAS  PubMed  Google Scholar 

  105. Korotkova TM, Sergeeva OA, Eriksson KS, Haas HL, Brown RE. Excitation of ventral tegmental area dopaminergic and nondopaminergic neurons by orexins/hypocretins. J Neurosci. 2003;23(1):7–11.

    CAS  PubMed  Google Scholar 

  106. Li Y, van den Pol AN. μ-Opioid receptor-mediated depression of the hypothalamic hypocretin/orexin arousal system. J Neurosci. 2008;28(11):2814–9. doi:10.1523/jneurosci.5447-07.2008.

    CAS  PubMed  Google Scholar 

  107. Richardson KA, Aston-Jones G. Lateral hypothalamic orexin/hypocretin neurons that project to ventral tegmental area are differentially activated with morphine preference. J Neurosci. 2012;32(11):3809–17. doi:10.1523/jneurosci.3917-11.2012.

    CAS  PubMed Central  PubMed  Google Scholar 

  108. Smith RJ, Aston-Jones G. Orexin / hypocretin 1 receptor antagonist reduces heroin self-administration and cue-induced heroin seeking. Eur J Neurosci. 2012;35(5):798–804. doi:10.1111/j.1460-9568.2012.08013.x.

    PubMed Central  PubMed  Google Scholar 

  109. Sharf R, Sarhan M, DiLeone RJ. Orexin mediates the expression of precipitated morphine withdrawal and concurrent activation of the nucleus accumbens shell. Biol Psychiatry. 2008;64(3):175–83. doi:10.1016/j.biopsych.2008.03.006.

    CAS  PubMed Central  PubMed  Google Scholar 

  110. Schmeichel BE, Vendruscolo LF, Misra KK, Schlosburg JE, Contet C, Grigoriadis DE, et al. Hypocretin-2 receptor antagonism dose-dependently reduces compulsive-like self-administration of heroin in rats allowed extended access. Neuroscience 2013. San Diego: Society for Neuroscience; 2013.

  111. Steiner MA, Lecourt H, Jenck F. The dual orexin receptor antagonist almorexant, alone and in combination with morphine, cocaine and amphetamine, on conditioned place preference and locomotor sensitization in the rat. Int J Neuropsychopharmacol. 2013;16:417–32. doi:10.1017/S1461145712000193.

    CAS  PubMed  Google Scholar 

  112. Lawrence AJ, Cowen MS, Yang H-J, Chen F, Oldfield B. The orexin system regulates alcohol-seeking in rats. Br J Pharmacol. 2006;148(6):752–9. doi:10.1038/sj.bjp.0706789.

    CAS  PubMed Central  PubMed  Google Scholar 

  113. Martin-Fardon R, Weiss F. Blockade of hypocretin receptor-1 preferentially prevents cocaine seeking: comparison with natural reward seeking. Neuroreport. 2014;25(7):485–8. doi:10.1097/WNR.0000000000000120.

    CAS  PubMed  Google Scholar 

  114. Richards J, Simms J, Steensland P, Taha S, Borgland S, Bonci A, et al. Inhibition of orexin-1/hypocretin-1 receptors inhibits yohimbine-induced reinstatement of ethanol and sucrose seeking in Long-Evans rats. Psychopharmacology. 2008;199(1):109–17. doi:10.1007/s00213-008-1136-5.

    CAS  PubMed Central  PubMed  Google Scholar 

  115. Anderson RI, Becker HC, Adams BL, Jesudason CD, Rorick-Kehn LM. Orexin-1 and orexin-2 receptor antagonists reduce ethanol self-administration in high-drinking rodent models. Front Neurosci. 2014;8. doi:10.3389/fnins.2014.00033.

  116. Shoblock J, Welty N, Aluisio L, Fraser I, Motley S, Morton K, et al. Selective blockade of the orexin-2 receptor attenuates ethanol self-administration, place preference, and reinstatement. Psychopharmacology. 2011;215(1):191–203. doi:10.1007/s00213-010-2127-x.

    CAS  PubMed  Google Scholar 

  117. Jupp B, Krstew E, Dezsi G, Lawrence AJ. Discrete cue-conditioned alcohol-seeking after protracted abstinence: pattern of neural activation and involvement of orexin1 receptors. Br J Pharmacol. 2011;162(4):880–9. doi:10.1111/j.1476-5381.2010.01088.x.

    CAS  PubMed Central  PubMed  Google Scholar 

  118. Brown RM, Khoo SY-S, Lawrence AJ. Central orexin (hypocretin) 2 receptor antagonism reduces ethanol self-administration, but not cue-conditioned ethanol-seeking, in ethanol-preferring rats. Int J Neuropsychopharmacol. 2013;16(9):2067–79. doi:10.1017/S1461145713000333.

