1
|
Gil-Martins E, Barbosa DJ, Borges F, Remião F, Silva R. Toxicodynamic insights of 2C and NBOMe drugs - Is there abuse potential? Toxicol Rep 2025; 14:101890. [PMID: 39867514 PMCID: PMC11762925 DOI: 10.1016/j.toxrep.2025.101890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
Drug use represents a prevalent and multifaceted societal problem, with profound implications for public health, social welfare, and economic stability. To circumvent strict international drug control regulations, there is a growing trend in the development and market introduction of novel psychoactive substances (NPS), encompassing a wide range of compounds with psychoactive properties. This includes, among other classes of drugs, the phenethylamines. Originally derived from natural sources, these drugs have garnered particular attention due to their psychedelic effects. They comprise a broad spectrum of compounds, including 2,5-dimethoxyphenylethylamine (2C) drugs and their corresponding N-(2,5-dimethoxybenzyl)phenethylamine (NBOMe). Psychedelics are conventionally perceived as having low addiction potential, although recent reports have raised concerns regarding this topic. These substances primarily interact with serotonin receptors, particularly the 5-HT2A subtype, resulting in alterations in sensory perception, mood, and introspective experiences. In addition to their psychedelic properties, 2C and NBOMe drugs have been associated with a multitude of adverse effects, such as cardiovascular complications and neurotoxicity. This manuscript provides a comprehensive review of the psychedelic pathways underlying 2C and NBOMe designer drugs, focusing on their interactions with serotonergic and other neurotransmitter systems, shedding light on their potential for abuse.
Collapse
Affiliation(s)
- Eva Gil-Martins
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Daniel José Barbosa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, Gandra, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Gandra, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Fernanda Borges
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Renata Silva
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
2
|
Bilel S, Miliano C, Corli G, Bassi M, Trusel M, Tonini R, De Luca MA, Marti M. Acute Effects of the Psychedelic Phenethylamine 25I-NBOMe in C57BL/6J Male Mice. Int J Mol Sci 2025; 26:2815. [PMID: 40141457 PMCID: PMC11943083 DOI: 10.3390/ijms26062815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
25I-NBOMe (4-Iodo-2,5-dimethoxy-N-(2-methoxybenzyl) phenethylamine) is a synthetic psychedelic compound abused for its ambiguous legal state as a counterfeit lysergic acid diethylamide (LSD). 25I-NBOMe acts as a selective agonist of 5HT2A receptors leading to hallucinations, intoxications, and fatalities. Here, we assessed the rewarding properties of 25I-NBOMe and its behavioral and neurotoxic acute effects on the central nervous system of C57BL/6J mice. We evaluated the dopamine (DA) levels using in vivo microdialysis in the nucleus accumbens (NAc) shell after 25I-NBOMe (0.1-1 mg/kg i.p.) injection. We also investigated the effects of 25I-NBOMe (0.1-1 mg/kg i.p.) on locomotor activity, reaction time, and prepulse inhibition. Moreover, we assessed the acute 25I-NBOMe (1 µM) effects on synaptic transmission and plasticity in the medial prefrontal cortex (mPFC) by using ex vivo electrophysiology. Our findings suggest that 25I-NBOMe affects the DA transmission in NAc shell at the highest dose tested, increases the reaction time within 30 min after the administration, and disrupts the PPI. In slices, it prevents long-term synaptic potentiation (LTP) in the mPFC, an effect that could not be reverted by the co-administration of the selective 5HT2A antagonist (MDL100907). Overall, these findings provide valuable new insights into the effects of 25I-NBOMe and the associated risks of its use.
Collapse
Affiliation(s)
- Sabrine Bilel
- Department of Translational Medicine, Section of Legal Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (S.B.); (G.C.); (M.B.)
| | - Cristina Miliano
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy;
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Giorgia Corli
- Department of Translational Medicine, Section of Legal Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (S.B.); (G.C.); (M.B.)
| | - Marta Bassi
- Department of Translational Medicine, Section of Legal Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (S.B.); (G.C.); (M.B.)
| | - Massimo Trusel
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, 16163 Genova, Italy (R.T.)
| | - Raffaella Tonini
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, 16163 Genova, Italy (R.T.)
| | | | - Matteo Marti
- Department of Translational Medicine, Section of Legal Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (S.B.); (G.C.); (M.B.)
