Abstract
Novel psychoactive substances (NPS) are new drugs of abuse. Over the last 10 years 50–100 new NPS have been detected for the first time each year. This has led to numerous deaths and challenges to healthcare providers and law‐makers worldwide. We review preclinical studies of NPS and discuss how these studies have influenced legislative decisions. We focus on the UK legal system but include experiences from Europe. We reviewed manuscripts from 2008–2019 and have summarised the in vitro and in vivo data on NPS, highlighting how these studies define pharmacological mechanisms and how they might predict harm in humans. We found that only a small percentage of NPS have been examined in preclinical studies. Most preclinical studies of NPS focus on basic pharmacological mechanisms (46% of studies reviewed) and/or addictive liability (32%) rather than toxicity and harm (24%). Very few preclinical studies into NPS include data from chronic dosing schedules (9%) or female rodents (4%). We conclude that preclinical studies can predict harm to humans, but most of the predictions are based on basic pharmacology rather than demonstrated toxicity. Some of these studies have been used to make changes to the law in the UK and Europe and perhaps, because of the paucity of toxicology data, most NPS have been placed in the highly dangerous schedule 1 or Class A category in the UK. We suggest that in silico studies and high throughput toxicology screens might be the most economical way forward to rapidly screen the health harms of NPS.
Keywords: abuse, legal high, legislation, novel psychoactive substance, toxicity
1. INTRODUCTION
Over the last decade, the drug scene has completely changed. In 2018 a new novel psychoactive substance (NPS) was reported to the European Commission Early Warning System every week, with 55 NPS reported in total. This is a much lower number than that reported in 2013–2015.1 Despite the fact that the observed decline could have resulted from many factors such as international legislation or possible lack of innovation, many substances continued to resurge globally,1 indicating a new established illicit repertoire. Other approaches employed to capture the true number of NPS that are globally available includes using a 24/7 web crawler such as the NPS.Finder.2 This novel approach showed that the online scenario includes over 4000 unique psychoactive molecules of interest to psychonauts, a number that is approximately 4‐fold the number being reported to the known NPS databases (European Monitoring Centre on Drugs and Drug Addiction [EMCDDA] and United Nations Office on Drugs and Crimes).2
In the UK, NPS were originally known as “psychoactive drugs which are not prohibited by the United Nations Single Convention on Narcotic Drugs or by the Misuse of Drugs Act 1971, and which people in the UK are seeking for intoxicant use”.3 They were therefore initially emerging to circumvent international legislation. In the UK, the Psychoactive Substances Act 2016 broadly defined them as “any substance, which is capable of producing a psychoactive effect”,4 covering any substance that is not controlled by the Misuse of Drugs Act 1971. Under the latter definition, the UK Home Office issued a forensic strategy to test for the psychoactivity of an unknown substance,5 which is key for prosecution in the UK.6 In this respect, in vitro testing was recommended by the Advisory Council on the Misuse of Drugs (ACMD) to prove if a substance is psychoactive. This mandates positive receptor and functional assays at 1 or more of the following receptors or transporters: https://www.guidetopharmacology.org/GRAC/ObjectDisplayForward?objectId=56 (cannabinoid receptor type 1), https://www.guidetopharmacology.org/GRAC/FamilyDisplayForward?familyId=72 (γ‐aminobutyric acid A receptor), https://www.guidetopharmacology.org/GRAC/ObjectDisplayForward?objectId=6 (serotonin 2A receptor), https://www.guidetopharmacology.org/GRAC/FamilyDisplayForward?familyId=75 (N‐methyl‐D‐aspartate receptor), https://www.guidetopharmacology.org/GRAC/ObjectDisplayForward?objectId=319 receptor and https://www.guidetopharmacology.org/GRAC/FamilyDisplayForward?familyId=176 (https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=940, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=5, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=484). However, since in vitro testing may not be suitable for all substances (e.g. nitrous oxide and solvents), in vivo testing has been recommended as an alternative. in vitro testing can predict human doses for stimulants and psychedelics, for example7 but cannot easily predict human pharmacokinetics or brain penetration. Information may also be retrieved from scientific literature if in vitro/in vivo testing cannot be performed.5
Despite the apparent decline in the numbers of emerging NPS, research is lagging behind the rapid changes in this complex market. Case reports including clinical and forensic cases, and emergency and mental health admissions have shown that NPS are associated with unpredictable acute adverse events, serious harms and even deaths.8 Knowledge of long‐term effects following chronic use of NPS is lacking and systems that capture harms resulting from NPS are still patchy.9
Animal models have been previously validated and have played pivotal roles in preclinical research informing the neurobiological, psychopathological, behavioural and aetiological aspects underlying drug addiction and acute/chronic drug use.10, 11 They have long been used to inform on the various phases of drug addiction including drug self‐administration patterns, behavioural criteria that define vulnerability to drug abuse, conditioned place preference, craving and relapse, escalation of drug‐taking, impulsivity, continued drug‐taking despite adverse drug reactions, physical dependence and other neuropsychopathological aspects, which lead from a voluntary drug‐taking to compulsive behaviours.10, 11, 12, 13, 14 Furthermore, preclinical studies have contributed to understanding the toxicity spectrum associated with some traditional drugs of abuse including knowledge of neurotoxic effects seen with https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=4804 15 and https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=4574,16 as well as non‐neurological toxic effects such as MDMA‐induced heart‐valve issues17 and bladder problems associated with https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=4233 use.18
Recent reviews of NPS included studies which focussed on toxicity such as Assi et al.19 who reviewed the literature between 2007–2015 for NPS toxicity, yielding 20 studies and concluded that the harmful effects of NPS could be severe or lethal. Tracy et al.20 summarised NPS‐induced health harms as deduced from available clinical data, while Evans‐Brown and Sedefov reviewed the NPS risk assessment protocols across the EU.21
Here we review the preclinical studies of NPS over the last 10 years and summarise the findings from the main classes of NPS. We then examine the evidence that has been used in changing the legal status of NPS focussing on the UK with some examples from Europe.
2. MATERIALS AND METHODS
A literature search was performed on PubMed and Web‐of‐Science (Medline), from January 2008 to November 2019. We employed database‐specific search strategies with multiple keywords utilising word truncation/wild card symbols and index terms as appropriate for each database. The literature search was not undertaken using individually named NPS due to their large number, up to 4000 by some estimates2; consequently, not all pertinent studies may be identified.
Each identified article was categorised into the following topics (based on title and abstract review): pharmacological and behavioural experiments (preclinical studies), human studies (clinical), detection and identification (forensic), legal status (legal), epidemiology, comments, letters, replies (reports), and reviews.
2.1. Search strategy and study selection of NPS
The following search terms were used in PubMed:
(((((((legal high[Title/Abstract]) OR novel psychoactive substance[Title/Abstract]) OR new psychoactive substance[Title/Abstract]) OR bath salt[Title/Abstract]) OR designer drug[Title/Abstract]) OR plant food[Title/Abstract])) AND ((((((((((in‐vitro[Title/Abstract]) OR in‐vivo[Title/Abstract]) OR cell[Title/Abstract]) OR slice[Title/Abstract]) OR anesthetized[Title/Abstract]) OR anaesthetised[Title/Abstract]) OR rat[Title/Abstract]) OR mouse[Title/Abstract]) OR murine[Title/Abstract]) OR rodent[Title/Abstract]) NOT review.
In Web‐of‐Science, we selected the Medline database with the following search terms:
TI = (legal highs* or novel psychoactive substance* or new psychoactive substance* or bath salt* or designer drug* or plant food*) and MH = (in‐vivo* or in‐vitro* or cell* or slice* or anesthetised* or rat* or mouse* or rodents*).
The literature search yielded 335 articles from both PubMed and Web‐of‐Science (Figure 1). An additional 22 articles were deemed to be relevant. Forty‐seven articles were excluded due to duplicates. In total, 310 articles were screened for inclusion, and 66 were excluded as they were deemed irrelevant; 205 articles were screened for eligibility, and 136 were excluded in the final screening because they did not meet the following criteria: (i) NPS was not a major outcome; (ii) the article was a clinical study; (iii) the article was a forensic study; or (iv) the article was a review. In total, 108 journal articles from the original search were used in the final analysis (Figure 1).
Figure 1.
Flow diagram showing how articles were screened after PubMed and Medline searches. NPS = novel psychoactive substance
2.2. Nomenclature of targets and ligands
Key protein targets and ligands in this article are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY.
3. SUMMARY OF RESULTS FROM NPS SEARCHES
After excluding ineligible manuscripts, we found 55 animal (in vitro or in vivo) studies examining cathinones, 25 studies on stimulants, only 8 studies on synthetic cannabinoid receptor agonists (SCRAs) and 20 studies on other NPS. We have grouped the types of studies undertaken into 4 broad areas: (i) behavioural models of addiction; (ii) neurochemistry/neuropharmacology of addiction; (iii) toxicity; and (iv) cognitive dysfunction.
3.1. Cathinones
The most commonly studied cathinone was MDPV (3,4‐methylenedioxypyrovalerone; 25 studies [45%]), followed by mephedrone and methylone (16 studies [29%]) and α‐PVP (α‐pyrrolidinovalerophenone; 7 studies [13%]). All other cathinones studied were only examined in 3 or fewer manuscripts: (Table 1). Most studies examined these substances in models of addiction such as increased locomotion, locomotor sensitisation, conditioned place preference, drug self‐administration, increased intracranial brain stimulation and withdrawal effects; there were 31 (56%) such studies. The next most common type of study were neurochemical assays associated with dopamine, serotonin or noradrenaline; these mostly examined changes in these transmitter levels or effects on their transporters; there were 17 such studies (31%). The next most common type of study was related to toxicity: cell death, cell toxicity, reactive oxygen species or hyperthermia, and there were 11 such studies (20%). Three studies (5%) examined cardiovascular issues such as blood pressure or heart rate, 1 (2%) looked at hepatotoxicity, 3 (5%) looked at cognitive deficits such as memory impairment and 1 study (2%) examined the effects of cathinones on rodent offspring. Only 2 studies (4%) examined chronic effects of cathinones and only 2 studies (4%) reported using female rodents.
Table 1.
Pre‐clinical studies examining cathinone novel psychoactive substance (NPS) pharmacology. The most recent manuscripts are presented first
Study | DOI | NPS | Dose/conc | Species/sex | Age/weight | Samples | Assay | Main results |
---|---|---|---|---|---|---|---|---|
Dias da Silva, D. et al., 201922 | 10.1007/s00204‐019‐02539‐x | Metaphedrone | 31 nM to 10mM 1 μM,10 μM, 100 μM and 500 μM | Male Wistar Han rats | 150–250 g | Hepatocytes | Cytotoxic assays | ↑ autophagic and apoptotic/necrotic mechanisms |
Gannon, B.M. et al., 201923 | 10.1007/s00213‐018‐5046‐x | MDPV and methylone | 0.32 mg/kg/inf | Male Sprague–Dawley rats | 275–300 g | Self‐administration | Methylone + caffeine results in an enhanced reinforcing effectiveness compared to methylone alone | |
Luethi, D. et al., 201924 | 10.1177/0269881119844185 | Methylone and MDPV | Human embryonic kidney 293 cells | Monoamine transporter assays |
↑ potency to inhibit NA uptake with methylone compared with DA and 5‐HT uptake. ↓ transporter inhibition potencies with N‐demethylation of methylone. = NA and DA uptake inhibition potencies with O‐demethylenation of methylone and MDPV. |
|||
Blough, B.E. et al., 201925 | 10.1007/s00213‐018‐5063‐9 | Methcathinone analogs | Male Sprague–Dawley rats | 300–400 g | Synaptosomes |
Compounds substituted at the 2‐position (ortho) were primarily dopaminergic Compounds Substituted at the 4‐position (Para) were found to be more serotonergic |
||
Zhou, X. et al., 201926 | 10.3390/ijms20071561 | 3‐methylmethcathinone, mephedrone, methylone, MDPV, α‐PVP, and naphyrone | C2C12 myoblasts | Cytotoxic assays | All cathinones showed cell membrane integrity, depleted ATP levels, and increased mitochondrial superoxide concentrations α‐PVP and naphyrone impaired basal and maximal cellular respiration | |||
Gerecsei, L.I. et al., 201927 | MDPV | Mice | Neural tube | Cytotoxic assays, western blotting |
↑ apoptotic cells in the pallium and in the subpallium ↓NR2B expression in the treated group |
|||
Kolesnikova, T.O. et al., 201928 | 10.1016/j.ntt.2019.02.001 | α‐PVP | 1, 5, 25 and 50 mg/L for 20 min 1, 5 and 10 mg/L for 7 d | Zebrafish | Drug exposure HPLC | α‐PVP psychostimulant effects at 5, 25 and 50 mg/L Hypolocomotion and repeated withdrawal after a 7‐day chronic treatment | ||
Mayer, F.P. et al., 201929 | 10.1016/j.neuropharm.2018.12.032 | Enantiomers of nor‐mephedrone, 4‐hydroxytolyl‐mephedrone (4‐OH‐mephedrone) and dihydro‐mephedrone | Human urine | Monoamine transporter assays |
↑ potency at SERT with S‐enantiomers of nor‐mephedrone and 4‐OH‐mephedrone than the corresponding R‐enantiomers. ↓ effectivity as releaser at SERT with R‐enantiomers. |
|||
Eshleman, A.J. et al., 201930 | 10.1007/s00213‐018‐5059‐5 | 22 cathinones, 6 benzofurans and 1 stimulant | Human embryonic kidney 293 cells | Monoamine transporter assays |
Most α‐pyrrolidinophenones had high hDAT potencies and selectivities 4‐cl‐ethcathinone and 3,4‐methylenedioxy‐N‐propylcathinone had higher hSERT selectivity Benzofurans had low hDAT selectivity and were releasers at hSERT |
|||
Yoon, H.S. et al., 201931 | 10.1016/j.neuint.2018.12.005 | α‐PVT | 20 mg/kg (i.p.) | Rats | Nucleus accumbens | Behavioural experiments, western blotting |
↑ locomotor sensitization ↓ phosphorylation levels of GSK3β |
|
Atehortua‐Martinez, L.A. et al., 201932 | 10.1177/0269881118822151 | MDPV | 3 mg/kg (i.p.) | Male Sprague–Dawley rats | 8 wk 250–300 g | Nucleus accumbens, caudate putamen and prefrontal cortex | Behavioural experiments, western blotting |
↑ stereotypies and open arm entries in the elevated plus maze with acute administration ↑ ΔFosB with chronic administration |