    CAS  PubMed  Google Scholar 

  119. Sharma R, Sahota P, Thakkar MM. Role of adenosine and the orexinergic perifornical hypothalamus in sleep-promoting effects of ethanol. Sleep. 2014;37(3):525–33. doi:10.5665/sleep.3490.

    PubMed  Google Scholar 

  120. Dayas CV, McGranahan TM, Martin-Fardon R, Weiss F. Stimuli linked to ethanol availability activate hypothalamic CART and orexin neurons in a reinstatement model of relapse. Biol Psychiatry. 2008;63(2):152–7. doi:10.1016/j.biopsych.2007.02.002.

    CAS  PubMed  Google Scholar 

  121. Srinivasan S, Simms JA, Nielsen CK, Lieske SP, Bito-Onon JJ, Yi H, et al. The dual orexin/hypocretin receptor antagonist, almorexant, in the ventral tegmental area attenuates ethanol self-administration. PLoS One. 2012;7(9):e44726. doi:10.1371/journal.pone.0044726.

    CAS  PubMed Central  PubMed  Google Scholar 

  122. Smith RJ, Tahsili-Fahadan P, Aston-Jones G. Orexin/hypocretin is necessary for context-driven cocaine-seeking. Neuropharmacology. 2010;58(1):179–84. doi:10.1016/j.neuropharm.2009.06.042.

    CAS  PubMed Central  PubMed  Google Scholar 

  123. Boutrel B, Kenny PJ, Specio SE, Martin-Fardon R, Markou A, Koob GF, et al. Role for hypocretin in mediating stress-induced reinstatement of cocaine-seeking behavior. Proc Natl Acad Sci USA. 2005;102(52):19168–73. doi:10.1073/pnas.0507480102.

    CAS  PubMed Central  PubMed  Google Scholar 

  124. Smith RJ, See RE, Aston-Jones G. Orexin/hypocretin signaling at the orexin 1 receptor regulates cue-elicited cocaine-seeking. Eur J Neurosci. 2009;30(3):493–503. doi:10.1111/j.1460-9568.2009.06844.x.

    PubMed Central  PubMed  Google Scholar 

  125. Rao Y, Mineur YS, Gan G, Wang AH, Liu Z-W, Wu X, et al. Repeated in vivo exposure of cocaine induces long-lasting synaptic plasticity in hypocretin/orexin-producing neurons in the lateral hypothalamus in mice. J Physiol. 2013;591(7):1951–66. doi:10.1113/jphysiol.2012.246983.

    CAS  PubMed Central  PubMed  Google Scholar 

  126. Harris GC, Aston-Jones G. Critical role for ventral tegmental glutamate in preference for a cocaine-conditioned environment. Neuropsychopharmacology. 2003;28(1):73–6. doi:10.1038/sj.npp.1300011.

    CAS  PubMed  Google Scholar 

  127. Borgland SL, Taha SA, Sarti F, Fields HL, Bonci A. Orexin A in the VTA is critical for the induction of synaptic plasticity and behavioral sensitization to cocaine. Neuron. 2006;49(4):589–601. doi:10.1016/j.neuron.2006.01.016.

    CAS  PubMed  Google Scholar 

  128. Borgland SL, Storm E, Bonci A. Orexin B/hypocretin 2 increases glutamatergic transmission to ventral tegmental area neurons. Eur J Neurosci. 2008;28(8):1545–56. doi:10.1111/j.1460-9568.2008.06397.x.

    CAS  PubMed  Google Scholar 

  129. Mahler SV, Smith RJ, Aston-Jones G. Interactions between VTA orexin and glutamate in cue-induced reinstatement of cocaine seeking in rats. Psychopharmacology. 2013;226(4):687–98. doi:10.1007/s00213-012-2681-5.

    CAS  PubMed Central  PubMed  Google Scholar 

  130. Moorman DE, Aston-Jones G. Orexin/hypocretin modulates response of ventral tegmental dopamine neurons to prefrontal activation: diurnal influences. J Neurosci. 2010;30(46):15585–99. doi:10.1523/jneurosci.2871-10.2010.

    CAS  PubMed Central  PubMed  Google Scholar 

  131. España RA, Oleson EB, Locke JL, Brookshire BR, Roberts DCS, Jones SR. The hypocretin–orexin system regulates cocaine self-administration via actions on the mesolimbic dopamine system. Eur J Neurosci. 2010;31(2):336–48. doi:10.1111/j.1460-9568.2009.07065.x.