- Collaborative Center for the Italian National Early Warning System, Department of Anti-Drug Policies, Presidency of the Council of Ministers, 00186 Roma, Italy
| |
Collapse
|
3
|
Alhosan N, Cavallo D, Santiago M, Kelly E, Henderson G. Slow dissociation kinetics of fentanyls and nitazenes correlates with reduced sensitivity to naloxone reversal at the μ-opioid receptor. Br J Pharmacol 2025; 182:969-987. [PMID: 39437833 DOI: 10.1111/bph.17376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND AND PURPOSE Fentanyls and nitazenes are μ-opioid receptor agonists responsible for a large number of opioid overdose deaths. Here, we determined the potency, dissociation kinetics and antagonism by naloxone at the μ receptor of several fentanyl and nitazene analogues, compared to morphine and DAMGO. EXPERIMENTAL APPROACH In vitro assays of G protein activation and signalling and arrestin recruitment were performed. AtT20 cells expressing μ receptors were loaded with a membrane potential dye and changes in fluorescence used to determine agonist potency, dissociation kinetics and susceptibility to antagonism by naloxone. BRET experiments were undertaken in HEK293T cells expressing μ receptors to assess Gi protein activation and β-arrestin 2 recruitment. KEY RESULTS The apparent rate of agonist dissociation from the μ receptor varied: morphine, DAMGO, alfentanil and fentanyl dissociated rapidly, whereas isotonitazene, etonitazene, ohmefentanyl and carfentanil dissociated slowly. Slowly dissociating agonists were more resistant to antagonism by naloxone. For carfentanil, the slow apparent rate of dissociation was not because of G protein receptor kinase-mediated arrestin recruitment as its apparent rate of dissociation was not increased by inhibition of G protein-coupled receptor kinases (GRKs) with Compound 101. The in vitro relative potencies of fentanyls and nitazenes compared to morphine were much lower than that previously observed in in vivo experiments. CONCLUSIONS AND IMPLICATIONS With fentanyls and nitazenes that slowly dissociate from the μ receptor, antagonism by naloxone is pseudo-competitive. In overdoses involving fentanyls and nitazenes, higher doses of naloxone may be required for reversal than those normally used to reverse heroin overdose.
Collapse
Affiliation(s)
- Norah Alhosan
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
- College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Damiana Cavallo
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Marina Santiago
- Macquarie Medical School, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Eamonn Kelly
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Graeme Henderson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| |
Collapse
|
4
|
Witley S, Edvardsson CE, Aranäs C, Tufvesson-Alm M, Stalberga D, Green H, Vestlund J, Jerlhag E. Des-acyl ghrelin reduces alcohol intake and alcohol-induced reward in rodents. Transl Psychiatry 2024; 14:277. [PMID: 38965230 PMCID: PMC11224403 DOI: 10.1038/s41398-024-02996-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/23/2024] [Accepted: 06/27/2024] [Indexed: 07/06/2024] Open
Abstract
The mechanisms contributing to alcohol use disorder (AUD) are complex and the orexigenic peptide ghrelin, which enhances alcohol reward, is implied as a crucial modulator. The major proportion of circulating ghrelin is however the non-octanoylated form of ghrelin, des-acyl ghrelin (DAG), whose role in reward processes is unknown. As recent studies show that DAG decreases food intake, we hypothesize that DAG attenuates alcohol-related responses in animal models. Acute and repeated DAG treatment dose-dependently decreased alcohol drinking in male and female rats. In these alcohol-consuming male rats, repeated DAG treatment causes higher levels of dopamine metabolites in the ventral tegmental area, an area central to reward processing. The role of DAG in reward processing is further supported as DAG prevents alcohol-induced locomotor stimulation, reward in the conditioned place preference paradigm, and dopamine release in the nucleus accumbens in male rodents. On the contrary, DAG does not alter the memory of alcohol reward or affect neurotransmission in the hippocampus, an area central to memory. Further, circulating DAG levels are positively correlated with alcohol drinking in female but not male rats. Studies were conducted in attempts to identify tentative targets of DAG, which currently are unknown. Data from these recombinant cell system revealed that DAG does not bind to either of the monoamine transporters, 5HT2A, CB1, or µ-opioid receptors. Collectively, our data show that DAG attenuates alcohol-related responses in rodents, an effect opposite to that of ghrelin, and contributes towards a deeper insight into behaviors regulated by the ghrelinergic signaling pathway.
