Log in

Potential of Cannabinoid Receptor Ligands as Treatment for Substance Use Disorders

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

Abstract

Substance use disorder (SUD) is a major public health crisis worldwide, and effective treatment options are limited. During the past 2 decades, researchers have investigated the impact of a variety of pharmacological approaches to treat SUD, one of which is the use of medical cannabis or cannabinoids. Significant progress was made with the discovery of rimonabant, a selective CB1 receptor (CB1R) antagonist (also an inverse agonist), as a promising therapeutic for SUDs and obesity. However, serious adverse effects such as depression and suicidality led to the withdrawal of rimonabant (and almost all other CB1R antagonists/inverse agonists) from clinical trials worldwide in 2008. Since then, much research interest has shifted to other cannabinoid-based strategies, such as peripheral CB1R antagonists/inverse agonists, neutral CB1R antagonists, allosteric CB1R modulators, CB2R agonists, fatty acid amide hydrolase (FAAH) inhibitors, monoacylglycerol lipase (MAGL) inhibitors, fatty acid binding protein (FABP) inhibitors, or nonaddictive phytocannabinoids with CB1R or CB2R-binding profiles, as new therapeutics for SUDs. In this article, we first review recent progress in research regarding the endocannabinoid systems, cannabis reward versus aversion, and the underlying receptor mechanisms. We then review recent progress in cannabinoid-based medication development for the treatment of SUDs. As evidence continues to accumulate, neutral CB1R antagonists (such as AM4113), CB2R agonists (JWH133, **e2-64), and nonselective phytocannabinoids (cannabidiol, β-caryophyllene, ∆9-tetrahydrocannabivarin) have shown great therapeutic potential for SUDs, as shown in experimental animals. Several cannabinoid-based medications (e.g., dronabinol, nabilone, PF-04457845) that entered clinical trials have shown promising results in reducing withdrawal symptoms in cannabis and opioid users.

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

Access this article

Subscribe and save

Springer+ Basic
EUR 32.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or Ebook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

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

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Rudd RA, Aleshire N, Zibbell JE, Gladden RM. Increases in drug and opioid overdose deaths—United States, 2000–2014. MMWR Morb Mortal Wkly Rep. 2016;64(50–51):1378–82.

    Article  PubMed  Google Scholar 

  2. Drug Overdose Deaths | Drug Overdose | CDC Injury Center [Internet]; 2018. https://www.cdc.gov/drugoverdose/data/statedeaths.html. Cited 17 May 2019.

  3. Jordan CJ, Cao J, Newman AH, ** Z-X. Progress in agonist therapy for substance use disorders: lessons learned from methadone and buprenorphine. Neuropharmacology. 2019. https://doi.org/10.1016/j.neuropharm.2019.04.015.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Jordan CJ, ** Z-X. Discovery and development of varenicline for smoking cessation. Expert Opin Drug Discov. 2018;13(7):671–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. ** Z-X, Gardner EL. Hypothesis-driven medication discovery for the treatment of psychostimulant addiction. Curr Drug Abuse Rev. 2008;1(3):303–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kalivas PW, Volkow ND. New medications for drug addiction hiding in glutamatergic neuroplasticity. Mol Psychiatry. 2011;16(10):974–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Filip M, Frankowska M, Sadakierska-Chudy A, Suder A, Szumiec L, Mierzejewski P, et al. GABAB receptors as a therapeutic strategy in substance use disorders: focus on positive allosteric modulators. Neuropharmacology. 2015;88:36–47.

    Article  CAS  PubMed  Google Scholar 

  8. Volkow ND, Jones EB, Einstein EB, Wargo EM. Prevention and treatment of opioid misuse and addiction: a review. JAMA Psychiatry. 2019;76(2):208–16.

    Article  PubMed  Google Scholar 

  9. Volkow ND, Wise RA, Baler R. The dopamine motive system: implications for drug and food addiction. Nat Rev Neurosci. 2017;18(12):741–52.

    Article  CAS  PubMed  Google Scholar 

  10. Ostroumov A, Dani JA. Inhibitory plasticity of mesocorticolimbic circuits in addiction and mental illness. Trends Neurosci. 2018;41(12):898–910.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Koob GF, Volkow ND. Neurobiology of addiction: a neurocircuitry analysis. Lancet Psychiatry. 2016;3(8):760–73.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Parsons LH, Hurd YL. Endocannabinoid signalling in reward and addiction. Nat Rev Neurosci. 2015;16(10):579–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Trigo JM, Le Foll B. Inhibition of monoacylglycerol lipase (MAGL) enhances cue-induced reinstatement of nicotine-seeking behavior in mice. Psychopharmacology (Berl). 2016;233(10):1815–22.

    Article  CAS  Google Scholar 

  14. Serrano A, Parsons LH. Endocannabinoid influence in drug reinforcement, dependence and addiction-related behaviors. Pharmacol Ther. 2011;132(3):215–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ligresti A, Cascio MG, Di Marzo V. Endocannabinoid metabolic pathways and enzymes. Curr Drug Targets CNS Neurol Disord. 2005;4(6):615–23.

    Article  CAS  PubMed  Google Scholar 

  16. Devane WA, Hanus L, Breuer A, Pertwee RG, Stevenson LA, Griffin G, et al. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science. 1992;258(5090):1946–9.

    Article  CAS  PubMed  Google Scholar 

  17. McPartland JM, Glass M, Pertwee RG. Meta-analysis of cannabinoid ligand binding affinity and receptor distribution: interspecies differences. Br J Pharmacol. 2007;152(5):583–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Pacher P, Bátkai S, Kunos G. The endocannabinoid system as an emerging target of pharmacotherapy. Pharmacol Rev. 2006;58(3):389–462.

    Article  CAS  PubMed  Google Scholar 

  19. Sugiura T, Waku K. 2-Arachidonoylglycerol and the cannabinoid receptors. Chem Phys Lipids. 2000;108(1–2):89–106.

    Article  CAS  PubMed  Google Scholar 

  20. Long JZ, Li W, Booker L, Burston JJ, Kinsey SG, Schlosburg JE, et al. Selective blockade of 2-arachidonoylglycerol hydrolysis produces cannabinoid behavioral effects. Nat Chem Biol. 2009;5(1):37–44.

    Article  CAS  PubMed  Google Scholar 

  21. Kaczocha M, Glaser ST, Deutsch DG. Identification of intracellular carriers for the endocannabinoid anandamide. Proc Natl Acad Sci USA. 2009;106(15):6375–80.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Oddi S, Fezza F, Pasquariello N, D’Agostino A, Catanzaro G, De Simone C, et al. Molecular identification of albumin and Hsp70 as cytosolic anandamide-binding proteins. Chem Biol. 2009;16(6):624–32.

    Article  CAS  PubMed  Google Scholar 

  23. Hermann A, Kaczocha M, Deutsch DG. 2-Arachidonoylglycerol (2-AG) membrane transport: history and outlook. AAPS J. 2006;8(2):E409–12.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Herkenham M, Lynn AB, Little MD, Johnson MR, Melvin LS, de Costa BR, et al. Cannabinoid receptor localization in brain. Proc Natl Acad Sci USA. 1990;87(5):1932–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Piomelli D. The molecular logic of endocannabinoid signalling. Nat Rev Neurosci. 2003;4(11):873–84.

    Article  CAS  PubMed  Google Scholar 

  26. Kano M, Ohno-Shosaku T, Hashimotodani Y, Uchigashima M, Watanabe M. Endocannabinoid-mediated control of synaptic transmission. Physiol Rev. 2009;89(1):309–80.

    Article  CAS  PubMed  Google Scholar 

  27. Castillo PE, Younts TJ, Chávez AE, Hashimotodani Y. Endocannabinoid signaling and synaptic function. Neuron. 2012;76(1):70–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Howlett AC, Blume LC, Dalton GD. CB(1) cannabinoid receptors and their associated proteins. Curr Med Chem. 2010;17(14):1382–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Freund TF, Katona I, Piomelli D. Role of endogenous cannabinoids in synaptic signaling. Physiol Rev. 2003;83(3):1017–66.

    Article  CAS  PubMed  Google Scholar 

  30. Stella N. Cannabinoid and cannabinoid-like receptors in microglia, astrocytes, and astrocytomas. Glia. 2010;58(9):1017–30.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Han J, Kesner P, Metna-Laurent M, Duan T, Xu L, Georges F, et al. Acute cannabinoids impair working memory through astroglial CB1 receptor modulation of hippocampal LTD. Cell. 2012;148(5):1039–50.

    Article  CAS  PubMed  Google Scholar 

  32. Oliveira da Cruz JF, Robin LM, Drago F, Marsicano G, Metna-Laurent M. Astroglial type-1 cannabinoid receptor (CB1): a new player in the tripartite synapse. Neuroscience. 2016;26(323):35–42.

    Article  CAS  Google Scholar 

  33. Navarrete M, Araque A. Endocannabinoids mediate neuron-astrocyte communication. Neuron. 2008;57(6):883–93.

    Article  CAS  PubMed  Google Scholar 

  34. Mothet JP, Parent AT, Wolosker H, Brady RO, Linden DJ, Ferris CD, et al. D-serine is an endogenous ligand for the glycine site of the N-methyl-d-aspartate receptor. Proc Natl Acad Sci USA. 2000;97(9):4926–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Metna-Laurent M, Marsicano G. Rising stars: modulation of brain functions by astroglial type-1 cannabinoid receptors. Glia. 2015;63(3):353–64.

    Article  PubMed  Google Scholar 

  36. Robin LM, Oliveira da Cruz JF, Langlais VC, Martin-Fernandez M, Metna-Laurent M, Busquets-Garcia A, et al. Astroglial CB1 receptors determine synaptic D-serine availability to enable recognition memory. Neuron. 2018;98(5):935–944.e5.

    Article  CAS  PubMed  Google Scholar 

  37. Calabrese EJ, Rubio-Casillas A. Biphasic effects of THC in memory and cognition. Eur J Clin Investig. 2018;48(5):e12920.

    Article  CAS  Google Scholar 

  38. Mailleux P, Vanderhaeghen JJ. Distribution of neuronal cannabinoid receptor in the adult rat brain: a comparative receptor binding radioautography and in situ hybridization histochemistry. Neuroscience. 1992;48(3):655–68.

    Article  CAS  PubMed  Google Scholar 

  39. Mátyás F, Yanovsky Y, Mackie K, Kelsch W, Misgeld U, Freund TF. Subcellular localization of type 1 cannabinoid receptors in the rat basal ganglia. Neuroscience. 2006;137(1):337–61.

    Article  CAS  PubMed  Google Scholar 

  40. Han X, He Y, Bi G-H, Zhang H-Y, Song R, Liu Q-R, et al. CB1 receptor activation on VgluT2-expressing glutamatergic neurons underlies Δ9-tetrahydrocannabinol (Δ9-THC)-induced aversive effects in mice. Sci Rep. 2017;7(1):12315.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Mackie K. Distribution of cannabinoid receptors in the central and peripheral nervous system. Handb Exp Pharmacol. 2005;168:299–325.

