Signalling profiles of a structurally diverse panel of synthetic cannabinoid receptor agonists

https://doi.org/10.1016/j.bcp.2020.113871Get rights and content

Abstract

Synthetic cannabinoid receptor agonists (SCRAs) represent the most rapidly proliferating class of “designer drugs” or “new psychoactive substances”. SCRAs offer unregulated alternatives to cannabis that evade routine drug tests, but their use is increasingly associated with severe toxicity and death worldwide. Little is currently known about SCRA molecular pharmacology, or the mechanisms underpinning their toxicity, although the effects are believed to be primarily mediated by the type 1 cannabinoid receptor (CB1). In this study, we aimed to characterise the signalling profiles of a structurally diverse panel of novel SCRAs at CB1. We compare SCRAs to traditional reference cannabinoids CP55,940, WIN55,212-2, and THC. The activity of the SCRAs was assessed in key receptor signalling and regulatory pathways, including cAMP production, translocation of β-arrestin 1 and 2, and receptor internalisation. The activity profiles of the ligands were also evaluated using operational analysis to identify ligand bias. Results revealed that SCRAs activities were relatively balanced in the pathways evaluated (compared to WIN55,212-2), although 5F-CUMYL-P7AICA and XLR-11 possessed partial efficacy in cAMP stimulation and β-arrestin translocation. Notably, the SCRAs showed distinct potency and efficacy profiles compared to THC. In particular, while the majority of SCRAs demonstrated robust β-arrestin translocation, cAMP stimulation, and internalisation, THC failed to elicit high efficacy responses in any of these assays. Further study is required to delineate if these pathways could contribute to SCRA toxicity in humans.

Introduction

A long history of recreational and medicinal use of cannabis (Cannabis sativa) prompted the investigation into, and the subsequent discovery of the endocannabinoid system. This included the study of the related Gαi/o protein-coupled receptors; type 1 cannabinoid receptor (CB1) [1], found abundantly throughout the central nervous system and type 2 cannabinoid receptor (CB2), located primarily in immune cells [2], [3]. Following the discovery and elucidation of the structure of the primary bioactive component of cannabis, Δ9-tetrahydrocannabinol (THC) [4], synthetic analogues of THC were developed for pharmacological interrogation of the endocannabinoid system for potential medicinal effects.

The THC-like in vivo effects of many synthetic cannabinoids has resulted in their diversion to recreational drugs in an attempt to create “legal” or unregulated alternatives to cannabis. In the 2000s, designer synthetic cannabinoids were included in herbal incense products branded “Spice” or “AK-47 24 Karat Gold”, which contained the compounds JWH-018 and JWH-073 [5], [6]. New generation synthetic cannabinoid receptor agonists (SCRAs) with no precedent in the scientific literature are constantly emerging, enabling producers to change the composition of compounds in order to circumvent laws [7]. SCRAs now represent the most rapidly proliferating class of New Psychoactive Substances (NPS), with over 260 reported to the United Nations Office on Drugs and Crime as of 2018 [8].

Synthetic cannabinoid use is a growing social and health issue. Cases of severe adverse effects and death linked to recreational use of SCRAs have been reported internationally, in contrast to the relatively low incidence of toxicity reported for cannabis [9], [10], [11]. However, molecular pharmacology data and potential mechanisms of toxicity for many of these compounds are currently limited. The apparently greater abuse liability and toxicity of SCRAs compared to cannabis, may be linked to their ability to potently and efficaciously activate cannabinoid receptors (Fig. 1). For example, when compared to traditional research agonists like CP55,940, THC induces potent CB1-mediated activation of ERK phosphorylation and inhibition of adenylate cyclase but with lower efficacy in these pathways [12], [13]. In contrast, indole or indazole derivatives detected in illicit synthetic preparations, such as 5F-CUMYL-PICA, AB-PINACA, AMB-FUBINACA and MDMB-FUBINACA display higher potency and efficacy at CB1-mediated inhibition of cAMP production [14], [15], [16]. This suggests the profile or pattern of responses produced by toxic SCRAs differs from THC, which may help explain their enhanced toxicity.

Interestingly, within the indole and indazole SCRA families, varying degrees of activity (and potentially toxicity) have been associated with specific structural features. For instance, the inclusion of an azaindole core (an indole analogue) in 5F-CUMYL-P7AICA conferred reduced CB1 affinity and functional activity compared to the indazole and indole cores of 5F-CUMYL PINACA and 5F-CUMYL PICA, respectively [17]. Similarly, terminal fluorination of the N-pentyl indole of XLR-11 was found to increase CB1 potency compared to the non-fluorinated analogue, UR-144 [18], [19]. Additionally, particular classes of compounds (indoles and indazole-3-carboxamides with amino acid derivative side-chains, like AMB-FUBINACA and XLR-11) seem to be more frequently encountered in cases of SCRA-related toxicity and death, in contrast to structurally-related compounds like 5F-CUMYL-PINACA and 5F-MDMB-PICA – despite all compounds being in the NPS marketplace and exhibiting equivalently high efficacy in in vitro signalling assays [9], [17], [19], [20]. While it is challenging to compare toxicities because the exact use and dose profiles of sufficient numbers of SCRAs are not known, this might suggest that small structural alterations between ligands may induce different active receptor conformations to differentially activate downstream signalling pathways (termed “biased agonism” or “functional selectivity”), which may have implications for toxicity in vivo [21]. Further insight into SCRA pharmacology may help identify such biased signalling profiles, and particular structural conformations or chemical features responsible for their effects.