Collapse
Affiliation(s)
- Sarah Witley
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Christian E Edvardsson
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Cajsa Aranäs
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Maximilian Tufvesson-Alm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Darta Stalberga
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Faculty of Medicine, Linköping University, Linköping, Sweden
| | - Henrik Green
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Faculty of Medicine, Linköping University, Linköping, Sweden
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden
| | - Jesper Vestlund
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
5
|
Herian M, Świt P. 25X-NBOMe compounds - chemistry, pharmacology and toxicology. A comprehensive review. Crit Rev Toxicol 2023; 53:15-33. [PMID: 37115704 DOI: 10.1080/10408444.2023.2194907] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Recently, a growing number of reports have indicated a positive effect of hallucinogenic-based therapies in different neuropsychiatric disorders. However, hallucinogens belonging to the group of new psychoactive substances (NPS) may produce high toxicity. NPS, due to their multi-receptors affinity, are extremely dangerous for the human body and mental health. An example of hallucinogens that have been lately responsible for many severe intoxications and deaths are 25X-NBOMes - N-(2-methoxybenzyl)-2,5-dimethoxy-4-substituted phenethylamines, synthetic compounds with strong hallucinogenic properties. 25X-NBOMes exhibit a high binding affinity to serotonin receptors but also to dopamine, adrenergic and histamine receptors. Apart from their influence on perception, many case reports point out systemic and neurological poisoning with these compounds. In humans, the most frequent side effects are tachycardia, anxiety, hypertension and seizures. Moreover, preclinical studies confirm that 25X-NBOMes cause developmental impairments, cytotoxicity, cardiovascular toxicity and changes in behavior of animals. Metabolism of NBOMes seems to be very complex and involves many metabolic pathways. This fact may explain the observed high toxicity. In addition, many analytical methods have been applied in order to identify these compounds and their metabolites. The presented review summarized the current knowledge about 25X-NBOMes, especially in the context of toxicity.
Collapse
Affiliation(s)
- Monika Herian
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Paweł Świt
- Institute of Chemistry, Faculty of Science and Technology, University of Silesia, Katowice, Poland
| |
Collapse
|
6
|
Xie B, Le Rouzic VP, Goldberg A, Tsai MHM, Chen L, Zhang T, Sinha A, Pan YX, Baumann MH, Shi L. Binding preference at the μ-opioid receptor underlies distinct pharmacology of cyclopropyl versus valeryl analogs of fentanyl. Neuropharmacology 2023; 227:109442. [PMID: 36731721 PMCID: PMC9974845 DOI: 10.1016/j.neuropharm.2023.109442] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/12/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023]
Abstract
Illicitly manufactured fentanyl is driving the current opioid crisis, and various fentanyl analogs are appearing in recreational drug markets worldwide. To assess the potential health risks posed by fentanyl analogs, it is necessary to understand structure-activity relationships for these compounds. Here we compared the pharmacology of two structurally related fentanyl analogs implicated in opioid overdose: cyclopropylfentanyl and valerylfentanyl. Cyclopropylfentanyl has a three-carbon ring attached to the carbonyl group on the fentanyl scaffold, whereas valerylfentanyl has a four-carbon chain at the same position. In vitro assays examining μ-opioid receptor (MOR) coupling to G proteins in CHO cells showed that cyclopropylfentanyl is a full agonist (EC50 = 8.6 nM, %Emax = 113%), with potency and efficacy similar to fentanyl (EC50 = 10.3 nM, %Emax = 113%). By contrast, valerylfentanyl is a partial agonist at MOR (EC50 = 179.8 nM, %Emax = 60%). Similar results were found in assays assessing MOR-mediated β-arrestin recruitment in HEK cells. In vivo studies in male CD-1 mice demonstrated that both fentanyl analogs induce naloxone-reversible antinociception and respiratory suppression, but cyclopropylfentanyl is 100-times more potent as an antinociceptive agent (ED50 = 0.04 mg/kg, s. c.) than valerylfentanyl (ED50 = 4.0 mg/kg, s. c.). Molecular simulation results revealed that the alkyl chain of valerylfentanyl cannot be well accommodated by the active state of MOR and may transition the receptor toward an inactive state, converting the fentanyl scaffold to a partial agonist. Taken together, our results suggest that cyclopropylfentanyl presents much greater risk of adverse effects when compared to valerylfentanyl. Moreover, the summed findings may provide clues to the design of therapeutic opioids with reduced adverse side effects.
