Notice: file_put_contents(): Write of 323527 bytes failed with errno=28 No space left on device in /opt/frankenphp/design.onmedianet.com/app/src/Arsae/CacheManager.php on line 36

Warning: http_response_code(): Cannot set response code - headers already sent (output started at /opt/frankenphp/design.onmedianet.com/app/src/Arsae/CacheManager.php:36) in /opt/frankenphp/design.onmedianet.com/app/src/Models/Response.php on line 17

Warning: Cannot modify header information - headers already sent by (output started at /opt/frankenphp/design.onmedianet.com/app/src/Arsae/CacheManager.php:36) in /opt/frankenphp/design.onmedianet.com/app/src/Models/Response.php on line 20
Dark Classics in Chemical Neuroscience: NBOMes - PMC Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Jun 13.
Published in final edited form as: ACS Chem Neurosci. 2019 Nov 12;11(23):3860–3869. doi: 10.1021/acschemneuro.9b00528

Dark Classics in Chemical Neuroscience: NBOMes

Christian B M Poulie, Anders A Jensen, Adam L Halberstadt, Jesper L Kristensen
PMCID: PMC9191638  NIHMSID: NIHMS1059581  PMID: 31657895

Abstract

N-Benzylphenethylamines, commonly known as NBOMes, are synthetic psychedelic compounds derived from the phenethylamine class of psychedelics (2C-X compounds), which originally have been derived from the naturally occurring alkaloid mescaline. Analogously to their parent compounds and other classical psychedelics, such as psilocybin and lysergic acid diethylamide (LSD), NBOMes are believed to exert their main pharmacological effects through activation of serotonin 2A (5-HT2A) receptors. Since their introduction as New Psychoactive Substances (NPSs) in 2010, NBOMes have been widely used for recreational purposes; this has resulted in numerous cases of acute toxicity, sometimes with lethal outcomes, leading to the classification of several NBOMes as Schedule I substances in 2013. However, in addition to their recreational use, the NBOMe class has yielded several important biochemical tools, including [11C]Cimbi-36, which is now being used in positron emission tomography (PET) studies of the 5-HT2A. and 5-HT2C receptors in the mammalian brain, and 25CN-NBOH, one of the most selective 5-HT2A receptor agonists developed to date. In this Review, the history, chemistry, structure–activity relationships, ADME (absorption, distribution, metabolism, and excretion) properties, and safety profiles of NBOMes will be outlined and discussed.

Keywords: N-Benzylphenethylamines, novel psychoactive substance, 5-HT2A receptor, psychedelics

Graphical Abstract

graphic file with name nihms-1059581-f0004.jpg

HISTORY

Psychedelics are compounds that induce perceptual alterations as well as effects on mood and cognition.1 In scientific literature, psychedelics have been classified according to a variety of different names. Psychotomimetics, one of the earliest terms, was coined to indicate that these compounds are capable of inducing a mental state similar to psychosis. Later, these compounds were classified as hallucinogens, a term still in widespread use2 also encompassing a wide range of mechanistically distinct drugs, such as the κ-opioid receptor agonist salvinorin A3 and the N-methyl-d-aspartate (NMDA) receptor antagonist phencyclidine (PCP).4 The term psychedelic, which was coined by Dr. Humphrey Osmond in 1956 and means “mind manifesting”. This term is usually reserved for compounds that are also referred to as serotonergic or classical hallucinogens.

The classical psychedelics exert their main pharmacological effects through activation of the serotonin 2A receptor (5-HT2AR); they can be subdivided into two main families based on their structural properties: indoleamines and phenylalkylamines. The indoleamine scaffold is present in both the lysergamide and tryptamine psychedelics, as exemplified by lysergic acid diethylamide (LSD)5 and psilocybin,6 respectively (Figure 1). The phenylalkylamine family can be subdivided into phenylisopropylamines (“psychedelic amphetamines”, including 2,5-dimethoxy-4-iodoamphetamine (DOI) and other members of the DOX class), the phenethylamines (most notably mescaline and 2C-X compounds such as 2C-B), and last the N-benzylphenethylamines (NBOMes) (which are the main focus of this Review) (Figure 1). The respective nomenclatures for members of these three groups are rooted in different principles. The “2C” code name refers to the two ethylene carbons that separate the phenyl ring from the primary amine, with the last letter in the acronym identifying the substituent in the 4-position, for example, bromine in the case of 2C-B (Figure 1). In the case of the NBOMes, the 25B in the name 25B-NBOMe refers to the position and identity of the substituents on the phenethylamine moiety (i.e., methoxy groups in the 2- and 5-positions and a bromine substituent in the 4-position), the NB stands for N-benzyl, and the OMe represents the methoxy substituent on the ortho-position of the benzyl moiety (Figure 1).

Figure 1.

Figure 1.

Overview and classification of the chemical scaffolds considered as classical hallucinogens. The core scaffolds are highlighted in red.

The history of the phenethylamine family of hallucinogens has spanned a period of at least 5500 years (Table 1), with the first evidence for the ceremonial use of the sacred cactus Lophophora williamsii, better known as peyote, dating back to 3780–3660 BCE.7 L. williamsii is a small bulbous and spineless cactus, containing mescaline as its primary alkaloidal constituent. In 1896, mescaline was isolated from peyote by Arthur Heffter,8 who was also the first to recognize the psychedelic properties of mescaline based on self-experiments, thus making mescaline the first chemically pure psychedelic substance.9 In the following years, a large amount of scientific research was conducted with mescaline, including the first reported synthesis of the compound by Ernst Späth, proving irrefutably that the compound is a β-phenethylamine,10 and the publication of Der Meskalinrausch (“The Mescaline Rush”) by Kurt Beringer, which provided a detailed description of its psychedelic effects.11 However, it was mainly the publication of The Doors of Perception by Aldous Huxley in 1954 that introduced the notion of mescaline as a psychedelic to the general public.12

Table 1.

Overview of noteworthy events in the history of the psychedelic phenethylamines

year noteworthy event
~3700 BCE traditional use of peyote, by Native Americans
1896–1897 Arthur Heffter, first isolation and self-experiments with mescaline
1919 Ernst Späth, First chemical synthesis of mescaline
1954 Aldous Huxley, publication of The Doors of Perception
1970 mescaline and peyote are placed in Schedule I of the Controlled Substance Act
1974–1975 Alexander T. Shulgin, first synthesis and self-experiments with 2C-B
1991 Alexander T. Shulgin and Ann Shulgin, Publication of PIHKAL: A Chemical Love Story
1994 2C-B is added to Schedule I of the Controlled Substance Act
1994 Richard A. Glennon et al., first synthesis and pharmacological evaluation of N-benzylphenethylamines
2003 Ralf Heim, first synthesis of 25B-NBOMe
2006 David E. Nichols et al., first systematic SAR study of the NBOMe class
2010 NBOMes sold online as NPS
2013 25I-NBOMe, 25B-NBOMe, and 25C-NBOMe are added to Schedule I of the Controlled Substance Act

Explorations into the putative therapeutic potential of mescaline and other psychedelics came to an abrupt halt when President Nixon signed “The Comprehensive Drug Abuse Prevention and Control Act” into law on October 27, 1970. Title II of this act, the “Controlled Substance Act” (CSA), classified many of the best-known psychedelics, including LSD, psilocybin, mescaline, and peyote, as Schedule I substances, thus severely restricting the use of these compounds in scientific research.13