    Article  CAS  Google Scholar 

  42. Fratta W, Fattore L. Molecular mechanisms of cannabinoid addiction. Curr Opin Neurobiol. 2013;23(4):487–92.

    Article  CAS  PubMed  Google Scholar 

  43. Gueudet C, Santucci V, Rinaldi-Carmona M, Soubrié P, Le Fur G. The CB1 cannabinoid receptor antagonist SR 141716A affects A9 dopamine neuronal activity in the rat. Neuroreport. 1995;6(10):1421–5.

    Article  CAS  PubMed  Google Scholar 

  44. De Luca MA, Bimpisidis Z, Melis M, Marti M, Caboni P, Valentini V, et al. Stimulation of in vivo dopamine transmission and intravenous self-administration in rats and mice by JWH-018, a Spice cannabinoid. Neuropharmacology. 2015;99:705–14.

    Article  CAS  PubMed  Google Scholar 

  45. De Luca MA, Castelli MP, Loi B, Porcu A, Martorelli M, Miliano C, et al. Native CB1 receptor affinity, intrinsic activity and accumbens shell dopamine stimulant properties of third generation SPICE/K2 cannabinoids: BB-22, 5F-PB-22, 5F-AKB-48 and STS-135. Neuropharmacology. 2016;105:630–8.

    Article  CAS  PubMed  Google Scholar 

  46. Mateo Y, Johnson KA, Covey DP, Atwood BK, Wang H-L, Zhang S, et al. Endocannabinoid actions on cortical terminals orchestrate local modulation of dopamine release in the nucleus accumbens. Neuron. 2017;96(5):1112–1126.e5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. French ED, Dillon K, Wu X. Cannabinoids excite dopamine neurons in the ventral tegmentum and substantia nigra. Neuroreport. 1997;8(3):649–52.

    Article  CAS  PubMed  Google Scholar 

  48. Braida D, Iosuè S, Pegorini S, Sala M. Delta9-tetrahydrocannabinol-induced conditioned place preference and intracerebroventricular self-administration in rats. Eur J Pharmacol. 2004;506(1):63–9.

    Article  CAS  PubMed  Google Scholar 

  49. Gardner EL. Endocannabinoid signaling system and brain reward: emphasis on dopamine. Pharmacol Biochem Behav. 2005;81(2):263–84.

    Article  CAS  PubMed  Google Scholar 

  50. Spiller KJ, Bi G-H, He Y, Galaj E, Gardner EL, ** Z-X. Cannabinoid CB1 and CB2 receptor mechanisms underlie cannabis reward and aversion in rats. Br J Pharmacol. 2019;176(9):1268–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Takahashi RN, Singer G. Self-administration of delta 9-tetrahydrocannabinol by rats. Pharmacol Biochem Behav. 1979;11(6):737–40.

    Article  CAS  PubMed  Google Scholar 

  52. Fattore L, Cossu G, Martellotta CM, Fratta W. Intravenous self-administration of the cannabinoid CB1 receptor agonist WIN 55,212-2 in rats. Psychopharmacology (Berl). 2001;156(4):410–6.

    Article  CAS  Google Scholar 

  53. Lefever TW, Marusich JA, Antonazzo KR, Wiley JL. Evaluation of WIN 55,212-2 self-administration in rats as a potential cannabinoid abuse liability model. Pharmacol Biochem Behav. 2014;118:30–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Spencer S, Neuhofer D, Chioma VC, Garcia-Keller C, Schwartz DJ, Allen N, et al. A model of Δ9-tetrahydrocannabinol self-administration and reinstatement that alters synaptic plasticity in nucleus accumbens. Biol Psychiatry. 2018;84(8):601–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Szabo B, Siemes S, Wallmichrath I. Inhibition of GABAergic neurotransmission in the ventral tegmental area by cannabinoids. Eur J Neurosci. 2002;15(12):2057–61.

    Article  PubMed  Google Scholar 

  56. Lupica CR, Riegel AC, Hoffman AF. Marijuana and cannabinoid regulation of brain reward circuits. Br J Pharmacol. 2004;143(2):227–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Lupica CR, Riegel AC. Endocannabinoid release from midbrain dopamine neurons: a potential substrate for cannabinoid receptor antagonist treatment of addiction. Neuropharmacology. 2005;48(8):1105–16.

    Article  CAS  PubMed  Google Scholar 

  58. Lupica CR, Hoffman AF. Cannabinoid disruption of learning mechanisms involved in reward processing. Learn Mem. 2018;25(9):435–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Maldonado R, Valverde O, Berrendero F. Involvement of the endocannabinoid system in drug addiction. Trends Neurosci. 2006;29(4):225–32.

    Article  CAS  PubMed  Google Scholar 

  60. Fattore L, Fadda P, Spano MS, Pistis M, Fratta W. Neurobiological mechanisms of cannabinoid addiction. Mol Cell Endocrinol. 2008;286(1–2 Suppl 1):S97–107.

    Article  CAS  PubMed  Google Scholar 

  61. Raft D, Gregg J, Ghia J, Harris L. Effects of intravenous tetrahydrocannabinol on experimental and surgical pain. Psychological correlates of the analgesic response. Clin Pharmacol Ther. 1977;21(1):26–33.

    Article  CAS  PubMed  Google Scholar 

  62. D’Souza DC, Perry E, MacDougall L, Ammerman Y, Cooper T, Wu Y-T, et al. The psychotomimetic effects of intravenous delta-9-tetrahydrocannabinol in healthy individuals: implications for psychosis. Neuropsychopharmacology. 2004;29(8):1558–72.

    Article  CAS  PubMed  Google Scholar 

  63. Gregg JM, Small EW, Moore R, Raft D, Toomey TC. Emotional response to intravenous delta9tetrahydrocannabinol during oral surgery. J Oral Surg. 1976;34(4):301–13.

    CAS  PubMed  Google Scholar 

  64. Farris SG, Zvolensky MJ, Boden MT, Bonn-Miller MO. Cannabis use expectancies mediate the relation between depressive symptoms and cannabis use among cannabis-dependent veterans. J Addict Med. 2014;8(2):130–6.

    Article  PubMed  Google Scholar 

  65. Tanda G, Munzar P, Goldberg SR. Self-administration behavior is maintained by the psychoactive ingredient of marijuana in squirrel monkeys. Nat Neurosci. 2000;3(11):1073–4.

    Article  CAS  PubMed  Google Scholar 

  66. Justinova Z, Tanda G, Redhi GH, Goldberg SR. Self-administration of delta9-tetrahydrocannabinol (THC) by drug naive squirrel monkeys. Psychopharmacology (Berl). 2003;169(2):135–40.

    Article  CAS  Google Scholar 

  67. Mansbach RS, Nicholson KL, Martin BR, Balster RL. Failure of Delta(9)-tetrahydrocannabinol and CP 55,940 to maintain intravenous self-administration under a fixed-interval schedule in rhesus monkeys. Behav Pharmacol. 1994;5(2):219–25.

    Article  CAS  PubMed  Google Scholar 

  68. John WS, Martin TJ, Nader MA. Behavioral determinants of cannabinoid self-administration in old world monkeys. Neuropsychopharmacology. 2017;42(7):1522–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Gardner EL, Paredes W, Smith D, Donner A, Milling C, Cohen D, et al. Facilitation of brain stimulation reward by delta 9-tetrahydrocannabinol. Psychopharmacology. 1988;96(1):142–4.

    Article  CAS  PubMed  Google Scholar 

  70. Lepore M, Liu X, Savage V, Matalon D, Gardner EL. Genetic differences in delta 9-tetrahydrocannabinol-induced facilitation of brain stimulation reward as measured by a rate-frequency curve-shift electrical brain stimulation paradigm in three different rat strains. Life Sci. 1996;58(25):PL365–72.

    Article  CAS  PubMed  Google Scholar 

  71. Katsidoni V, Kastellakis A, Panagis G. Biphasic effects of Δ9-tetrahydrocannabinol on brain stimulation reward and motor activity. Int J Neuropsychopharmacol. 2013;16(10):2273–84.

    Article  CAS  PubMed  Google Scholar 

  72. Vlachou S, Nomikos GG, Stephens DN, Panagis G. Lack of evidence for appetitive effects of Delta 9-tetrahydrocannabinol in the intracranial self-stimulation and conditioned place preference procedures in rodents. Behav Pharmacol. 2007;18(4):311–9.

    Article  CAS  PubMed  Google Scholar 

  73. Kwilasz AJ, Negus SS. Dissociable effects of the cannabinoid receptor agonists Δ9-tetrahydrocannabinol and CP55940 on pain-stimulated versus pain-depressed behavior in rats. J Pharmacol Exp Ther. 2012;343(2):389–400.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Negus SS, Miller LL. Intracranial self-stimulation to evaluate abuse potential of drugs. Pharmacol Rev. 2014;66(3):869–917.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Wiebelhaus JM, Grim TW, Owens RA, Lazenka MF, Sim-Selley LJ, Abdullah RA, et al. Δ9-tetrahydrocannabinol and endocannabinoid degradative enzyme inhibitors attenuate intracranial self-stimulation in mice. J Pharmacol Exp Ther. 2015;352(2):195–207.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Panagis G, Vlachou S, Nomikos GG. Behavioral pharmacology of cannabinoids with a focus on preclinical models for studying reinforcing and dependence-producing properties. Curr Drug Abuse Rev. 2008;1(3):350–74.

    Article  CAS  PubMed  Google Scholar 

  77. Vlachou S, Panagis G. Regulation of brain reward by the endocannabinoid system: a critical review of behavioral studies in animals. Curr Pharm Des. 2014;20(13):2072–88.

    Article  CAS  PubMed  Google Scholar 

  78. Castañeda E, Moss DE, Oddie SD, Whishaw IQ. THC does not affect striatal dopamine release: microdialysis in freely moving rats. Pharmacol Biochem Behav. 1991;40(3):587–91.

    Article  PubMed  Google Scholar 

  79. Wang H-L, Qi J, Zhang S, Wang H, Morales M. Rewarding effects of optical stimulation of ventral tegmental area glutamatergic neurons. J Neurosci. 2015;35(48):15948–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Manzanares J, Cabañero D, Puente N, García-Gutiérrez MS, Grandes P, Maldonado R. Role of the endocannabinoid system in drug addiction. Biochem Pharmacol. 2018;157:108–21.

    Article  CAS  PubMed  Google Scholar 

  81. Jordan CJ, ** Z-X. Progress in brain cannabinoid CB2 receptor research: from genes to behavior. Neurosci Biobehav Rev. 2019;98:208–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. ** Z-X, Peng X-Q, Li X, Song R, Zhang H-Y, Liu Q-R, et al. Brain cannabinoid CB2 receptors modulate cocaine’s actions in mice. Nat Neurosci. 2011;14(9):1160–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Zhang H-Y, Gao M, Liu Q-R, Bi G-H, Li X, Yang H-J, et al. Cannabinoid CB2 receptors modulate midbrain dopamine neuronal activity and dopamine-related behavior in mice. Proc Natl Acad Sci USA. 2014;111(46):E5007–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Zhang H-Y, Gao M, Shen H, Bi G-H, Yang H-J, Liu Q-R, et al. Expression of functional cannabinoid CB2 receptor in VTA dopamine neurons in rats. Addict Biol. 2017;22(3):752–65.