Colon‐Perez, L.M. et al., 201833 | 10.1016/j.neuropharm.2018.04.031 | MDPV | 1 and 3 mg/kg (i.p.) | Male long Evans rats | 250–300 g | Whole brain striatum | Functional magnetic resonance imaging immunoblotting DAT assay social interaction test ultrasonic vocalization intracranial self‐stimulation |
↓ strength of correlated neural activity at 1 h ↓ striatal DAT at 24 h and caused a shift in subcellular levels and distribution of DAT, TH and VMAT2 ↓ ICSS thresholds at 1 h ↑ 50 kHz USV calls at 1 h ↓ social interactions |
Oliver, C.F. et al., 201834 | 10.1016/j.drugalcdep.2018.01.013 | MDPV | 2 mg/kg (i.p.) | Male Sprague–Dawley rats | 275–300 g | Locomotor activity, CPP, ultrasonic vocalization | AMD3100 reduced MDPV‐induced locomotor activation, conditioned place preference and modulated MDPV‐induced increase at 50 k‐Hz USV calls. | |
Luethi, D. et al., 201835 | 10.1016/j.neuropharm.2017.07.026 | 5‐IT, 4‐MA, 3‐MMC, N‐methyl‐2‐AI and MMAI | Human embryonic kidney 293 cells | Monoamine transporter assays |
4‐MA, and MMAI entactogenic effects And 3‐MMC, 5‐IT, and N‐methyl‐2‐AI have stimulant‐type properties |
|||
Siedlecka‐Kroplewska, K. et al., 201836 | 10.1007/s12640‐018‐9898‐y | 3‐Fluoromethcathinone | 10 μM | Mice | Hippocampal HT22 cell line | Flow cytometry, western blotting |
↑ intracellular production of reactive oxygen species ↑ formation of autophagic vacuoles ↓ level of p62/SQSTM1 protein |
|
Zwartsen, A. et al., 201837 | 10.1016/j.neuro.2018.03.007 | PMMA, α‐PVP, methylone and MDPV | PMMA = 10‐1000 μM α‐PVP = 1–300 μM Methylone = 1–1000 μM MDPV = 1–1000 μM | Male Wistar rats | Postnatal day 0–1 | Cortical neurons culture | Multi‐well microelectrodes arrays | ↓ the weighted mean firing rate and weighted mean burst rate |
Gerecsei. Et al., 201838 | 10.3389/fnins.2018.00027 | MDPV | 10 mg/kg (s.c.) | C57Bl/6 J mice | Posterior intralaminar complex of the thalamus medial preoptic nucleus | Open field test, grip strength test, force plate actometry, pup retrieval test, nest building of pregnant mothers, in situ hybridization |
↓ birth rate, survival of offspring and maternal care in the drug‐treated animals ↑ locomotor activity of the pups in the MDPV treated group |
|
Gannon, B.M. et al., 201739 | 10.1097/FBP.0000000000000315 | MDPV and α‐PVP | 0.032 mg/kg/infusion (i.v.) | Male Sprague–Dawley rats | 275–300 g | Self‐administration | Both enantiomers of MDPV and α‐PVP function as highly effective reinforcers. | |
Philogene‐Khalid, H.L. et al., 201740 | 10.1021/acschemneuro.7b00212 | R‐MEPH and S‐MEPH | 0.25, 0.50, 2.00 mg/kg/inf (i.v.) | Rats | Self‐administration ultrasonic vocalization | ↑ break points rats trained to self‐administer R‐MEPH and greatest rates of 50 kHz ultrasonic vocalization than S‐MEPH | ||
Nelson, K.H. et al., 201741 | 10.1016/j.pbb.2017.04.003 | α‐PVP | 0.3, 1 and 3 mg/kg (i.p.) | Male Sprague–Dawley rats | 321–445 g | Conditioned place preference, conditioned taste avoidance | α‐PVP induced dose‐dependent taste avoidance as well as significant increases in time spent on the drug‐paired side (no dose‐dependent) | |
Gannon, B.M. et al., 201742 | 10.1016/j.drugalcdep.2017.06.031 | MDPV | 0.03, 0.10, 0.30, and 1.00 mg/mL | Male NIH Swiss mice | 20–25 g | Two‐bottle choice procedures, radiotelemetry, conditioned place preference |
↑MDPV preference solution when was paired with quinine but not with water ↑ escalate consumption with chronic (10 d) access at 0.30 mg/mL MDPV ↑ CPP at 0.30 mg/mL MDPV with a magnitude similar to the preference observed following intraperitoneal administration of MDPV ↑ locomotor activity at 0.1–1.0 mg/mL MDPV |
|
Lantz, S.M. et al., 201743 | 10.1016/j.neulet.2017.06.059 | Phthalimide | 10–1000 μM | Rats | PC12 cells | Cytotoxicity assay |
↑death cell after CP exposition CP alters mitochondrial function and DA and 5‐HT levels |
|
Luethi, D. et al., 201744 | 10.1016/j.tox.2017.06.004 | Bupropion, MDPV, mephedrone and naphyrone | 0.5–2 mM | HepG2 cells HepaRG cells | Cytotoxicity assay | Bupropion, MDPV, mephedrone and naphyrone are mitochondrial toxicants | ||
Grecco, G.G. et al., 201745 | 10.1016/j.taap.2017.05.010 | Methylone, butylone and pentylone | 20 mg/kg (s.c) | Male Sprague–Dawley rats | Blood samples CSF samples | Microdialysis HPLC |
↑Cmax and AUC0‐∞, and the longest t1/2 in the plasma with pentylone ↑Cmax and AUC0‐∞ in the CNS with methylone and butylone |
|
Elmore, J.S. et al., 201746 | 10.1038/npp.2016.213 | Methylone | 3, 6 and 12 mg/kg | Male Sprague–Dawley rats | 250–300 g | Synaptosomes | Locomotor activity and temperature monoamine transporter assays in vivo microdialysis |
↑ motor activation at 12 mg/kg methylone ↓ core temperature at 3 and 6 mg/kg but showed biphasic effects at 12 mg/kg Methylone acted as a fully efficacious substrate‐type releaser at DAT, NET and SERT ↑ brain extracellular dopamine and 5‐HT in vivo |
McLaughlin, G. et al., 201747 | 10.1002/dta.2053 | Mexedrone | Dose–response curve | Male Sprague–Dawley rats | 250–300 g | Synaptosomes | Monoamine transporter assays | Mexedrone and N‐methoxymephedrone showed comparable potency at DAT but the latter compound was more potent at NET and SERT |
Schindler, C.W. et al., 201648 | 10.1111/bph.13640 | MDPV | 0.3–3.0 mg/kg | Male Sprague–Dawley rats | Telemetry | S(+) enantiomer appeared to mediate the cardiovascular effects of MDPV | ||
Berquist, M.D. et al., 201649 | 10.1016/j.drugalcdep.2016.05.001. | 4‐MMC and MDPV | 0.5 mg/kg (i.p.) 0.5, 1.0, or 2.0 mg/kg (i.p.) | Male Sprague–Dawley rats | Locomotor activity | Locomotor responses sensitize to MDPV and to certain mixtures of MDPV and 4‐MMC following repeated dosing | ||
Schindler, C.W. et al., 201650 | 10.1007/s00213‐015‐4057‐0. | MDPV and methylone | 0.03 mg/kg/inj 0.3 or 0.5 mg/kg/inj | Male Sprague–Dawley rats | 300–400 g | Self‐administration microdialysis studies | MDPV self‐administration was acquired rapidly compare with methylone. MDPV (0.1 and 0.3 mg/kg) increased extracellular dopamine while i.v. methylone (1 and 3 mg/kg) increased extracellular dopamine and 5‐HT | |
Valente, M.J. et al., 201651 | 10.1093/toxsci/kfw105 | Methylone, MDPV, pentedrone and 4‐MEC | 0.05 to 10 or 20mM | Male Wistar rats | 210–250 g | HepaRG | Cytotoxic assay |
MDPV and pentedrone were the most cytotoxic. All cathinones triggered significant caspase activation and apoptosis |
Colon‐Perez, L.M. et al., 201652 | 10.1038/npp.2016.40 | MDPV | 0.3, 1.0, or 3.0 mg/kg | Male long Evans rats | 250–300 g | Prefrontal cortex | Functional magnetic resonance imaging |
MDPV dose‐dependently reduced functional connectivity between frontal cortical and striatal areas Dopamine receptor blockade did not prevent the MDPV‐induced decrease in functional connectivity. |
Gannon, B.M. et al., 201653 | 10.1124/jpet.115.229500 | MDPV | 0.01, 0.03, 0.10, 0.30, 1, 3, 10 and 30 mg/kg (i.p.) | Male NIH Swiss mice | 20 – 25 g | Drug discrimination, radiotelemetry |
S(+)‐MDPV was most potent to fully substitution for the cocaine training dose ↑locomotion after doses of S(+)‐MDPV and racemic MDPV |
|
López‐Arnau, R. et al., 201554 | 10.1016/j.taap.2015.03.015 | Mephedrone | 25 mg/kg (s.c.) | Male Sprague–Dawley rats adolescent | 115–130 g 5 wk | Hippocampus, striatum and frontal cortex | Radioligand binding experiments, western blotting, Morris water maze |
↓ densities of dopamine and serotonin transporters without microgliosis ↓ expression of tyrosine hydroxylase and tryptophan hydroxylase 2. Impairment of the reference memory in the Morris water maze |
Kiyatkin, E.A. et al., 201555 | 10.1038/npp.2014.191 | MDPV and Methylone | 1, 3, and 9 mg/kg (s.c.) 0.1, 0.3, and 1.0 mg/kg (s.c.) | Male long Evans rats | 3–4 mo | Social interaction | Methylone and MDPV dose‐dependently increased brain temperature | |
Hutchinson, C.V. et al., 201556 | 10.1016/j.neulet.2015.03.021 | Mephedrone | 1 or 10 μM | Flatworm planaria | Conditioned place preference hypo‐locomotion |
↑ preference to cocaine at both 1 and 10 μM ↓ locomotion after mephedrone withdrawal |
||
Lopez‐Arnau, R. et al., 201457 | 10.1177/0269881114548439 | Methylone | 4 × 20 mg/kg (s.c.) | Male Sprague–Dawley rats | 125–175 g 46 wk | Hippocampus, striatum and frontal cortex | Morris water maze, microglia activation |
Hyperthermia, Memory loss |
Adam, A. et al., 201458 | 10.1016/j.neuro.2014.07.004 | MDPV | 10 mg/kg | Male and female mice C57 | 7 d and 16 wk | Coronal brain slices | Caspase‐3 |
↑ neuronal apoptosis in young but not adult ↑ locomotion |
Den Hollander, B. et al., 201459 | 10.1093/toxsci/kfu108 | β‐Keto amphetamine and 4‐MMC | Male mice C57BL/6 J | 8 wk | SH‐SY5Y neuroblastoma cells | Cytotoxicity assay | Cytotoxic | |
Opacka‐Juffry, J. et al., 201460 | 10.1016/j.pnpbp.2014.04.009 | Mephedrone | 0.3–30 μM | Male Wistar rats | 8 wk | Nucleus accumbens | RTI‐121 binding, voltammetry | Displaced RTI‐121 and caused reverse transport of DA |
Simmler, L.D. et al., 201461 | 10.1016/j.neuropharm.2013.11.008 | Methedrone, 4‐MEC, 3‐FMC, pentylone, ethcathinone, buphedrone, pentedrone and N,N‐dimethylcathinone | Human embryonic kidney 293 cells | Monoamine transporter assay |
All the cathinones were potent NA uptake inhibitors but different in DA vs 5‐HT None of the cathinones bound to rodent trace amine‐associated receptor 1 |
|||
Kaizaki, 201462 | α‐PVP | 25 mg/kg oral | Male balbC mice | 8 wk | Locomotion microdialysis | ↑ release of DA and locomotion | ||
Marusich, J.A. et al., 201463 | 10.1016/j.neuropharm.2014.02.016. | MDPV, α‐PVP, α‐PBP and α‐PPP | 0.3–3.0 mg/kg MDPV, 1.0–10.0 mg/kg α‐PVP, 1.0–10.0 mg/kg α‐PBP, and 3.0–30.0 mg/kg α‐PPP | Male Sprague–Dawley rats | 300–400 g | Synaptosomes | Transporter uptake and release assays locomotor activity |
α‐PVP is a potent uptake blocker at dopamine and norepinephrine transporters α‐PBP and α‐PPP are also catecholamine transporter blockers but display reduced potency ↑ locomotor activity with all of them |
Watterson, L.R. et al., 201464 | 10.1093/ijnp/pyu014 | α‐PVP and 4‐MEC | 1, 3, 10, 30, mg/kg 0.1, 0.3, 1, and 3 mg/kg | Male Sprague–Dawley rats | 250 g | Intracranial self‐stimulation in rats | ↑ intracranial self‐stimulation threshold reductions similar to that of methamphetamine | |
Bonano, J.S. et al., 201465 | 10.1007/s00213‐013‐3223‐5. | MDPV, methylone andmephedrone | 0.32, 1 and 3.2 1, 3.2 and 10 | Male Sprague–Dawley rats | 314–387 g | Intracranial self‐stimulation | Methcathinone was the most potent compound, and MDPV was the longest acting compound | |
Gregg, R.A. et al., 201366 | 10.1097/FBP.0000000000000006. | Mephedrone | 15 mg/kg (i.p.) | Male Sprague–Dawley rats | 260–290 g | Locomotor activity | ↑ cocaine‐induced locomotor activation by prior MEPH exposure | |
Gregg, R.A. et al., 201367 | 10.1016/j.drugalcdep.2013.06.014. | Mephedrone | 15 mg/kg and 30 mg/kg | Male Sprague–Dawley rats | 260–290 g | Sensitization paradigms | ↑ repetitive movement by MEPH challenge compared to acute MEPH exposure in both paradigms | |
Gatch, M.B. et al., 201368 | 10.1097/FBP.0b013e328364166d. | MDPV, methylone, mephedrone, naphyrone, flephedrone and butylone | 1, 3, 10 or 30 mg/kg 0.3, 1, 3, 10 or 30 mg/kg 3, 10, 30 or 100 mg/kg | Male Swiss–Webster mice | 10 wk |
Locomotor activity Discriminative stimulus effects |
MDPV and naphyrone produced locomotor stimulant effects that lasted much longer than cocaine or methamphetamine all compounds fully substituted for the discriminative stimulus effects of cocaine and methamphetamine | |
Cameron, K. et al., 201369 | 10.1007/s00213‐013‐2967‐2. | MDPV andmephedrone | Female Xenopus laevis | Oocytes | Electrophysiological recordings |
Mephedrone is a dopamine‐releasing agent and MDPV behaves as a cocaine‐like reuptake inhibitor of dopamine. |
||
Baumann, M.H. et al., 201370 | 10.1038/npp.2012.204 | MDPV | 0.1–0.3 mg/kg, (i.v.) 0.1–3.0 mg/kg, (s.c.) | Male Sprague–Dawley rats male CB57/BL6 mice | 300–400 g 25–35 g | Synaptosomes striatum nucleus accumbens | Uptake and release assays, fast‐scan cyclic voltammetry, in vivo microdialysis, telemetry, locomotor activity |
↑ amplitude of the dopamine signal ↑ extracellular concentrations of dopamine in the nucleus accumbens ↑ locomotor activation, tachycardia, and hypertension |
den Hollander, B. et al., 201371 | 10.1016/j.pbb.2012.10.006 | Methylone or mephedrone | 30 mg/kg, twice daily for 4 d | Male C57BL/J6 mice male Wistar rats | 8 wk | Frontal cortex, striatum and hippocampus | Behavioural tests, neurotransmitter and transporters levels |
↓ working memory with MEPH ↑ body temperature ↓ 5‐HT levels in the frontal cortex, striatum and hippocampus with methylone |
Lopez‐Arnau, R. et al., 201372 | 10.1016/j.pnpbp.2013.04.007 | Methylone | 10–30 mg/kg (i.v. or oral) | Male Sprague–Dawley rats | 225–250 g | Locomotion + pharmacokinetic analysis | ↑locomotion | |
Cozzi, N.V. et al., 201373 | 10.1016/j.ejphar.2012.11.008 | Methcathinone, 2‐TFMAP, 3‐TFMAP and 4‐TFMAP | Methcathinone: 0.3 mg/kg and 1.0 mg/kg 4‐TFMAP: 1.0 mg/kg and 3.0 mg/kg | Male Sprague–Dawley rats | 300–350 g | Human platelets HEK293 cells C6 glioma cells Synaptosomes Accumbens | Monoamine transporter assay Microdialysis locomotor activity |
↑ uptake inhibition and release with 3‐TFMAP and 4–TFMAP at SERT compared with methcathinone ↑ 5‐HT extracellular level with 4‐TFMAP but doesn't affect the locomotor activity |
Lisek, R., 201274 | 10.1016/j.drugalcdep.2012.04.021. | Mephedrone | 3, 5, 10, 30 mg/kg (i.p.) 30 mg/kg (i.p.) | Male Sprague–Dawley rats CD‐1 mice | 225–275 g 25 – 30 g | Locomotor activity conditioned place preference |
↑ ambulatory activity in rats and was inhibited by pretreatment with SCH 23390 and enhanced by pretreatment with sulpiride ↑ CPP by MEPH |
|
Meng, H. et al., 201275 | 10.1016/j.toxlet.2011.10.010 | Mephedrone | 0.3–15 mg/kg | Chinese hamster Guinea pigs | Chinese hamster ovary cells single ventricular myocytes | Patch‐clamp myocyte action potential echocardio | ↑HR and ↑BP | |
Baumann, M.H. et al., 2012 70 | 10.1038/npp.2011.304 | Mephedrone and Methylone | 0.3 and 1.0 mg/kg 3.0 and 10.0 mg/kg, (s.c.) 3 doses | Male Sprague–Dawley rats | 300–350 g | Synaptosomes | Monoamine transporter assay microdialysis locomotion activity |
Mephedrone and methylone similar to MDMA in potency and selectivity for monoamine transporters. ↑ extracellular DA and 5‐HT levels after mephedrone and methylone i.v administration in accumbens ↑ locomotor activity and stereotypy after 1 mg/kg mephedrone dose ↑ locomotor activity after 0.3 mg/kg and 1 mg/kg methylone doses. |
BP = blood pressure; CPP = conditioned place preference; DA = dopamine; DAT = dopamine transporter; HR = heart rate; HPLC = high‐performance liquid chromatography; ICSS = intraccranial self‐stimulation; i.p. = intraperitoneal; i.v. = intravenous; MEPH = mephedrone; MDPV = Methylenedioxypyrovalerone; MMAI = 5‐Methoxy‐6‐methyl‐2‐aminoindane; NA = noradrenaline; NET = noradrenaline transporter; N‐methyl‐2‐AI = N‐methyl‐2‐aminoindane; PMMA = para‐Methoxy‐N‐methylamphetamine; SERT = serotonin transporter; s.c. = subcutaneous; TFMAP = CF3 substituted methcathinone analogues; USV = ultrasonic vocalisation; 3‐FMC = 3‐Fluoromethcathinone; 3‐MMC = 3‐Methylmethcathinone; 4‐MA = 4‐methylamphetamine; 4‐MEC = 4‐Methylethcathinone; 5‐HT = serotonin; 5‐IT = 5‐(2‐Aminopropyl)indole; αPBP = α‐Pyrrolidinobutiophenone; αPPP = α‐Pyrrolidinopropiophenone; αPVP = α‐Pyrrolidinovalerophenone; αPVT = α‐Pyrrolidinopentiothiophenone.