Collapse
Affiliation(s)
- Bing Xie
- Computational Chemistry and Molecular Biophysics Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Valerie P Le Rouzic
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Alexander Goldberg
- Computational Chemistry and Molecular Biophysics Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Meng-Hua M Tsai
- Computational Chemistry and Molecular Biophysics Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Li Chen
- Computational Chemistry and Molecular Biophysics Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Tiffany Zhang
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Antara Sinha
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Ying-Xian Pan
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA; Department of Anesthesiology, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Michael H Baumann
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
7
|
Deventer MH, Persson M, Laus A, Pottie E, Cannaert A, Tocco G, Gréen H, Stove CP. Off-target activity of NBOMes and NBOMe analogs at the µ opioid receptor. Arch Toxicol 2023; 97:1367-1384. [PMID: 36853332 DOI: 10.1007/s00204-023-03465-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/16/2023] [Indexed: 03/01/2023]
Abstract
New psychoactive substances (NPS) are introduced on the illicit drug market at a rapid pace. Their molecular targets are often inadequately elucidated, which contributes to the delayed characterization of their pharmacological effects. Inspired by earlier findings, this study set out to investigate the µ opioid receptor (MOR) activation potential of a large set of psychedelics, substances which typically activate the serotonin (5-HT2A) receptor as their target receptor. We observed that some substances carrying the N-benzyl phenethylamine (NBOMe) structure activated MOR, as confirmed by both the NanoBiT® βarr2 recruitment assay and the G protein-based AequoScreen® Ca2+ release assay. The use of two orthogonal systems proved beneficial as some aspecific, receptor independent effects were found for various analogs when using the Ca2+ release assay. The specific 'off-target' effects at MOR could be blocked by the opioid antagonist naloxone, suggesting that these NBOMes occupy the same common opioid binding pocket as conventional opioids. This was corroborated by molecular docking, which revealed the plausibility of multiple interactions of 25I-NBOMe with MOR, similar to those observed for opioids. Additionally, structure-activity relationship findings seen in vitro were rationalized in silico for two 25I-NBOMe isomers. Overall, as MOR activity of these psychedelics was only noticed at high concentrations, we consider it unlikely that for the tested compounds there will be a relevant opioid toxicity in vivo at physiologically relevant concentrations. However, small modifications to the original NBOMe structure may result in a panel of more efficacious and potent MOR agonists, potentially exhibiting a dual MOR/5-HT2A activation potential.