    PubMed Central  PubMed  Google Scholar 

  132. Hollander J, Pham D, Fowler C, Kenny PJ. Hypocretin-1 receptors regulate the reinforcing and reward-enhancing effects of cocaine: Pharmacological and behavioral genetics evidence. Front Behav Neurosci. 2012;6. doi:10.3389/fnbeh.2012.00047.

  133. Yeoh JW, James MH, Jobling P, Bains JS, Graham BA, Dayas CV. Cocaine potentiates excitatory drive in the perifornical/lateral hypothalamus. J Physiol. 2012;590(16):3677–89. doi:10.1113/jphysiol.2012.230268.

    CAS  PubMed Central  PubMed  Google Scholar 

  134. Zhou L, Ghee SM, Chan C, Lin L, Cameron MD, Kenny PJ, et al. Orexin-1 receptor mediation of cocaine seeking in male and female rats. J Pharmacol Exp Ther. 2012;340(3):801–9. doi:10.1124/jpet.111.187567.

    CAS  PubMed Central  PubMed  Google Scholar 

  135. Yeoh JW, Campbell EJ, James MH, Graham BA, Dayas CV. Orexin antagonists for neuropsychiatric disease: progress and potential pitfalls. Front Neurosci. 2014;8:36. doi:10.3389/fnins.2014.00036.

    PubMed Central  PubMed  Google Scholar 

  136. Zhou L, Smith RJ, Do PH, Aston-Jones G, See RE. Repeated orexin 1 receptor antagonism effects on cocaine seeking in rats. Neuropharmacology. 2012;63(7):1201–7. doi:10.1016/j.neuropharm.2012.07.044.

    CAS  PubMed Central  PubMed  Google Scholar 

  137. Hollander JA, Lu Q, Cameron MD, Kamenecka TM, Kenny PJ. Insular hypocretin transmission regulates nicotine reward. Proc Natl Acad Sci. 2008;105(49):19480–5. doi:10.1073/pnas.0808023105.

    CAS  PubMed Central  PubMed  Google Scholar 

  138. Plaza-Zabala A, Flores A, Martin-Garcia E, Saravia R, Maldonado R, Berrendero F. A role for hypocretin/orexin receptor-1 in cue-induced reinstatement of nicotine-seeking behavior. Neuropsychopharmacology. 2013;38(9):1724–36. doi:10.1038/npp.2013.72.

    CAS  PubMed  Google Scholar 

  139. Plaza-Zabala A, Martín-García E, de Lecea L, Maldonado R, Berrendero F. Hypocretins regulate the anxiogenic-like effects of nicotine and induce reinstatement of nicotine-seeking behavior. J Neurosci. 2010;30(6):2300–10. doi:10.1523/jneurosci.5724-09.2010.

    PubMed Central  PubMed  Google Scholar 

  140. Plaza-Zabala A, Flores Á, Maldonado R, Berrendero F. Hypocretin/orexin signaling in the hypothalamic paraventricular nucleus is essential for the expression of nicotine withdrawal. Biol Psychiatry. 2012;71(3):214–23. doi:10.1016/j.biopsych.2011.06.025.

    CAS  PubMed  Google Scholar 

  141. Flores Á, Maldonado R, Berrendero F. The hypocretin/orexin receptor-1 as a novel target to modulate cannabinoid reward. Biol Psychiatry. 2014;75(6):499–507. doi:10.1016/j.biopsych.2013.06.012.

    CAS  PubMed  Google Scholar 

  142. Cristino L, Busetto G, Imperatore R, Ferrandino I, Palomba L, Silvestri C, et al. Obesity-driven synaptic remodeling affects endocannabinoid control of orexinergic neurons. Proc Natl Acad Sci. 2013;110(24):E2229–38. doi:10.1073/pnas.1219485110.

    CAS  PubMed Central  PubMed  Google Scholar 

  143. Rotter A, Bayerlein K, Hansbauer M, Weiland J, Sperling W, Kornhuber J, et al. Orexin A expression and promoter methylation in patients with cannabis dependence in comparison to nicotine-dependent cigarette smokers and nonsmokers. Neuropsychobiology. 2012;66(2):126–33. doi:10.1159/000339457.

    CAS  PubMed  Google Scholar 

  144. Vittoz NM, Schmeichel B, Berridge CW. Hypocretin /orexin preferentially activates caudomedial ventral tegmental area dopamine neurons. Eur J Neurosci. 2008;28(8):1629–40. doi:10.1111/j.1460-9568.2008.06453.x.