    Article  CAS  PubMed  Google Scholar 

  85. Foster DJ, Wilson JM, Remke DH, Mahmood MS, Uddin MJ, Wess J, et al. Antipsychotic-like effects of M4 positive allosteric modulators are mediated by CB2 receptor-dependent inhibition of dopamine release. Neuron. 2016;91(6):1244–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Delis F, Polissidis A, Poulia N, Justinova Z, Nomikos GG, Goldberg SR, et al. Attenuation of cocaine-induced conditioned place preference and motor activity via cannabinoid CB2 receptor agonism and CB1 receptor antagonism in rats. Int J Neuropsychopharmacol. 2017;20(3):269–78.

    CAS  PubMed  Google Scholar 

  87. Zhang H-Y, Bi G-H, Li X, Li J, Qu H, Zhang S-J, et al. Species differences in cannabinoid receptor 2 and receptor responses to cocaine self-administration in mice and rats. Neuropsychopharmacology. 2015;40(4):1037–51.

    Article  CAS  PubMed  Google Scholar 

  88. Aracil-Fernández A, Trigo JM, García-Gutiérrez MS, Ortega-Álvaro A, Ternianov A, Navarro D, et al. Decreased cocaine motor sensitization and self-administration in mice overexpressing cannabinoid CB2 receptors. Neuropsychopharmacology. 2012;37(7):1749–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Gamaleddin I, Zvonok A, Makriyannis A, Goldberg SR, Le Foll B. Effects of a selective cannabinoid CB2 agonist and antagonist on intravenous nicotine self administration and reinstatement of nicotine seeking. PLoS One. 2012;7(1):e29900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Solinas M, Panlilio LV, Goldberg SR. Exposure to delta-9-tetrahydrocannabinol (THC) increases subsequent heroin taking but not heroin’s reinforcing efficacy: a self-administration study in rats. Neuropsychopharmacology. 2004;29(7):1301–11.

    Article  CAS  PubMed  Google Scholar 

  91. Colombo G, Serra S, Brunetti G, Gomez R, Melis S, Vacca G, et al. Stimulation of voluntary ethanol intake by cannabinoid receptor agonists in ethanol-preferring sP rats. Psychopharmacology (Berl). 2002;159(2):181–7.

    Article  CAS  Google Scholar 

  92. Linsenbardt DN, Boehm SL. Agonism of the endocannabinoid system modulates binge-like alcohol intake in male C57BL/6J mice: involvement of the posterior ventral tegmental area. Neuroscience. 2009;164(2):424–34.

    Article  CAS  PubMed  Google Scholar 

  93. Caillé S, Alvarez-Jaimes L, Polis I, Stouffer DG, Parsons LH. Specific alterations of extracellular endocannabinoid levels in the nucleus accumbens by ethanol, heroin, and cocaine self-administration. J Neurosci. 2007;27(14):3695–702.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Wang H, Treadway T, Covey DP, Cheer JF, Lupica CR. Cocaine-induced endocannabinoid mobilization in the ventral tegmental area. Cell Rep. 2015;12(12):1997–2008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Le Foll B, Gorelick DA, Goldberg SR. The future of endocannabinoid-oriented clinical research after CB1 antagonists. Psychopharmacology. 2009;205(1):171–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Maccioni P, Colombo G, Carai MAM. Blockade of the cannabinoid CB1 receptor and alcohol dependence: preclinical evidence and preliminary clinical data. CNS Neurol Disord Drug Targets. 2010;9(1):55–9.

    Article  CAS  PubMed  Google Scholar 

  97. Gamaleddin IH, Trigo JM, Gueye AB, Zvonok A, Makriyannis A, Goldberg SR, et al. Role of the endogenous cannabinoid system in nicotine addiction: novel insights. Front Psychiatry. 2015;6:41.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Stern CAJ, de Carvalho CR, Bertoglio LJ, Takahashi RN. Effects of cannabinoid drugs on aversive or rewarding drug-associated memory extinction and reconsolidation. Neuroscience. 2018;01(370):62–80.

    Article  CAS  Google Scholar 

  99. Rinaldi-Carmona M, Barth F, Héaulme M, Shire D, Calandra B, Congy C, et al. SR141716A, a potent and selective antagonist of the brain cannabinoid receptor. FEBS Lett. 1994;350(2–3):240–4.

    Article  CAS  PubMed  Google Scholar 

  100. Mato S, Pazos A, Valdizán EM. Cannabinoid receptor antagonism and inverse agonism in response to SR141716A on cAMP production in human and rat brain. Eur J Pharmacol. 2002;443(1–3):43–6.

    Article  CAS  PubMed  Google Scholar 

  101. Le Foll B, Goldberg SR. Cannabinoid CB1 receptor antagonists as promising new medications for drug dependence. J Pharmacol Exp Ther. 2005;312(3):875–83.

    Article  CAS  PubMed  Google Scholar 

  102. Wiskerke J, Pattij T, Schoffelmeer ANM, De Vries TJ. The role of CB1 receptors in psychostimulant addiction. Addict Biol. 2008;13(2):225–38.

    Article  CAS  PubMed  Google Scholar 

  103. Cahill K, Ussher MH. Cannabinoid type 1 receptor antagonists for smoking cessation. Cochrane Database Syst Rev. 2011;3:CD005353.

    Google Scholar 

  104. Gaal LFV, Rissanen AM, Scheen AJ, Ziegler O, Rössner S. Effects of the cannabinoid-1 receptor blocker rimonabant on weight reduction and cardiovascular risk factors in overweight patients: 1-year experience from the RIO-Europe study. Lancet. 2005;365(9468):1389–97.

    Article  CAS  PubMed  Google Scholar 

  105. Elrashidi MY, Ebbert JO. Emerging drugs for the treatment of tobacco dependence: 2014 update. Expert Opin Emerg Drugs. 2014;19(2):243–60.

    Article  CAS  PubMed  Google Scholar 

  106. Sloan ME, Gowin JL, Ramchandani VA, Hurd YL, Le Foll B. The endocannabinoid system as a target for addiction treatment: trials and tribulations. Neuropharmacology. 2017;15(124):73–83.

    Article  CAS  Google Scholar 

  107. Christensen R, Kristensen PK, Bartels EM, Bliddal H, Astrup AV. A meta-analysis of the efficacy and safety of the anti-obesity agent Rimonabant. Ugeskr Laeg. 2007;169(50):4360–3.

    PubMed  Google Scholar 

  108. Sam AH, Salem V, Ghatei MA. Rimonabant: from RIO to ban. J Obes. 2011;2011:432607.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Morgan CJA, Das RK, Joye A, Curran HV, Kamboj SK. Cannabidiol reduces cigarette consumption in tobacco smokers: preliminary findings. Addict Behav. 2013;38(9):2433–6.

    Article  PubMed  Google Scholar 

  110. Chorvat RJ. Peripherally restricted CB1 receptor blockers. Bioorg Med Chem Lett. 2013;23(17):4751–60.

    Article  CAS  PubMed  Google Scholar 

  111. Cluny NL, Vemuri VK, Chambers AP, Limebeer CL, Bedard H, Wood JT, et al. A novel peripherally restricted cannabinoid receptor antagonist, AM6545, reduces food intake and body weight, but does not cause malaise, in rodents. Br J Pharmacol. 2010;161(3):629–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Kunos G, Tam J. The case for peripheral CB1 receptor blockade in the treatment of visceral obesity and its cardiometabolic complications. Br J Pharmacol. 2011;163(7):1423–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Chorvat RJ, Berbaum J, Seriacki K, McElroy JF. JD-5006 and JD-5037: peripherally restricted (PR) cannabinoid-1 receptor blockers related to SLV-319 (Ibipinabant) as metabolic disorder therapeutics devoid of CNS liabilities. Bioorg Med Chem Lett. 2012;22(19):6173–80.

    Article  CAS  PubMed  Google Scholar 

  114. Tam J, Cinar R, Liu J, Godlewski G, Wesley D, Jourdan T, et al. Peripheral cannabinoid-1 receptor inverse agonism reduces obesity by reversing leptin resistance. Cell Metab. 2012;16(2):167–79.

    Article  CAS  PubMed  Google Scholar 

  115. Tai S, Nikas SP, Shukla VG, Vemuri K, Makriyannis A, Järbe TUC. Cannabinoid withdrawal in mice: inverse agonist vs neutral antagonist. Psychopharmacology (Berl). 2015;232(15):2751–61.

    Article  CAS  Google Scholar 

  116. Godlewski G, Cinar R, Coffey NJ, Liu J, Jourdan T, Mukhopadhyay B, et al. Targeting peripheral CB1 receptors reduces ethanol intake via a gut–brain axis. Cell Metab. 2019;29(6):1320–1333.e8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. ** Z-X, Spiller K, Pak AC, Gilbert J, Dillon C, Li X, et al. Cannabinoid CB1 receptor antagonists attenuate cocaine’s rewarding effects: experiments with self-administration and brain-stimulation reward in rats. Neuropsychopharmacology. 2008;33(7):1735–45.

    Article  CAS  PubMed  Google Scholar 

  118. He X-H, Jordan CJ, Vemuri K, Bi G-H, Zhan J, Gardner EL, et al. Cannabinoid CB1 receptor neutral antagonist AM4113 inhibits heroin self-administration without depressive side effects in rats. Acta Pharmacol Sin. 2019;40:365–73.

    Article  CAS  PubMed  Google Scholar 

  119. Gardner EL, Gamaleddin I, Manzanares RJ, Rodrigues FF. The endocannabinoid system: useful targets for anti-addiction treatments? Subst Abuse. 2013;34:324–5.

    Google Scholar 

  120. Gueye AB, Pryslawsky Y, Trigo JM, Poulia N, Delis F, Antoniou K, et al. The CB1 neutral antagonist AM4113 retains the therapeutic efficacy of the inverse agonist rimonabant for nicotine dependence and weight loss with better psychiatric tolerability. Int J Neuropsychopharmacol. 2016. https://doi.org/10.1093/ijnp/pyw068.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Alvarado M, Decara J, Luque MJ, Hernandez-Folgado L, Gómez-Cañas M, Gómez-Ruiz M, et al. Novel antiobesity agents: synthesis and pharmacological evaluation of analogues of Rimonabant and of LH21. Bioorg Med Chem. 2013;21(7):1708–16.

    Article  CAS  PubMed  Google Scholar 

  122. Seltzman HH, Maitra R, Bortoff K, Henson J, Reggio PH, Wesley D, et al. Metabolic profiling of CB1 neutral antagonists. Methods Enzymol. 2017;593:199–215.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Salamone JD, McLaughlin PJ, Sink K, Makriyannis A, Parker LA. Cannabinoid CB1 receptor inverse agonists and neutral antagonists: effects on food intake, food-reinforced behavior and food aversions. Physiol Behav. 2007;91(4):383–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Sink KS, McLaughlin PJ, Wood JAT, Brown C, Fan P, Vemuri VK, et al. The novel cannabinoid CB1 receptor neutral antagonist AM4113 suppresses food intake and food-reinforced behavior but does not induce signs of nausea in rats. Neuropsychopharmacology. 2008;33(4):946–55.