Collapse
Affiliation(s)
- Marie H Deventer
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| | - Mattias Persson
- Department of Forensic Genetic and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden
| | - Antonio Laus
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Eline Pottie
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| | - Annelies Cannaert
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| | - Graziella Tocco
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Henrik Gréen
- Department of Forensic Genetic and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden.,Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Christophe P Stove
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium.
| |
Collapse
|
8
|
Finlay DB, Mackie W, Webb HDJ, Thomsen LR, Nimick M, Rosengren RJ, Marusich JA, Glass M, Wiley JL. The piperazine analogue para-fluorophenylpiperazine alters timing of the physiological effects of the synthetic cannabinoid receptor agonist AMB-FUBINACA, without changing its discriminative stimulus, signalling effects, or metabolism. Pharmacol Biochem Behav 2023; 223:173530. [PMID: 36805861 PMCID: PMC10020421 DOI: 10.1016/j.pbb.2023.173530] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
AMB-FUBINACA is a synthetic cannabinoid receptor agonist (SCRA), which has been associated with substantial abuse and health harm since 2016 in many countries including New Zealand. A characteristic of AMB-FUBINACA use in New Zealand has included the observation that forensic samples (from autopsies) and drugs seized by police have often been found to contain para-fluorophenylpiperazine (pFPP), a relatively little-characterised piperazine analogue that has been suggested to act through 5HT1a serotonin receptors. In the current study, we aimed to characterise the interactions of these two agents in rat physiological endpoints using plethysmography and telemetry, and to examine whether pFPP altered the subjective effects of AMB-FUBINACA in mice trained to differentiate a cannabinoid (THC) from vehicle. Though pFPP did not alter the ability of AMB-FUBINACA to substitute for THC, it did appear to abate some of the physiological effects of AMB-FUBINACA in rats by delaying the onset of AMB-FUBINACA-mediated hypothermia and shortening duration of bradycardia. In HEK cells stably expressing the CB1 cannabinoid receptor, 5HT1a, or both CB1 and 5HT1a, cAMP signalling was recorded using a BRET biosensor (CAMYEL) to assess possible direct receptor interactions. Although low potency pFPP agonism at 5HT1a was confirmed, little evidence for signalling interactions was detected in these assays: additive or synergistic effects on potency or efficacy were not detected between pFPP and AMB-FUBINACA-mediated cAMP inhibition. Experiments utilising higher potency, classical 5HT1a ligands (agonist 8OH-DPAT and antagonist WAY100635) also failed to reveal evidence for mutual CB1/5HT1a interactions or cross-antagonism. Finally, the ability of pFPP to alter the metabolism of AMB-FUBINACA in rat and human liver microsomes into its primary carboxylic acid metabolite via carboxylesterase-1 was assessed by HPLC; no inhibition was detected. Overall, the effects we have observed do not suggest that increased harm/toxicity would result from the combination of pFPP and AMB-FUBINACA.
Collapse
Affiliation(s)
- David B Finlay
- Department of Pharmacology & Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.
| | - Warwick Mackie
- Department of Pharmacology & Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Hunter D J Webb
- Department of Pharmacology & Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Lucy R Thomsen
- Department of Pharmacology & Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Mhairi Nimick
- Department of Pharmacology & Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Rhonda J Rosengren
- Department of Pharmacology & Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | | | - Michelle Glass
- Department of Pharmacology & Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | | |
Collapse
|
9
|
Vandeputte MM, Vasudevan L, Stove CP. In vitro functional assays as a tool to study new synthetic opioids at the μ-opioid receptor: Potential, pitfalls and progress. Pharmacol Ther 2022; 235:108161. [DOI: 10.1016/j.pharmthera.2022.108161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/30/2022] [Accepted: 02/14/2022] [Indexed: 10/19/2022]
|
10
|
Pottie E, Stove CP. In vitro assays for the functional characterization of (psychedelic) substances at the serotonin receptor 5-HT 2A R. J Neurochem 2022; 162:39-59. [PMID: 34978711 DOI: 10.1111/jnc.15570] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/24/2021] [Accepted: 12/29/2021] [Indexed: 12/20/2022]
Abstract
Serotonergic psychedelics are substances that induce alterations in mood, perception, and thought, and have the activation of serotonin (5-HT) 2A receptors (5-HT2A Rs) as a main pharmacological mechanism. Besides their appearance on the (illicit) drug market, e.g. as new psychoactive substances, their potential therapeutic application is increasingly explored. This group of substances demonstrates a broad structural variety, leading to insufficiently described structure-activity relationships, hence illustrating the need for better functional characterization. This review therefore elaborates on the in vitro molecular techniques that have been used the most abundantly for the characterization of (psychedelic) 5-HT2A R agonists. More specifically, this review covers assays to monitor the canonical G protein signaling pathway (e.g. measuring G protein recruitment/activation, inositol phosphate accumulation, or Ca2+ mobilization), assays to monitor non-canonical G protein signaling (such as arachidonic acid release), assays to monitor β-arrestin recruitment or signaling, and assays to monitor receptor conformational changes. In particular, focus lies on the mechanism behind the techniques, and the specific advantages and challenges that are associated with these. Additionally, several variables are discussed that one should consider when attempting to compare functional outcomes from different studies, both linked to the specific assay mechanism and linked to its specific execution, as these may heavily impact the assay outcome.