    PubMed Central  PubMed  Google Scholar 

  145. Peciña S, Berridge KC. Hedonic hot spot in nucleus accumbens shell: where do μ-opioids cause increased hedonic impact of sweetness? J Neurosci. 2005;25(50):11777–86. doi:10.1523/jneurosci.2329-05.2005.

    PubMed  Google Scholar 

  146. Peciña S, Berridge KC. Dopamine or opioid stimulation of nucleus accumbens similarly amplify cue-triggered ‘wanting’ for reward: entire core and medial shell mapped as substrates for PIT enhancement. Eur J Neurosci. 2013;37(9):1529–40. doi:10.1111/ejn.12174.

    PubMed Central  PubMed  Google Scholar 

  147. Castro DC, Berridge KC. Opioid hedonic hotspot in nucleus accumbens shell: mu, delta, and kappa maps for enhancement of sweetness “liking” and “wanting”. J Neurosci. 2014;34(12):4239–50. doi:10.1523/jneurosci.4458-13.2014.

    PubMed  Google Scholar 

  148. Carelli RM, Wondolowski J. Anatomic distribution of reinforcer selective cell firing in the core and shell of the nucleus accumbens. Synapse. 2006;59(2):69–73. doi:10.1002/syn.20217.

    CAS  PubMed  Google Scholar 

  149. Robinson DL, Carelli RM. Distinct subsets of nucleus accumbens neurons encode operant responding for ethanol versus water. Eur J Neurosci. 2008;28(9):1887–94. doi:10.1111/j.1460-9568.2008.06464.x.

    PubMed Central  PubMed  Google Scholar 

  150. Cameron CM, Carelli RM. Cocaine abstinence alters nucleus accumbens firing dynamics during goal-directed behaviors for cocaine and sucrose. Eur J Neurosci. 2012;35(6):940–51. doi:10.1111/j.1460-9568.2012.08024.x.

    PubMed Central  PubMed  Google Scholar 

  151. Patyal R, Woo EY, Borgland SL. Local hypocretin-1 modulates terminal dopamine concentration in the nucleus accumbens shell. Front Behav Neurosci. 2012;6. doi:10.3389/fnbeh.2012.00082.

  152. Ho C-Y, Berridge KC. An orexin hotspot in ventral pallidum amplifies hedonic ‘liking’ for sweetness. Neuropsychopharmacology. 2013;38(9):1655–64. doi:10.1038/npp.2013.62.

    CAS  PubMed  Google Scholar 

  153. Corbit LH, Balleine BW. The role of prelimbic cortex in instrumental conditioning. Behav Brain Res. 2003;146(1–2):145–57. doi:10.1016/j.bbr.2003.09.023.

    PubMed  Google Scholar 

  154. McFarland K, Kalivas PW. The circuitry mediating cocaine-induced reinstatement of drug-seeking behavior. J Neurosci. 2001;21(21):8655–63.

    CAS  PubMed  Google Scholar 

  155. Capriles N, Rodaros D, Sorge RE, Stewart J. A role for the prefrontal cortex in stress- and cocaine-induced reinstatement of cocaine seeking in rats. Psychopharmacology. 2003;168(1–2):66–74. doi:10.1007/s00213-002-1283-z.

    CAS  PubMed  Google Scholar 

  156. McLaughlin J, See RE. Selective inactivation of the dorsomedial prefrontal cortex and the basolateral amygdala attenuates conditioned-cued reinstatement of extinguished cocaine-seeking behavior in rats. Psychopharmacology. 2003;168(1–2):57–65. doi:10.1007/s00213-002-1196-x.

    CAS  PubMed  Google Scholar 

  157. Willcocks AL, McNally GP. The role of medial prefrontal cortex in extinction and reinstatement of alcohol-seeking in rats. Eur J Neurosci. 2013;37(2):259–68. doi:10.1111/ejn.12031.

    PubMed  Google Scholar 

  158. Vittoz NM, Berridge CW. Hypocretin/orexin selectively increases dopamine efflux within the prefrontal cortex: involvement of the ventral tegmental area. Neuropsychopharmacology. 2005;31(2):384–95. doi:10.1038/sj.npp.1300807.

    Google Scholar 

  159. Öngür D, Price JL. The organization of networks within the orbital and medial prefrontal cortex of rats, monkeys and humans. Cereb Cortex. 2000;10(3):206–19. doi:10.1093/cercor/10.3.206.

    PubMed  Google Scholar 

  160. Engelmann JM, Versace F, Robinson JD, Minnix JA, Lam CY, Cui Y, et al. Neural substrates of smoking cue reactivity: a meta-analysis of fMRI studies. NeuroImage. 2012;60(1):252–62. doi:10.1016/j.neuroimage.2011.12.024.