By that point, several analogues of mescaline had been prepared and evaluated in man (Scheme 1), including the amphetamine analogues 3,4,5-trimethoxyamphetamine (TMA)14,15 and 2,4,5-trimethoxyamphetamine (TMA-2).16 The potency increase observed with TMA and especially TMA-2 relative to mescaline led Alexander T. Shulgin to synthesize the more potent and long-acting compound 2,5-dimethoxy-4-methylamphetamine (DOM).17 The psychedelic potency of DOM was increased even further by replacing the para-methyl group with an iodine or bromine atom, yielding 2,5-dimethoxy-4-iodoamphetamine (DOI) and 2,5-dimethoxy-4-bromoamphetamine (DOB), respectively.18 In 1974, Shulgin translated this strategy back to the phenethylamine family with the synthesis of 2,5-dimethoxy-4-bromophenethylamine (2C-B),19 which he found to be a strong hallucinogen in a series of self-experiments conducted during 1974 and 1975 (the drug was described as “beautifully effective”).20 During the late 1970s and early 1980s, 2,5-dimethoxy-4-methylphenethylamine (2C-D), another compound from this class, received considerable attention from psychiatrists as a psychotherapeutic adjunct, most notably Hanscarl Leuner, who worked with 2C-D extensively under the code name LE-25 and pioneered the concept of psychedelic therapy.21 However, 2C-B was emergency scheduled by the Drug Enforcement Administration (DEA) in 1994, due to its appearance on the recreational drug market as a replacement for 3,4-methyl enedioxy methamphetamine (MDMA) (which had been scheduled in 1985). At that time, 2C-B was still being legally manufactured by the German company Drittewelle under the trade name of Erox and sold in Dutch “head-shops” under the name Nexus. In March 2001, the UN Commission on Narcotic Drugs added 2C-B to Schedule II of the Convention on Psychotropic Substances.

Scheme 1.

Scheme 1.

Evolution of the Phenethylamine Scaffold, from Mescaline to 25B-NBOMe

The same year that 2C-B was sch in the United States, Richard A. Glennon and co-workers reported that substitution of the primary amine of 2C-B with a benzyl moiety yielded an significant increase in 5-HT2AR binding affinity.22 Further structural optimization as part of the doctoral thesis of Ralf Heim23 and by David E. Nichols and co-workers24 revealed that the addition of an ortho-methoxy or -hydroxy group to the N-benzyl moiety increased 5-HT2AR affinity even further. In 2010 the first reports of online vendors distributing compounds from this class as designer drugs or New Psychoactive Substances (NPS) emerged, with 25I-NBOMe and 25B-NBOMe being the first compounds from this class to appear.25 Before long, accounts of toxicity and lethal overdoses from these drugs appeared, which resulted in the scheduling of these compounds by the DEA in November 2013.

SCIENTIFIC RELEVANCE OF NBOMes

In recent years, there has been a resurgence of scientific research into serotonergic psychedelics. Although a large amount of clinical research was conducted with LSD and other hallucinogens in the 1950s and 1960s, virtually all clinical research with these substances ceased during the early 1970s due to legal restrictions codified in the Controlled Substance Act; only recently has research with these drugs resumed. Most of this new work has focused on psilocybin, which has shown promising effects in pilot studies focusing on several therapeutic indications, including depression and anxiety,2630 substance abuse,3133 and obsessive-compulsive disorder.34 Notably, the U.S. Food and Drug Administration (FDA) has designated psilocybin as a Breakthrough Therapy for treatment-resistant depression. There has also been renewed interest in the use of LSD as a potential therapeutic agent.35,36 The reader is referred to other review articles for a more in-depth discussion of the remarkable findings on the therapeutic potential of psychedelic compounds in general.27,3745 Psilocybin has high affinity for a number of serotonergic receptors besides 5-HT2AR,46 and LSD possesses high affinity for an even wider range of monoaminergic receptors (see Table 2).47 However, the activation of 5-HT2ARs is considered essential for the psychedelic effects, as well as the apparent therapeutic potential of both of these drugs. Thus, ligands capable of activating the 5-HT2AR selectively would be important pharmacological tools. Given the inherent selectivity exhibited by many of the NBOMes for 5-HT2R over other serotonergic and monoaminergic receptors, they are obvious leads in the search for novel agents that could be applicable in investigations of the in vivo effects arising from selective activation of the 5-HT2AR.

Table 2.

Receptor Affinity Profiles of Classic Serotonergic Psychedelics at Selected Monoaminergic Receptors and Transportersa

LSDb psilocine DMTf mescalineb 2C-Bb 25B-NBOMeg 25CN-NBOHh
5-HT1A 3.0  567  183   4600 240   1,255   >10 000
5-HT1B 4c  220  129   n.d.i n.d.   >10 000   >10 000
5-HT1D 14d  36  39   n.d. n.d.   1472   >10 000
5-HT1E 93c  52  517   n.d. n.d.   >10 000   >10 000
5-HT2A 4.2  107  127   6300 8.6   0.8   2.2
5-HT2B 120  4.6  184   >20 000 130   0.5   58
5-HT2C 15  97  360   1700 47   1.7   50
5-HT5 9c  84  2135   n.d. n.d.   4087   >10 000
5-HT6 7c  57  464   n.d. n.d.   n.d.   573
5-HT7 7c  3.5  206   n.d. n.d.   4720   >10 000
H1 1100  305  n.d.   >25 000 14 000   n.d.   >10 000
α 2A 670  1379  n.d.   >15 000 8200   3551   803
α 2B 12  1894  n.d.   1400 320   1264   1226
D1 310  >10 000  n.d.   >14 000 12 000   >10 000   >10 000
D2 25  >10 000  n.d.   >10 000 2200   >10 000   >10 000
D3 96  2645  n.d.   >17 000 10 000   718   >10 000
D4 56c  >10 000  n.d.   n.d. n.d.   2253   >10 000
D5 340c  >10 000  n.d.   n.d. n.d.   >10 000   >10 000
SERT >30 000  3801  n.d.   >30 000 9,700   388   >10 000
DAT >30 000  >10 000  n.d.   >30 000 >30 000   >10 000   >10 000
NET >30 000  >10 000  n.d.   >30 000 >30 000   1,718   >10 000
a

All Ki data in nM for cloned human receptors, unless otherwise noted.

b

Reference 62.

c

Reference 57 (cloned rat receptors).

d

Reference 63 (human cortex).

e

Reference 56.

f

Reference 64.

g

Reference 65.

h

Reference 59.

i

n.d. = no data.

CHEMICAL SYNTHESIS

2,5-Dimethoxybenzaldehyde (1) is a common commercial starting point for the synthesis of compounds from the 2C-X and NBOMe classes (Scheme 2).19,51 The β-nitrostyrene (2) is prepared by the condensation of nitromethane with benzaldehyde 1. The transformation to the β-phenethylamine 3 is typically accomplished by reduction, using hydrogen and palladium on activated charcoal or another suitable reducing agent. β-Phenethylamine 3 is typically brominated at this stage. Alternatively, the 4-bromo group can be introduced at the benzaldehyde stage to avoid performing a late stage bromination (not shown).50 All NBOMe-class compounds are usually prepared from the corresponding 2C-X compound by reductive amination of the appropriate benzaldehyde.49,50

Scheme 2. Synthesis of NBOMes, Exemplified by the Synthesis of 25B-NBOMe via 2C-B4850a.

Scheme 2.

aReagents and conditions: (i) CH3NO2, NH4OH, 70 °C, 2 h, quant; (ii) H2, Pd/C, MeOH, r.t., 1 h, 99%; (iii) Br2, AcOH, r.t., 16 h, 99%; (iv) 2-methoxybenzaldehyde, Et3N, EtOH, reflux, 3 h, followed by NaBH4, r.t., 30 min, 85%.