Collapse
Affiliation(s)
- Eline Pottie
- Laboratory of Toxicology, Faculty of Pharmaceutical Sciences, Department of Bioanalysis, Ghent University, Ghent, Belgium
| | - Christophe P Stove
- Laboratory of Toxicology, Faculty of Pharmaceutical Sciences, Department of Bioanalysis, Ghent University, Ghent, Belgium
| |
Collapse
|
11
|
Kronstrand R, Norman C, Vikingsson S, Biemans A, Valencia Crespo B, Edwards D, Fletcher D, Gilbert N, Persson M, Reid R, Semenova O, Al Teneiji F, Wu X, Dahlén J, NicDaéid N, Tarbah F, Sutcliffe OB, McKenzie C, Gréen H. The metabolism of the synthetic cannabinoids ADB-BUTINACA and ADB-4en-PINACA and their detection in forensic toxicology casework and infused papers seized in prisons. Drug Test Anal 2021; 14:634-652. [PMID: 34811926 DOI: 10.1002/dta.3203] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/08/2021] [Accepted: 11/08/2021] [Indexed: 12/13/2022]
Abstract
Early warning systems detect new psychoactive substances (NPS), while dedicated monitoring programs and routine drug and toxicology testing identify fluctuations in prevalence. We report the increasing prevalence of the synthetic cannabinoid receptor agonist (SCRA) ADB-BUTINACA (N-[1-amino-3,3-dimethyl-1-oxobutan-2-yl]-1-butyl-1H-indazole-3-carbox-amide). ADB-BUTINACA was first detected in a seizure in Sweden in 2019, and we report its detection in 13 routine Swedish forensic toxicology cases soon after. In January 2021, ADB-BUTINACA was detected in SCRA-infused papers seized in Scottish prisons and has rapidly increased in prevalence, being detected in 60.4% of the SCRA-infused papers tested between January and July 2021. In this work, ADB-BUTINACA was incubated with human hepatocytes (HHeps), and 21 metabolites were identified in vitro, 14 being detected in authentic case samples. The parent drug and metabolites B9 (mono-hydroxylation on the n-butyl tail) and B16 (mono-hydroxylation on the indazole ring) are recommended biomarkers in blood, while metabolites B4 (dihydrodiol formation on the indazole core), B9, and B16 are suitable biomarkers in urine. ADB-4en-PINACA (N-[1-amino-3,3-dimethyl-1-oxobutan-2-yl]-1-[pent-4-en-1-yl]-1H-indazole-3-carboxamide) was detected in Scottish prisons in December 2020, but, unlike ADB-BUTINACA, prevalence has remained low. ADB-4en-PINACA was incubated with HHeps, and 11 metabolites were identified. Metabolites E3 (dihydrodiol formed in the tail moiety) and E7 (hydroxylation on the linked/head group) are the most abundant metabolites in vitro and are suggested as urinary biomarkers. The in vitro potencies of ADB-BUTINACA (EC50 , 11.5 nM and ADB-4en-PINACA (EC50 , 11.6 nM) are similar to that of MDMB-4en-PINACA (EC50 , 4.3 nM). A third tert-leucinamide SCRA, ADB-HEXINACA was also detected in prison samples and warrants further investigation.