    PubMed Central  PubMed  Google Scholar 

  161. Kirouac GJ, Parsons MP, Li S. Orexin (hypocretin) innervation of the paraventricular nucleus of the thalamus. Brain Res. 2005;1059(2):179–88. doi:10.1016/j.brainres.2005.08.035.

    CAS  PubMed  Google Scholar 

  162. Pasumarthi RK, Fadel J. Activation of orexin/hypocretin projections to basal forebrain and paraventricular thalamus by acute nicotine. Brain Res Bull. 2008;77(6):367–73. doi:10.1016/j.brainresbull.2008.09.014.

    CAS  PubMed Central  PubMed  Google Scholar 

  163. Choi DL, Davis JF, Magrisso IJ, Fitzgerald ME, Lipton JW, Benoit SC. Orexin signaling in the paraventricular thalamic nucleus modulates mesolimbic dopamine and hedonic feeding in the rat. Neuroscience. 2012;210:243–8. doi:10.1016/j.neuroscience.2012.02.036.

    CAS  PubMed Central  PubMed  Google Scholar 

  164. Kelley AE, Baldo BA, Pratt WE. A proposed hypothalamic–thalamic–striatal axis for the integration of energy balance, arousal, and food reward. J Comp Neurol. 2005;493(1):72–85. doi:10.1002/cne.20769.

    CAS  PubMed  Google Scholar 

  165. Parsons MP, Li S, Kirouac GJ. The paraventricular nucleus of the thalamus as an interface between the orexin and CART peptides and the shell of the nucleus accumbens. Synapse. 2006;59(8):480–90. doi:10.1002/syn.20264.

    CAS  PubMed  Google Scholar 

  166. James MH, Charnley JL, Levi EM, Jones E, Yeoh JW, Smith DW, et al. Orexin-1 receptor signalling within the ventral tegmental area, but not the paraventricular thalamus, is critical to regulating cue-induced reinstatement of cocaine-seeking. Int J Neuropsychopharmacol. 2011;14(05):684–90. doi:10.1017/S1461145711000423.

    CAS  PubMed  Google Scholar 

  167. Martin-Fardon R, Zorrilla EP, Ciccocioppo R, Weiss F. Role of innate and drug-induced dysregulation of brain stress and arousal systems in addiction: focus on corticotropin-releasing factor, nociceptin/orphanin FQ, and orexin/hypocretin. Brain Res. 2010;1314:145–61. doi:10.1016/j.brainres.2009.12.027.

    CAS  PubMed  Google Scholar 

  168. Li Y, Li S, Wei C, Wang H, Sui N, Kirouac GJ. Changes in emotional behavior produced by orexin microinjections in the paraventricular nucleus of the thalamus. Pharmacol Biochem Behav. 2010;95(1):121–8. doi:10.1016/j.pbb.2009.12.016.

    CAS  PubMed  Google Scholar 

  169. Li Y, Wang H, Qi K, Chen X, Li S, Sui N, et al. Orexins in the midline thalamus are involved in the expression of conditioned place aversion to morphine withdrawal. Physiol Behav. 2011;102(1):42–50. doi:10.1016/j.physbeh.2010.10.006.

    CAS  PubMed  Google Scholar 

  170. James MH, Dayas CV. What about me…? The PVT: A role for the paraventricular thalamus (PVT) in drug-seeking behaviour. Front Behav Neurosci. 2013;7:18. doi:10.3389/fnbeh.2013.00018.

    PubMed Central  PubMed  Google Scholar 

  171. Jupp B, Krivdic B, Krstew E, Lawrence AJ. The orexin1 receptor antagonist SB-334867 dissociates the motivational properties of alcohol and sucrose in rats. Brain Res. 2011;1391:54–9. doi:10.1016/j.brainres.2011.03.045.

    CAS  PubMed  Google Scholar 

  172. Martin-Fardon R, Weiss F. N-(2-methyl-6-benzoxazolyl)-N′-1,5-naphthyridin-4-yl urea (SB334867), a hypocretin receptor-1 antagonist, preferentially prevents ethanol seeking: comparison with natural reward seeking. Addict Biol. 2014;19(2):233–6. doi:10.1111/j.1369-1600.2012.00480.x.

    CAS  PubMed  Google Scholar 

  173. Hamlin AS, Newby J, McNally GP. The neural correlates and role of D1 dopamine receptors in renewal of extinguished alcohol-seeking. Neuroscience. 2007;146(2):525–36. doi:10.1016/j.neuroscience.2007.01.063.