STRUCTURE–ACTIVITY RELATIONSHIPS OF THE NBOMes

The Phenethylamine Moiety.

The 2,4,5-trisubstituted phenyl ring has been intensively investigated in both 2C-X and NBOMe analogues, exemplified by 2C-B and 25B-NBOMe.52 In comparison, phenethylamines containing a 3,4,5-trisubstition pattern, such as in mescaline, have substantially reduced potency at the 5-HT2AR. The 2,4,6-trisubstition pattern in combination with an α-methyl group has been reported to give compounds with moderate 5-HT2AR activity.17 Of these positions, the 4-position in both 2,4,5- and 3,4,5-trisubstituded compounds is the most tolerant to a range of substituents, with a preference for small or nonbulky lipophilic moieties up to 5–6 carbons in length.53 A plethora of ligands with methoxy-groups in the 2,5-positions have been investigated, in large part due to the ease of their synthesis. Modification of the 4-position substituent can markedly alter the agonist efficacy of the 2C-X and NBOMe analogues, with bulky or extended linear substituents converting partial agonists into competitive antagonists at 5-HT2AR (Figure 2).54

Figure 2.

Figure 2.

Overview of the structure–activity relationship of NBOMes at the 5-HT2AR.

The Primary Amine.

While, for many years, it was believed that any type of substitution on the primary amine of the phenylethylamine scaffold would result in a significant loss of 5-HT2AR activity, addition of an N-benzyl moiety to the scaffold was surprisingly found to increase affinity and agonist potency at the receptor.22 Interestingly, the effect of adding an N-benzyl group was more pronounced for phenethylamines displaying modest affinities for 5-HT2AR, whereas the binding properties of analogues that already possessed high affinity at the receptor were not as substantially altered. Phenethylamines containing an N-benzyl moiety also exhibit some structure–activity relationship (SAR) differences compared to the 2C-X compounds. In contrast to the 2C-X analogues, the absence of either a lipophilic substituent in the 4-position or the 5-methoxy group in NBOMes is not detrimental for their 5-HT2AR affinity.50 Conversely, the presence of the α-methyl group in NBOMes reduces both affinity and efficacy.24 This suggests that the NBOMe scaffold could target the orthosteric site in 5-HT2AR in a somewhat different binding mode compared to members of the DOX and 2C-X phenylethylamine classes.

The Benzyl Moiety.

The substitution pattern on the benzyl moiety has also been investigated; H-bond acceptors have the most significant influence on 5-HT2AR affinity, with an observed substitution trend of ortho > meta > para. Ortho-fluoro substitutions are also tolerated, as well as introduction of small fused heterocycles into the scaffold.49,55

PHARMACOLOGY

It has been recognized that most of the psychological and therapeutic effects produced by serotoninergic psychedelics are mediated by activation of 5-HT2AR.42,43 Compounds from the tryptamine class are usually considered to be selective for serotonin receptors over other monoaminergic receptors, but they are fairly promiscuous agonists within the serotonin receptor family (Table 2).56 Ergolines are generally non-selective and target a broad range of monoaminergic receptors with high binding affinities (Table 2).57 In general, the most selective agonists for 5-HT2AR and 5-HT2CR come from the phenylalkylamine class, with compounds from the amphetamine and phenethylamine class usually displaying substantially higher binding affinities for these receptors compared to other receptors for serotonin and other monoamines.

The psychedelic phenethylamines typically exhibit less than 5–10-fold selectivity for 5-HT2A over 5-HT2C receptors.53,54 Notable exceptions include (S,S)-DMBMPP and 25CN-NBOH (Figure 3), which are the most selective 5-HT2AR agonists published to date.49,5861 (S,S)-DMBMPP exhibits more than 100-fold higher binding affinity for the 5-HT2AR over 5-HT2CR, and 25CN-NBOH has substantially higher agonist potencies at 5-HT2AR compared to 5-HT2CR.

Figure 3.

Figure 3.

Chemical structures of DMBMPP and 25CN-NBOH.

The availability of 25CN-NBOH, DMPMPP, and other 5-HT2AR-selective agonists with good pharmacokinetic properties will be instrumental for the future delineation of the role played by the receptor in the therapeutic effects of classical psychedelics. The hallucinogenic properties are also believed to be inherently linked to augmentation of 5-HT2AR signaling, which constitutes a major potential obstacle to the realization of the therapeutic potential of 5-HT2AR agonists. Thus, an important new frontier in this field will be needed to establish whether or not the psychedelic properties and the therapeutic effects arising from 5-HT2AR activation can be separated in specific agonists. In this respect, it is interesting to note that in contrast to most other 5-HT2AR agonists, lisuride, a close structural analogue of LSD and a potent 5-HT2AR agonist, does not induce a head twitch response in rodents or produce psychedelic effects in man.6668 Over the past decade, research has focused on the fact that GPCRs can potentially signal through multiple intracellular pathways and that some compounds (biased agonists) can selectively activate a specific subset of these pathways.69 If it is possible to develop biased 5-HT2AR agonists with pathway-specific effects, then it will be interesting to probe the effects of these compounds in vivo.

EFFECT OF NBOMes IN IN VIVO BEHAVIORAL MODELS

Administration of 5-HT2AR agonists to rodents induces a rapid paroxysmal head rotation known as the head twitch response (HTR).70,71 In rats, the HTR often involves both the head and trunk, hence in that species the behavior is also called a wet-dog shake (WDS).72 The HTR is widely used as a behavioral proxy in rodents for hallucinogen effects in humans because it is one of the few behavioral effects that can reliably distinguish hallucinogenic and nonhallucinogenic 5-HT2AR agonists, such as LSD and lisuride, respectively.70,73

Various compounds from the NBOMe class have been tested in the HTR paradigm. After subcutaneous administration, 25I-NBOMe induces the HTR in C57BL/6J mice with an ED50 of 0.17 μmol/kg.74 In comparison to 25I-NBOMe, 2C-I has 18-fold lower potency (ED50 = 2.42 μmol/kg SC), which is consistent with their relative binding affinities and agonist potencies at 5-HT2AR.74 Similar findings with 25I-NBOMe and 2C-I have been reported in Sprague–Dawley (SD) rats.74 Likewise, 25C-NBOMe induces WDS in SD rats with 26-fold higher potency than 2C-C.75 25B-NBOMe induces the HTR in NMRI mice when administered at 0.5 mg/kg IP but not at 0.05 mg/kg, meaning it has about the same potency as 25I-NBOMe.76 25CN-NBOH (ED50 = 1.03 μmol/kg SC)59 also induces the HTR in C57BL/6J mice but has relatively lower potency compared to other NBOMe compounds. Importantly, experiments have confirmed that 5-HT2A antagonists block the HTR induced by 25I-NBOMe and other N-benzyl-substituted phenethylamines.58,61,74,75

The HTR potency of NBOMes derived from 2C-I is dependent on the identity and position of the substituent on the N-benzyl ring. According to HTR studies performed in C57BL/6J mice, rearranging the ortho-methoxy group of 25I-NBOMe to the meta-position reduced its potency 55-fold (25I-NB3OMe: ED50 = 9.36 μmol/kg SC), whereas the paramethoxy analogue 25I-NB4OMe showed no evidence of activity at doses up to 30 mg/kg.77 The presence of a halogen substituent is also detrimental for activity; replacing the ortho-methoxy moiety with bromine produced a 26-fold potency reduction (25I-NBBr: ED50 = 4.50 μmol/kg SC). Rearrangement of the bromine atom to either the meta- or para-position was not tolerated, and no HTR was observed with 25I-NB3Br or 25I-NB4Br at doses up to 30 mg/kg. The N-(2,3-methylenedioxybenzyl) analogue 25I-NBMD (ED50 = 2.36 μmol/kg SC)74 is significantly less potent than 25I-NBOMe and shows the same potency as the nonbenzylated parent compound 2C-I. Therefore, for N-benzylphenethylamines, the highest potency in the HTR assay is associated with an ortho-substituent on the benzyl ring, especially if the substituent contains an oxygen atom.