    Article  CAS  PubMed  Google Scholar 

  125. Chambers AP, Vemuri VK, Peng Y, Wood JT, Olszewska T, Pittman QJ, et al. A neutral CB1 receptor antagonist reduces weight gain in rat. Am J Physiol Regul Integr Comp Physiol. 2007;293(6):R2185–93.

    Article  CAS  PubMed  Google Scholar 

  126. Järbe TUC, LeMay BJ, Olszewska T, Vemuri VK, Wood JT, Makriyannis A. Intrinsic effects of AM4113, a putative neutral CB1 receptor selective antagonist, on open-field behaviors in rats. Pharmacol Biochem Behav. 2008;91(1):84–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Balla A, Dong B, Shilpa BM, Vemuri K, Makriyannis A, Pandey SC, et al. Cannabinoid-1 receptor neutral antagonist reduces binge-like alcohol consumption and alcohol-induced accumbal dopaminergic signaling. Neuropharmacology. 2018;15(131):200–8.

    Article  CAS  Google Scholar 

  128. Schindler CW, Redhi GH, Vemuri K, Makriyannis A, Le Foll B, Bergman J, et al. Blockade of nicotine and cannabinoid reinforcement and relapse by a cannabinoid CB1-receptor neutral antagonist AM4113 and inverse agonist rimonabant in squirrel monkeys. Neuropsychopharmacology. 2016;41(9):2283–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Kangas BD, Delatte MS, Vemuri VK, Thakur GA, Nikas SP, Subramanian KV, et al. Cannabinoid discrimination and antagonism by CB(1) neutral and inverse agonist antagonists. J Pharmacol Exp Ther. 2013;344(3):561–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Wills KL, Vemuri K, Kalmar A, Lee A, Limebeer CL, Makriyannis A, et al. CB1 antagonism: interference with affective properties of acute naloxone-precipitated morphine withdrawal in rats. Psychopharmacology (Berl). 2014;231(22):4291–300.

    Article  CAS  Google Scholar 

  131. Jagerovic N, Hernandez-Folgado L, Alkorta I, Goya P, Navarro M, Serrano A, et al. Discovery of 5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-3-hexyl-1h-1,2,4-triazole, a novel in vivo cannabinoid antagonist containing a 1,2,4-triazole motif. J Med Chem. 2004;47(11):2939–42.

    Article  CAS  PubMed  Google Scholar 

  132. Chen RZ, Frassetto A, Lao JZ, Huang R-RC, **ao JC, Clements MJ, et al. Pharmacological evaluation of LH-21, a newly discovered molecule that binds to cannabinoid CB1 receptor. Eur J Pharmacol. 2008;584(2–3):338–42.

    Article  CAS  PubMed  Google Scholar 

  133. Pavon FJ, Bilbao A, Hernández-Folgado L, Cippitelli A, Jagerovic N, Abellán G, et al. Antiobesity effects of the novel in vivo neutral cannabinoid receptor antagonist 5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-3-hexyl-1H-1,2,4-triazole–LH 21. Neuropharmacology. 2006;51(2):358–66.

    Article  CAS  PubMed  Google Scholar 

  134. Pavón FJ, Serrano A, Pérez-Valero V, Jagerovic N, Hernández-Folgado L, Bermúdez-Silva FJ, et al. Central versus peripheral antagonism of cannabinoid CB1 receptor in obesity: effects of LH-21, a peripherally acting neutral cannabinoid receptor antagonist, in Zucker rats. J Neuroendocrinol. 2008;20(Suppl 1):116–23.

    Article  CAS  PubMed  Google Scholar 

  135. Alonso M, Serrano A, Vida M, Crespillo A, Hernandez-Folgado L, Jagerovic N, et al. Anti-obesity efficacy of LH-21, a cannabinoid CB1 receptor antagonist with poor brain penetration, in diet-induced obese rats. Br J Pharmacol. 2012;165(7):2274–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Gardner EL, Bi G-H, Thakur GA, Makriyannis A, Seltzman HH, He X-Y, et al. Preclinical evaluation of neutral cannabinoid CB1 receptor antagonists and cannabinoid CB1 receptor negative allosteric modulators for treating drug addiction. Int J Neuropsychopharmacol. 2016. Report No.: Meeting Abstract-PM288.

  137. Price MR, Baillie GL, Thomas A, Stevenson LA, Easson M, Goodwin R, et al. Allosteric modulation of the cannabinoid CB1 receptor. Mol Pharmacol. 2005;68(5):1484–95.

    Article  CAS  PubMed  Google Scholar 

  138. **g L, Qiu Y, Zhang Y, Li J-X. Effects of the cannabinoid CB1 receptor allosteric modulator ORG 27569 on reinstatement of cocaine- and methamphetamine-seeking behavior in rats. Drug Alcohol Depend. 2014;1(143):251–6.

    Article  CAS  Google Scholar 

  139. Gamage TF, Ignatowska-Jankowska BM, Wiley JL, Abdelrahman M, Trembleau L, Greig IR, et al. In-vivo pharmacological evaluation of the CB1-receptor allosteric modulator Org-27569. Behav Pharmacol. 2014;25(2):182–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Ding Y, Qiu Y, **g L, Thorn DA, Zhang Y, Li J-X. Behavioral effects of the cannabinoid CB1 receptor allosteric modulator ORG27569 in rats. Pharmacol Res Perspect. 2014;2(6):e00069.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Hofer SC, Ralvenius WT, Gachet MS, Fritschy J-M, Zeilhofer HU, Gertsch J. Localization and production of peptide endocannabinoids in the rodent CNS and adrenal medulla. Neuropharmacology. 2015;98:78–89.

    Article  CAS  PubMed  Google Scholar 

  142. Bauer M, Chicca A, Tamborrini M, Eisen D, Lerner R, Lutz B, et al. Identification and quantification of a new family of peptide endocannabinoids (Pepcans) showing negative allosteric modulation at CB1 receptors. J Biol Chem. 2012;287(44):36944–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Petrucci V, Chicca A, Glasmacher S, Paloczi J, Cao Z, Pacher P, et al. Pepcan-12 (RVD-hemopressin) is a CB2 receptor positive allosteric modulator constitutively secreted by adrenals and in liver upon tissue damage. Sci Rep. 2017;7(1):9560.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Gomes I, Grushko JS, Golebiewska U, Hoogendoorn S, Gupta A, Heimann AS, et al. Novel endogenous peptide agonists of cannabinoid receptors. FASEB J. 2009;23(9):3020–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Ferrante C, Recinella L, Leone S, Chiavaroli A, Di Nisio C, Martinotti S, et al. Anorexigenic effects induced by RVD-hemopressin(α) administration. Pharmacol Rep. 2017;69(6):1402–7.

    Article  CAS  PubMed  Google Scholar 

  146. Leone S, Recinella L, Chiavaroli A, Martinotti S, Ferrante C, Mollica A, et al. Emotional disorders induced by Hemopressin and RVD-hemopressin(α) administration in rats. Pharmacol Rep. 2017;69(6):1247–53.

    Article  CAS  PubMed  Google Scholar 

  147. Atwood BK, Mackie K. CB2: a cannabinoid receptor with an identity crisis. Br J Pharmacol. 2010;160(3):467–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Lanciego JL, Barroso-Chinea P, Rico AJ, Conte-Perales L, Callén L, Roda E, et al. Expression of the mRNA coding the cannabinoid receptor 2 in the pallidal complex of Macaca fascicularis. J Psychopharmacol (Oxford). 2011;25(1):97–104.

    Article  CAS  Google Scholar 

  149. Sierra S, Luquin N, Rico AJ, Gómez-Bautista V, Roda E, Dopeso-Reyes IG, et al. Detection of cannabinoid receptors CB1 and CB2 within basal ganglia output neurons in macaques: changes following experimental parkinsonism. Brain Struct Funct. 2015;220(5):2721–38.

    Article  CAS  PubMed  Google Scholar 

  150. Ignatowska-Jankowska BM, Muldoon PP, Lichtman AH, Damaj MI. The cannabinoid CB2 receptor is necessary for nicotine-conditioned place preference, but not other behavioral effects of nicotine in mice. Psychopharmacology (Berl). 2013;229(4):591–601.

    Article  CAS  Google Scholar 

  151. Navarrete F, Rodríguez-Arias M, Martín-García E, Navarro D, García-Gutiérrez MS, Aguilar MA, et al. Role of CB2 cannabinoid receptors in the rewarding, reinforcing, and physical effects of nicotine. Neuropsychopharmacology. 2013;38(12):2515–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Liu Q-R, Canseco-Alba A, Zhang H-Y, Tagliaferro P, Chung M, Dennis E, et al. Cannabinoid type 2 receptors in dopamine neurons inhibits psychomotor behaviors, alters anxiety, depression and alcohol preference. Sci Rep. 2017;7(1):17410.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Ortega-Álvaro A, Ternianov A, Aracil-Fernández A, Navarrete F, García-Gutiérrez MS, Manzanares J. Role of cannabinoid CB2 receptor in the reinforcing actions of ethanol. Addict Biol. 2015;20(1):43–55.

    Article  CAS  PubMed  Google Scholar 

  154. Powers MS, Breit KR, Chester JA. Genetic versus pharmacological assessment of the role of cannabinoid type 2 receptors in alcohol reward-related behaviors. Alcohol Clin Exp Res. 2015;39(12):2438–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Bystrowska B, Frankowska M, Smaga I, Pomierny-Chamioło L, Filip M. effects of cocaine self-administration and its extinction on the rat brain cannabinoid CB1 and CB2 receptors. Neurotox Res. 2018;34:547–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Bystrowska B, Frankowska M, Smaga I, Niedzielska-Andres E, Pomierny-Chamioło L, Filip M. cocaine-induced reinstatement of cocaine seeking provokes changes in the endocannabinoid and N-acylethanolamine levels in rat brain structures. Molecules. 2019;24(6):E1125.

    Article  CAS  PubMed  Google Scholar 

  157. Huffman JW. CB2 receptor ligands. Mini Rev Med Chem. 2005;5(7):641–9.

    Article  CAS  PubMed  Google Scholar 

  158. Ma Z, Gao F, Larsen B, Gao M, Luo Z, Chen D, et al. Mechanisms of cannabinoid CB2 receptor-mediated reduction of dopamine neuronal excitability in mouse ventral tegmental area. EBioMedicine. 2019;42:225–37.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Canseco-Alba A, Schanz N, Sanabria B, Zhao J, Lin Z, Liu Q-R, et al. Behavioral effects of psychostimulants in mutant mice with cell-type specific deletion of CB2 cannabinoid receptors in dopamine neurons. Behav Brain Res. 2018;30(360):286–97.

    Google Scholar 

  160. Valenzano KJ, Tafesse L, Lee G, Harrison JE, Boulet JM, Gottshall SL, et al. Pharmacological and pharmacokinetic characterization of the cannabinoid receptor 2 agonist, GW405833, utilizing rodent models of acute and chronic pain, anxiety, ataxia and catalepsy. Neuropharmacology. 2005;48(5):658–72.