Collapse
Affiliation(s)
- Robert Kronstrand
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden.,Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Caitlyn Norman
- Leverhulme Research Centre for Forensic Science, School of Science and Engineering, University of Dundee, Dundee, UK
| | - Svante Vikingsson
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden.,Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden.,RTI International, Research Triangle, North Carolina, USA
| | - Anoek Biemans
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Bryan Valencia Crespo
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Darren Edwards
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, UK
| | - Daniel Fletcher
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, UK.,BioAscent, Motherwell, UK
| | - Nicolas Gilbert
- Department of Natural Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| | - Mattias Persson
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden
| | - Robert Reid
- Leverhulme Research Centre for Forensic Science, School of Science and Engineering, University of Dundee, Dundee, UK
| | - Olga Semenova
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, UK
| | - Faisal Al Teneiji
- Leverhulme Research Centre for Forensic Science, School of Science and Engineering, University of Dundee, Dundee, UK.,General Department of Forensic Science and Criminology, Toxicology Department, Dubai Police, Dubai, United Arab Emirates
| | - Xiongyu Wu
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Johan Dahlén
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Niamh NicDaéid
- Leverhulme Research Centre for Forensic Science, School of Science and Engineering, University of Dundee, Dundee, UK
| | - Fuad Tarbah
- General Department of Forensic Science and Criminology, Toxicology Department, Dubai Police, Dubai, United Arab Emirates
| | - Oliver B Sutcliffe
- Department of Natural Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| | - Craig McKenzie
- Leverhulme Research Centre for Forensic Science, School of Science and Engineering, University of Dundee, Dundee, UK.,Chiron AS, Trondheim, Norway
| | - Henrik Gréen
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden.,Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
12
|
Rudin D, Liechti ME, Luethi D. Molecular and clinical aspects of potential neurotoxicity induced by new psychoactive stimulants and psychedelics. Exp Neurol 2021; 343:113778. [PMID: 34090893 DOI: 10.1016/j.expneurol.2021.113778] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/25/2021] [Accepted: 05/29/2021] [Indexed: 12/20/2022]
Abstract
New psychoactive stimulants and psychedelics continue to play an important role on the illicit new psychoactive substance (NPS) market. Designer stimulants and psychedelics both affect monoaminergic systems, although by different mechanisms. Stimulant NPS primarily interact with monoamine transporters, either as inhibitors or as substrates. Psychedelic NPS most potently interact with serotonergic receptors and mediate their mind-altering effects mainly through agonism at serotonin 5-hydroxytryptamine-2A (5-HT2A) receptors. Rarely, designer stimulants and psychedelics are associated with potentially severe adverse effects. However, due to the high number of emerging NPS, it is not possible to investigate the toxicity of each individual substance in detail. The brain is an organ particularly sensitive to substance-induced toxicity due to its high metabolic activity. In fact, stimulant and psychedelic NPS have been linked to neurological and cognitive impairments. Furthermore, studies using in vitro cell models or rodents indicate a variety of mechanisms that could potentially lead to neurotoxic damage in NPS users. Cytotoxicity, mitochondrial dysfunction, and oxidative stress may potentially contribute to neurotoxicity of stimulant NPS in addition to altered neurochemistry. Serotonin 5-HT2A receptor-mediated toxicity, oxidative stress, and activation of mitochondrial apoptosis pathways could contribute to neurotoxicity of some psychedelic NPS. However, it remains unclear how well the current preclinical data of NPS-induced neurotoxicity translate to humans.