Drug discrimination is another behavioral paradigm used to study hallucinogen effects. It was first shown in 1971 that rats can be trained to discriminate the interoceptive stimulus effects of mescaline and LSD from saline.78 Many other serotonergic hallucinogens have been used as training drugs, including DOM, DOB, DOI, psilocybin, 5-MeO–DMT, and DPT.7984 The interoceptive stimulus effects produced by serotonergic hallucinogens appear to be rather uniform in nature because they reliably produce cross-generalization in rats. Recently, 25B-NBOMe, 25C-NBOMe, and 25I-NBOMe were evaluated in SD rats trained to discriminate 0.5 mg/kg DOM (administered by the IP route).85 25B-NBOMe and 25C-NBOMe produced full substitution, indicating they have DOM-like stimulus effects. Evaluation of the response to 25I-NBOMe was confounded by the fact that higher doses disrupted operant responding, but it did produce a high degree of partial substitution (a maximum of 74% drug-appropriate responding occurred at 2.5 mg/kg IP).

Most drug discrimination studies with serotonergic hallucinogens have been conducted in rats, although mice have also been used in some studies. For example, mice have been trained to discriminate LSD,8688 DOI,89 R-(−)-DOI,58 and R-(−)-DOB.90 Interestingly, there are apparently significant differences between the interoceptive stimulus effects of hallucinogens in rats vs mice.8789 For example, although the discriminative stimulus induced by DOI in rats appears to be exclusively mediated by 5-HT2A,91,92 the 5-HT2C receptor may contribute to the stimulus effects of the drug in mice.89 In light of these species-specific differences, it is interesting to note that 25CN-NBOH did not fully substitute when tested in NIH Swiss mice trained to discriminate 0.3 mg/kg IP R-(−)-DOI, producing a maximum of 55% drug-appropriate responding.58 Although it is possible that 25CN-NBOH does not produce hallucinogen-like interoceptive stimulus effects, the absence of full substitution in mice could also potentially reflect the relatively high selectivity of 25CN-NBOH for 5-HT2AR. The reported selectivity of 25CN-NBOH for 5-HT2A over 5-HT2C varies depending on the experimental conditions,49,59,60 but 25CN-NBOH is clearly more selective than DOI, which is only ~5-fold and 12-fold selective for 5-HT2A over 5-HT2C at the human and murine receptors, respectively.9395 If, as noted above, the 5-HT2C receptor contributes to the DOI stimulus in mice, then a 5-HT2A-selective agonist such as 25CN-NBOH may not produce full substitution in that species. Importantly, 25CN-NBOH is anticipated to fully substitute for DOI in rats, but the prediction needs to be tested experimentally.

NBOMes AS NEUROIMAGING TOOLS

Positron emission tomography (PET) is a highly sensitive imaging tool that finds widespread use in basic research and drug discovery, where it can be used to determine receptor availability and to measure occupancy by drug molecules.96 The requirements for PET ligands and therapeutic drugs are very different:97 High affinity and selectivity, low nonspecific binding, and reversible kinetics are key requirements for imaging purposes. The NBOMes seem to possess many of these characteristics, and thus they have been investigated as PET ligands for 5-HT2AR. After screening several members of the NBOMe class, [11C]25B-NBOMe was found to have very good properties as a 5-HT2AR PET ligand, despite having limited (~15-fold) selectivity for 5-HT2AR over 5-HT2CR. [11C]25B-NBOMe has been evaluated as a PET ligand in pigs,65,76,98 primates (Macaca mulatta),99,100 and humans;101,102 several PET centers around the world are currently using this ligand, under the code name [11C]-CIMBI-36, to investigate the clinical relevance of 5-HT2AR. Several attempts have been made to develop an 18F-based PET tracer from the NBOMe-class, since having a PET ligand based on this radionuclide would broaden its clinical applicability, but so far these attempts have been unsuccessful.103105 These results highlight the fact that even very small changes to the structure of a molecule can have dramatic effects on its behavior in vivo.

ADME

Only limited data are available on the human absorption, distribution, metabolism, and excretion (ADME) properties of NBOMes, due to the ethical and legal concerns associated with their use. Therefore, there is a great need for controlled experiments on the properties of these compounds, for both scientific as well as toxicological reasons. For instance, metabolites may have toxic pharmacological properties and/or produce dangerous drug–drug interactions with either pharmaceuticals or other illicit substances.

The microsomal stability of several NBOMes has been determined in human liver microsomes (HLMs).50 The average intrinsic clearance (Clint) was 6 (1.9–14) L/kg/h, which is much higher than the corresponding Clint values for 2C-X compounds (average clearance = 0.51 L/kg/h) and mescaline (0.19 L/kg/h).106 Thus, NBOMes are generally metabolized at a much faster rate than their parent phenethylamines, indicating that their lack of per oral bioavailability is a consequence of extensive first pass metabolism. In a follow-up study, five phase I metabolites of 25B-NBOMe were identified. All three O-demethylated metabolites (4–6) were found in approximately the same abundancies, as were the hydroxylated metabolites (7–8), albeit with relatively lower abundancy. 2C-B, the N-debenzylated metabolite, was also detected, but it also had lower abundancy.107

Studies of other NBOMes have shown that there is a plentitude of overlap between the metabolites of different members of this group.108 CYP2D6 is the main contributor to the hepatic clearance of most NBOMes, as exemplified by 25B-NBOMe, where the 2D6 isoenzyme is responsible for 69% of the clearance.109 Other cytochrome P450s involved in the phase I metabolism of these compounds include CYP1A2, CYP2B6, CYP2C19, and CYP3A4.109,110 Detailed studies of the phase I and phase II metabolites have found a large array of minor metabolites, ranging up to 69 unique metabolites (Scheme 3).108119

Scheme 3.

Scheme 3.

Putative Phase I Metabolic Pathways for 25B-NBOMe in Humans

SAFETY

Many instances of acute toxicity due to NBOMe exposure have been reported; currently, there are over 50 confirmed cases.120 Of these cases, at least 9 were fatalities,121125 of which at least 2 were direct 25B-NBOMe poisonings.126,127 Including those 2 fatalities, a total of 18 cases of acute 25B-NBOMe toxicity have been reported. In addition to causing hallucinations, other common side effects produced by NBOMes include severe agitation, confusion, diaphoresis, hypertension, tachycardia, and hyperthermia. Less common, but more severe are seizures, rhabdomyolysis, metabolic acidosis, renal failure, multiorgan failure, and coma.128132 The in vitro neurotoxicity of both 25B-NBOMe and 25C-NBOMe has also been investigated, and these compounds were found to inhibit neuronal activity and cell viability, respectively.133,134 Additionally, 25D-NBOMe and 25C-NBOMe were shown to have potential cardiotoxic effects, as both compounds prolonged QT intervals in rat ECG studies. Furthermore, 25D-NBOMe also inhibited K+-channels in a hERG assay.135

CONCLUSIONS

In conclusion, numerous cases have occurred where recreational use of NBOMes caused severe toxic effects, necessitating emergency medical intervention (which unfortunately did not always prevent a fatal outcome); the fact that in some of these cases NBOMes had been taken at moderate doses should serve as warning about the recreational use of these substances. The origin of the NBOMe-induced toxicity is not well understood, and certain individuals may show idiosyncratic responses. Nonetheless, this class of psychedelic compounds, in particular 25B-NBOMe and 25CN-NBOH, have provided the scientific community with new tools to selectively investigate 5-HT2A receptors in preclinical and clinical research. Given the recent surge in clinical research with classical psychedelics, NBOMes have emerged as an important class of compounds that will allow researchers to focus on the specific effects produced by 5-HT2AR activation. Furthermore, certain members of the NBOMe class may have unique clinical or therapeutic effects compared to existing psychedelic compounds due to their relatively high selectivity for the 5-HT2AR.