    Article  CAS  PubMed  Google Scholar 

  161. Adamczyk P, Miszkiel J, McCreary AC, Filip M, Papp M, Przegaliński E. The effects of cannabinoid CB1, CB2 and vanilloid TRPV1 receptor antagonists on cocaine addictive behavior in rats. Brain Res. 2012;20(1444):45–54.

    Article  CAS  Google Scholar 

  162. Wiley JL, Beletskaya ID, Ng EW, Dai Z, Crocker PJ, Mahadevan A, et al. Resorcinol derivatives: a novel template for the development of cannabinoid CB(1)/CB(2) and CB(2)-selective agonists. J Pharmacol Exp Ther. 2002;301(2):679–89.

    Article  CAS  PubMed  Google Scholar 

  163. Alavi MS, Hosseinzadeh H, Shamsizadeh A, Roohbakhsh A. The effect of O-1602, an atypical cannabinoid, on morphine-induced conditioned place preference and physical dependence. Pharmacol Rep. 2016;68(3):592–7.

    Article  CAS  PubMed  Google Scholar 

  164. Zhang M, Dong L, Zou H, Li J, Li Q, Wang G, et al. Effects of cannabinoid type 2 receptor agonist AM1241 on morphine-induced antinociception, acute and chronic tolerance, and dependence in mice. J Pain. 2018;19(10):1113–29.

    Article  CAS  PubMed  Google Scholar 

  165. Li A-L, Lin X, Dhopeshwarkar AS, Thomaz AC, Carey LM, Liu Y, et al. Cannabinoid CB2 agonist AM1710 differentially suppresses distinct pathological pain states and attenuates morphine tolerance and withdrawal. Mol Pharmacol. 2019;95(2):155–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Grenald SA, Young MA, Wang Y, Ossipov MH, Ibrahim MM, Largent-Milnes TM, et al. Synergistic attenuation of chronic pain using mu opioid and cannabinoid receptor 2 agonists. Neuropharmacology. 2017;116:59–70.

    Article  CAS  PubMed  Google Scholar 

  167. Ibrahim MM, Deng H, Zvonok A, Cockayne DA, Kwan J, Mata HP, et al. Activation of CB2 cannabinoid receptors by AM1241 inhibits experimental neuropathic pain: pain inhibition by receptors not present in the CNS. Proc Natl Acad Sci USA. 2003;100(18):10529–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Yang P, Wang L, Feng R, Almehizia AA, Tong Q, Myint KZ, et al. Novel triaryl sulfonamide derivatives as selective cannabinoid receptor 2 inverse agonists and osteoclast inhibitors: discovery, optimization, and biological evaluation. J Med Chem. 2013;56:2045–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Jordan C, Feng XW, Bi G-H, Liang Y, Han X, **e X-Q, et al. **e2-64 is a promising cannabinoid CB2 receptor ligand that reduces cocaine abuse-related behaviors in rodents. Addict Biol. 2019. (in press).

  170. Melis M, Pistis M, Perra S, Muntoni AL, Pillolla G, Gessa GL. Endocannabinoids mediate presynaptic inhibition of glutamatergic transmission in rat ventral tegmental area dopamine neurons through activation of CB1 receptors. J Neurosci. 2004;24(1):53–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Panlilio LV, Justinova Z, Goldberg SR. Inhibition of FAAH and activation of PPAR: New approaches to the treatment of cognitive dysfunction and drug addiction. Pharmacol Ther. 2013;138(1):84–102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Deutsch DG. A personal retrospective: elevating anandamide (AEA) by targeting fatty acid amide hydrolase (FAAH) and the fatty acid binding proteins (FABPs). Front Pharmacol [Internet]; 2016. Cited 6 Mar 2019.

  173. Alexander JP, Cravatt BF. Mechanism of carbamate inactivation of FAAH: implications for the design of covalent inhibitors and in vivo functional probes for enzymes. Chem Biol. 2005;12(11):1179–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Justinova Z, Mangieri RA, Bortolato M, Chefer SI, Mukhin AG, Clapper JR, et al. Fatty acid amide hydrolase inhibition heightens anandamide signaling without producing reinforcing effects in primates. Biol Psychiatry. 2008;64(11):930–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Adamczyk P, McCreary AC, Przegalinski E, Mierzejewski P, Bienkowski P, Filip M. The effects of fatty acid amide hydrolase inhibitors on maintenance of cocaine and food self-administration and on reinstatement of cocaine-seeking and food-taking behavior in rats. J Physiol Pharmacol. 2009;60(3):119–25.

    CAS  PubMed  Google Scholar 

  176. Chauvet C, Nicolas C, Thiriet N, Lardeux MV, Duranti A, Solinas M. Chronic stimulation of the tone of endogenous anandamide reduces cue- and stress-induced relapse in rats. Int J Neuropsychopharmacol. 2014. https://doi.org/10.1093/ijnp/pyu025.

    Article  PubMed  PubMed Central  Google Scholar 

  177. Solinas M, Panlilio LV, Tanda G, Makriyannis A, Matthews SA, Goldberg SR. Cannabinoid agonists but not inhibitors of endogenous cannabinoid transport or metabolism enhance the reinforcing efficacy of heroin in rats. Neuropsychopharmacology. 2005;30(11):2046–57.

    Article  CAS  PubMed  Google Scholar 

  178. McCallum AL, Limebeer CL, Parker LA. Reducing endocannabinoid metabolism with the fatty acid amide hydrolase inhibitor, URB597, fails to modify reinstatement of morphine-induced conditioned floor preference and naloxone-precipitated morphine withdrawal-induced conditioned floor avoidance. Pharmacol Biochem Behav. 2010;96(4):496–500.

    Article  CAS  PubMed  Google Scholar 

  179. Manwell LA, Satvat E, Lang ST, Allen CP, Leri F, Parker LA. FAAH inhibitor, URB-597, promotes extinction and CB(1) antagonist, SR141716, inhibits extinction of conditioned aversion produced by naloxone-precipitated morphine withdrawal, but not extinction of conditioned preference produced by morphine in rats. Pharmacol Biochem Behav. 2009;94(1):154–62.

    Article  CAS  PubMed  Google Scholar 

  180. Ramesh D, Ross GR, Schlosburg JE, Owens RA, Abdullah RA, Kinsey SG, et al. Blockade of endocannabinoid hydrolytic enzymes attenuates precipitated opioid withdrawal symptoms in mice. J Pharmacol Exp Ther. 2011;339(1):173–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Shahidi S, Hasanein P. Behavioral effects of fatty acid amide hydrolase inhibition on morphine withdrawal symptoms. Brain Res Bull. 2011;86(1–2):118–22.

    Article  CAS  PubMed  Google Scholar 

  182. Scherma M, Panlilio LV, Fadda P, Fattore L, Gamaleddin I, Le Foll B, et al. Inhibition of anandamide hydrolysis by cyclohexyl carbamic acid 3′-carbamoyl-3-yl ester (URB597) reverses abuse-related behavioral and neurochemical effects of nicotine in rats. J Pharmacol Exp Ther. 2008;327(2):482–90.

    Article  CAS  PubMed  Google Scholar 

  183. Forget B, Guranda M, Gamaleddin I, Goldberg SR, Le Foll B. Attenuation of cue-induced reinstatement of nicotine seeking by URB597 through cannabinoid CB1 receptor in rats. Psychopharmacology (Berl). 2016;233(10):1823–8.

    Article  CAS  Google Scholar 

  184. Forget B, Coen KM, Le Foll B. Inhibition of fatty acid amide hydrolase reduces reinstatement of nicotine seeking but not break point for nicotine self-administration—comparison with CB(1) receptor blockade. Psychopharmacology (Berl). 2009;205(4):613–24.

    Article  CAS  Google Scholar 

  185. Justinova Z, Panlilio LV, Moreno-Sanz G, Redhi GH, Auber A, Secci ME, et al. Effects of fatty acid amide hydrolase (FAAH) inhibitors in non-human primate models of nicotine reward and relapse. Neuropsychopharmacology. 2015;40(9):2185–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Cippitelli A, Astarita G, Duranti A, Caprioli G, Ubaldi M, Stopponi S, et al. Endocannabinoid regulation of acute and protracted nicotine withdrawal: effect of FAAH inhibition. PLoS One. 2011;6(11):e28142.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Melis M, Pillolla G, Luchicchi A, Muntoni AL, Yasar S, Goldberg SR, et al. Endogenous fatty acid ethanolamides suppress nicotine-induced activation of mesolimbic dopamine neurons through nuclear receptors. J Neurosci. 2008;28(51):13985–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Luchicchi A, Lecca S, Carta S, Pillolla G, Muntoni AL, Yasar S, et al. Effects of fatty acid amide hydrolase inhibition on neuronal responses to nicotine, cocaine and morphine in the nucleus accumbens shell and ventral tegmental area: involvement of PPAR-alpha nuclear receptors. Addict Biol. 2010;15(3):277–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Blednov YA, Cravatt BF, Boehm SL, Walker D, Harris RA. Role of endocannabinoids in alcohol consumption and intoxication: studies of mice lacking fatty acid amide hydrolase. Neuropsychopharmacology. 2007;32(7):1570–82.

    Article  CAS  PubMed  Google Scholar 

  190. Basavarajappa BS, Yalamanchili R, Cravatt BF, Cooper TB, Hungund BL. Increased ethanol consumption and preference and decreased ethanol sensitivity in female FAAH knockout mice. Neuropharmacology. 2006;50(7):834–44.

    Article  CAS  PubMed  Google Scholar 

  191. Cippitelli A, Cannella N, Braconi S, Duranti A, Tontini A, Bilbao A, et al. Increase of brain endocannabinoid anandamide levels by FAAH inhibition and alcohol abuse behaviours in the rat. Psychopharmacology (Berl). 2008;198(4):449–60.

    Article  CAS  Google Scholar 

  192. Zhou Y, Schwartz BI, Giza J, Gross SS, Lee FS, Kreek MJ. Blockade of alcohol escalation and “relapse” drinking by pharmacological FAAH inhibition in male and female C57BL/6J mice. Psychopharmacology (Berl). 2017;234(19):2955–70.

    Article  CAS  Google Scholar 

  193. Solinas M, Justinova Z, Goldberg SR, Tanda G. Anandamide administration alone and after inhibition of fatty acid amide hydrolase (FAAH) increases dopamine levels in the nucleus accumbens shell in rats. J Neurochem. 2006;98(2):408–19.

    Article  CAS  PubMed  Google Scholar 

  194. Gobbi G, Bambico FR, Mangieri R, Bortolato M, Campolongo P, Solinas M, et al. Antidepressant-like activity and modulation of brain monoaminergic transmission by blockade of anandamide hydrolysis. Proc Natl Acad Sci USA. 2005;102(51):18620–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Wiley JL, Walentiny DM, Wright MJ, Beardsley PM, Burston JJ, Poklis JL, et al. Endocannabinoid contribution to Δ9-tetrahydrocannabinol discrimination in rodents. Eur J Pharmacol. 2014;15(737):97–105.