Collapse
Affiliation(s)
- Deborah Rudin
- Division of Clinical Pharmacology and Toxicology, University Hospital Basel and University of Basel, Basel, Switzerland; Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Dino Luethi
- Division of Clinical Pharmacology and Toxicology, University Hospital Basel and University of Basel, Basel, Switzerland; Institute of Pharmacology, Medical University of Vienna, Vienna, Austria; Institute of Applied Physics, TU Wien, Vienna, Austria.
| |
Collapse
|
13
|
Pham DNK, Chadeayne AR, Golen JA, Manke DR. 5-Meth-oxy- N, N-di- n-propyl-tryptamine (5-MeO-DPT): freebase and fumarate. Acta Crystallogr E Crystallogr Commun 2021; 77:522-526. [PMID: 34026257 PMCID: PMC8100262 DOI: 10.1107/s2056989021003753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 04/07/2021] [Indexed: 11/10/2022]
Abstract
The solid-state structures of the synthetic psychedelic 5-meth-oxy-N,N-di-n-propyl-tryptamine (5-MeO-DPT) {systematic name: N-[2-(5-meth-oxy-1H-indol-3-yl)eth-yl]-N-propyl-propan-1-amine}, C17H25N2O, and its fumarate salt, bis-(5-meth-oxy-N,N-di-n-propyl-tryptammonium) fumarate (systematic name: bis-{N-[2-(5-meth-oxy-1H-indol-3-yl)eth-yl]-N-propyl-propan-1-aminium} but-2-ene-dio-ate), 2C17H25N2O+·C4H2O4 2-, are reported. The freebase has a single tryptamine mol-ecule in the asymmetric unit. The mol-ecules are linked together by N-H⋯N hydrogen bonds in zigzag chains along the [010] direction. The fumarate salt has a single tryptammonium cation and half of a fumarate dianion in the asymmetric unit. The tryptammonium and fumarate ions are held together in one-dimensional chains by a series of N-H⋯O hydrogen bonds. These chains are combinations of R 4 4(22) rings, and C 2 2(14) and C 4 4(28) parallel chains along [001].
Collapse
Affiliation(s)
- Duyen N. K. Pham
- University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA
| | | | - James A. Golen
- University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA
| | - David R. Manke
- University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA
| |
Collapse
|
14
|
Pharmacokinetics and pharmacodynamics of cyclopropylfentanyl in male rats. Psychopharmacology (Berl) 2021; 238:3629-3641. [PMID: 34613431 PMCID: PMC8629808 DOI: 10.1007/s00213-021-05981-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/06/2021] [Indexed: 01/29/2023]
Abstract
BACKGROUND Illicitly manufactured fentanyl and its analogs are a major driving force behind the ongoing opioid crisis. Cyclopropylfentanyl is a fentanyl analog associated with many overdose deaths, but limited knowledge is available about its pharmacology. In the present study, we developed a bioanalytical method for the determination of cyclopropylfentanyl and its main metabolite cyclopropylnorfentanyl and evaluated pharmacokinetic-pharmacodynamic relationships in rats. METHOD An ultra-high performance liquid chromatography tandem mass spectrometry (UHPLC-MS/MS) method was developed and validated for determination of cyclopropylfentanyl and cyclopropylnorfentanyl in rat plasma. Male Sprague-Dawley rats fitted with jugular catheters and temperature transponders received cyclopropylfentanyl (30, 100, and 300 μg/kg) or saline subcutaneously. Blood specimens were withdrawn over an 8-h time period, along with measurements of pharmacodynamic endpoints. RESULTS The analytical method was validated, and both analytes exhibited a low limit of quantification (15 pg/mL). Cyclopropylfentanyl caused dose-related increases in hot plate latency (ED50 = 48 µg/kg) and catalepsy (ED50 = 87 µg/kg) and produced long-lasting hypothermia at the highest dose. Plasma cyclopropylfentanyl rose rapidly in a dose-related fashion, reaching maximal concentration (Cmax) after 15-28 min, whereas metabolite Cmax occurred later at 45-90 min. Cyclopropylfentanyl Cmax values were similar to concentrations measured in non-fatal intoxications in humans; however, differences in parent drug: metabolite ratio indicated possible interspecies variance in metabolism. CONCLUSION Our study shows that cyclopropylfentanyl produces typical opioid-like effects in male rats. Cyclopropylfentanyl displays much greater analgesic potency when compared to morphine, suggesting that cyclopropylfentanyl poses increased overdose risk for unsuspecting users.
Collapse
|