3.2. Stimulants
The most studied NPS stimulants were https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?tab=biology&ligandId=115 (3‐trifluoromethylphenylpiperazine), N‐benzylpiperazine and 4,4′‐DMAR (4,4′‐dimethylaminorex), which were all studied in 4 manuscripts (16%). The other stimulants were only studied in 3 or fewer manuscripts (Table 2). The NPS stimulants were mostly studied using neurochemical assays at dopamine, serotonin or noradrenaline systems (16 studies [64%]) with 6 studies (24%) examining toxicity or hyperthermia, 3 studies (12%) looked at the behavioural effects using models of addiction and a single study used assays related to the cardiovascular system. Thus, similar to the cathinones, the vast majority of studies examined either behavioural or neurochemical indices of addiction and very few looked at acute toxic effects. No studies examined chronic effects of stimulants and no studies reported using female rodents.
Table 2.
Preclinical studies examining stimulant novel psychoactive substance (NPS) pharmacology. The most recent manuscripts are presented first
Study | DOI | NPS | Dose/concentration | Species/sex | Age | Sample | Assay | Main results |
---|---|---|---|---|---|---|---|---|
Rickli, A. et al., 201976 | 10.1016/j.neuro.2019.02.011 | 4‐MAR, 4,4′‐DMAR, and 3,4‐DMAR | Human embryonic kidney 293 cells | Monoamine transporter assays |
4,4′‐DMAR potently inhibited all monoamine transporters. 4‐MAR preferentially inhibited NET and DAT 3,4‐DMAR only weakly inhibited NET |
|||
Cai, W.T. et al., 201977 | 10.1016/j.neuint.2019.104487 | Methiopropamine | 5 mg/kg (i.p.) | Rats | Nucleus accumbens | ↑number of total spine density | ||
Maier, J. et al., 201878 | 10.1016/j.neuropharm.2018.06.018 | 4,4′‐DMAR | Dose–response curve | Human/rat | Human embryonic kidney 293 cells human striatal tissue rat pheochromocytoma cells (rPC12) | Uptake inhibition assays transporter release assays receptor and transporter binding and activation assays |
↓ uptake mediated by human DAT, NET or SERT Release assays identified 4,4′‐DMAR as a substrate type releaser, capable of inducing Transporter‐mediated reverse transport via DAT, NET and SERT ↓ rat and human isoforms of VMAT2 at a potency like MDMA |
|
Davidson, C. et al., 201879 | 10.3389/fpsyt.2018.00149 | 3,4‐CTMP and ethylphenidate | 1, 10 and 100 nM/1 and 10 μM | Male Wistar rats | 8 wk | Nucleus accumbens and stria terminalis | Fast cyclic voltammetry (DA and NA) |
↑ evoked dopamine and noradrenaline efflux with methylphenidate (10 μM) ↑ evoked dopamine and noradrenaline efflux with 3,4‐CTMP (0.1 and 1 μM) ↑ evoked dopamine and noradrenaline efflux with ethylphenidate (1 μM) |
Luethi, D. et al., 201880 | 10.1016/j.neuropharm.2017.08.020 | N‐benzylethylphenidate, 3,4‐dichloroethylphenidate, 3,4‐dichloromethylphenidate, ethylnaphthidate, ethylphenidate, 4‐fluoromethylphenidate, isopropylphenidate, 4‐methylmethylphenidate, methylmorphenate, and propylphenidate | Human embryonic kidney 293 cells | Monoamine transporter assays |
↑ inhibition NAT and DAT with all the drugs No cytotoxicity was observed after drug treatment at assay concentrations. |
|||
McLaughlin,G. et al., 201881 | 10.1002/dta.2396 | 4‐MPM/3‐MPM | Dose–response curve | Male Sprague–Dawley rats | 250–300 g | Caudate (for DAT assays) or whole brain minus cerebellum and caudate (for NET and SERT assays) | Monoamine transporter assays | 2‐MPM and 3‐MPM will exhibit stimulant properties like the parent compound phenmetrazine, whereas 4‐MPM may display entactogen properties more similar to MDMA. |
Zwartsen, A. et al., 201882 | 10.1016/j.neuro.2018.03.007 | BZP and TFMPP | BZP = 1–1000 μM TFMPP = 1–1000 μM | Male Wistar rats | Postnatal day 0–1 | Cortical neurons culture | Multiwell microelectrodes arrays | ↓ the weighted mean firing rate and weighted mean burst rate |
Mayer, F.P. et al., 201883 | 10.1016/j.neuropharm.2017.10.006 | 3‐FPM, 2‐FPM and 4‐FPM | Dose–response curve | Male Sprague–Dawley rats | Human embryonic kidney 293 cells Synaptosomes | Monoamine transporter assays patch clamp |
2‐, 3‐ and 4‐FPM inhibit uptake mediated by DAT and NET All FPM raised concentration‐dependent release of monoamines from rat brain synaptosomes |
|
Sahai, M.A. et al., 201784 | 10.1016/j.pnpbp.2016.11.004 | 5‐MAPB | 1,3,10 and 30 μM | Male Wistar rats | 8 wk | Striatum accumbens | Radioligand binding fast cyclic voltammetry |
5‐MAPB reduces the rate of dopamine reuptake 5‐MAPB binds to the DAT and displace RTI‐121 as DAT ligand |
Shimshoni, J.A. et al., 201785 | 10.1016/j.taap.2017.01.018 | MEAI | 10 and 30 mg/kg | Male Sprague–Dawley rats | Striatum primary neurons human primary hepatocytes | Safety profile cytotoxic assay | Good safety profile in rats at 10 and 30 mg/kg cytotoxic effect at 500 and 1000 mg/L concentrations | |
McLaughlin, G. et al., 201786 | 10.1002/dta.2167 | 4F‐MPH | Dose–response curve | Male Sprague–Dawley rats | 250–300 g | Synaptosomes | Monoamine transporter assays |
↑ potencies determined for blockage of dopamine uptake and norepinephrine uptake In (±)‐threo isomer ↓ potent at the dopamine transporter and norepinephrine transporter in MPH |
Yoon, H.S. et al., 201687 | 10.1016/j.bbr.2016.05.060 | Methiopropamine | i.p. 0.2, 1.0, or 5.0 mg/kg | Male Sprague–Dawley rats | 220–250 g | Locomotor activity |
↑ sensitized locomotor activity the group that was pre‐exposed at 5 mg/kg of MPA MPA‐induced locomotor sensitization was inhibited by a preinjection of a dopamine D2 receptor antagonist |
|
Marusich, J.A. et al., 201688 | 10.1016/j.neuropharm.2015.09.004 | 5‐IT and 6‐IT | 1.0–10.0 mg/kg | Male Sprague–Dawley rats | 300–400 g | Synaptosomes | Locomotor activity monoamine transporter assays |
5‐IT displayed greater potency for release at DAT over SERT, while 6‐IT displayed greater potency for release at SERT over DAT 5‐IT produced locomotor stimulation |
Simmler, L.D. et al., 201689 | 10.1124/jpet.115.229765 | 101 compounds: amphetamines, phenethylamines, aminoindanes, cathinones, ephedrines, tryptamines, piperazines and pipradrols | 10 pM to 10 μM | Human embryonic kidney 293 cells rat and mouse TAAR1 | Radioligand binding assay functional TAAR1 activity | Species differences in activity at TAAR1 among the highly active ligands, with a rank order of rat > mouse > human | ||
Asaoka, N. et al., 201690 | 5F‐ADB | 1 μM | Midbrain dopaminergic neurons | Electrophysiological recordings | 5F‐ADB significantly increase the spontaneous firing rate in dopaminergic but nor in serotoninergic neuron | |||
Persona, K. et al., 201691 | 10.1007/s12640‐016‐9604‐x | N‐Benzylpiperazine | 0.1, 0.3, 1, 3, 10, 30, 100, 300, and 1000 μg/mL 10, 30, 100, and 300 μg/mL | Human glioblastoma cells (LN‐18) | Cytotoxicity assay, real‐time polymerase chain reaction gene expression analysis | ↑ LDH levels ↑ mitochondrial membrane potential, ↓ ATP and ↑ reactive oxygen species production, ↑ levels of DNA damage marker (8‐OHdG) and activation of caspases: −3 and − 9. | ||
Arbo, M.D. et al., 201692 | 10.1007/s00204‐016‐1665‐3 | 1‐benzylpiperazine, 1 (3,4methylenedioxybenzyl) piperazine, 1‐(3‐trifluoromethylphenyl) piperazine and 1‐(4‐methoxyphenyl) piperazine | 625, 210 and 0.5 μM for BZP 35, 12 and 0.5 μM for TFMPP 522, 175 and 0.5 μM for MeOPP 467, 160 and 0.5 μM for MDBP | Male Wistar rats | 300–400 g | Hepatocytes | Cytotoxicity assay, gene array | Key enzymes of cholesterol biosynthesis, glycoprotein transmembrane NMB and fatty acid desaturase 1 were upregulated by all 4 piperazine drugs. Betaine‐homocysteine‐S‐methyltransferase 2 were downregulated all 4 piperazine derivatives |
Arbo, M.D. et al., 201693 | 10.1002/jat.3153 | N‐ benzylpiperazine, 1‐(3‐trifluoromethylphenyl) piperazine, 1‐(4‐methoxyphenyl)piperazine and 1‐(3,4‐methylenedioxybenzyl)piperazine | 500 or 1000 μM BZP 5, 50 or 100 μM TFMPP 250 or 500 μM MeOPP or MDBP | SH‐SY5Y cell | Cytotoxic assay | 1‐(3‐trifluoromethylphenyl) piperazine was the most cytotoxic | ||
Rubio, M. et al., 201594 | 10.1016/j.neulet.2015.01.075 | 4‐Methylamphetamine (4‐MA) | s.c. 2.5, 5 and 10 mg/kg | Male Sprague–Dawley rats | 125–175 g | Locomotor activity, radiotelemetry | ↑dose‐dependent manner, locomotor activity dose‐dependent hypothermic response to 4‐MA | |
Rickli, A. et al., 201595 | 10.1016/j.euroneuro.2014.12.012 | Para‐halogenated amphetamines and pyrovalerone cathinones | Human embryonic kidney 293 cells | Monoamine transporter assays | ↑ 5‐HT properties in 4‐methyl, 4‐ethyl, and 4‐bromo groups 3,4‐methylenedioxypyrovalerone, pyrovalerone, α‐pyrrolidinovalerophenone, 3,4‐methylenedioxy‐α‐pyrrolidinopropiophenone and 3,4‐methylenedioxy‐α‐pyrrolidinobutiophenone potently inhibited NET and DAT but not SERT | |||
McLaughlin, G. et al., 201596 | 10.1002/dta.1732 | MDMAR and DMAR | 1 nM – 10 μM | Male Sprague–Dawley rats | 250–300 g | Synaptosomes | Synaptosomal uptake/release | More efficacious than MDMA |
Arbo, M.D. et al., 201497 | 10.1016/j.toxlet.2014.06.031 | N‐ benzylpiperazine, 1‐(3‐trifluoromethylphenyl)piperazine, 1‐(4‐methoxyphenyl)piperazine and 1‐(3,4‐methylenedioxybenzyl)piperazine | H9c2 rat cardiac cell line | Cytotoxic assay | TFMPP seems to be the most potent cytotoxic compound | |||
Brandt, S.D. et al., 201498 | 10.1002/dta.1668 | 4,4′‐DMAR | 0.3 nM – 10 μM | Male Sprague–Dawley rats | 250–300 g | Synaptosomes | Monoamine transporters assays | ↑ release of DA, NA, 5‐HT. More potent than AMPH at 5‐HT |
Dawson, P. et al., 201499 | 10.1016/j.pnpbp.2013.08.013 | 5‐APB | 0.3–30 μM | Male Sprague–Dawley rats | 8 wk | Prefrontal cortex, nucleus accumbens and caudate–putamen | Binding and voltammetry in slices. Fundus and aorta preps | Displaced both DAT and 5‐HT2 ligands and ↑ DA efflux. ↑ contraction in fundus and aorta |
Davidson, C. et al., 2012100 | 10.1177/0269881111430733 | Desoxypipradrol | 1‐10 μM | Male Sprague–Dawley rats | 8 wk | Nucleus accumbens | Voltammetry in slices | Bigger increase in DA vs cocaine |
BZP = Benzylpiperazine; DA = dopamine; NA = noradrenaline; DAT = dopamine transporter; i.p. = intraperitoneal; MEAI = 5‐methoxy‐2‐aminoindane; NET = noradrenaline transporter; SERT = serotonin transporter; s.c. = subcutaneous; TFMPP = 3‐Trifluoromethylphenylpiperazine; 3‐FPM = 3‐Fluorophenmetrazine; 3‐MPM = 3‐methoxyphenmetrazine; 3,4‐CTMP = 3,4‐dichloromethylphenidate; 4F‐MPH = 4‐fluoromethylphenidate; 4‐MAR = 4‐Methylaminorex; 4‐MPM = 4‐Methylphenmetrazine; 4,4‐DMAR = 4,4′‐Dimethylaminorex; 5‐APB = 5‐(2‐aminopropyl)benzofuran; 5‐HT = serotonin; 5‐IT = 5‐(2‐Aminopropyl)indole; 5‐MAPB = 5‐(2‐methylaminopropyl)benzofuran.