Funding

A.L.H. is supported by NIDA (DA041336) and the Veteran’s Administration VISN 22 Mental Illness Research, Education, and Clinical Centre. J.L.K. acknowledges generous support from the Lundbeck Foundation.

ABBREVIATIONS

5HT2AR

serotonin 2A receptor

5HT2CR

serotonin 2C receptor

NPS

novel psychoactive substance

SAR

structure–activity relationship

Footnotes

The authors declare no competing financial interest.

REFERENCES

  • (1).Halberstadt AL (2015) Recent advances in the neuropsychopharmacology of serotonergic hallucinogens. Behav. Brain Res 277, 99–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (2).Nichols DE (2016) Psychedelics. Pharmacol. Rev 68, 264–355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (3).Roth BL, et al. (2002) Salvinorin A: A potent naturally occurring nonnitrogenous opioid selective agonist. Proc. Natl. Acad. Sci. U. S. A 99, 11934–11939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (4).Anis NA, Berry SC, Burton NR, and Lodge D (1983) The dissociative anaesthetics, ketamine and phencyclidine, selectively reduce excitation of central mammalian neurones by N-methylaspartate. Br. J. Pharmacol 79, 565–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (5).Nichols DE (2018) Dark Classics in Chemical Neuroscience: Lysergic Acid Diethylamide (LSD). ACS Chem. Neurosci 9, 2331–2343. [DOI] [PubMed] [Google Scholar]
  • (6).Geiger HA, Wurst MG, and Daniels RN (2018) DARK Classics in Chemical Neuroscience: Psilocybin. ACS Chem. Neurosci 9, 2438–2447. [DOI] [PubMed] [Google Scholar]
  • (7).El-Seedi HR, Smet P. A. G. M. De, Beck O, Possnert G, and Bruhn JG (2005) Prehistoric peyote use: Alkaloid analysis and radiocarbon dating of archaeological specimens of Lophophora from Texas. J. Ethnopharmacol 101, 238–242. [DOI] [PubMed] [Google Scholar]
  • (8).Heffter A (1896) Ueber Cacteenalkaloïde. Ber. Dtsch. Chem. Ges 29, 216–227. [Google Scholar]
  • (9).Heffter A (1898) Ueber Pellote. Naunyn-Schmiedeberg’s Arch. Pharmacol 40, 385–429. [Google Scholar]
  • (10).Späth E (1919) Über die Anhalonium-Alkaloide I. Anhalin und Mezcalin. Monatsh. Chem 40, 129–154. [Google Scholar]
  • (11).Beringer K (1927) Der Meskalinrausch, Springer-Verlag, Berlin, Heidelberg, Germany. [Google Scholar]
  • (12).Huxley A (1954) The Doors of Perception, Harper & Row, New York. [Google Scholar]
  • (13).Sessa B (2018) The 21st century psychedelic renaissance: heroic steps forward on the back of an elephant. Psychopharmacology (Berl) 235, 551–560. [DOI] [PubMed] [Google Scholar]
  • (14).Shulgin AT, Bunnell S, and Sargent T (1961) The Psychotomimetic Properties of 3,4,5-Trimethoxyamphetamine. Nature 189, 1011–1012. [Google Scholar]
  • (15).Peretz DI, Smythies JR, and Gibson WC (1955) A New Hallucinogen: 3,4,5-Trimethoxyphenyl-β-Aminopropane. J. Ment. Sci 101, 317–329. [PubMed] [Google Scholar]
  • (16).Shulgin AT (1964) Psychotomimetic amphetamines: Methoxy 3,4-dialkoxyamphetamines. Experientia 20, 366–367. [DOI] [PubMed] [Google Scholar]
  • (17).Shulgin AT, Sargent T, and Naranjo C (1969) Structure-Activity Relationships of One-Ring Psychotomimetics. Nature 221, 537–541. [DOI] [PubMed] [Google Scholar]
  • (18).Shulgin AT (1981) DOB. J. Psychoact. Drugs 13, 99–99. [DOI] [PubMed] [Google Scholar]
  • (19).Shulgin AT, and Carter MF (1975) Centrally Active Phenethylamines. Psychopharmacol. Commun 1, 93–98. [PubMed] [Google Scholar]
  • (20).Shulgin AT A.T. Shulgin’s Pharmacology Notebook 1. [Google Scholar]
  • (21).Leuner H (1981) Halluzinogene: Psychische Grenzzustände in Forschung und Psychotherapie, Hogrefe AG, Bern, Germany. [Google Scholar]
  • (22).Glennon RA, et al. (1994) Influence of Amine Substituents on 5-HT2A versus 5-HT2C Binding of Phenylalkyl- and Indolylalkylamines. J. Med. Chem 37, 1929–1935. [DOI] [PubMed] [Google Scholar]
  • (23).Heim R (2003) Synthese und Pharmakologie potenter 5-HT2A-Rezeptor Agonisten mit N-2-Methoxybenzyl-Partialstruktur. Entwicklung eines neuen Struktur-Wirkungskonzepts, PhD thesis, Freie Universität Berlin, Berlin, Germany. [Google Scholar]
  • (24).Braden MR, Parrish JC, Naylor JC, and Nichols DE (2006) Molecular Interaction of Serotonin 5-HT2A Receptor Residues Phe339(6.51) and Phe340(6.52) with Superpotent N-Benzyl Phenethylamine Agonists. Mol. Pharmacol 70, 1956–1964. [DOI] [PubMed] [Google Scholar]
  • (25).Poklis JL, Raso SA, Alford KN, Poklis A, and Peace MR (2015) Analysis of 25I-NBOMe, 25B-NBOMe, 25C-NBOMe and Other Dimethoxyphenyl- N -[(2-Methoxyphenyl) Methyl]-Ethanamine Derivatives on Blotter Paper. J. Anal. Toxicol 39, 617–623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (26).Grob CS, et al. (2011) Pilot Study of Psilocybin Treatment for Anxiety in Patients With Advanced-Stage Cancer. Arch. Gen. Psychiatry 68, 71–78. [DOI] [PubMed] [Google Scholar]
  • (27).Carhart-Harris RL, et al. (2016) Psilocybin with psychological support for treatment-resistant depression: an open-label feasibility study. Lancet Psychiatry 3, 619–627. [DOI] [PubMed] [Google Scholar]
  • (28).Griffiths RR, et al. (2016) Psilocybin produces substantial and sustained decreases in depression and anxiety in patients with life-threatening cancer: A randomized double-blind trial. J. Psychopharmacol 30, 1181–1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (29).Carhart-Harris RL, et al. (2017) Psilocybin for treatment-resistant depression: fMRI-measured brain mechanisms. Sci. Rep 7, 13187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (30).Reiche S, et al. (2018) Serotonergic hallucinogens in the treatment of anxiety and depression in patients suffering from a life-threatening disease: A systematic review. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 81, 1–10. [DOI] [PubMed] [Google Scholar]
  • (31).Garcia-Romeu A, Griffiths R, and Johnson M (2015) Psilocybin-Occasioned Mystical Experiences in the Treatment of Tobacco Addiction. Curr. Drug Abuse Rev 7, 157–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (32).Johnson MW, Garcia-Romeu A, Johnson PS, and Griffiths RR (2017) An online survey of tobacco smoking cessation associated with naturalistic psychedelic use. J. Psychopharmacol 31, 841–850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (33).Bogenschutz MP, et al. (2015) Psilocybin-assisted treatment for alcohol dependence: A proof-of-concept study. J. Psychopharmacol 29, 289–299. [DOI] [PubMed] [Google Scholar]