    CAS  PubMed  Google Scholar 

  174. Hamlin AS, Blatchford KE, McNally GP. Renewal of an extinguished instrumental response: neural correlates and the role of D1 dopamine receptors. Neuroscience. 2006;143(1):25–38. doi:10.1016/j.neuroscience.2006.07.035.

    CAS  PubMed  Google Scholar 

  175. Cason AM, Aston-Jones G. Role of orexin/hypocretin in conditioned sucrose-seeking in rats. Psychopharmacology. 2013;226(1):155–65. doi:10.1007/s00213-012-2902-y.

    CAS  PubMed Central  PubMed  Google Scholar 

  176. Cason AM, Aston-Jones G. Attenuation of saccharin-seeking in rats by orexin/hypocretin receptor 1 antagonist. Psychopharmacology. 2013;228(3):499–507. doi:10.1007/s00213-013-3051-7.

    CAS  PubMed  Google Scholar 

  177. Borgland SL, Chang S-J, Bowers MS, Thompson JL, Vittoz N, Floresco SB, et al. Orexin A/hypocretin-1 selectively promotes motivation for positive reinforcers. J Neurosci. 2009;29(36):11215–25. doi:10.1523/jneurosci.6096-08.2009.

    CAS  PubMed Central  PubMed  Google Scholar 

  178. Moorman DE, Aston-Jones G. Orexin-1 receptor antagonism decreases ethanol consumption and preference selectively in high-ethanol—preferring Sprague-Dawley rats. Alcohol. 2009;43(5):379–86. doi:10.1016/j.alcohol.2009.07.002.

    CAS  PubMed Central  PubMed  Google Scholar 

  179. Brown RM, Lawrence AJ. Ascending orexinergic pathways and alcohol-seeking. Curr Opin Neurobiol. 2013;23(4):467–72. doi:10.1016/j.conb.2013.02.014.

    CAS  PubMed  Google Scholar 

  180. Thorpe AJ, Kotz CM. Orexin A in the nucleus accumbens stimulates feeding and locomotor activity. Brain Res. 2005;1050(1–2):156–62. doi:10.1016/j.brainres.2005.05.045.

    CAS  PubMed  Google Scholar 

  181. Thorpe AJ, Cleary JP, Levine AS, Kotz CM. Centrally administered orexin A increases motivation for sweet pellets in rats. Psychopharmacology. 2005;182(1):75–83. doi:10.1007/s00213-005-0040-5.

    CAS  PubMed  Google Scholar 

  182. Dugovic C, Shelton JE, Aluisio LE, Fraser IC, Jiang X, Sutton SW, et al. Blockade of orexin-1 receptors attenuates orexin-2 receptor antagonism-induced sleep promotion in the rat. J Pharmacol Exp Ther. 2009;330(1):142–51. doi:10.1124/jpet.109.152009.

    CAS  PubMed  Google Scholar 

  183. Morairty SR, Revel FG, Malherbe P, Moreau J-L, Valladao D, Wettstein JG, et al. Dual hypocretin receptor antagonism is more effective for sleep promotion than antagonism of either receptor alone. PLoS One. 2012;7(7):e39131. doi:10.1371/journal.pone.0039131.

    CAS  PubMed Central  PubMed  Google Scholar 

  184. Russell SH, Small CJ, Kennedy AR, Stanley SA, Seth A, Murphy KG, et al. Orexin A interactions in the hypothalamo-pituitary gonadal axis. Endocrinology. 2001;142(12):5294–302. doi:10.1210/endo.142.12.8558.

    CAS  PubMed  Google Scholar 

  185. Hoch M, Hoever P, Alessi F, Marjason J, Dingemanse J. Pharmacokinetics and tolerability of almorexant in Japanese and Caucasian healthy male subjects. Pharmacology. 2011;88(3–4):121–6. doi:10.1159/000330098.

    CAS  PubMed  Google Scholar 

  186. Hoever P, Hay J, Rad M, Cavallaro M, van Gerven JM, Dingemanse J. Tolerability, pharmacokinetics, and pharmacodynamics of single-dose almorexant, an orexin receptor antagonist, in healthy elderly subjects. J Clin Psychopharmacol. 2013;33(3):363–70. doi:10.1097/JCP.0b013e31828f5a7a.

    CAS  PubMed  Google Scholar 

  187. Hoyer D, Dürst T, Fendt M, Jacobson LH, Betschart C, Hintermann S, et al. Distinct effects of IPSU and suvorexant on mouse sleep architecture. Front Neurosci. 2013;7. doi:10.3389/fnins.2013.00235.