3.3. SCRAs
There have been very few preclinical studies on SCRAs over the last 10 years with only 10 or so SCRAs examined in 8 studies. These include JWH and CP compounds while AKB48 was the most studied SCRA (3 studies [38%]). Despite the lack of studies, it is clear that the types of assay differ from both the cathinone and stimulant research. In the SCRA manuscripts, 5 studies (63%) examined cell toxicity, 5 studies (63%) examined their neuropharmacology, 1 study looked at cardiovascular effects and 1 study looked at models of addiction. Thus, in contrast to the cathinone and stimulant studies, there was only 1 study looking models of addiction and proportionally more studies examined toxicity (Table 3).
Table 3.
Preclinical studies examining synthetic cannabinoid receptor agonist novel psychoactive substance (NPS) pharmacology. The most recent manuscripts are presented first
Study | DOI | NPS | Dose/conc | Species/sex | Age | Sample | Assay | Main results |
---|---|---|---|---|---|---|---|---|
Bilel, S. et al., 2019101 | 10.3389/fnins.2019.01163 | AKB48 | 0.25 mg/kg to 3 mg/kg | Male rats | Nucleus accumbens | Pharmacological and behavioural effects |
↑ DA release in the nucleus accumbens shell at 0.25 mg/kg ↓ startle/PPI response at 3 mg/kg ↑ hypothermia, analgesia, and catalepsy at 3 mg/kg ↑impaired place preference and hypolocomotion at 0.5 mg/kg |
|
Banister, S.D. et al., 2019102 | 10.1002/dta.2491 | SCRAs 5F‐CUMYL‐PICA, 5F‐CUMYL‐PINACA and 5F‐CUMYL‐P7AICA | 0.1, 0.3, 1, and 3 mg/kg (i.p.) | Male C57BL/6 J mice | 21.2–29 g | Human embryonic kidney 293 cells AtT20 adenocarcinoma cells | Binding assay functional activity assay in vivo pharmacological assessment |
All compounds were potent CB1 agonists 5F‐CUMYL‐P7AICA induced hypothermia |
Kim, S. et al., 2019103 | 10.3390/molecules24163000 | AKB48 | 0.1 to 100 μM | Human liver microsomes LLC‐PK1‐MDR1 LLC‐PK1‐mock cells HEK293 cells HEK293‐mock cells | Cytochrome P450 assays |
↑inhibition of CYP3A4 and UGT1A9 activities |
||
Kevin, R.C. et al., 2019104 | 10.3389/fphar.2019.00595 | CUMYL‐4CN‐BINACA | 0.03, 0.1, 0.3, and 1 mg/kg (i.v.) | Male C57BL/6 J mice | Mouse AtT20FlpIn neuroblastoma cells human embryonic kidney 293 cells | Radioligand binding assay receptor functional assay biotelemetry locomotor activity |
Potent CB1 receptor agonist ↑ pro‐convulsant effects at 0.3 mg/kg |
|
Domoto, M. et al., 2018105 | 10.1007/s00213‐018‐4933‐5 | 5F‐AMB | 300 nM | Male and female C57BL/6 J mice | 4–6 wk | Medial prefrontal cortex | Patch‐clamp | ↓ excitatory and inhibitory transmission in mPFC L5 pyramidal neurons via the activation of CB1 receptors located in presynaptic terminals |
De Luca, M.A. et al., 2016106 | 10.1016/j.neuropharm.2015.11.017 | BB‐22, 5F‐PB‐22, 5F‐AKB‐48 and STS‐135 | BB‐22: 0.003–0.1 mg/kg 5F‐PB‐22: 0.01 mg/kg 5F‐AKB‐48: 0.1 mg/kg STS‐135: 0.15 mg/kg | Male Sprague–Dawley rats C57BL/J6 and CB1knockout mice | 200–300 g 17–20 g | Nucleus accumbens medial prefrontal cortex | Binding assay in vivo microdialysis |
5F‐AKB‐48 and STS‐135 had higher Ki for CB1 binding. ↑ DA in the accumbens shell with all compounds |
Yun, J. et al., 2016107 | 10.1039/c6tx00259e | JWH‐030 | 0.1, 1, 10, or 100 μM 0.001–2500 μM 30 μM i.v. 0.5 mg/kg | Male Sprague–Dawley rats male New Zealand white rabbits | 7 wk 2890–3380 g | H9c2 cells rabbit Purkinje fibers | Cytotoxic assay patch clamp action potential duration (APD) electrocardiogram | JWH‐030 was more cytotoxic than JWH‐210, JWH‐250 and RCS4 JWH‐030 to block the hERG channel JWH‐030 significantly reduced the APD at 90% repolarization JWH‐030 prolonged the QT interval in rats |
Tomiyama, K. 2014108 | 10.1016/j.taap.2013.10.028 | CP‐55,940, CP‐47,497, CP‐47,497‐C8, HU‐210, JWH‐018, JWH‐210, AM‐2201, and MAM‐2201 | 1–30 μM for 0.5–3 h | ICR mouse | 15‐d gestation | Primary neuronal cell culture | Forebrain cell cultures for caspase‐3 | Apoptosis |
AKB‐48 = N‐(1‐adamantyl)‐1‐pentyl‐1H‐indazole‐3‐carboxamide; AM2201 = [1‐(5‐fluoropentyl)indol‐3‐yl]‐naphthalen‐1‐ylmethanone; BB‐22 = quinolin‐8‐yl 1‐(cyclohexylmethyl)indole‐3‐carboxylate; CUMYL‐4CN‐BENICA = 1‐(4‐cyanobutyl)‐N‐(2‐phenylpropan‐2‐yl)indazole‐3‐carboxamide; CP47497 = 2‐[(1R,3S)‐3‐hydroxycyclohexyl]‐5‐(2‐methyloctan‐2‐yl)phenol; CP55940 = 2‐[(1R,2R,5R)‐5‐hydroxy‐2‐(3‐hydroxypropyl)cyclohexyl]‐5‐(2‐methyloctan‐2‐yl)phenol; DA = dopamine; HU‐210 = 1,1‐dimethylheptyl‐ 11‐hydroxytetrahydrocannabinol; JWH‐018 = naphthalen‐1‐yl‐(1‐pentylindol‐3‐yl)methanone; JWH‐030 = naphthalen‐1‐yl‐(1‐pentylpyrrol‐3‐yl)methanone; JWH‐210 = (4‐ethylnaphthalen‐1‐yl)‐(1‐pentylindol‐3‐yl)methanone; NA = noradrenaline; DAT = dopamine transporter; MAM2201 = [1‐(5‐fluoropentyl)indol‐3‐yl]‐(4‐methylnaphthalen‐1‐yl)methanone; NET = noradrenaline transporter; P7‐AICA = pyrrolo[2,3‐b]pyridine‐3‐carboxamide; SERT = serotonin transporter; STS‐135 = N‐(1‐adamantyl)‐1‐(5‐fluoropentyl)indole‐3‐carboxamide; i.p. = intraperitoneal; i.v. = intravenous; 5F‐AKB‐48 = N‐(1‐adamantyl)‐1‐(5‐fluoropentyl)indazole‐3‐carboxamide; 5F‐AMB = methyl 2‐[[1‐(5‐fluoropentyl)indazole‐3‐carbonyl]amino]‐3‐methylbutanoate; 5F‐CUMYL PICA = 1‐(5‐fluoropentyl)‐N‐(2‐phenylpropan‐2‐yl)indole‐3‐carboxamide; 5F‐PB‐22 = quinolin‐8‐yl 1‐(5‐fluoropentyl)indole‐3‐carboxylate.
We note previous studies on SCRAs from outside our search period (January 2008–December 2018), such as Randall et al.109 In this article, we focussed only on studies from the last 10 years, which include the so‐called third generation SCRAs.110, 111
3.4. Other NPS
There are relatively few studies on other NPS (20 studies) with the most studied drugs being methoxetamine and 25B‐NBOMe (both 4 studies, 20%). Most of the assays used are to examine the neuropharmacology of the drugs (12 studies [60%]) with only 1 manuscript examining effects of the drugs in models of abuse. Four studies (20%) looked at the toxicity of these drugs, 2 studies (10%) found cognitive dysfunction, 1 found bladder and renal toxicity, and 1 looked at cardiovascular effects (Table 4).
Table 4.
Preclinical studies examining other novel psychoactive substance (NPS) pharmacology. The most recent manuscripts are presented first
Study | DOI | NPS | Dose/conc | Species/sex | Age | Sample | Assay | Main results |
---|---|---|---|---|---|---|---|---|
Luethi, D. et al., 2019112 | 10.1016/j.ejphar.2019.05.014 | 2C‐BI derivatives | Human cells | Monoamine transporter assays | 2C‐BI‐8 and 2C‐BI‐12 activated serotonin 5‐HT2A and 5‐HT2B receptors at sub‐μM concentrations | |||
Wallach, J. et al., 2019113 | 10.1016/j.ejphar.2019.172427 | Fluorolintane and its 5 aryl‐fluorine‐substituted isomers | Rats | Hippocampal slices | Monoamine transporter assays field‐recording electrophysiology Behavioural test |
↑affinity for NMDA receptors ↓ long‐term potentiation ↓NMDA receptor‐induced field excitatory postsynaptic potentials ↓ PPI |
||
Costa, G. et al., 2019114 | 10.1016/j.neuropharm.2018.10.031 | Methoxetamine | 0.1–0.5 mg/kg, i.p., × 5 d | Rats | Ultrasonic vocalizations Behavioural test |
Pre‐treatment with MXE impair alterations in the elevated plus maze, marble burying and novel object ↑ neurotoxicity |
||
Yoon, K.S. et al., 2019115 | 10.1007/s12012‐018‐9489‐4 | Methoxetamine | 0.1–500 μM | Mice | H9c2 cells | Cytotoxic assays |
↓ cell viability and PAK‐1 mRNA levels at 10 μM ↓beating rate of primary cardiomyocytes at 100 μM |
|
Halberstadt,A.L. et al., 2019116 | 10.1016/j.neuropharm.2018.10.037 | DOB, 2C‐B and benzodifuranyl and tetrahydrobenzodifuranyl analogs | C57BL/6 J mice | Head twitch response (HTR) assay |
DOB and 2C‐B induced the HTR ↑potency of DOB‐DFLY and 2C‐B‐DFLY than DOB and 2C‐B 2C‐I‐FLY, 2C‐E‐FLY and 2C‐EF‐FLY active in the HTR assay but had low potency |
|||
Herian, M. et al., 2019117 | 10.1007/s12640‐019‐00033‐x | 25I‐NBOMe | 0.3, 1, 3, and 10 mg/kg | Male Wistar‐Han rats | 280–350 g | Frontal cortex | Microdialysis | Inverted U‐shaped dose–response curve on extracellular DA and 5‐HT levels U‐shaped dose–response curve on GLU levels |
Zwartsen, A. et al., 201838 | 10.1016/j.neuro.2018.03.007 | 2C‐B, 25B‐NBOMe | 2C‐B = 1–300 μM 25B‐NBOMe = 0.01–30 μM | Male Wistar rats | Postnatal day 0–1 | Cortical neurons culture | Multi‐well microelectrodes arrays | ↓ the weighted mean firing rate and weighted mean burst rate |
Shintani‐Ishida, K. et al., 2018118 | 10.1111/1556‐4029.13583 | 25B‐NBOMe | i.p. 0.5 mg/kg | Male Sprague–Dawley rats | 8 wk | Blood samples lung, heart, kidney or brain tissue homogenate | HPLC | ↑ 25B‐NBOMe concentration in blood samples after 6 hours 25B‐NBOMe accumulated primarily in the lung |
Cha, H.J. et al.,2018119 | 10.1016/j.neulet.2018.04.009 | 4‐chloro‐2,5‐dimethoxyamphetamine and AH‐7921 | DOC: 0.1, 0.3, and 0.5 mg/kg AH‐7921: 0.1, 0.3, and 1 mg/kg | Male Sprague–Dawley rats C57BL/6 mice | 180–220 g 15–20 g |
Conditioned place preference Self‐administration |
↑ preference with both drugs at 0.3 mg/kg ↑ number of responses to the active lever in the self‐administration test at 0.01 mg/kg |
|
Luethi, D. et al., 20187 | 10.1016/j.ejphar.2017.12.012 | Diclofensine, diphenidine, and methoxphenidine | Human embryonic kidney 293 cells | Monoamine transporter assays | Diclofensine bound to adrenergic, dopamine, serotonin and trace amine‐associated receptors. Diphenidine bound to adrenergic α1A and α2A receptors and serotonin 5‐hydroxytryptamine 1A (5‐HT1A) and 5‐HT2A receptors. Methoxphenidine bound to adrenergic α2A and serotonin 5‐HT2A and 5‐HT2C receptors | |||
Hondebrink, L. et al., 2017120 | 10.1016/j.neuropharm.2017.04.035 | Methoxetamine | 0.1 M | Male Wistar rats | Postnatal day 1 |
Rat primary cortical cells Human SH‐SY5Y cells Human embryonic kidney 293 cells |
Cell intracellular calcium imaging Multi‐well microelectrodes arrays Monoamine transporter assays |
↑ the glutamate‐evoked increase in [Ca2+] in rat primary cortical cells with 10 μM methoxetamine ↓ the K + ‐ and acetylcholine‐evoked increase in [Ca2+]I in human SH‐SY5Y cells with 10 μM methoxetamine ↓ spontaneous neural activity between 10‐100 μM methoxetamine ↓ uptake via monoamine transporters (DAT, NET and SERT) |
Kang, H. et al., 2017121 | 10.1016/j.neuropharm.2016.08.004 | Ephenidine | Dose–response curve 1 and 10 μM | Male rats (Wistar and Sprague Dawley) | 3–10 wk | Whole rat brain CA1 of rat hippocampal slices hippocampal pyramidal cells | Receptor binding assays, extracellular recording of field, excitatory postsynaptic potentials patch clamp |
Ephenidine acts at the PCP site of the NMDA receptor and has lower affinity for the dopamine and noradrenaline transporters ↑ inhibition of the NMDA receptor mediated fEPSP at 10 μM 10 μM blocked NMDA receptor‐mediated EPSCs |
Rickli, A. et al., 2016122 | 10.1016/j.euroneuro.2016.05.001 | DiPT, 4‐OH‐DiPT, 4‐OH‐MET, 5‐MeO‐AMT, and 5‐MeO‐MiPT lysergic acid diethylamide, psilocin, N,N‐dimethyltryptamine and mescaline | Monoamine transporter assay | ↓ binding 5‐HT2A with all the tryptamines and psilocin and DMT compared with LSD DMT, DiPT, 4‐OH‐DiPT and 4‐OH‐MET, interacted partially with the norepinephrine transporter LSD but not the tryptamines interacted with adrenergic and dopaminergic receptors | ||||
Wallach, J. et al., 2016123 | 10.1371/journal.Pone.0157021 | DPH, 2‐MXP and 3‐ and 4‐MeO‐ isomers and 2‐cl‐diphenidine (2‐cl‐DPH) | 1uM and 10uM 1.25,2.5,5 and 10 mg/kg (s.c) | Male Wistar rats male Sprague–Dawley rats | 9–10 wk 250–275 g | Whole brain hippocampal slices |
Binding studies, monoamine reuptake inhibition assays, in vitro field excitatory postsynaptic potential PPI |
DPH and 2‐MXP, were found to be relatively selective NMDAR antagonists and inhibited NMDAR mediated field EPSPs DPH and 2‐MXP significantly inhibited PPI |
Rickli, A. et al., 2015 124 | 10.1016/j.neuropharm.2015.08.034 | 2C drugs | Human cells | Monoamine transporter assay | NBOMe drugs were very potent 5‐HT2A receptor agonists 2C drugs increased the binding affinity at serotonergic 5‐HT2A, 5‐HT2C, adrenergic α1, dopaminergic D1–3, and histaminergic H1 receptors and monoamine transporters but reduced binding to 5‐HT1A receptors and TAAR1 | |||
Blough, B.E. et al., 2014125 | 10.1007/s00213‐014‐3557‐7 | Tryptamines (21 compounds) | Rats | Synaptosomes | Monoamine transporter assay | All tryptamines were 5‐HT2A agonists. N‐ethyltryptamine was the greatest 5‐HT releaser and 5‐MeO‐MIPT was the weakest 5‐HT uptake inhibitor. | ||
Dargan, P.I. et al., 2014126 | 10.3109/15563650.2014.892605 | Methoxetamine | i.p 30 mg/kg x 3 mo | Mice | 2–5 mo | Bladder and kidneys | CD4 and Sirius red staining | Bladder and renal toxicity |
Paulke, A. et al., 2013127 | 10.1016/j.jep.2013.04.044 | Argyreia nervosa and LSA | In silico and radioligand assays | Lower affinity than LSD but clear affinity at 5‐HT1A, 5‐HT2, and α2 | ||||
Compton, D.M. et al., 2011128 | 10.1016/j.physbeh.2011.01.021 | 5‐MeO‐DIPT | 6 x 5 or 20 mg/kg | Male long Evans rats | 35 or 48 d | Variety of behavioural tests | Cognitive deficits |
AH7921 = 3,4‐dichloro‐N‐[[1‐(dimethylamino)cyclohexyl]methyl]benzamide;hydrochloride; DA = dopamine; DAT = dopamine transporter; DiPT = N‐[2‐(1H‐indol‐3‐yl)ethyl]‐N‐propan‐2‐ylpropan‐2‐amine; DOB = 1‐(4‐bromo‐2,5‐dimethoxyphenyl)propan‐2‐amine; DPH = diphenidine (1‐(1,2‐diphenylethyl)piperidine); HPLC = high‐performance liquid chromatography; LSA = lysergic acid amide; NA = noradrenaline; NET = noradrenaline transporter; PPI = prepulse inhibition; SERT = serotonin transporter; 2C‐B = 2,5‐dimethoxy‐4‐bromophenethylamine; 25I‐NBOMe = 2‐(4‐iodo‐2,5‐dimethoxyphenyl)‐N‐[(2‐methoxyphenyl)methyl]ethanamine; 2‐MXP = 2‐methoxydiphenidine (1‐[1‐(2‐methoxyphenyl)‐2‐phenylethyl]piperidine); 4HO‐DiPT = 4‐Hydroxy‐N,N‐diisopropyltryptamine; 4HO‐MET = 4‐hydroxy‐N‐methyl‐N‐ethyltryptamine; 5‐MeO‐AMT = 1‐(5‐methoxy‐1H‐indol‐3‐yl)propan‐2‐amine; 5‐MeO‐DiPT = N‐[2‐(5‐methoxy‐1H‐indol‐3‐yl)ethyl]‐N‐propan‐2‐ylpropan‐2‐amine; 5‐MeO‐MiPT = N‐[2‐(5‐methoxy‐1H‐indol‐3‐yl)ethyl]‐N‐methylpropan‐2‐amine.