    Article  CAS  Google Scholar 

  196. Kathuria S, Gaetani S, Fegley D, Valiño F, Duranti A, Tontini A, et al. Modulation of anxiety through blockade of anandamide hydrolysis. Nat Med. 2003;9(1):76–81.

    Article  CAS  PubMed  Google Scholar 

  197. Piomelli D, Tarzia G, Duranti A, Tontini A, Mor M, Compton TR, et al. Pharmacological profile of the selective FAAH inhibitor KDS-4103 (URB597). CNS Drug Rev. 2006;12(1):21–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Huggins JP, Smart TS, Langman S, Taylor L, Young T. An efficient randomised, placebo-controlled clinical trial with the irreversible fatty acid amide hydrolase-1 inhibitor PF-04457845, which modulates endocannabinoids but fails to induce effective analgesia in patients with pain due to osteoarthritis of the knee. Pain. 2012;153(9):1837–46.

    Article  CAS  PubMed  Google Scholar 

  199. Li GL, Winter H, Arends R, Jay GW, Le V, Young T, et al. Assessment of the pharmacology and tolerability of PF-04457845, an irreversible inhibitor of fatty acid amide hydrolase-1, in healthy subjects. Br J Clin Pharmacol. 2012;73(5):706–16.

    Article  CAS  PubMed  Google Scholar 

  200. Pawsey S, Wood M, Browne H, Donaldson K, Christie M, Warrington S. Safety, tolerability and pharmacokinetics of FAAH inhibitor V158866: a double-blind, randomised, placebo-controlled phase i study in healthy volunteers. Drugs R D. 2016;16(2):181–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Kaur R, Sidhu P, Singh S. What failed BIA 10-2474 phase I clinical trial? Global speculations and recommendations for future Phase I trials. J Pharmacol Pharmacother. 2016;7(3):120–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Kerbrat A, Ferré J-C, Fillatre P, Ronzière T, Vannier S, Carsin-Nicol B, et al. Acute neurologic disorder from an inhibitor of fatty acid amide hydrolase. N Engl J Med. 2016;375(18):1717–25.

    Article  CAS  PubMed  Google Scholar 

  203. Janssen Research & Development, LLC Voluntarily Suspends Dosing in Phase 2 Clinical Trials of Experimental Treatment for Mood Disorders | Janssen [Internet]; 2016. https://web.archive.org/web/20160125052230/http://www.janssen.com/janssen-research-development-llc-voluntarily-suspends-dosing-phase-2-clinical-trials-experimental. Cited 12 May 2019.

  204. FAAH inhibitor safety under microscope after Bial drug trial death [Internet]. in-pharmatechnologist.com. https://www.in-pharmatechnologist.com/Article/2016/01/19/FAAH-inhibitor-safety-under-microscope-after-Bial-drug-trial-death. Cited 12 May 2019.

  205. van Esbroeck ACM, Janssen APA, Cognetta AB, Ogasawara D, Shpak G, van der Kroeg M, et al. Activity-based protein profiling reveals off-target proteins of the FAAH inhibitor BIA 10-2474. Science. 2017;356(6342):1084–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. FDA finds drugs under investigation in the U.S. related to French BIA 10-2474 drug do not pose similar safety risks. FDA [Internet]; 2018. http://www.fda.gov/drugs/drug-safety-and-availability/fda-finds-drugs-under-investigation-us-related-french-bia-10-2474-drug-do-not-pose-similar-safety. Cited 6 Aug 2019.

  207. Postnov A, Schmidt ME, Pemberton DJ, de Hoon J, van Hecken A, van den Boer M, et al. Fatty acid amide hydrolase inhibition by JNJ-42165279: a multiple-ascending dose and a positron emission tomography study in healthy volunteers. Clin Transl Sci. 2018;11(4):397–404.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Wagenlehner FME, van Till JWO, Houbiers JGA, Martina RV, Cerneus DP, Melis JHJM, et al. Fatty acid amide hydrolase inhibitor treatment in men with chronic prostatitis/chronic pelvic pain syndrome: an adaptive double-blind, randomized controlled trial. Urology. 2017;103:191–7.

    Article  PubMed  Google Scholar 

  209. D’Souza DC, Cortes-Briones J, Creatura G, Bluez G, Thurnauer H, Deaso E, et al. Efficacy and safety of a fatty acid amide hydrolase inhibitor (PF-04457845) in the treatment of cannabis withdrawal and dependence in men: a double-blind, placebo-controlled, parallel group, phase 2a single-site randomised controlled trial. Lancet Psychiatry. 2019;6(1):35–45.

    Article  PubMed  Google Scholar 

  210. Dinh TP, Kathuria S, Piomelli D. RNA interference suggests a primary role for monoacylglycerol lipase in the degradation of the endocannabinoid 2-arachidonoylglycerol. Mol Pharmacol. 2004;66(5):1260–4.

    Article  CAS  PubMed  Google Scholar 

  211. Long JZ, Nomura DK, Cravatt BF. Characterization of monoacylglycerol lipase inhibition reveals differences in central and peripheral endocannabinoid metabolism. Chem Biol. 2009;16(7):744–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Long JZ, Nomura DK, Vann RE, Walentiny DM, Booker L, ** X, et al. Dual blockade of FAAH and MAGL identifies behavioral processes regulated by endocannabinoid crosstalk in vivo. Proc Natl Acad Sci USA. 2009;106(48):20270–5.

    Article  PubMed  PubMed Central  Google Scholar 

  213. Nomura DK, Morrison BE, Blankman JL, Long JZ, Kinsey SG, Marcondes MCG, et al. Endocannabinoid hydrolysis generates brain prostaglandins that promote neuroinflammation. Science. 2011;334(6057):809–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Mulvihill MM, Nomura DK. Therapeutic potential of monoacylglycerol lipase inhibitors. Life Sci. 2013;92(8–9):492–7.

    Article  CAS  PubMed  Google Scholar 

  215. Nader J, Rapino C, Gennequin B, Chavant F, Francheteau M, Makriyannis A, et al. Prior stimulation of the endocannabinoid system prevents methamphetamine-induced dopaminergic neurotoxicity in the striatum through activation of CB2 receptors. Neuropharmacology. 2014;87:214–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Blanco E, Pavón FJ, Palomino A, Luque-Rojas MJ, Serrano A, Rivera P, et al. Cocaine-induced behavioral sensitization is associated with changes in the expression of endocannabinoid and glutamatergic signaling systems in the mouse prefrontal cortex. Int J Neuropsychopharmacol. 2014. https://doi.org/10.1093/ijnp/pyu024.

    Article  PubMed  PubMed Central  Google Scholar 

  217. Li W, Zhang C-L, Qiu Z-G. Differential expression of endocannabinoid system-related genes in the dorsal hippocampus following expression and reinstatement of morphine conditioned place preference in mice. Neurosci Lett. 2017;16(643):38–44.

    Article  CAS  Google Scholar 

  218. Schlosburg JE, Carlson BLA, Ramesh D, Abdullah RA, Long JZ, Cravatt BF, et al. Inhibitors of endocannabinoid-metabolizing enzymes reduce precipitated withdrawal responses in THC-dependent mice. AAPS J. 2009;11(2):342–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Muldoon PP, Chen J, Harenza JL, Abdullah RA, Sim-Selley LJ, Cravatt BF, et al. Inhibition of monoacylglycerol lipase reduces nicotine withdrawal. Br J Pharmacol. 2015;172(3):869–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Gamage TF, Ignatowska-Jankowska BM, Muldoon PP, Cravatt BF, Damaj MI, Lichtman AH. Differential effects of endocannabinoid catabolic inhibitors on morphine withdrawal in mice. Drug Alcohol Depend. 2015;1(146):7–16.

    Article  CAS  Google Scholar 

  221. Wilkerson JL, Ghosh S, Mustafa M, Abdullah RA, Niphakis MJ, Cabrera R, et al. The endocannabinoid hydrolysis inhibitor SA-57: intrinsic antinociceptive effects, augmented morphine-induced antinociception, and attenuated heroin seeking behavior in mice. Neuropharmacology. 2017;01(114):156–67.

    Article  CAS  Google Scholar 

  222. Di Marzo V, Fontana A, Cadas H, Schinelli S, Cimino G, Schwartz JC, et al. Formation and inactivation of endogenous cannabinoid anandamide in central neurons. Nature. 1994;372(6507):686–91.

    Article  PubMed  Google Scholar 

  223. Beltramo M, Stella N, Calignano A, Lin SY, Makriyannis A, Piomelli D. Functional role of high-affinity anandamide transport, as revealed by selective inhibition. Science. 1997;277(5329):1094–7.

    Article  CAS  PubMed  Google Scholar 

  224. Hillard CJ, Edgemond WS, Jarrahian A, Campbell WB. Accumulation of N-arachidonoylethanolamine (anandamide) into cerebellar granule cells occurs via facilitated diffusion. J Neurochem. 1997;69(2):631–8.

    Article  CAS  PubMed  Google Scholar 

  225. Fegley D, Kathuria S, Mercier R, Li C, Goutopoulos A, Makriyannis A, et al. Anandamide transport is independent of fatty-acid amide hydrolase activity and is blocked by the hydrolysis-resistant inhibitor AM1172. Proc Natl Acad Sci USA. 2004;101(23):8756–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Gianessi CA, Groman SM, Thompson SL, Jiang M, van der Stelt M, Taylor JR. Endocannabinoid contributions to alcohol habits and motivation: relevance to treatment. Addict Biol. 2019;6:e12768.

    Article  Google Scholar 

  227. Cippitelli A, Bilbao A, Gorriti MA, Navarro M, Massi M, Piomelli D, et al. The anandamide transport inhibitor AM404 reduces ethanol self-administration. Eur J Neurosci. 2007;26(2):476–86.

    Article  PubMed  Google Scholar 

  228. Gamaleddin I, Guranda M, Scherma M, Fratta W, Makriyannis A, Vadivel SK, et al. AM404 attenuates reinstatement of nicotine seeking induced by nicotine-associated cues and nicotine priming but does not affect nicotine- and food-taking. J Psychopharmacol. 2013;27(6):564–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Scherma M, Justinová Z, Zanettini C, Panlilio LV, Mascia P, Fadda P, et al. The anandamide transport inhibitor AM404 reduces the rewarding effects of nicotine and nicotine-induced dopamine elevations in the nucleus accumbens shell in rats. Br J Pharmacol. 2012;165(8):2539–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Vlachou S, Nomikos GG, Panagis G. Effects of endocannabinoid neurotransmission modulators on brain stimulation reward. Psychopharmacology (Berl). 2006;188(3):293–305.

    Article  CAS  Google Scholar 

  231. Vlachou S, Stamatopoulou F, Nomikos GG, Panagis G. Enhancement of endocannabinoid neurotransmission through CB1 cannabinoid receptors counteracts the reinforcing and psychostimulant effects of cocaine. Int J Neuropsychopharmacol. 2008;11(7):905–23.

    Article  CAS  PubMed  Google Scholar 

  232. Schindler CW, Scherma M, Redhi GH, Vadivel SK, Makriyannis A, Goldberg SR, et al. Self-administration of the anandamide transport inhibitor AM404 by squirrel monkeys. Psychopharmacology (Berl). 2016;233(10):1867–77.