  • (34).Moreno FA, Wiegand CB, Taitano EK, and Delgado PL (2006) Safety, tolerability, and efficacy of psilocybin in 9 patients with obsessive-compulsive disorder. J. Clin. Psychiatry 67, 1735–1740. [DOI] [PubMed] [Google Scholar]
  • (35).Gasser P, et al. (2014) Safety and Efficacy of Lysergic Acid Diethylamide-Assisted Psychotherapy for Anxiety Associated With Life-threatening Diseases. J. Nerv. Ment. Dis 202, 513–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (36).Gasser P, Kirchner K, and Passie T (2015) LSD-assisted psychotherapy for anxiety associated with a life-threatening disease: A qualitative study of acute and sustained subjective effects. J. Psychopharmacol 29, 57–68. [DOI] [PubMed] [Google Scholar]
  • (37).Vollenweider FX, and Kometer M (2010) The neurobiology of psychedelic drugs: Implications for the treatment of mood disorders. Nat. Rev. Neurosci 11, 642–651. [DOI] [PubMed] [Google Scholar]
  • (38).Bogenschutz MP, and Pommy JM (2012) Therapeutic mechanisms of classic hallucinogens in the treatment of addictions: from indirect evidence to testable hypotheses. Drug Test. Anal 4, 543–555. [DOI] [PubMed] [Google Scholar]
  • (39).Garcia-Romeu A, Kersgaard B, and Addy PH (2016) Clinical applications of hallucinogens: A review. Exp. Clin. Psychopharmacol 24, 229–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (40).Rucker JJH, Jelen LA, Flynn S, Frowde KD, and Young AH (2016) Psychedelics in the treatment of unipolar mood disorders: A systematic review. J. Psychopharmacol 30, 1220–1229. [DOI] [PubMed] [Google Scholar]
  • (41).Szabo A (2015) Psychedelics and Immunomodulation: Novel Approaches and Therapeutic Opportunities. Front. Immunol 6, 358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (42).Nichols DE, Johnson MW, and Nichols CD (2017) Psychedelics as Medicines: An Emerging New Paradigm. Clin. Pharmacol. Ther 101, 209–219. [DOI] [PubMed] [Google Scholar]
  • (43).Kyzar EJ, Nichols CD, Gainetdinov RR, Nichols DE, and Kalueff AV. (2017) Psychedelic Drugs in Biomedicine. Trends Pharmacol. Sci 38, 992–1005. [DOI] [PubMed] [Google Scholar]
  • (44).Carhart-Harris RL, and Goodwin GM (2017) The Therapeutic Potential of Psychedelic Drugs: Past, Present, and Future. Neuropsychopharmacology 42, 2105–2113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (45).Belouin SJ, and Henningfield JE (2018) Psychedelics: Where we are now, why we got here, what we must do. Neuropharmacology 142, 7–19. [DOI] [PubMed] [Google Scholar]
  • (46).Lee H-M, and Roth BL (2012) Hallucinogen actions on human brain revealed. Proc. Natl. Acad. Sci. U. S. A 109, 1820–1821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (47).Passie T, Halpern JH, Stichtenoth DO, Emrich HM, and Hintzen A (2008) The Pharmacology of Lysergic Acid Diethylamide: A Review. CNS Neurosci. Ther 14, 295–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (48).Karlsen M, Liu H, Berg T, Johansen JE, and Hoff BH (2014) Synthesis of [13C6]-labelled phenethylamine derivatives for drug quantification in biological samples. J. Labelled Compd. Radiopharm 57, 378–387. [DOI] [PubMed] [Google Scholar]
  • (49).Hansen M, et al. (2014) Synthesis and Structure-Activity Relationships of N -Benzyl Phenethylamines as 5-HT 2A/2C Agonists. ACS Chem. Neurosci 5, 243–249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (50).Leth-Petersen S, et al. (2016) 5-HT2A /5-HT2C Receptor Pharmacology and Intrinsic Clearance of N-Benzylphenethylamines Modified at the Primary Site of Metabolism. ACS Chem. Neurosci 7, 1614–1619. [DOI] [PubMed] [Google Scholar]
  • (51).Shulgin AT, and Shulgin A (1991) PIHKAL, Phenethylamines I Have Known and Loved. A Chemical Love Story, Transform Press, Berkeley, CA. [Google Scholar]
  • (52).Nichols DE (2012) Structure-activity relationships of serotonin 5-HT 2A agonists. Wiley Interdiscip. Rev. Membr. Transp. Signal 1, 559–579. [Google Scholar]
  • (53).Blaazer AR, Smid P, and Kruse CG (2008) Structure-Activity Relationships of Phenylalkylamines as Agonist Ligands for 5-HT 2A Receptors. ChemMedChem 3, 1299–1309. [DOI] [PubMed] [Google Scholar]
  • (54).Nichols DE (1981) Structure-Activity Relationships of Phenethylamine Hallucinogens. J. Pharm. Sci 70, 839–849. [DOI] [PubMed] [Google Scholar]
  • (55).Hansen M, et al. (2015) Synthesis and pharmacological evaluation of N-benzyl substituted 4-bromo-2,5-dimethoxyphenethylamines as 5-HT2A/2C partial agonists. Bioorg. Med. Chem 23, 3933–3937. [DOI] [PubMed] [Google Scholar]
  • (56).Halberstadt AL, and Geyer MA (2011) Multiple receptors contribute to the behavioral effects of indoleamine hallucinogens. Neuropharmacology 61, 364–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (57).Nichols DE, Frescas S, Marona-Lewicka D, and Kurrasch-Orbaugh DM (2002) Lysergamides of isomeric 2,4-dimethylazetidines map the binding orientation of the diethylamide moiety in the potent hallucinogenic agent N,N-diethyllysergamide (LSD). J. Med. Chem 45, 4344–4349. [DOI] [PubMed] [Google Scholar]
  • (58).Fantegrossi WE, et al. (2015) Hallucinogen-like effects of 2-([2-(4-cyano-2,5-dimethoxyphenyl) ethylamino]methyl)phenol (25CN-NBOH), a novel N-benzylphenethylamine with 100-fold selectivity for 5-HT2A receptors, in mice. Psychopharmacology (Berl). 232, 1039–1047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (59).Halberstadt AL, et al. (2016) Effect of 5-HT 2A and 5-HT 2C receptors on temporal discrimination by mice. Neuropharmacology 107, 364–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (60).Jensen AA, et al. (2017) Detailed Characterization of the In Vitro Pharmacological and Pharmacokinetic Properties of N -(2-Hydroxybenzyl)-2,5-Dimethoxy-4-Cyanophenylethylamine (25CN-NBOH), a Highly Selective and Brain-Penetrant 5-HT 2A Receptor Agonist. J. Pharmacol. Exp. Ther 361, 441–453. [DOI] [PubMed] [Google Scholar]
  • (61).Buchborn T, Lyons T, and Knöpfel T (2018) Tolerance and Tachyphylaxis to Head Twitches Induced by the 5-HT2A Agonist 25CN-NBOH in Mice. Front. Pharmacol 9, 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (62).Rickli A, et al. (2015) Receptor interaction profiles of novel N-2-methoxybenzyl (NBOMe) derivatives of 2,5-dimethoxy-substituted phenethylamines (2C drugs). Neuropharmacology 99, 546–553. [DOI] [PubMed] [Google Scholar]
  • (63).Pierce PA, and Peroutka SJ (1989) Hallucinogenic drug interactions with neurotransmitter receptor binding sites in human cortex. Psychopharmacology (Berl) 97, 118–122. [DOI] [PubMed] [Google Scholar]