3.5. Overview of preclinical data on NPS (more formal and supported)
Taken together we can see that most recent studies on NPS examined the reinforcing effects or addictive liability of NPS, whether through behavioural models (32%) or neurotransmitter changes such as dopamine levels (46%). Taking NPS for their rewarding effects is a motivation for use; other reasons for using NPS including self‐medicating to treat withdrawal symptoms from other drugs or the availability when traditional drugs are scarce or supplementing the illicit drug use by trying to induce synergistic or additive effects. From a legislative point of view, it might be useful to know if a drug was more or less addictive and this criterion is used by the EU for banning NPS.129 Further, more resources, whether it be policing, research, healthcare or education could be targeted at those NPS predicted to be more addictive. The next most common type of study examined cell toxicity (24%). This is useful as highly toxic substances should clearly be banned and this criterion is also used by the EU. Interestingly, the effects of these drugs, which might result in acute hospitalisation, for example cardiotoxicity, seizure activity, hyperthermia or death, have hardly been studied at all in NPS (all <5%).
Only 4% of studies included female rodent samples and only 9% included chronic dosing studies. The age range of most studies was appropriate with most (72%) using rodents, which were adolescent or in early adulthood (100–299 g) and only 22% of studies examining older rodents (>300 g).
4. PREDICTIVE VALIDITY OF NPS TESTS IN ANIMAL TISSUE
As detailed above, when NPS started flooding the market over the last decade or so, preclinical tests were exploited to compare these substances to their traditional counterpart, describing them as https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=2286 ‐like or amphetamine‐like substances.60, 130 Unlike known illicit substances such as https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=4803 and MDMA (3,4‐methylenedioxymethamphetamine), the pharmacology related to emerging NPS is scarce and may not suitably be extrapolated from existing knowledge. For example, when the risk assessment of 2C‐I (2,5‐dimethoxy‐4‐iodophenethylamine) was carried out in 2003, a speculative comparison was made with the phenethylamine analogue 2C‐B (2,5‐dimethoxy‐4‐bromophenethylamine) and the amphetamine analogue DOB (https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=163), as they both have similar chemical structures to 2C‐I but with a bromine instead of iodine. It was deemed inappropriate to compare or extrapolate data related to the structurally similar MDMA, PMA (paramethoxyamphetamine) and 4‐MTA (4‐methylthioamphetamine) due to the absence of the 2,5‐dimethoxy functional group.131
The validity of animal models in understanding the harmful effects of illicit substances in humans is well established and therefore, can provide a predictive validity for NPS‐related harms. For instance, ketamine has been associated with bladder toxicity.132 Similar to ketamine, animal models showed significant bladder and renal toxicity in rodents following the administration of 30 mg/kg methoxetamine intraperitoneally.126 Symptoms included inflammatory cell infiltration, tubular cell necrosis, glomerular damage and increased micturition frequency bladder dysfunction.126 , 133
Benzofurans such as 5‐APB (5‐(2‐aminopropyl)benzofuran), 5‐APDB (5‐(2‐aminopropyl)‐2,3‐dihydrobenzofuran), 6‐APB (6‐(2‐aminopropyl)benzofuran), 6‐APDB (5‐(2‐aminopropyl)‐2,3‐dihydrobenzofuran), 4‐APB (4‐(2‐aminopropyl)benzofuran), 7‐APB (7‐(2‐aminopropyl)benzofuran), 5‐EAPB (5‐(2‐ethylaminopropyl)benzofuran) and 5‐MAPDB (1‐(2,3‐dihydrobenzofuran‐5‐yl)‐N‐methylpropan‐2‐amine) are all structurally similar to MDMA.134 Similar to MDMA, they were found to activate the 5‐HT2B receptor (Dawson et al., 2014), which induces heart valve fibrosis124, 135, 136 and inhibit dopamine transporters (DAT).84, 99
MDPV (3,4‐methylanedioxypyrovalerone) is a cathinone derivative with a nitrogen atom in the pyrrolidine ring and a 3,4‐methylenedioxy group on the phenyl ring similar to MDMA.137 in vitro and in vivo rodent models showed that MDPV blocks the dopamine and norepinephrine transporters in a similar way to the pyrovalerone analogues. It is more potent at both the dopamine and norepinephrine transporters and less potent blocking serotonin in a similar way to cocaine.137, 138 The potent blockade of dopamine and epinephrine stipulates that MDPV has the potential of inducing a high risk of abuse/addiction, and life‐threatening cardiovascular stimulation including tachycardia and hypertension respectively, more than cocaine.137, 138 This was consistent with in vitro data assessing blood–brain barrier permeability, in vivo microdialysis and in vivo locomotor activity testing in rats.130, 137, 138
One problem with preclinical studies is whether or not the toxic dose or concentration from the preclinical study reflects clinical doses. There is a wealth of literature on MDMA toxicity in the 5‐HT system, but this has been criticised because the doses needed to show neurotoxicity in animals may be far above the clinically relevant doses.139
Taken together, we can see that preclinical studies can be used to predict health harms. Some researchers have gone a step further, using in silico studies to predict addictive liability of NPS. The benzofurans 5‐APB and 5‐MAPB were predicted to bind to DAT in a similar way to MDMA,84 while ketamine‐like NPS diphenidine and methoxphenidine were predicted to bind differently at DAT.140
5. EVIDENCE USED IN UK LEGISLATION AND IN EUROPE
5.1. Evidence used in the UK
In the UK, the government takes advice on issues around drugs of abuse from the ACMD who risk assess emerging substances and issue subsequent recommendations.141 The ACMD also advises the government on the control of drugs and drugs requiring a temporary class drug order. In their first NPS report, on BZP (benzylpiperazine), they recommended that the drug and some of its 1‐phenyl and 1‐benzyl derivatives be brought under the Misuse of Drugs Act (1971): “The ACMD considers that the harms and misuse of BZP and substituted piperazines (identified in Annex 4) are commensurate with Class C, under schedule 2, part III, of the Misuse of Drugs Act (1971); and should be scheduled under Schedule I of the Misuse of Drugs Regulations (2001) (having no recognised medicinal use).” This decision was based on a report by the EMCDDA.142 In this report there is evidence from preclinical studies in the 1970s and 80s that BZP had addictive liability and had similar properties to amphetamine or MDMA. These studies were undertaken as the drug was being examined for antidepressant effects. More recent data from the 2000s are focused on human use and in particular BZP use in New Zealand where it was a popular party drug. These data suggest that the drug should not be used by people susceptible to seizures or in those with cardiac conditions. It is difficult to attribute blame to a particular drug in many of the human studies as in most cases the drug takers, although being shown to have BZP in their systems, may have consumed numerous other substances. Thus, although the UK government banned BZP, as it was obliged to do by the European Commission, the direct evidence for toxicity was limited because much of the data came from polydrug users. Nevertheless, its known pharmacological profile and its suspected effects on humans clearly merited some sort of control.
In another early publication, this time advice to the then Home Secretary Theresa May, on D2PM (Ivory wave) which “… typically cause prolonged agitation (lasting up to 5 days after drug use which is sometimes severe, requiring physical restraint), paranoia, hallucinations and myoclonus (muscle spasms/twitches)”, the ACMD recommended that there was an immediate ban on the import of 2‐DPMP. This advice was followed a year later by a report on desoxypipradrol,3 the ACMD could again rely on data from previous studies as desoxypipradrol had been tested for narcolepsy. After considering information from hospital emergency rooms, published research, drug company data and coroners reports the advice was to bring desoxypipradrol and its associated drugs D2PM and 2‐diphenylmethylpyrrolidine under the Misuse of Drugs Act (1971). The clinical evidence used to ban desoxypipradrol included 3 studies: a case report of a man who had taken D2PM with agitation and chest pains143; 5 case reports have shown the presence of D2PM rather than desoxypipradrol and the victims exhibited signs of agitation, anxiety and insomnia but not any sympathomimetic toxicity144; 2 case reports involving polydrug abusers, again presented with agitation, anxiety and insomnia but without increased heart rate, hypertension or hyperthermia.145 None of these studies actually confirmed desoxypipradrol use. Recent preclinical data have shown that desoxypipradrol was more potent than cocaine at inhibiting dopamine reuptake.100 Perhaps the best evidence to ban desoxypipradrol came from a report by Ciba‐Geigy in the 1950s showing that desoxypipradrol tended to have a much lower 50% lethal dose in animals than amphetamine or methamphetamine.
The ACMD went on to provide a number of reports and advice on various NPS including cathinones (2010), methoxetamine (2012), benzofurans (2013), N‐BOMe, AH‐7921 (3,4‐dochloro‐N‐[[1‐(dimethylamino)cyclohexyl]‐benzamide), tryptamines, MT‐45 (1‐cyclohexyl‐4‐(1,2‐diphenylethyl)‐piperazine) and 4,4'DMAR (all 2014), MPA (methiopropamine; 2016), third generation SCRAs (2016), https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=7236‐like NPS (2017) and 2–4‐dinitrophenol (2019). Not all NPS had a previous history as a potential medicine, where there were previous preclinical and clinical studies on the compound. For example, see the EMCDDA risk assessments where the pharmacodynamic profiles of emerging substances such as SCRAs on other pharmacological targets other than CB1 is very limited. This is particularly true for AB‐CHMINACA (N‐(1‐amino‐3‐methyl‐1‐oxobutan‐2‐yl)‐1‐(cyclohexylmethyl)‐1H‐indazole‐3‐carboxamide), ADB‐CHMINACA (N‐(1‐amino‐3,3‐dimethyl‐1‐oxobutan‐2‐yl)‐1‐(cyclohexylmethyl)‐1H‐indazole‐3‐carboxamide), 5F‐MDMB‐PINACA (5F‐ADB or methyl‐2‐{[1‐(5‐fluoropentyl)‐1H‐indazole3‐carbonyl]amino}‐3,3‐dimethylbutanoate), CUMYL‐4CN‐BINACA (1‐(4‐cyanobutyl)‐N‐(2‐phenylpropan‐2‐yl)indazole‐3‐carboxamide) and many more.
In May 2016, the UK government adopted a different approach aimed at limiting the supply of NPS and capturing substances that escaped the Misuse of Drugs Act 19714 rather than on evidence‐based risk assessments. However, as explained above, implementation of the Act is highly reliant on in vitro databases of a representative set of molecules.6 This is quite limited given the time taken to develop these libraries and the limitations of these libraries when facing emerging previously unseen substances.
Take together we can see that preclinical evidence has previously played an important role in deciding to ban NPS in the UK and continues to support the implementation of the new legislation. The clinical evidence, mostly case reports, is somewhat unreliable due to polydrug use and comorbidity, amongst other issues. Much of the preclinical evidence only suggested potential harms, rather than showing actual toxicity, and is dogged by the obvious need to extrapolate from animals to humans.