    Article  CAS  Google Scholar 

  233. Levin FR, Mariani JJ, Brooks DJ, Pavlicova M, Cheng W, Nunes EV. Dronabinol for the treatment of cannabis dependence: a randomized, double-blind, placebo-controlled trial. Drug Alcohol Depend. 2011;116(1–3):142–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Herrmann ES, Cooper ZD, Bedi G, Ramesh D, Reed SC, Comer SD, et al. Effects of zolpidem alone and in combination with nabilone on cannabis withdrawal and a laboratory model of relapse in cannabis users. Psychopharmacology (Berl). 2016;233(13):2469–78.

    Article  CAS  Google Scholar 

  235. Vandrey R, Stitzer ML, Mintzer MZ, Huestis MA, Murray JA, Lee D. The dose effects of short-term dronabinol (oral THC) maintenance in daily cannabis users. Drug Alcohol Depend. 2013;128(1–2):64–70.

    Article  CAS  PubMed  Google Scholar 

  236. Haney M, Cooper ZD, Bedi G, Vosburg SK, Comer SD, Foltin RW. Nabilone decreases marijuana withdrawal and a laboratory measure of marijuana relapse. Neuropsychopharmacology. 2013;38(8):1557–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Levin FR, Mariani JJ, Pavlicova M, Brooks D, Glass A, Mahony A, et al. Dronabinol and lofexidine for cannabis use disorder: a randomized, double-blind, placebo-controlled trial. Drug Alcohol Depend. 2016;1(159):53–60.

    Article  CAS  Google Scholar 

  238. Haney M, Hart CL, Vosburg SK, Comer SD, Reed SC, Foltin RW. Effects of THC and lofexidine in a human laboratory model of marijuana withdrawal and relapse. Psychopharmacology (Berl). 2008;197(1):157–68.

    Article  CAS  Google Scholar 

  239. Budney AJ, Vandrey RG, Hughes JR, Moore BA, Bahrenburg B. Oral delta-9-tetrahydrocannabinol suppresses cannabis withdrawal symptoms. Drug Alcohol Depend. 2007;86(1):22–9.

    Article  CAS  PubMed  Google Scholar 

  240. Haney M, Hart CL, Vosburg SK, Nasser J, Bennett A, Zubaran C, et al. Marijuana withdrawal in humans: effects of oral THC or divalproex. Neuropsychopharmacology. 2004;29(1):158–70.

    Article  CAS  PubMed  Google Scholar 

  241. Jicha CJ, Lofwall MR, Nuzzo PA, Babalonis S, Elayi SC, Walsh SL. Safety of oral dronabinol during opioid withdrawal in humans. Drug Alcohol Depend. 2015;1(157):179–83.

    Article  CAS  Google Scholar 

  242. Bisaga A, Sullivan MA, Glass A, Mishlen K, Pavlicova M, Haney M, et al. The effects of dronabinol during detoxification and the initiation of treatment with extended release naltrexone. Drug Alcohol Depend. 2015;1(154):38–45.

    Article  CAS  Google Scholar 

  243. Lofwall MR, Babalonis S, Nuzzo PA, Elayi SC, Walsh SL. Opioid withdrawal suppression efficacy of oral dronabinol in opioid dependent humans. Drug Alcohol Depend. 2016;1(164):143–50.

    Article  CAS  Google Scholar 

  244. Samanta D. Cannabidiol: a review of clinical efficacy and safety in epilepsy. Pediatr Neurol. 2019;96:24–9.

    Article  PubMed  Google Scholar 

  245. Maroon J, Bost J. Review of the neurological benefits of phytocannabinoids. Surg Neurol Int. 2018;9:91.

    Article  PubMed  PubMed Central  Google Scholar 

  246. Bisogno T, Hanuš L, De Petrocellis L, Tchilibon S, Ponde DE, Brandi I, et al. Molecular targets for cannabidiol and its synthetic analogues: effect on vanilloid VR1 receptors and on the cellular uptake and enzymatic hydrolysis of anandamide. Br J Pharmacol. 2001;134(4):845–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  247. Thomas A, Baillie GL, Phillips AM, Razdan RK, Ross RA, Pertwee RG. Cannabidiol displays unexpectedly high potency as an antagonist of CB1 and CB2 receptor agonists in vitro. Br J Pharmacol. 2007;150(5):613–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  248. Pertwee RG. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: Δ9-tetrahydrocannabinol, cannabidiol and Δ9-tetrahydrocannabivarin. Br J Pharmacol. 2008;153(2):199–215.

    Article  CAS  PubMed  Google Scholar 

  249. Martínez-Pinilla E, Varani K, Reyes-Resina I, Angelats E, Vincenzi F, Ferreiro-Vera C, et al. Binding and signaling studies disclose a potential allosteric site for cannabidiol in cannabinoid CB2 receptors. Front Pharmacol. 2017;8:744.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Navarro G, Reyes-Resina I, Rivas-Santisteban R, Sánchez de Medina V, Morales P, Casano S, et al. Cannabidiol skews biased agonism at cannabinoid CB1 and CB2 receptors with smaller effect in CB1–CB2 heteroreceptor complexes. Biochem Pharmacol. 2018;157:148–58.

    Article  CAS  PubMed  Google Scholar 

  251. Tham M, Yilmaz O, Alaverdashvili M, Kelly MEM, Denovan-Wright EM, Laprairie RB. Allosteric and orthosteric pharmacology of cannabidiol and cannabidiol-dimethylheptyl at the type 1 and type 2 cannabinoid receptors. Br J Pharmacol. 2019;176(10):1455–69.

    Article  CAS  PubMed  Google Scholar 

  252. Luján MÁ, Castro-Zavala A, Alegre-Zurano L, Valverde O. Repeated Cannabidiol treatment reduces cocaine intake and modulates neural proliferation and CB1R expression in the mouse hippocampus. Neuropharmacology. 2018;143:163–75.

    Article  CAS  PubMed  Google Scholar 

  253. Galaj E, Bi G-H, Yang H-J, ** Z-X. Cannabidiol attenuates the rewarding effects of cocaine by CB2, 5-TH1A and TRPV1 receptor mechanisms. Neuropharmacology. 2019. https://doi.org/10.1016/j.neuropharm.2019.107740.

    Article  PubMed  PubMed Central  Google Scholar 

  254. Hay GL, Baracz SJ, Everett NA, Roberts J, Costa PA, Arnold JC, et al. Cannabidiol treatment reduces the motivation to self-administer methamphetamine and methamphetamine-primed relapse in rats. J Psychopharmacol (Oxford). 2018;32(12):1369–78.

    Article  CAS  Google Scholar 

  255. Viudez-Martínez A, García-Gutiérrez MS, Navarrón CM, Morales-Calero MI, Navarrete F, Torres-Suárez AI, et al. Cannabidiol reduces ethanol consumption, motivation and relapse in mice. Addict Biol. 2018;23(1):154–64.

    Article  CAS  PubMed  Google Scholar 

  256. Ren Y, Whittard J, Higuera-Matas A, Morris CV, Hurd YL. Cannabidiol, a nonpsychotropic component of cannabis, inhibits cue-induced heroin seeking and normalizes discrete mesolimbic neuronal disturbances. J Neurosci. 2009;29(47):14764–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  257. Bi G-H, Galaj E, He Y, ** Z-X. Cannabidiol inhibits sucrose self-administration by CB1 and CB2 receptor mechanisms in rodents. Addict Biol. 2019;19:e12783.

    Google Scholar 

  258. Katsidoni V, Anagnostou I, Panagis G. Cannabidiol inhibits the reward-facilitating effect of morphine: involvement of 5-HT1A receptors in the dorsal raphe nucleus. Addict Biol. 2013;18(2):286–96.

    Article  CAS  PubMed  Google Scholar 

  259. Markos JR, Harris HM, Gul W, ElSohly MA, Sufka KJ. Effects of cannabidiol on morphine conditioned place preference in mice. Planta Med. 2018;84(4):221–4.

    Article  CAS  PubMed  Google Scholar 

  260. Parker LA, Burton P, Sorge RE, Yakiwchuk C, Mechoulam R. Effect of low doses of delta9-tetrahydrocannabinol and cannabidiol on the extinction of cocaine-induced and amphetamine-induced conditioned place preference learning in rats. Psychopharmacology (Berl). 2004;175(3):360–6.

    Article  CAS  Google Scholar 

  261. Gonzalez-Cuevas G, Martin-Fardon R, Kerr TM, Stouffer DG, Parsons LH, Hammell DC, et al. Unique treatment potential of cannabidiol for the prevention of relapse to drug use: preclinical proof of principle. Neuropsychopharmacology. 2018;43(10):2036–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. de Carvalho CR, Takahashi RN. Cannabidiol disrupts the reconsolidation of contextual drug-associated memories in Wistar rats. Addict Biol. 2017;22(3):742–51.

    Article  CAS  PubMed  Google Scholar 

  263. Karimi-Haghighi S, Haghparast A. Cannabidiol inhibits priming-induced reinstatement of methamphetamine in REM sleep deprived rats. Prog Neuropsychopharmacol Biol Psychiatry. 2018;02(82):307–13.

    Article  CAS  Google Scholar 

  264. Bhargava HN. Effect of some cannabinoids on naloxone-precipitated abstinence in morphine-dependent mice. Psychopharmacology (Berl). 1976;49(3):267–70.

    Article  CAS  Google Scholar 

  265. Vilela LR, Gomides LF, David BA, Antunes MM, Diniz AB, Moreira F de A, et al. Cannabidiol rescues acute hepatic toxicity and seizure induced by cocaine. Mediators Inflamm [Internet]; 2015. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4427116/. Cited 2 Mar 2019.

  266. Taylor L, Gidal B, Blakey G, Tayo B, Morrison G. A phase I, randomized, double-blind, placebo-controlled, single ascending dose, multiple dose, and food effect trial of the safety, tolerability and pharmacokinetics of highly purified cannabidiol in healthy subjects. CNS Drugs. 2018;32(11):1053–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  267. Schoedel KA, Szeto I, Setnik B, Sellers EM, Levy-Cooperman N, Mills C, et al. Abuse potential assessment of cannabidiol (CBD) in recreational polydrug users: a randomized, double-blind, controlled trial. Epilepsy Behav. 2018;1(88):162–71.

    Article  Google Scholar 

  268. Manini AF, Yiannoulos G, Bergamaschi MM, Hernandez S, Olmedo R, Barnes AJ, et al. Safety and pharmacokinetics of oral cannabidiol when administered concomitantly with intravenous fentanyl in humans. J Addict Med. 2015;9:204–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  269. Dalton WS, Martz R, Lemberger L, Rodda BE, Forney RB. Influence of cannabidiol on delta-9-tetrahydrocannabinol effects. Clin Pharmacol Ther. 1976;19(3):300–9.

    Article  CAS  PubMed  Google Scholar 

  270. Karniol IG, Shirakawa I, Kasinski N, Pfeferman A, Carlini EA. Cannabidiol interferes with the effects of delta 9—tetrahydrocannabinol in man. Eur J Pharmacol. 1974;28(1):172–7.