This study has characterised the molecular pharmacology of a structurally-diverse panel of novel synthetic cannabinoids at CB1 (Fig. 2). The activity profiles of select compounds were compared in a battery of in vitro assays; cAMP production, β-arrestin translocation and receptor internalisation. To explore differences in the activity profiles of the synthetic cannabinoids, evaluation of functional selectivity was also performed to allow comparisons between the phytocannabinoid THC, and classical research cannabinoids, CP55,940 and WIN55,212-2.

Section snippets

Drugs

CP55,940 was purchased from Cayman Chemical Company (Ann Arbour, MI, USA); forskolin and WIN55,212-2 were obtained from Tocris Bioscience (Bristol, UK); (−)-trans9- tetrahydrocannabinol (THC) was purchased from THC Pharm GmbH (Frankfurt, Germany). Synthetic cannabinoids were provided by Dr Samuel Banister (University of Sydney, Australia), with the exception of 4-cyano CUMYL-BUTINACA (4CN-CUMYL-BUTINACA), AB-CHMINACA, AB-FUBINACA and XLR-11, which were obtained from Cayman Chemical Company

SCRAs display full efficacy in cAMP signalling

The canonical signalling pathway of CB1 involves coupling to Gαi proteins, resulting in inhibition of adenylate cyclase and consequent reduction in cAMP concentration [2], [31]. To determine CB1 agonist behaviour, we examined the ability of a select group of SCRAs to inhibit adenylate cyclase using a real-time BRET-CAMYEL assay in HEK 293 3HA-hCB1 cells. Congruent with previous studies [12], [26], we found WIN55,212-2, CP55,940, and THC generated concentration-dependent inhibition of

Discussion

The increasing emergence of novel SCRAs as recreational drugs raises concerns surrounding their pharmacological action and subsequent effects upon human consumption [38]. SCRA-related fatalities and hospitalisation are a growing global public health problem [39], with over 70 deaths linked to synthetic cannabinoid use in New Zealand in the past two years [40]. The current study has utilised several methods to explore the molecular pharmacology of synthetic cannabinoids and found that a diverse

Acknowledgements

MP and JJM are recipients of the University of Otago Doctoral Scholarship. DBF is supported by a Lottery Health Research Postdoctoral Fellowship. Research was supported by grants from the Maurice and Phyllis Paykel Trust and Health Research Council of New Zealand to MG.

Conflicts of interest

The authors declare no conflicts of interest.

Author contributions

MP designed and performed experiments, analysed data and wrote the paper. JJM and DBF performed experiments, assisted with data analysis, and revised drafts of the manuscript. JAJ provided the arrestin assay. SDB synthesised compounds used in experiments. NG contributed to experimental design and oversaw aspects of the project. MG oversaw all elements of the project, obtained the funding, and reviewed drafts of the paper. All authors reviewed and approved the final version of the paper.

References (64)

  • K. Mackie

    Distribution of cannabinoid receptors in the central and peripheral nervous system

    Handb. Exp. Pharmacol.

    (2005)
  • L.A. Matsuda et al.

    Structure of a cannabinoid receptor and functional expression of the cloned cDNA

    Nature

    (1990)
  • S. Munro et al.

    Molecular characterization of a peripheral receptor for cannabinoids

    Nature

    (1993)
  • Y. Gaoni et al.

    Isolation, structure, and partial synthesis of an active constituent of hashish

    J. Am. Chem. Soc.

    (1964)
  • V. Auwarter et al.

    'Spice' and other herbal blends: harmless incense or cannabinoid designer drugs?

    J. Mass Spectrom.

    (2009)
  • R. Lindigkeit et al.

    Spice: a never ending story?

    Foren. Sci. Int.

    (2009)
  • S.D. Banister et al.

    The chemistry and pharmacology of synthetic cannabinoid receptor agonist new psychoactive substances: evolution

    Handb. Exp. Pharmacol.

    (2018)
  • United Nation Office on Drugs and Crime. World Drug Report 2017....
  • L.M. Labay et al.

    Synthetic cannabinoid drug use as a cause or contributory cause of death

    Foren. Sci. Int.

    (2016)
  • M. Hermanns-Clausen et al.