5.2. Overview of evidence used in Europe
In Europe, the process has been quite similar to that described above for the UK. The EMCDDA have published numerous reports and advice on NPS. These started with BZP (2007, 2009), Spice (2009), 4‐MA (2012), MDPV, methoxetamine, AH7921, 25I‐NBOMe, 4,4′‐DMAR and MT‐45 (2014) and more recently several reports on SCRAs and synthetic opioids. In their report129 on “New Psychoactive Substances in Europe; Legislation and Prosecution; Current challenges and Solutions” they describe 4 broad attempts of member countries controlling NPS. First, by trying to control NPS under laws around medicinal products, but this was thrown out by the Centre for Justice for the European Union on the grounds that NPS were not medicines. Second, some countries have tried to use existing laws around existing consumer safety laws. Third, existing drug laws have been modified by using group definitions of some NPS. Fourth, new laws have been developed, such as the UK Psychoactive Substances Act 2016 described above. In these new laws, the criteria used to define a psychoactive substance are often different and most use an element around harm or threat to health (including dependence) and a criterion that the drug has a psychoactive effect. Clearly, without preclinical data, and preferably human data, it is difficult to say with any certainty that an NPS has a psychoactive effect or could cause harm. Some countries also define psychoactivity quite clearly in their legislation with Ireland requiring significant mental disturbance or change and Hungary and Portugal requiring the NPS to have “effects similar to established drugs of abuse and a likelihood to cause dependence”.129
The EMCDDA have published 22 risk assessments for NPS. The most recent assessment will be used as an example. This was a risk assessment of the synthetic opioid cyclopropylfentanyl. The EMCDDA and Europol examined the available information based upon the following criteria: (i) the amount of the material seized; (ii) evidence of organised crime involvement; (iii) evidence of international trafficking; (iv) analogy with better‐studied compounds; (v) evidence of the potential for further (rapid) spread; and (vi) evidence of cases of serious intoxication or fatalities. They reported a pharmacological description of a few studies mostly limited to examining the effects of this NPS at μ‐opioid receptors in vitro, where it was shown to be more potent than https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=1627 or https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=1626 146 and a single animal study, in mice, suggesting analgesic properties.147 The health risks were suggested to come from accidental overdose and no acute or chronic toxicity studies had been carried out, nor had its dependence liability been examined. It was assumed that this NPS would have toxicity similar to morphine or fentanyl. The report described deaths associated with cyclopropylfentanyl, which included 78 in Sweden, 3 in the UK, 1 in Norway and over 100 in the USA,146 although in nearly all cases users had recently used multiple drugs. The report was submitted to the European Commission and Council of the European Union, who decided that cyclopropylfentanyl should be subject to control measures across member states. Thus, the EU banned this NPS based on very little direct pharmacological or toxicology data and include information on criminality as a criterion to ban the substance.
6. CONCLUSIONS
Clearly it would be beneficial to know as much as possible about each NPS, however scientific curiosity needs to be tempered by the reality of the current drug scene where the vast majority of problems are caused by relatively few established drugs of abuse. It is possible that some NPS may join these established drugs of abuse as major players in the morbidity and mortality associated with recreational drug use and it is these most commonly used NPS that we should focus upon. However, horizon scanning for the next MCAT/mephedrone or Spice/SCRA is also important. We have summarised the preclinical data on NPS above and given examples of these data being used in UK and EU legislation. It is reasonable to ask if the changes in legislation have been evidence‐based? Nutt and colleagues have published in this area for established drugs of abuse and concluded that UK and Australian scheduling of drugs of abuse is not particularly evidence based. They highlight the easy access to nicotine and alcohol, despite these drugs being ranked as highly dangerous. By contrast, ecstasy, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=17 and magic mushrooms, despite being ranked very low on the danger scale, are schedule 1, class A drugs in the UK.148 It is difficult to say that the UK blanket ban4 on psychoactive substances is evidenced based. The vast majority of NPS have not been examined at all while those that have been examined, as described above, tend to be looked at for their addictive effects rather than toxic effects. In addition, studies examining the long‐term effects of NPS are very rare and these types of studies would be best placed from which to extrapolate health harms. Nevertheless, the ban appears to be having some of the desired effects as recent evidence suggests fewer problems associated with NPS.5 The ACMD and EMCDDA reports are generally evidence based but limited to a relatively small number of NPS or families of NPS and a relatively small number of supporting studies. Having said that, given the very large number of NPS (up to 4000 by some estimations,2 there is little chance of all of these drugs being looked at in detail. The best that we can realistically hope for is putting many of these drugs through some high throughput screens for receptor/transporter binding120, 149 and high throughput toxicity assays.150 An alternative approach is in silico testingi.e. modelling the effects of the drugs at receptors or transporters of interest. We have done this for some ketamine‐like NPS including diphenidine, methoxphenidine, MDMA, and some similar NPS including 5‐APB and 5‐MAPB.84, 140 We also suggest that future studies focus more on measures of toxicity (e.g. neurotoxicity, vasoconstriction, hepatotoxicity, seizures and hyperthermia) as this has clearly been overlooked with most studies focussing on rodent behavioural or neurochemical (dopamine) measures of dependence. Finally, there are very few preclinical studies using tissues from female rodents. This is important as drug effects do show sexual differentiation.151 Thus, in order to get an accurate overview of NPS pharmacology, more studies using female tissue are needed.
COMPETING INTERESTS
There are no competing interests to declare.
Santos‐Toscano R, Guirguis A, Davidson C. How preclinical studies have influenced novel psychoactive substance legislation in the UK and Europe. Br J Clin Pharmacol. 2020;86:452–481. 10.1111/bcp.14224
REFERENCES
- 1. EMCDDA . European Drug Report. 2019.
- 2. Schifano F, Napoletano F, Chiappini S, et al. New/emerging psychoactive substances and associated psychopathological consequences. Psychol Med. 2019;1‐13. [DOI] [PubMed] [Google Scholar]
- 3. ACMD (2011) Novel Psychoactive Substances report 2011. https://www.gov.uk/government/publications/novel-psychoactive-substances-report-2011
- 4. PSA 2016. Psychoactive Substances Act 2016 http://www.legislation.gov.uk/ukpga/2016/2/contents/enacted
- 5. Home Office . Drugs and Alcohol Unit/Centre for Applied Science and Technology. 2016. Psychoactive Substances Act 2016 Forensic Strategy. Home Office: UK.
- 6.CPS 2018 Crown Prosecution Service (2018). https://www.cps.gov.uk/legal-guidance/psychoactive-substances
- 7. Luethi D, Hoener MC, Liechti ME. Effects of the new psychoactive substances diclofensine, diphenidine, and methoxphenidine on monoaminergic systems. Eur J Pharmacol. 2018. Jan;819:242‐247. [DOI] [PubMed] [Google Scholar]
- 8. Schifano F, Orsolini L, Duccio Papanti G, Corkery JM. Novel psychoactive substances of interest for psychiatry. World Psychiatry. 2015;14(1):15‐26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Sumnall HR, Evans‐Brown M, McVeigh J. Social, policy, and public health perspectives on new psychoactive substances. Drug Test Anal. 2011;3(7–8):515‐523. [DOI] [PubMed] [Google Scholar]
- 10. Belin‐Rauscent A, Belin D. Animal models of drug addiction. Addictions‐From Pathophysiology to Treatment. 2012; 1‐30.
- 11. Gurney SMR, Scott KS, Kacinko SL, Presley BC, Logan BK. Pharmacology, toxicology, and adverse effects of synthetic cannabinoid drugs. Forensic Sci Rev. 2014;26(1):53‐78. [PubMed] [Google Scholar]
- 12. Lynch WJ, Nicholson KL, Dance ME, Morgan RW, Foley PL. Animal models of substance abuse and addiction: implications for science, animal welfare, and society. Comp Med. 2010;60(3):177‐188. [PMC free article] [PubMed] [Google Scholar]
- 13. Nutt D, Mclellan AT. Improve addiction treatment and policies? Public Health Rev. 2013;35(2):1‐12. [Google Scholar]
- 14. Shippenberg TS, Koob GF. (2002). Recent advances in animal models of drug addiction and alcoholism. Neuropsychopharmacology: The fifth generation of progress, 13;81‐97.
- 15. Davidson C, Gow AJ, Lee TH, Ellinwood EH. Methamphetamine neurotoxicity: necrotic and apoptotic mechanisms and relevance to human abuse and treatment. Brain Res Brain Res Rev. 2001;36(1):1‐22. [DOI] [PubMed] [Google Scholar]
- 16. Halpin LE, Collins SA, Yamamoto BK. Neurotoxicity of methamphetamine and 3,4‐methylenedioxymethamphetamine. Life Sci. 2014;97(1):37‐44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Cosyns B, Droogmans S, Rosenhek R, Lancellotti P. Drug‐induced valvular heart disease. Heart. 2013;99(1):7‐12. [DOI] [PubMed] [Google Scholar]
- 18. Myers FAJ, Bluth MH, Cheung WW. Ketamine: a cause of urinary tract dysfunction. Clin Lab Med. 2016;36(4):721‐744. [DOI] [PubMed] [Google Scholar]
- 19. Assi S, Gulyamova N, Ibrahim K, Kneller P, Osselton D. Profile, effects, and toxicity of novel psychoactive substances: a systematic review of quantitative studies. Hum Psychopharmacol. 2017;32(3):e2607. [DOI] [PubMed] [Google Scholar]
- 20. Tracy DK, Wood DM, Baumeister D. Novel psychoactive substances: types, mechanisms of action, and effects. BMJ. 2017;356:i6848. [DOI] [PubMed] [Google Scholar]
- 21. Evans‐Brown M, Sedefov R. Responding to new psychoactive substances in the European Union: early warning, risk assessment, and control measures. Handb Exp Pharmacol. 2018;252:3‐49. [DOI] [PubMed] [Google Scholar]
- 22. Dias D, Ferreira B, Roque R, Rita B, Tomás R, Almeida D. De. The new psychoactive substance 3 ‐ methylmethcathinone (3 ‐ MMC or metaphedrone) induces oxidative stress, apoptosis, and autophagy in primary rat hepatocytes at human ‐ relevant concentrations. Arch Toxicol [Internet]. 2019;93(9):2617‐2634. 10.1007/s00204-019-02539-x [DOI] [PubMed] [Google Scholar]
- 23. Gannon BM, Mesmin MP, Sulima A, Rice KC, Collins GT. Behavioral economic analysis of the reinforcing effects of “bath salts” mixtures: studies with MDPV, methylone, and caffeine in male Sprague‐Dawley rats. Psychopharmacology (Berl). 2019;236(3):1031‐1041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Luethi D, Kolaczynska KE, Walter M, et al. Metabolites of the ring‐substituted stimulants MDMA, methylone and MDPV differentially affect human monoaminergic systems. 2019; [DOI] [PMC free article] [PubMed]
- 25. Blough BE, Decker AM, Landavazo A, et al. The dopamine, serotonin and norepinephrine releasing activities of a series of methcathinone analogs in male rat brain synaptosomes. Psychopharmacology (Berl). 2019;236(3):915‐924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Zhou X, Luethi D, Sanvee GM, Bouitbir J, Liechti ME, Krähenbühl S. Molecular Toxicological Mechanisms of Synthetic Cathinones on C2C12 Myoblasts. 2019; [DOI] [PMC free article] [PubMed]
- 27. Gerecsei LI, Balazsa T, Echevarria D, Adam A, Zachar G, Csillag A. Selective neuronal death following exposure to methylenedioxypyrovalerone is accompanied by an inhibition of NMDA receptor NR2B subunit expression. Acta Neurobiol Exp (Wars). 2019;79(1):92‐100. [PubMed] [Google Scholar]
- 28. Kolesnikova TO, Khatsko SL, Eltsov OS, Shevyrin VA, Kalueff AV. When fish take a bath: psychopharmacological characterization of the effects of a synthetic cathinone bath salt “flakka” on adult zebrafish. Neurotoxicol Teratol [Internet]2019/02/20. 2019;73:15‐21. Available from: https://www.ncbi.nlm.nih.gov/pubmed/30796953 [DOI] [PubMed] [Google Scholar]
- 29. Mayer FP, Cintulova D, Pittrich DA, et al. Stereochemistry of phase‐1 metabolites of mephedrone determines their effectiveness as releasers at the serotonin transporter. Neuropharmacology. 2019;148:199‐209. [DOI] [PubMed] [Google Scholar]
- 30. Eshleman AJ, Nagarajan S, Wolfrum KM, et al. Structure‐activity relationships of bath salt components: substituted cathinones and benzofurans at biogenic amine transporters. Psychopharmacology (Berl). 2019;236(3):939‐952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Yoon HS, Ku MJ, Cai WT, Kim J‐H. A novel synthetic cathinone, α‐pyrrolidinopentiothiophenone (PVT), produces locomotor sensitization in rat: implications for GSK3β connections in the nucleus accumbens core. Neurochem Int [Internet]2018/12/18. 2019. Mar;124:25‐30. Available from: https://www.ncbi.nlm.nih.gov/pubmed/30571998. [DOI] [PubMed] [Google Scholar]
- 32. Atehortua‐Martinez LA, Masniere C, Campolongo P, et al. Acute and chronic neurobehavioral effects of the designer drug and bath salt constituent 3,4‐methylenedioxypyrovalerone in the rat. J Psychopharmacol. 2019;33(3):392‐405. [DOI] [PubMed] [Google Scholar]
- 33. Colon‐Perez LM, Pino JA, Saha K, et al. Functional connectivity, behavioral and dopaminergic alterations 24 hours following acute exposure to synthetic bath salt drug methylenedioxypyrovalerone. Neuropharmacology. 2018;137:178‐193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Oliver CF, Simmons SJ, Nayak SU, Smith GR, Reitz AB, Rawls SM. Chemokines and ‘bath salts’: CXCR4 receptor antagonist reduces rewarding and locomotor‐stimulant effects of the designer cathinone MDPV in rats. Drug Alcohol Depend [Internet]. 2018;186:75‐79. Available from: 10.1016/j.drugalcdep.2018.01.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Luethi D, Kolaczynska KE, Docci L, Krahenbuhl S, Hoener MC, Liechti ME. Pharmacological profile of mephedrone analogs and related new psychoactive substances. Neuropharmacology. 2018;134(Pt A):4‐12. [DOI] [PubMed] [Google Scholar]