  • (64).Keiser MJ, et al. (2009) Predicting new molecular targets for known drugs. Nature 462, 175–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (65).Ettrup A, et al. (2011) Radiosynthesis and in vivo evaluation of a series of substituted 11C-phenethylamines as 5-HT2A agonist PET tracers. Eur. J. Nucl. Med. Mol. Imaging 38, 681–693. [DOI] [PubMed] [Google Scholar]
  • (66).Nichols DE (2004) Hallucinogens. Pharmacol. Ther 101, 131–181. [DOI] [PubMed] [Google Scholar]
  • (67).Carbonaro TM, et al. (2015) The role of 5-HT2A, 5-HT2C and mGlu2 receptors in the behavioral effects of tryptamine hallucinogens N,N-dimethyltryptamine and N,N-diisopropyltryptamine in rats and mice. Psychopharmacology (Berl). 232, 275–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (68).Rogawski MA, and Aghajanian GK (1979) Response of central monoaminergic neurons to lisuride: Comparison with LSD. Life Sci. 24, 1289–1297. [DOI] [PubMed] [Google Scholar]
  • (69).Tan L, Yan W, McCorvy JD, and Cheng J (2018) Biased Ligands of G Protein-Coupled Receptors (GPCRs): Structure-Functional Selectivity Relationships (SFSRs) and Therapeutic Potential. J. Med. Chem 61, 9841–9878. [DOI] [PubMed] [Google Scholar]
  • (70).Halberstadt AL, and Geyer MA (2013) Characterization of the head-twitch response induced by hallucinogens in mice. Psychopharmacology (Berl). 227, 727–739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (71).Canal CE, and Morgan D (2012) Head-twitch response in rodents induced by the hallucinogen 2,5-dimethoxy-4-iodoamphetamine: a comprehensive history, a re-evaluation of mechanisms, and its utility as a model. Drug Test. Anal 4, 556–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (72).Bedard P, and Pycock CJ (1977) ‘Wet-dog’ shake behaviour in the rat: a possible quantitative model of central 5-hydroxytryptamine activity. Neuropharmacology 16, 663–670. [DOI] [PubMed] [Google Scholar]
  • (73).González-Maeso J, et al. (2007) Hallucinogens Recruit Specific Cortical 5-HT2A Receptor-Mediated Signaling Pathways to Affect Behavior. Neuron 53, 439–452. [DOI] [PubMed] [Google Scholar]
  • (74).Halberstadt AL, and Geyer MA (2014) Effects of the hallucinogen 2,5-dimethoxy-4-iodophenethylamine (2C-I) and superpotent N-benzyl derivatives on the head twitch response. Neuropharmacology 77, 200–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (75).Elmore JS, et al. (2018) Comparative neuropharmacology of N-(2-methoxybenzyl)-2,5-dimethoxyphenethylamine (NBOMe) hallucinogens and their 2C counterparts in male rats. Neuropharmacology 142, 240–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (76).Ettrup A, et al. (2013) Preclinical Safety Assessment of the 5-HT2A Receptor Agonist PET Radioligand [11C]Cimbi-36. Mol. Imaging Biol 15, 376–383. [DOI] [PubMed] [Google Scholar]
  • (77).Nichols DE, et al. (2015) N-Benzyl-5-methoxytryptamines as Potent Serotonin 5-HT2 Receptor Family Agonists and Comparison with a Series of Phenethylamine Analogues. ACS Chem. Neurosci 6, 1165–1175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (78).Hirschhorn ID, and Winter JC (1971) Mescaline and lysergic acid diethylamide (LSD) as discriminative stimuli. Psychopharmacologia 22, 64–71. [DOI] [PubMed] [Google Scholar]
  • (79).Glennon RA, Rosecrans JA, Young R, and Gaines J (1979) Hallucinogens as a discriminative stimuli: generalization of DOM to a 5-methoxy-N, N-dimethyltryptamine stimulus. Life Sci. 24, 993–997. [DOI] [PubMed] [Google Scholar]
  • (80).Glennon RA (1986) Discriminative stimulus properties of the serotonergic agent 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI). Life Sci. 39, 825–830. [DOI] [PubMed] [Google Scholar]
  • (81).Glennon RA (1988) Site-selective serotonin agonists as discriminative stimuli. Psychopharmacol. Ser 4, 15–31. [DOI] [PubMed] [Google Scholar]
  • (82).Young R, Glennon RA, and Rosecrans JA (1980) Discriminative stimulus properties of the hallucinogenic agent DOM. Commun. Psychopharmacol 4, 501–506. [PubMed] [Google Scholar]
  • (83).Winter JC, Rice KC, Amorosi DJ, and Rabin RA (2007) Psilocybin-induced stimulus control in the rat. Pharmacol., Biochem. Behav 87, 472–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (84).Fantegrossi WE, et al. (2008) Hallucinogen-like effects of N,N-dipropyltryptamine (DPT): possible mediation by serotonin 5-HT1A and 5-HT2A receptors in rodents. Pharmacol., Biochem. Behav 88, 358–365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (85).Gatch MB, Dolan SB, and Forster MJ (2017) Locomotor and discriminative stimulus effects of four novel hallucinogens in rodents. Behav. Pharmacol 28, 375–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (86).Krall CM, Richards JB, Rabin RA, and Winter JC (2008) Marked decrease of LSD-induced stimulus control in serotonin transporter knockout mice. Pharmacol., Biochem. Behav 88, 349–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (87).Winter JC, et al. (2005) The stimulus properties of LSD in C57BL/6 mice. Pharmacol., Biochem. Behav 81, 830–837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (88).Benneyworth MA, Smith RL, Barrett RJ, and Sanders-Bush E. (2005) Complex discriminative stimulus properties of (+)lysergic acid diethylamide (LSD) in C57Bl/6J mice. Psychopharmacology (Berl). 179, 854–862. [DOI] [PubMed] [Google Scholar]
  • (89).Smith RL, Barrett RJ, and Sanders-Bush E (2003) Discriminative stimulus properties of 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane [(±)DOI] in C57BL/6J mice. Psychopharmacology (Berl). 166, 61–68. [DOI] [PubMed] [Google Scholar]
  • (90).Benneyworth MA, Smith RL, and Sanders-Bush E (2008) Chronic Phenethylamine Hallucinogen Treatment Alters Behavioral Sensitivity to a Metabotropic Glutamate 2/3 Receptor Agonist. Neuropsychopharmacology 33, 2206–2216. [DOI] [PubMed] [Google Scholar]
  • (91).Smith RL, Canton H, Barrett RJ, and Sanders-Bush E (1998) Agonist properties of N,N-dimethyltryptamine at serotonin 5-HT2A and 5-HT2C receptors. Pharmacol., Biochem. Behav 61, 323–330. [DOI] [PubMed] [Google Scholar]
  • (92).Schreiber R, Brocco M, and Millan MJ (1994) Blockade of the discriminative stimulus effects of DOI by MDL 100,907 and the ‘atypical’ antipsychotics, clozapine and risperidone. Eur. J. Pharmacol 264, 99–102. [DOI] [PubMed] [Google Scholar]