    Acute toxicity due to the confirmed consumption of synthetic cannabinoids: clinical and laboratory findings

    Addiction

    (2013)
  • W.E. Fantegrossi et al.

    Distinct pharmacology and metabolism of K2 synthetic cannabinoids compared to Delta(9)-THC: mechanism underlying greater toxicity?

    Life Sci.

    (2014)
  • E. Khajehali et al.

    Biased agonism and biased allosteric modulation at the CB1 cannabinoid receptor

    Mol. Pharmacol.

    (2015)
  • D.W. Bonhaus et al.

    Dual activation and inhibition of adenylyl cyclase by cannabinoid receptor agonists: evidence for agonist-specific trafficking of intracellular responses

    J. Pharmacol. Exp. Ther.

    (1998)
  • W.J. Costain et al.

    Pharmacological characterization of emerging synthetic cannabinoids in HEK293T cells and hippocampal neurons

    Eur. J. Pharmacol.

    (2016)
  • T.F. Gamage et al.

    Molecular and behavioral pharmacological characterization of abused synthetic cannabinoids MMB- and MDMB-FUBINACA, MN-18, NNEI, CUMYL-PICA, and 5-Fluoro-CUMYL-PICA

    J. Pharmacol. Exp. Ther.

    (2018)
  • D.B. Finlay et al.

    Do toxic synthetic cannabinoid receptor agonists have signature in vitro activity profiles? A case study of AMB-FUBINACA

    ACS Chem. Neurosci.

    (2019)
  • S.D. Banister et al.

    Synthesis and pharmacology of new psychoactive substance 5F-CUMYL-P7AICA, a scaffold hopping analogue of synthetic cannabinoid receptor agonists 5F-CUMYL-PICA and 5F-CUMYL-PINACA

    Drug Test Anal.

    (2018)
  • J.L. Wiley et al.

    Cannabinoids in disguise: Delta9-tetrahydrocannabinol-like effects of tetramethylcyclopropyl ketone indoles

    Neuropharmacology

    (2013)
  • S.D. Banister et al.

    Effects of bioisosteric fluorine in synthetic cannabinoid designer drugs JWH-018, AM-2201, UR-144, XLR-11, PB-22, 5F-PB-22, APICA, and STS-135

    ACS Chem. Neurosci.

    (2015)
  • S.D. Banister et al.

    Pharmacology of Valinate and tert-Leucinate Synthetic Cannabinoids 5F-AMBICA, 5F-AMB, 5F-ADB, AMB-FUBINACA, MDMB-FUBINACA, MDMB-CHMICA, and Their Analogues

    ACS Chem. Neurosci.

    (2016)
  • E. Wouters et al.

    Insights into biased signaling at cannabinoid receptors: synthetic cannabinoid receptor agonists

    Biochem. Pharmacol.

    (2019)
  • E.E. Cawston et al.

    Real-time characterization of cannabinoid receptor 1 (CB1) allosteric modulators reveals novel mechanism of action

    Br. J. Pharmacol.

    (2013)
  • B.K. Atwood et al.

    Expression of G protein-coupled receptors and related proteins in HEK293, AtT20, BV2, and N18 cell lines as revealed by microarray analysis

    BMC Genomics

    (2011)
  • L.I. Jiang et al.

    Use of a cAMP BRET sensor to characterize a novel regulation of cAMP by the sphingosine 1-phosphate/G13 pathway

    J. Biol. Chem.

    (2007)
  • M.S. Ibsen et al.

    Cannabinoid CB1 and CB2 receptor-mediated arrestin translocation: species, subtype, and agonist-dependence

    Front. Pharmacol.

    (2019)
  • D.B. Finlay et al.

    Galphas signalling of the CB1 receptor and the influence of receptor number

    Br. J. Pharmacol.

    (2017)
  • N.L. Grimsey et al.

    A novel high-throughput assay for the quantitative assessment of receptor trafficking

    Clin. Exp. Pharmacol. Physiol.

    (2008)
  • D.B. Finlay et al.

    GPR18 undergoes a high degree of constitutive trafficking but is unresponsive to N-Arachidonoyl Glycine

    PeerJ

    (2016)
  • E. van der Westhuizen et al.

    Quantification of ligand bias for clinically relevant beta2-adrenergic receptor ligands: implications for drug taxonomy

    Mol. Pharmacol.

    (2014)
  • X. Zhu et al.

    An evaluation of the operational model when applied to quantify functional selectivity

    Br. J. Pharmacol.

    (2018)
  • C.C. Felder et al.

    Comparison of the pharmacology and signal transduction of the human cannabinoid CB1 and CB2 receptors

    Mol. Pharmacol.

    (1995)
  • M. Glass et al.

    Agonist selective regulation of G proteins by cannabinoid CB(1) and CB(2) receptors

    Mol. Pharmacol.

    (1999)
  • Cited by (0)

    View full text