- 36. Siedlecka‐Kroplewska K, Wrońska A, Stasiłojć G, Kmieć Z. The designer drug 3‐Fluoromethcathinone induces oxidative stress and activates autophagy in HT22 neuronal cells. Neurotox Res. 2018;34(3):388‐400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Zwartsen A, Hondebrink L, Westerink RH. Neurotoxicity screening of new psychoactive substances (NPS): effects on neuronal activity in rat cortical cultures using microelectrode arrays (MEA). Neurotoxicology [Internet]. 2018;66:87‐97. Available from: 10.1016/j.neuro.2018.03.007 [DOI] [PubMed] [Google Scholar]
- 38. Gerecsei LI, Csillag A, Zachar G, et al. Gestational exposure to the synthetic cathinone methylenedioxypyrovalerone results in reduced maternal care and behavioral alterations in mouse pups. Front Neurosci. 2018;12:1‐14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Gannon BM, Rice KC. CGT. Reinforcing effects of abused‘bath salts'constituents3,4‐methylenedioxypyrovalerone and α‐pyrrolidinopentiophenone and their enantiomers. Behav Pharmacol. 2017;28(7):578‐581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Philogene‐Khalid HL, Simmons SJ, Nayak S, et al. Stereoselective differences between the reinforcing and motivational effects of cathinone‐derived 4‐Methylmethcathinone (Mephedrone) in self‐administering rats. ACS Chem Nerosci. 2017;8(12):2648‐2654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Nelson KH, Hempel BJ, Clasen MM, Rice KC, Riley AL. Conditioned taste avoidance, conditioned place preference and hyperthermia induced by the second generation ‘bath salt’ α‐pyrrolidinopentiophenone (α‐PVP). Pharmacol Biochem Behav. 2017;156:48‐55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Gannon BM, Russell LN, Modi MS, Rice KC, Fantegrossi WE. Effects of orally self‐administered bath salt constituent 3,4‐methylenedioxypyrovalerone (MDPV) in mice. Drug Alcohol Depend [Internet]. 2017;179:408‐415. Available from: 10.1016/j.drugalcdep.2017.06.031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Lantz SM, Rosas‐Hernandez H, Cuevas E, et al. Monoaminergic toxicity induced by cathinone phthalimide: an in vitro study. Neurosci Lett [Internet]. 2017;655:76‐81. Available from: 10.1016/j.neulet.2017.06.059 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Luethi D, Liechti ME, Krahenbuhl S. Mechanisms of hepatocellular toxicity associated with new psychoactive synthetic cathinones. Toxicology. 2017;387:57‐66. [DOI] [PubMed] [Google Scholar]
- 45. Grecco GG, Kisor DF, Magura JS, Sprague JE. Impact of common clandestine structural modifications on synthetic cathinone “bath salt” pharmacokinetics. Toxicol Appl Pharmacol. 2017;328:18‐24. [DOI] [PubMed] [Google Scholar]
- 46. Elmore JS, Dillon‐Carter O, Partilla JS, et al. Pharmacokinetic profiles and Pharmacodynamic effects for Methylone and its metabolites in rats. Neuropsychopharmacology [Internet]. 2017;42(3):649‐660. Available from: 10.1038/npp.2016.213 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. McLaughlin G, Morris N, Kavanagh PV, et al. Synthesis, characterization and monoamine transporter activity of the new psychoactive substance mexedrone and its N‐methoxy positional isomer, N‐methoxymephedrone. Drug Test Anal. 2017;9(3):358‐368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Schindler CW, Thorndike EB, Suzuki M, Rice KC, Baumann MH. Pharmacological mechanisms underlying the cardiovascular effects of the “bath salt” constituent 3,4‐methylenedioxypyrovalerone (MDPV). Br J Pharmacol. 2016;173(24):3492‐3501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Berquist MD 2nd, Traxler HK, Mahler AM, Baker LE. Sensitization to the locomotor stimulant effects of “bath salt” constituents, 4‐methylmethcathinone (4‐MMC) and 3,4‐methylenedioxypyrovalerone (MDPV), in male Sprague‐Dawley rats. Drug Alcohol Depend. 2016;164:128‐134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Schindler CW, Thorndike EB, Goldberg SR, et al. Reinforcing and neurochemical effects of the “bath salts” constituents 3,4‐methylenedioxypyrovalerone (MDPV) and 3,4‐methylenedioxy‐N‐methylcathinone (methylone) in male rats. Psychopharmacology (Berl). 2016;233(10):1981‐1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Valente MJ, Araújo AM, Bastos M d L, et al. Characterization of hepatotoxicity mechanisms triggered by designer cathinone drugs (β‐keto amphetamines). Toxicol Sci. 2016;153(1):89‐102. [DOI] [PubMed] [Google Scholar]
- 52. Colon‐Perez LM, Tran K, Thompson K, et al. The psychoactive designer drug and bath salt constituent MDPV causes widespread disruption of brain functional connectivity. Neuropsychopharmacology [Internet]. 2016;41(9):2352‐2365. Available from: 10.1038/npp.2016.40 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Gannon BM, Williamson A, Suzuki M, Rice KC, Fantegrossi WE. Stereoselective effects of abused “bath salt” constituent 3,4‐methylenedioxypyrovalerone in mice: drug discrimination, locomotor activity, and thermoregulation. J Pharmacol Exp Ther. 2016;356(3):615‐623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Lopez‐Arnau R, Martinez‐Clemente J, Pubill D, Escubedo E, Camarasa J. Serotonergic impairment and memory deficits in adolescent rats after binge exposure of methylone. J Psychopharmacol. 2014;28(11):1053‐1063. [DOI] [PubMed] [Google Scholar]
- 55. Kiyatkin EA, Kim AH, Wakabayashi KT, Baumann MH, Shaham Y. Effects of social interaction and warm ambient temperature on brain hyperthermia induced by the designer drugs Methylone and MDPV. Neuropsychopharmacology [Internet]. 2015;40(2):436‐445. Available from: 10.1038/npp.2014.191 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Hutchinson CV, Prados J, Davidson C. Persistent conditioned place preference to cocaine and withdrawal hypo‐locomotion to mephedrone in the flatworm planaria. Neurosci Lett [Internet]. 2015;593:19‐23. Available from: 10.1016/j.neulet.2015.03.021 [DOI] [PubMed] [Google Scholar]
- 57. Lopez‐Arnau R, Martinez‐Clemente J, Carbo M l, Pubill D, Escubedo E, Camarasa J. An integrated pharmacokinetic and pharmacodynamic study of a new drug of abuse, methylone, a synthetic cathinone sold as “bath salts”. Prog Neuropsychopharmacol Biol Psychiatry. 2013;45:64‐72. [DOI] [PubMed] [Google Scholar]
- 58. Adam A, Gerecsei LI, Lepesi N, Csillag A. Apoptotic effects of the “designer drug” methylenedioxypyrovalerone (MDPV) on the neonatal mouse brain. Neurotoxicology. 2014;44:231‐236. [DOI] [PubMed] [Google Scholar]
- 59. Den Hollander B, Rozov S, Linden AM, Uusi‐Oukari M, Ojanperä I, Korpi ER. Long‐term cognitive and neurochemical effects of “bath salt” designer drugs methylone and mephedrone. Pharmacol Biochem Behav [Internet]. 2013;103(3):501‐509. Available from: 10.1016/j.pbb.2012.10.006 [DOI] [PubMed] [Google Scholar]
- 60. Opacka‐Juffry J, Pinnell T, Patel N, Bevan M, Meintel M, Davidson C. Stimulant mechanisms of cathinones ‐ effects of mephedrone and other cathinones on basal and electrically evoked dopamine efflux in rat accumbens brain slices. Prog Neuropsychopharmacol Biol Psychiatry. 2014;54:122‐130. [DOI] [PubMed] [Google Scholar]
- 61. Simmler LD, Rickli A, Hoener MC, Liechti ME. Monoamine transporter and receptor interaction profiles of a new series of designer cathinones. Neuropharmacology [Internet]2013/11/22. 2014;79:152‐160. Available from: https://www.ncbi.nlm.nih.gov/pubmed/24275046 [DOI] [PubMed] [Google Scholar]
- 62. Kaizaki A, Tanaka S, Numazawa S. New recreational drug 1‐phenyl‐2‐(1‐pyrrolidinyl)‐1‐pentanone (alpha‐PVP) activates central nervous system via dopaminergic neuron. J Toxicol Sci. 2014;39(1):1‐6. [DOI] [PubMed] [Google Scholar]
- 63. Marusich JA, Antonazzo KR, Wiley JL, Blough BE, Partilla JS, MHB . Pharmacology of novel synthetic stimulants structurally related to the “bath salts” constituent 3,4‐methylenedioxypyrovalerone (MDPV). Neuropharmacology. 2014;87:206‐213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Watterson LR, Burrows BT, Hernandez RD, et al. Effects of alpha‐pyrrolidinopentiophenone and 4‐methyl‐N‐ethylcathinone, two synthetic cathinones commonly found in second‐generation “bath salts,” on intracranial self‐stimulation thresholds in rats. Int J Neuropsychopharmacol. 2014;18(1). 10.1093/ijnp/pyu014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Bonano JS, Glennon RA, De Felice LJ, Banks ML, Negus SS. Abuse‐related and abuse‐limiting effects of methcathinone and the synthetic “bath salts” cathinone analogs methylenedioxypyrovalerone (MDPV), methylone and mephedrone on intracranial self‐stimulation in rats. Psychopharmacology (Berl). 2014;231(1):199‐207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Gregg RA, Tallarida CS, Reitz AB, Rawls SM. Mephedrone interactions with cocaine: prior exposure to the “bath salt” constituent enhances cocaine‐induced locomotor activation in rats. Behav Pharmacol. 2013;24(8):684‐688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Gregg RA, Tallarida CS, Reitz A, McCurdy C, Rawls SM. Mephedrone (4‐methylmethcathinone), a principal constituent of psychoactive bath salts, produces behavioral sensitization in rats. Drug Alcohol Depend. 2013;133(2):746‐750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Gatch MB, Taylor CM, Forster MJ. Locomotor stimulant and discriminative stimulus effects of “bath salt” cathinones. Behav Pharmacol. 2013;24(5–6):437‐447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Cameron K, Kolanos R, Vekariya R, De Felice L, Glennon RA. Mephedrone and methylenedioxypyrovalerone (MDPV), major constituents of “bath salts,” produce opposite effects at the human dopamine transporter. Psychopharmacology (Berl). 2013;227(3):493‐499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Baumann MH, Jr MAA, Partilla JS, et al. The designer Methcathinone analogs, Mephedrone and Methylone, are substrates for monoamine transporters in brain tissue. Neuropsychopharmacology [Internet]. 2011;37(5):1192‐1203. Available from: 10.1038/npp.2011.304 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.10.1016/j.pbb.2012.10.006 Den Hollander, 2013.
- 72. López‐Arnau R, Martínez‐Clemente J, Rodrigo T, Pubill D, Camarasa J, Escubedo E. Neuronal changes and oxidative stress in adolescent rats after repeated exposure to mephedrone. Toxicol Appl Pharmacol [Internet]. 2015;286(1):27‐35. Available from: 10.1016/j.taap.2015.03.015 [DOI] [PubMed] [Google Scholar]
- 73. Cozzi NV, Brandt SD, Daley PF, et al. Pharmacological examination of trifluoromethyl ring‐substituted methcathinone analogs. Eur J Pharmacol. 2014;699(0):180‐187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Lisek R, Xu W, Yuvasheva E, et al. Mephedrone ('bath salt’) elicits conditioned place preference and dopamine‐sensitive motor activation. Drug Alcohol Depend. 2012;126(1–2):257‐262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Meng H, Cao J, Kang J, et al. Mephedrone, a new designer drug of abuse, produces acute hemodynamic effects in the rat. Toxicol Lett. 2012;208(1):62‐68. [DOI] [PubMed] [Google Scholar]
- 76. Rickli A, Kolaczynska K, Hoener MC, Liechti ME. Pharmacological characterization of the aminorex analogs 4‐MAR, 4,4′‐DMAR, and 3,4‐DMAR. Neurotoxicology. 2019;72:95‐100. [DOI] [PubMed] [Google Scholar]
- 77. Cai WT, Yoon HS, Lee S, Kim J‐H. Repeated exposure to methiopropamine increases dendritic spine density in the rat nucleus accumbens core. Neurochem Int. 2019;129104487. [DOI] [PubMed] [Google Scholar]
- 78. Maier J, Mayer FP, Luethi D, et al. The psychostimulant (±)‐cis‐4,4′‐dimethylaminorex (4,4′‐DMAR) interacts with human plasmalemmal and vesicular monoamine transporters. Neuropharmacology. 2018;138:282‐291. [DOI] [PubMed] [Google Scholar]
- 79. Davidson C, Raby CAR, Barrese V, Ramsey J. In vitro neurochemical assessment of methylphenidate and its “legal high” analogs 3,4‐CTMP and ethylphenidate in rat nucleus accumbens and bed nucleus of the stria terminalis. Front Psych. 2018;9:1‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Luethi D, Kaeser PJ, Brandt SD, Krahenbuhl S, Hoener MC, Liechti ME. Pharmacological profile of methylphenidate‐based designer drugs. Neuropharmacology. 2018;134(Pt A):133‐140. [DOI] [PubMed] [Google Scholar]
- 81. McLaughlin G, Baumann MH, Kavanagh PV, et al. Synthesis, analytical characterization, and monoamine transporter activity of the new psychoactive substance 4‐methylphenmetrazine (4‐MPM), with differentiation from its ortho‐ and meta‐ positional isomers. Drug Test Anal. 2018;10(9):1404‐1416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Zwartsen A, Verboven AHA, van Kleef RGDM, Wijnolts FMJ, Westerink RHS, Hondebrink L. Measuring inhibition of monoamine reuptake transporters by new psychoactive substances (NPS) in real‐time using a high‐throughput, fluorescence‐based assay. Toxicol Vitr [Internet]. 2017;45:60‐71. Available from: 10.1016/j.tiv.2017.05.010 [DOI] [PubMed] [Google Scholar]
- 83. Mayer FP, Burchardt NV, Decker AM, et al. Fluorinated phenmetrazine “legal highs” act as substrates for high‐affinity monoamine transporters of the SLC6 family. Neuropharmacology [Internet]. 2018;134:149‐157. Available from: 10.1016/j.neuropharm.2017.10.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Sahai MA, Davidson C, Khelashvili G, et al. Combined in vitro and in silico approaches to the assessment of stimulant properties of novel psychoactive substances ‐ the case of the benzofuran 5‐MAPB. Prog Neuropsychopharmacol Biol Psychiatry. 2017;75:1‐9. [DOI] [PubMed] [Google Scholar]
- 85. Shimshoni JA, Winkler I, Edery N, Golan E, van Wettum R, Nutt D. Toxicological evaluation of 5‐methoxy‐2‐aminoindane (MEAI): binge mitigating agent in development. Toxicol Appl Pharmacol [Internet]2017/02/04. 2017;319:59‐68. Available from: https://www.ncbi.nlm.nih.gov/pubmed/28167221 [DOI] [PubMed] [Google Scholar]
- 86. McLaughlin G, Morris N, Kavanagh PV, et al. Analytical characterization and pharmacological evaluation of the new psychoactive substance 4‐fluoromethylphenidate (4F‐MPH) and differentiation between the (±)‐threo and (±)‐erythro diastereomers. Drug Test Anal. 2017;9(3):347‐357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Yoon HS, Cai WT, Lee YH, Park KT, Lee YS, Kim JH. The expression of methiopropamine‐induced locomotor sensitization requires dopamine D2, but not D1, receptor activation in the rat. Behav Brain Res [Internet]. 2016;311:403‐407. Available from: 10.1016/j.bbr.2016.05.060 [DOI] [PubMed] [Google Scholar]
- 88. Marusich JA, Antonazzo KR, Blough BE, et al. The new psychoactive substances 5‐(2‐aminopropyl)indole (5‐IT) and 6‐(2‐aminopropyl)indole (6‐IT) interact with monoamine transporters in brain tissue. Neuropharmacology. 2016;101:68‐75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Simmler LD, Buchy D, Chaboz S, Hoener MC, Liechti ME. In vitro characterization of psychoactive substances at rat, Mouse, and Human Trace Amine‐Associated Receptor 1 s. 2016;134–144. [DOI] [PubMed]
- 90. Asaoka N, Kawai H, Nishitani N, et al. A new designer drug 5F‐ADB activates midbrain dopaminergic neurons but not serotonergic neurons. J Toxicol Sci. 2016;41(6):813‐816. [DOI] [PubMed] [Google Scholar]