  188. Sifferlen T, Koberstein R, Cottreel E, Boller A, Weller T, Gatfield J, et al. Structure-activity relationship studies and sleep-promoting activity of novel 1-chloro-5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine derivatives as dual orexin receptor antagonists. Part 2. Bioorg Med Chem Lett. 2013;23(13):3857–63. doi:10.1016/j.bmcl.2013.04.071.

    CAS  PubMed  Google Scholar 

  189. Uslaner JM, Tye SJ, Eddins DM, Wang X, Fox SV, Savitz AT, et al. Orexin receptor antagonists differ from standard sleep drugs by promoting sleep at doses that do not disrupt cognition. Sci Transl Med. 2013;5(179):179ra44. doi:10.1126/scitranslmed.3005213.

    PubMed  Google Scholar 

  190. Lutter M, Krishnan V, Russo SJ, Jung S, McClung CA, Nestler EJ. Orexin signaling mediates the antidepressant-like effect of calorie restriction. J Neurosci. 2008;28(12):3071–5. doi:10.1523/jneurosci.5584-07.2008.

    CAS  PubMed Central  PubMed  Google Scholar 

  191. Nocjar C, Zhang J, Feng P, Panksepp J. The social defeat animal model of depression shows diminished levels of orexin in mesocortical regions of the dopamine system, and of dynorphin and orexin in the hypothalamus. Neuroscience. 2012;218:138–53. doi:10.1016/j.neuroscience.2012.05.033.

    CAS  PubMed  Google Scholar 

  192. Johnson PL, Truitt W, Fitz SD, Minick PE, Dietrich A, Sanghani S, et al. A key role for orexin in panic anxiety. Nat Med. 2010;16(1):111–5. doi:10.1038/nm.2075.

    CAS  PubMed Central  PubMed  Google Scholar 

  193. Hirota K, Kushikata T, Kudo M, Kudo T, Smart D, Matsuki A. Effects of central hypocretin-1 administration on hemodynamic responses in young-adult and middle-aged rats. Brain Res. 2003;981(1–2):143–50. doi:10.1016/S0006-8993(03)03002-6.

    CAS  PubMed  Google Scholar 

  194. Li A, Hindmarch CCT, Nattie EE, Paton JFR. Antagonism of orexin receptors significantly lowers blood pressure in spontaneously hypertensive rats. J Physiol. 2013;591(17):4237–48. doi:10.1113/jphysiol.2013.256271.

    CAS  PubMed  Google Scholar 

  195. Ramirez AD, Gotter AL, Fox SV, Tannenbaum PL, Yao L, Tye SJ, et al. Dual orexin receptor antagonists show distinct effects on locomotor performance, ethanol interaction and sleep architecture relative to gamma-aminobutyric acid-A receptor modulators. Front Neurosci. 2013;7. doi:10.3389/fnins.2013.00254.

  196. Regard JB, Sato IT, Coughlin SR. Anatomical profiling of G protein-coupled receptor expression. Cell. 2008;135(3):561–71. doi:10.1016/j.cell.2008.08.040.

    CAS  PubMed Central  PubMed  Google Scholar 

  197. Faedo S, Perdonà E, Antolini M, di Fabio R, Merlo Pich E, Corsi M. Functional and binding kinetic studies make a distinction between OX1 and OX2 orexin receptor antagonists. Eur J Pharmacol. 2012;692(1–3):1–9. doi:10.1016/j.ejphar.2012.07.007.

    CAS  PubMed  Google Scholar 

  198. McElhinny CJ Jr, Lewin AH, Mascarella SW, Runyon S, Brieaddy L, Carroll FI. Hydrolytic instability of the important orexin 1 receptor antagonist SB-334867: possible confounding effects on in vivo and in vitro studies. Bioorg Med Chem Lett. 2012;22(21):6661–4. doi:10.1016/j.bmcl.2012.08.109.

    CAS  PubMed  Google Scholar 

  199. Zhang G-C, Mao L-M, Liu X-Y, Wang JQ. Long-lasting up-regulation of orexin receptor type 2 protein levels in the rat nucleus accumbens after chronic cocaine administration. J Neurochem. 2007;103(1):400–7. doi:10.1111/j.1471-4159.2007.04748.x.

    CAS  PubMed  Google Scholar 

  200. Voorhees C, Cunningham C. Involvement of the orexin/hypocretin system in ethanol conditioned place preference. Psychopharmacology. 2011;214(4):805–18. doi:10.1007/s00213-010-2082-6.