    Article  CAS  PubMed  Google Scholar 

  271. Zuardi AW, Shirakawa I, Finkelfarb E, Karniol IG. Action of cannabidiol on the anxiety and other effects produced by delta 9-THC in normal subjects. Psychopharmacology (Berl). 1982;76(3):245–50.

    Article  CAS  Google Scholar 

  272. Haney M, Malcolm RJ, Babalonis S, Nuzzo PA, Cooper ZD, Bedi G, et al. Oral cannabidiol does not alter the subjective, reinforcing or cardiovascular effects of smoked cannabis. Neuropsychopharmacology. 2016;41(8):1974–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  273. Solowij N, Broyd S, Greenwood L-M, van Hell H, Martelozzo D, Rueb K, et al. A randomised controlled trial of vaporised Δ9-tetrahydrocannabinol and cannabidiol alone and in combination in frequent and infrequent cannabis users: acute intoxication effects. Eur Arch Psychiatry Clin Neurosci. 2019;269(1):17–35.

    Article  PubMed  Google Scholar 

  274. Allsop DJ, Copeland J, Lintzeris N, Dunlop AJ, Montebello M, Sadler C, et al. Nabiximols as an agonist replacement therapy during cannabis withdrawal: a randomized clinical trial. JAMA Psychiatry. 2014;71(3):281–91.

    Article  CAS  PubMed  Google Scholar 

  275. Trigo JM, Soliman A, Quilty LC, Fischer B, Rehm J, Selby P, et al. Nabiximols combined with motivational enhancement/cognitive behavioral therapy for the treatment of cannabis dependence: a pilot randomized clinical trial. PLoS One. 2018;13(1):e0190768.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  276. Mediavilla V, Steinemann S. Essential oil of Cannabis sativa L strains. J Int Hemp Assoc. 1997;4:82–4.

    Google Scholar 

  277. Sharma C, Al Kaabi JM, Nurulain SM, Goyal SN, Kamal MA, Ojha S. Polypharmacological properties and therapeutic potential of β-caryophyllene: a dietary phytocannabinoid of pharmaceutical promise. Curr Pharm Des. 2016;22(21):3237–64.

    Article  CAS  PubMed  Google Scholar 

  278. Corey EJ, Mitra RB, Hisashi U. Total synthesis of d, l-caryophyllene and d, l-isocaryophyllene. J Am Chem Soc. 1964;86(3):485–92.

    Article  CAS  Google Scholar 

  279. Gertsch J, Leonti M, Raduner S, Racz I, Chen J-Z, **e X-Q, et al. Beta-caryophyllene is a dietary cannabinoid. Proc Natl Acad Sci USA. 2008;105(26):9099–104.

    Article  PubMed  PubMed Central  Google Scholar 

  280. Varga ZV, Matyas C, Erdelyi K, Cinar R, Nieri D, Chicca A, et al. β-Caryophyllene protects against alcoholic steatohepatitis by attenuating inflammation and metabolic dysregulation in mice. Br J Pharmacol. 2018;175(2):320–34.

    Article  CAS  PubMed  Google Scholar 

  281. Liu H, Yang G, Tang Y, Cao D, Qi T, Qi Y, et al. Physicochemical characterization and pharmacokinetics evaluation of β-caryophyllene/β-cyclodextrin inclusion complex. Int J Pharm. 2013;450(1–2):304–10.

    Article  CAS  PubMed  Google Scholar 

  282. Schmitt D, Levy R, Carroll B. Toxicological evaluation of β-caryophyllene oil: subchronic toxicity in rats. Int J Toxicol. 2016;35(5):558–67.

    Article  CAS  PubMed  Google Scholar 

  283. Oliveira GL da S, Machado KC, Machado KC, da Silva APDSCL, Feitosa CM, de Castro Almeida FR. Non-clinical toxicity of β-caryophyllene, a dietary cannabinoid: absence of adverse effects in female Swiss mice. Regul Toxicol Pharmacol. 2018;92:338–46.

  284. Al Mansouri S, Ojha S, Al Maamari E, Al Ameri M, Nurulain SM, Bahi A. The cannabinoid receptor 2 agonist, β-caryophyllene, reduced voluntary alcohol intake and attenuated ethanol-induced place preference and sensitivity in mice. Pharmacol Biochem Behav. 2014;124:260–8.

    Article  CAS  PubMed  Google Scholar 

  285. Rose JE, Behm FM. Inhalation of vapor from black pepper extract reduces smoking withdrawal symptoms. Drug Alcohol Depend. 1994;34(3):225–9.

    Article  CAS  PubMed  Google Scholar 

  286. He Y, Galaj E, Bi GH, Wang XF, Gardner EL, ** Z-X. Beta-caryophyllene: a dietary cannabis terpene, inhibits nicotine-taking and nicotine-seeking behavior in rodents. Br J Pharmacol. 2019 (in press).

  287. Gill EW, Paton WDM, Pertwee RG. Preliminary experiments on the chemistry and pharmacology of cannabis. Nature. 1970;228:134–6.

    Article  CAS  PubMed  Google Scholar 

  288. Bolognini D, Costa B, Maione S, Comelli F, Marini P, Di Marzo V, et al. The plant cannabinoid Delta9-tetrahydrocannabivarin can decrease signs of inflammation and inflammatory pain in mice. Br J Pharmacol. 2010;160(3):677–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  289. McPartland JM, Duncan M, Di Marzo V, Pertwee RG. Are cannabidiol and Δ(9)-tetrahydrocannabivarin negative modulators of the endocannabinoid system? A systematic review. Br J Pharmacol. 2015;172(3):737–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  290. Thomas A, Stevenson LA, Wease KN, Price MR, Baillie G, Ross RA, et al. Evidence that the plant cannabinoid Delta9-tetrahydrocannabivarin is a cannabinoid CB1 and CB2 receptor antagonist. Br J Pharmacol. 2005;146(7):917–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  291. Pertwee RG, Thomas A, Stevenson LA, Ross RA, Varvel SA, Lichtman AH, et al. The psychoactive plant cannabinoid, Delta9-tetrahydrocannabinol, is antagonized by Delta8- and Delta9-tetrahydrocannabivarin in mice in vivo. Br J Pharmacol. 2007;150(5):586–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  292. Dennis I, Whalley BJ, Stephens GJ. Effects of Δ9-tetrahydrocannabivarin on [35S]GTPγS binding in mouse brain cerebellum and piriform cortex membranes. Br J Pharmacol. 2008;154(6):1349–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  293. Hill AJ, Weston SE, Jones NA, Smith I, Bevan SA, Williamson EM, et al. Δ9-Tetrahydrocannabivarin suppresses in vitro epileptiform and in vivo seizure activity in adult rats. Epilepsia. 2010;51(8):1522–32.

    Article  CAS  PubMed  Google Scholar 

  294. Cascio MG, Zamberletti E, Marini P, Parolaro D, Pertwee RG. The phytocannabinoid, Δ9-tetrahydrocannabivarin, can act through 5-HT1A receptors to produce antipsychotic effects. Br J Pharmacol. 2015;172(5):1305–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  295. De Petrocellis L, Ligresti A, Moriello AS, Allarà M, Bisogno T, Petrosino S, et al. Effects of cannabinoids and cannabinoid-enriched Cannabis extracts on TRP channels and endocannabinoid metabolic enzymes. Br J Pharmacol. 2011;163(7):1479–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  296. Janssens A, Silvestri C, Martella A, Vanoevelen JM, Di Marzo V, Voets T. Δ9-tetrahydrocannabivarin impairs epithelial calcium transport through inhibition of TRPV5 and TRPV6. Pharmacol Res. 2018;136:83–9.

    Article  CAS  PubMed  Google Scholar 

  297. Anavi-Goffer S, Baillie G, Irving AJ, Gertsch J, Greig IR, Pertwee RG, et al. Modulation of l-α-lysophosphatidylinositol/GPR55 mitogen-activated protein kinase (MAPK) signaling by cannabinoids. J Biol Chem. 2012;287(1):91–104.

    Article  CAS  PubMed  Google Scholar 

  298. Riedel G, Fadda P, McKillop-Smith S, Pertwee RG, Platt B, Robinson L. Synthetic and plant-derived cannabinoid receptor antagonists show hypophagic properties in fasted and non-fasted mice. Br J Pharmacol. 2009;156(7):1154–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  299. Wargent ET, Zaibi MS, Silvestri C, Hislop DC, Stocker CJ, Stott CG, et al. The cannabinoid Δ(9)-tetrahydrocannabivarin (THCV) ameliorates insulin sensitivity in two mouse models of obesity. Nutr Diabetes. 2013;27(3):e68.

    Article  CAS  Google Scholar 

  300. Bátkai S, Mukhopadhyay P, Horváth B, Rajesh M, Gao RY, Mahadevan A, et al. Δ8-Tetrahydrocannabivarin prevents hepatic ischaemia/reperfusion injury by decreasing oxidative stress and inflammatory responses through cannabinoid CB2 receptors. Br J Pharmacol. 2012;165(8):2450–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  301. ** Z-X, Muldoon P, Wang XF, Bi G-H, Damaj IM, Lichtman AH, et al. Δ8-Tetrahydrocannabivarin has potent anti-nicotine effects in multiple rodent models of nicotine dependence. Br J Pharmacol. 2019. https://doi.org/10.1111/bph.14844.

    Article  PubMed  PubMed Central  Google Scholar 

  302. Englund A, Atakan Z, Kralj A, Tunstall N, Murray R, Morrison P. The effect of five day dosing with THCV on THC-induced cognitive, psychological and physiological effects in healthy male human volunteers: a placebo-controlled, double-blind, crossover pilot trial. J Psychopharmacol (Oxford). 2016;30(2):140–51.

    Article  CAS  Google Scholar 

  303. Jadoon KA, Ratcliffe SH, Barrett DA, Thomas EL, Stott C, Bell JD, et al. Efficacy and safety of cannabidiol and tetrahydrocannabivarin on glycemic and lipid parameters in patients with type 2 diabetes: a randomized, double-blind, placebo-controlled, parallel group pilot study. Diabetes Care. 2016;39(10):1777–86.

    Article  CAS  PubMed  Google Scholar 

  304. Tudge L, Williams C, Cowen PJ, McCabe C. Neural effects of cannabinoid CB1 neutral antagonist tetrahydrocannabivarin on food reward and aversion in healthy volunteers. Int J Neuropsychopharmacol. 2014. https://doi.org/10.1093/ijnp/pyu094.

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Zheng-**ong **.

Ethics declarations

Funding

This research was supported by the Intramural Research Program of the National Institute on Drug Abuse, National Institutes of Health, USA.

Conflict of interest

Ewa Galaj and Zheng-**ong ** have no conflicts of interest that are directly relevant to the content of this article.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Galaj, E., **, ZX. Potential of Cannabinoid Receptor Ligands as Treatment for Substance Use Disorders. CNS Drugs 33, 1001–1030 (2019). https://doi.org/10.1007/s40263-019-00664-w

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40263-019-00664-w

Navigation