- 91. Persona K, Polus A, Góralska J, Gruca A, Dembińska‐Kieć A, Piekoszewski W. An in vitro study of the neurotoxic effects of N‐Benzylpiperazine: a designer drug of abuse. Neurotox Res. 2016;29(4):558‐568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Arbo MD, Melega S, Stöber R, et al. Hepatotoxicity of piperazine designer drugs: up‐regulation of key enzymes of cholesterol and lipid biosynthesis. Arch Toxicol. 2016;90(12):3045‐3060. [DOI] [PubMed] [Google Scholar]
- 93. Arbo MD, Silva R, Barbosa DJ, et al. In vitro neurotoxicity evaluation of piperazine designer drugs in differentiated human neuroblastoma SH‐SY5Y cells. J Appl Toxicol. 2016;36(1):121‐130. [DOI] [PubMed] [Google Scholar]
- 94. Rubio M, López‐Arnau R, Pubill D, Escubedo E, Camarasa J. Serotonin is involved in the psychostimulant and hypothermic effect of 4‐methylamphetamine in rats. Neurosci Lett [Internet]. 2015;590:68‐73.Available from: 10.1016/j.neulet.2015.01.075 [DOI] [PubMed] [Google Scholar]
- 95. Rickli A, Hoener MC, Liechti ME. Monoamine transporter and receptor interaction profiles of novel psychoactive substances: Para‐halogenated amphetamines and pyrovalerone cathinones. Eur Neuropsychopharmacol. 2015;25(3):365‐376. [DOI] [PubMed] [Google Scholar]
- 96. McLaughlin G, Morris N, Kavanagh PV, et al. Synthesis, characterization, and monoamine transporter activity of the new psychoactive substance 3′,4′‐methylenedioxy‐4‐methylaminorex (MDMAR). Drug Test Anal. 2015;7(7):555‐564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Arbo MD, Silva R, Barbosa DJ, et al. Piperazine designer drugs induce toxicity in cardiomyoblast h9c2 cells through mitochondrial impairment. Toxicol Lett [Internet]. 2014;229(1):178‐189Available from:. 10.1016/j.toxlet.2014.06.031 [DOI] [PubMed] [Google Scholar]
- 98. Brandt SD, Baumann MH, Partilla JS, et al. Characterization of a novel and potentially lethal designer drug (+/−)‐cis‐Para‐methyl‐4‐methylaminorex (4,4′‐DMAR, or'Serotoni’). Drug Test Anal. 2014;6(7–8):684‐695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Dawson P, Opacka‐Juffry J, Moffatt JD, et al. The effects of benzofury (5‐APB) on the dopamine transporter and 5‐HT2‐dependent vasoconstriction in the rat. Prog Neuropsychopharmacol Biol Psychiatry. 2014;48:57‐63. [DOI] [PubMed] [Google Scholar]
- 100. Davidson C, Ramsey J. Desoxypipradrol is more potent than cocaine on evoked dopamine efflux in the nucleus accumbens. J Psychopharmacol. 2011;26(7):1036‐1041. [DOI] [PubMed] [Google Scholar]
- 101. Bilel S, Tirri M, Arfè R, et al. Pharmacological and behavioral effects of the synthetic cannabinoid AKB48 in rats. Front Neurosci [Internet]. 2019;13:1163 Available from: https://www.ncbi.nlm.nih.gov/pubmed/31736697 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Banister SD, Adams A, Kevin RC, et al. Synthesis and pharmacology of new psychoactive substance 5F‐CUMYL‐P7AICA, a scaffold‐ hopping analog of synthetic cannabinoid receptor agonists 5F‐CUMYL‐PICA and 5F‐CUMYL‐PINACA. Drug Test Anal. 2019;11(2):279‐291. [DOI] [PubMed] [Google Scholar]
- 103. Kim S, Choi W, Kwon M, et al. In vitro inhibitory Effects of APINACA on human major cytochrome P450, UDP‐glucuronosyltransferase enzymes, and drug transporters. Molecules. 2019;24:3000. 10.3390/molecules24163000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Kevin RC. CUMYL‐4CN‐BINACA is an efficacious and potent pro‐ Convulsant synthetic cannabinoid receptor agonist. Front Pharmacol. 2019;10:1‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Domoto M, Sasase H, Wada S, et al. The synthetic cannabinoid 5F‐AMB changes the balance between excitation and inhibition of layer V pyramidal neurons in the mouse medial prefrontal cortex. Psychopharmacology (Berl). 2018;235(8):2367‐2376. [DOI] [PubMed] [Google Scholar]
- 106. De Luca MA, Castelli MP, Loi B, 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;105:630‐638. [DOI] [PubMed] [Google Scholar]
- 107. Yun J, Yoon KS, Lee TH, et al. Synthetic cannabinoid, JWH‐030, induces QT prolongation through hERG channel inhibition. Toxicol Res (Camb). 2016;5(6):1663‐1671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Tomiyama K, Funada M. Cytotoxicity of synthetic cannabinoids on primary neuronal cells of the forebrain: the involvement of cannabinoid CB1 receptors and apoptotic cell death. Toxicol Appl Pharmacol. 2014;274(1):17‐23. [DOI] [PubMed] [Google Scholar]
- 109. Randall MD, Kendall DA, O'Sullivan S. The complexities of the cardiovascular actions of cannabinoids. Br J Pharmacol. 2004;142(1):20‐26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Davidson C, Opacka‐Juffry J, Arevalo‐Martin A, Garcia‐Ovejero D, Molina‐Holgado E, Molina‐Holgado F. (2017) Advances in pharmacology, Chapter Four ‐ Spicing Up Pharmacology: A Review of Synthetic Cannabinoids From Structure to Adverse Events http://www.sciencedirect.com/science/journal/10543589/80/supp/C, 135–168. [DOI] [PubMed]
- 111. Zanda MT, Fattore L. Old and new synthetic cannabinoids: lessons from animal models. Drug Metab Rev. 2018;50(1):54‐64. [DOI] [PubMed] [Google Scholar]
- 112. Luethi D, Widmer R, Trachsel D, Hoener MC, Liechti ME. Monoamine receptor interaction profiles of 4‐aryl‐substituted 2,5‐dimethoxyphenethylamines (2C‐BI derivatives). Eur J Pharmacol. 2019;855:103‐111. [DOI] [PubMed] [Google Scholar]
- 113. Wallach J, Colestock T, Agramunt J, et al. Pharmacological characterizations of the “legal high” fluorolintane and isomers. Eur J Pharmacol. 2019;857: 172427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Costa G, Serra M, Pintori N, et al. The novel psychoactive substance methoxetamine induces persistent behavioral abnormalities and neurotoxicity in rats. Neuropharmacology [Internet]. 2018/10/23. 2019;144:219‐232. Available from: https://www.ncbi.nlm.nih.gov/pubmed/30366005 [DOI] [PubMed] [Google Scholar]
- 115. Yoon KS, Gu SM, Lamichhane S, et al. Methoxetamine induces cytotoxicity in H9c2 cells: possible role of p21 protein (Cdc42/Rac)‐activated kinase 1. Cardiovasc Toxicol [Internet]. 2019;19(3):229‐236. Available from: 10.1007/s12012-018-9489-4 [DOI] [PubMed] [Google Scholar]
- 116. Halberstadt AL, Chatha M, Stratford A, Grill M, Brandt SD. Comparison of the behavioral responses induced by phenylalkylamine hallucinogens and their tetrahydrobenzodifuran (“FLY”) and benzodifuran (“DragonFLY”) analogs. Neuropharmacology [Internet]2018/10/29. 2019;144:368‐376. Available from: https://www.ncbi.nlm.nih.gov/pubmed/30385253 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Herian M, Wojtas A, Kamińska K, Świt P, Wach A, Gołembiowska K. Hallucinogen‐like action of the novel designer drug 25I‐NBOMe and its effect on cortical neurotransmitters in rats. Neurotox Res. 2019;36(1):91‐100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Shintani‐Ishida K, Saka K, Nakamura M, Yoshida KI, Ikegaya H. Experimental study on the postmortem redistribution of the substituted phenethylamine, 25B‐NBOMe. J Forensic Sci. 2018;63:588‐591. [DOI] [PubMed] [Google Scholar]
- 119. Cha HJ, Jeon SY, Jang HJ, Shin J, Kim YH, Suh SK. Rewarding and reinforcing effects of 4‐chloro‐2,5‐dimethoxyamphetamine and AH‐7921 in rodents. Neurosci Lett [Internet]. 2018;676(April):66‐70. Available from: 10.1016/j.neulet.2018.04.009 [DOI] [PubMed] [Google Scholar]
- 120. Hondebrink L, Kasteel EEJ, Tukker AM, Wijnolts FMJ, Verboven AHA, Westerink RHS. Neuropharmacological characterization of the new psychoactive substance methoxetamine. Neuropharmacology [Internet]. 2017;123(2017):1‐9. Available from: 10.1016/j.neuropharm.2017.04.035 [DOI] [PubMed] [Google Scholar]
- 121. Kang H, Park P, Bortolotto ZA, et al. Ephenidine: a new psychoactive agent with ketamine‐like NMDA receptor antagonist properties. Neuropharmacology [Internet]. 2017;112(Pt A):144‐149. Available from: 10.1016/j.neuropharm.2016.08.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Rickli A, Moning OD, Hoener MC, Liechti ME. Receptor interaction profiles of novel psychoactive tryptamines compared with classic hallucinogens. Eur Neuropsychopharmacol [Internet]2016/05/20. 2016;26(8):1327‐1337. Available from: https://www.ncbi.nlm.nih.gov/pubmed/27216487 [DOI] [PubMed] [Google Scholar]
- 123. Wallach J, Kang H, Colestock T, et al. Pharmacological investigations of the dissociative “legal highs” Diphenidine, Methoxphenidine and analogues. PLoS One. 2016;11(6):1‐17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Rickli A, Kopf S, Hoener MC, Liechti ME. Pharmacological profile of novel psychoactive benzofurans. Br J Pharmacol. 2015;172(13):3412‐3425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Blough BE, Landavazo A, Decker AM, Partilla JS, Baumann MH, Rothman RB. Interaction of psychoactive tryptamines with biogenic amine transporters and serotonin receptor subtypes. Psychopharmacology (Berl). 2014;231(21):4135‐4144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Dargan PI, Tang HC, Liang W, Wood DM, Yew DT. Three months of methoxetamine administration is associated with significant bladder and renal toxicity in mice. Clin Toxicol (Phila). 2014;52(3):176‐180. [DOI] [PubMed] [Google Scholar]
- 127. Paulke A, Kremer C, Wunder C, et al. Argyreia nervosa (Burm. F.): receptor profiling of lysergic acid amide and other potential psychedelic LSD‐like compounds by computational and binding assay approaches. J Ethnopharmacol. 2013;148(2):492‐497. [DOI] [PubMed] [Google Scholar]
- 128. Compton DM, Dietrich KL, Selinger MC, Testa EK. 5‐methoxy‐N,N‐di (iso)propyltryptamine hydrochloride (foxy)‐induced cognitive deficits in rat after exposure in adolescence. Physiol Behav. 2011;103(2):203‐209. [DOI] [PubMed] [Google Scholar]
- 129. EMCDDA . New psychoactive substances in Europe. 2016.
- 130. Simmler LD, Buser TA, Donzelli M, et al. Pharmacological characterization of designer cathinones in vitro. Br J Pharmacol. 2013;168(2):458‐470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. EMCDDA . Report on the risk assessment of 2C‐I, 2C‐T‐2 and 2C‐T‐7 in the framework of the joint action on new synthetic drugs. 2004.
- 132. Chu PS, Kwok SC, Lam KM, et al. 'Street ketamine'‐associated bladder dysfunction: a report of ten cases. Hong Kong Med J. 2007;13(4):311‐313. [PubMed] [Google Scholar]
- 133. Wang Q, Wu Q, Wang J, et al. Ketamine analog Methoxetamine induced inflammation and dysfunction of bladder in rats. Int J Mol Sci. 2017;18(1):117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Greene SL. Benzofurans and benzodifurans In: Dargan PI, Wood DM, eds. Novel Psychoactive Substances. Amsterdam: Elsevier; 2013:283‐392. [Google Scholar]
- 135. Bhattacharyya S, Schapira AH, Mikhailidis DP, Davar J. Drug‐induced fibrotic valvular heart disease. Lancet (London, England). 2009;374(9689):577‐585. [DOI] [PubMed] [Google Scholar]
- 136. Setola V, Hufeisen SJ, Grande‐Allen KJ, et al. 3,4‐methylenedioxymethamphetamine (MDMA, “ecstasy”) induces fenfluramine‐like proliferative actions on human cardiac valvular interstitial cells in vitro. Mol Pharmacol. 2003;63(6):1223‐1229. [DOI] [PubMed] [Google Scholar]
- 137. EMCDDA–Europol (2013) EMCDDA–Europol 2012 Annual Report on the implementation of Council Decision 2005/387/JHA (New drugs in Europe, 2012), Implementation reports. http://www.emcdda.europa.eu/system/files/publications/819/TDAS14001ENN_466653.pdf
- 138. Baumann MH, Partilla JS, Lehner KR, et al. Powerful cocaine‐like actions of 3,4‐methylenedioxypyrovalerone (MDPV), a principal constituent of psychoactive “bath salts” products. Neuropsychopharmacology [Internet]. 2013;38(4):552‐562. Available from: 10.1038/npp.2012.204 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Baumann MH, Wang X, Rothman RB. 3,4‐Methylenedioxymethamphetamine (MDMA) neurotoxicity in rats: a reappraisal of past and present findings. Psychopharmacology (Berl). 2007;189(4):407‐424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Sahai MA, Davidson C, Dutta N, Opacka‐Juffry J. Mechanistic insights into the stimulant properties of novel psychoactive substances (NPS) and their discrimination by the dopamine transporter‐in Silico and in vitro exploration of dissociative Diarylethylamines. Brain Sci. 2018;8(4):63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.ACMD: https://www.gov.uk/search/policy-papers-and-consultations?organisations%5B%5D=advisory-council-on-the-misuse-of-drugs&parent=advisory-council-on-the-misuse-of-drugs
- 142. EMCDDA (2007) Report on the risk assessment of BZP in the framework of the Council decision on new psychoactive substances http://www.emcdda.europa.eu/system/files/publications/560/EMCDDA_risk_assessment_8_132618.pdf
- 143. Lidder S, Dargan P, Sexton M, et al. Cardiovascular toxicity associated with recreational use of diphenylprolinol (diphenyl‐2‐pyrrolidinemethanol [D2PM]). J Med Toxicol. 2008;4(3):167‐169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Wood DM, Puchnarewicz M, Johnston A, Dargan PI. A case series of individuals with analytically confirmed acute diphenyl‐2‐pyrrolidinemethanol (D2PM) toxicity. Eur J Clin Pharmacol. 2012;68(4):349‐353. [DOI] [PubMed] [Google Scholar]
- 145. Wood DM, Puchnarewicz M, Holt DW, Ramsey J, Dargan PI. Detection of the precursor benzophenone in individuals who have used legal highs containing diphenyl‐2‐pyyrrolidinemethanol (D2PM). Basic Clin Pharmacol Toxicol. 2011;109(Suppl. 1):86 10.1111/j.1742-7843.2011.00722.x [DOI] [Google Scholar]
- 146. United States Drug Enforcement Administration (US DEA) (2017), Cyclopropyl fentanyl. Background information and evaluation of ‘three factor analysis’ (factors 4, 5, and 6) for temporary scheduling. Drug and chemical evaluation section, diversion control division, drug enforcement administration, Washington, DC October 2017. Available at: https://www.regulations.gov/document?D=DEA-2017-0013-0003
- 147. Janssen PAJ, Van der Eycken CAM. The chemical anatomy of potent morphine‐like analgesics In: Burger A, ed. Drugs affecting the central nervous system. Vol.2 Inc, New York: Marcel Dekker; 1968:25‐60. [Google Scholar]
- 148. Nutt DJ, King LA, Phillips LD. Drug harms in the UK: a multicriteria decision analysis. Lancet (London, England). 2010;376(9752):1558‐1565. [DOI] [PubMed] [Google Scholar]
- 149. Iversen L, Gibbons S, Treble R, Setola V, Huang X‐P, Roth BL. Neurochemical profiles of some novel psychoactive substances. Eur J Pharmacol. 2013;700(1–3):147‐151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Araujo AM, Valente MJ, Carvalho M, et al. Raising awareness of new psychoactive substances: chemical analysis and in vitro toxicity screening of “legal high” packages containing synthetic cathinones. Arch Toxicol. 2015;89(5):757‐771. [DOI] [PubMed] [Google Scholar]
- 151. Becker JB. Sex differences in addiction. Dialogues Clin Neurosci. 2016;18(4):395‐402. [DOI] [PMC free article] [PubMed] [Google Scholar]