    CAS  PubMed Central  PubMed  Google Scholar 

  201. Plaza-Zabala A, Li X, Milovanovic M, Loweth JA, Maldonado R, Berrendero F, et al. An investigation of interactions between hypocretin/orexin signaling and glutamate receptor surface expression in the rat nucleus accumbens under basal conditions and after cocaine exposure. Neurosci Lett. 2013;557, Part B:101–6. doi:10.1016/j.neulet.2013.10.038.

    Google Scholar 

  202. Gozzi A, Turrini G, Piccoli L, Massagrande M, Amantini D, Antolini M, et al. Functional magnetic resonance imaging reveals different neural substrates for the effects of orexin-1 and orexin-2 receptor antagonists. PLoS One. 2011;6(1):e16406. doi:10.1371/journal.pone.0016406.

    CAS  PubMed Central  PubMed  Google Scholar 

  203. Miller WR, Walters ST, Bennett ME. How effective is alcoholism treatment in the United States? J Stud Alcohol. 2001;62(2):211–20.

    CAS  PubMed  Google Scholar 

  204. O’Brien CP. A range of research-based pharmacotherapies for addiction. Science. 1997;278(5335):66–70. doi:10.1126/science.278.5335.66.

    PubMed  Google Scholar 

  205. Miller WR, Manuel JK. How large must a treatment effect be before it matters to practitioners? An estimation method and demonstration. Drug Alcohol Rev. 2008;27(5):524–8. doi:10.1080/09595230801956165.

    PubMed  Google Scholar 

  206. Ketter TA, Citrome L, Wang PW, Culver JL, Srivastava S. Treatments for bipolar disorder: can number needed to treat/harm help inform clinical decisions? Acta Psychiatr Scand. 2011;123(3):175–89. doi:10.1111/j.1600-0447.2010.01645.x.

    CAS  PubMed  Google Scholar 

  207. Citrome L. Levomilnacipran for major depressive disorder: a systematic review of the efficacy and safety profile for this newly approved antidepressant—what is the number needed to treat, number needed to harm and likelihood to be helped or harmed? Int J Clin Pract. 2013;67(11):1089–104. doi:10.1111/ijcp.12298.

    CAS  PubMed  Google Scholar 

  208. Citrome L. Vortioxetine for major depressive disorder: a systematic review of the efficacy and safety profile for this newly approved antidepressant—what is the number needed to treat, number needed to harm and likelihood to be helped or harmed? Int J Clin Pract. 2014;68(1):60–82. doi:10.1111/ijcp.12350.

    CAS  PubMed  Google Scholar 

  209. Rösner S, Hackl-Herrwerth A, Leucht S, Lehert P, Vecchi S, Soyka M. Acamprosate for alcohol dependence. Cochrane Database Syst Rev. 2010;2010(9). doi:10.1002/14651858.CD004332.pub2.

  210. Bouton ME, Winterbauer NE, Todd TP. Relapse processes after the extinction of instrumental learning: Renewal, resurgence, and reacquisition. Behav Processes. 2012;90(1):130–41. doi:10.1016/j.beproc.2012.03.004.

    PubMed Central  PubMed  Google Scholar 

  211. Delamater AR, Westbrook RF. Psychological and neural mechanisms of experimental extinction: a selective review. Neurobiol Learn Mem. 2014;108:38–51. doi:10.1016/j.nlm.2013.09.016.

    PubMed  Google Scholar 

  212. Callander GE, Olorunda M, Monna D, Schuepbach E, Langenegger D, Betschart C, et al. Kinetic properties of ‘dual’ orexin receptor antagonists at OX1R and OX2R orexin receptors. Front Neurosci. 2013;7. doi:10.3389/fnins.2013.00230.

  213. Neubig RR, Spedding M, Kenakin T, Christopoulos A. International Union of Pharmacology Committee on Receptor Nomenclature and Drug Classification. XXXVIII. Update on terms and symbols in quantitative pharmacology. Pharmacol Rev. 2003;55(4):597–606. doi:10.1124/pr.55.4.4.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

RMB and SYK are supported by the National Health and Medical Research Council of Australia. SYK is supported by an Australian Postgraduate Award; RMB is supported by a Peter Doherty Fellowship. The authors declare that they have no conflicts of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Robyn Mary Brown.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Khoo, S.YS., Brown, R.M. Orexin/Hypocretin Based Pharmacotherapies for the Treatment of Addiction: DORA or SORA?. CNS Drugs 28, 713–730 (2014). https://doi.org/10.1007/s40263-014-0179-x

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40263-014-0179-x

Keywords

Navigation