  • (93).Pigott A, et al. (2012) trans −2-(2,5-Dimethoxy-4-iodophenyl)cyclopropylamine and trans −2-(2,5-dimethoxy-4-bromophenyl)cyclopropylamine as potent agonists for the 5-HT 2 receptor family. Beilstein J. Org. Chem 8, 1705–1709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (94).Almaula N, Ebersole BJ, Ballesteros JA, Weinstein H, and Sealfon SC (1996) Contribution of a helix 5 locus to selectivity of hallucinogenic and nonhallucinogenic ligands for the human 5-hydroxytryptamine2A and 5-hydroxytryptamine2C receptors: direct and indirect effects on ligand affinity mediated by the same locus. Mol. Pharmacol 50, 34–42. [PubMed] [Google Scholar]
  • (95).Canal CE, Booth RG, and Morgan D (2013) Support for 5-HT2C receptor functional selectivity in vivo utilizing structurally diverse, selective 5-HT2C receptor ligands and the 2,5-dimethoxy-4-iodoamphetamine elicited head-twitch response model. Neuropharmacology 70, 112–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (96).Piel M, Vernaleken I, and Rösch F (2014) Positron Emission Tomography in CNS Drug Discovery and Drug Monitoring. J. Med. Chem 57, 9232–9258. [DOI] [PubMed] [Google Scholar]
  • (97).Van de Bittner GC, Ricq EL, and Hooker JM (2014) A philosophy for CNS radiotracer design. Acc. Chem. Res 47, 3127–3134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (98).Jørgensen LM, et al. (2017) Cerebral 5-HT release correlates with [11C]Cimbi36 PET measures of 5-HT2A receptor occupancy in the pig brain. J. Cereb. Blood Flow Metab 37, 425–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (99).Finnema SJ, et al. (2014) Characterization of [11C]Cimbi-36 as an agonist PET radioligand for the 5-HT2A and 5-HT2C receptors in the nonhuman primate brain. NeuroImage 84, 342–353. [DOI] [PubMed] [Google Scholar]
  • (100).Yang K-C, et al. (2017) Fenfluramine Reduces [11C]Cimbi-36 Binding to the 5-HT2A Receptor in the Nonhuman Primate Brain. Int. J. Neuropsychopharmacol 20, 683–691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (101).Ettrup A, et al. (2016) Serotonin 2A receptor agonist binding in the human brain with [11C]Cimbi-36: Test-retest reproducibility and head-to-head comparison with the antagonist [18F]altanserin. NeuroImage 130, 167–174. [DOI] [PubMed] [Google Scholar]
  • (102).Ettrup A, et al. (2014) Serotonin 2A Receptor Agonist Binding in the Human Brain with [11C]Cimbi-36. J. Cereb. Blood Flow Metab 34, 1188–1196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (103).Herth MM, et al. (2016) Synthesis and evaluation of 18F-labeled 5-HT2A receptor agonists as PET ligands. Nucl. Med. Biol 43, 455–462. [DOI] [PubMed] [Google Scholar]
  • (104).Petersen IN, et al. (2016) Convergent 18 F-labeling and evaluation of N -benzyl-phenethylamines as 5-HT 2A receptor PET ligands. Bioorg. Med. Chem 24, 5353–5356. [DOI] [PubMed] [Google Scholar]
  • (105).Petersen IN, et al. (2017) 18 F-Labelling of electron rich iodonium ylides: application to the radiosynthesis of potential 5-HT 2A receptor PET ligands. Org. Biomol. Chem 15, 4351–4358. [DOI] [PubMed] [Google Scholar]
  • (106).Leth-Petersen S, et al. (2014) Correlating the Metabolic Stability of Psychedelic 5-HT2A Agonists with Anecdotal Reports of Human Oral Bioavailability. Neurochem. Res 39, 2018–2023. [DOI] [PubMed] [Google Scholar]
  • (107).Leth-Petersen S, et al. (2016) Metabolic Fate of Hallucinogenic NBOMes. Chem. Res. Toxicol 29, 96–100. [DOI] [PubMed] [Google Scholar]
  • (108).Wohlfarth A, et al. (2017) 25C-NBOMe and 25I-NBOMe metabolite studies in human hepatocytes, in vivo mouse and human urine with high-resolution mass spectrometry. Drug Test. Anal 9, 680–698. [DOI] [PubMed] [Google Scholar]
  • (109).Caspar AT, Meyer MR, and Maurer HH (2018) Human cytochrome P450 kinetic studies on six N −2-methoxybenzyl (NBOMe)-derived new psychoactive substances using the substrate depletion approach. Toxicol. Lett 285, 1–8. [DOI] [PubMed] [Google Scholar]
  • (110).Nielsen LM, et al. (2017) Characterization of the hepatic cytochrome P450 enzymes involved in the metabolism of 25I-NBOMe and 25I-NBOH. Drug Test. Anal 9, 671–679. [DOI] [PubMed] [Google Scholar]
  • (111).Boumrah Y, et al. (2016) In vitro characterization of potential CYP- and UGT-derived metabolites of the psychoactive drug 25B-NBOMe using LC-high resolution MS. Drug Test. Anal 8, 248–256. [DOI] [PubMed] [Google Scholar]
  • (112).Caspar AT, et al. (2015) Studies on the metabolism and toxicological detection of the new psychoactive designer drug 2-(4-iodo-2,5-dimethoxyphenyl)-N-[(2-methoxyphenyl)methyl]-ethanamine (25I-NBOMe) in human and rat urine using GC-MS, LC-MSn, and LC-HR-MS/MS. Anal. Bioanal. Chem 407, 6697–6719. [DOI] [PubMed] [Google Scholar]
  • (113).Seo H, Kim IS, Kim Y-H, Yoo HH, and Hong J (2019) Metabolic profile determination of 25N-NBOMe in human liver microsomes by liquid chromatography-quadrupole time-of-flight mass spectrometry. Int. J. Legal Med 133, 833–841. [DOI] [PubMed] [Google Scholar]
  • (114).Caspar AT, Meyer MR, Westphal F, Weber AA, and Maurer HH (2018) Nano liquid chromatography-high-resolution mass spectrometry for the identification of metabolites of the two new psychoactive substances N-(ortho-methoxybenzyl)-3,4-dimethoxyamphetamine and N-(ortho-methoxybenzyl)-4-methylmethamphetamine. Talanta 188, 111–123. [DOI] [PubMed] [Google Scholar]
  • (115).Grafinger KE, Stahl K, Wilke A, König S, and Weinmann W (2018) In vitro phase I metabolism of three phenethylamines 25D-NBOMe, 25E-NBOMe and 25N-NBOMe using microsomal and microbial models. Drug Test. Anal 10, 1607–1626. [DOI] [PubMed] [Google Scholar]
  • (116).Ketha H, Webb M, Clayton L, and Li S (2017) Gas Chromatography Mass Spectrometry (GC-MS) for Identification of Designer Stimulants Including 2C Amines, NBOMe Compounds, and Cathinones in Urine. Current Protocols in Toxicology, 4.43.1–4.43.10. [DOI] [PubMed] [Google Scholar]
  • (117).Richter LHJ, Maurer HH, and Meyer MR (2017) New psychoactive substances: Studies on the metabolism of XLR-11, AB-PINACA, FUB-PB-22, 4-methoxy-α-PVP, 25-I-NBOMe, and meclonazepam using human liver preparations in comparison to primary human hepatocytes, and human urine. Toxicol. Lett 280, 142–150. [DOI] [PubMed] [Google Scholar]
  • (118).Temporal K-DH, Scott KS, Mohr ALA, and Logan BK (2017) Metabolic Profile Determination of NBOMe Compounds Using Human Liver Microsomes and Comparison with Findings in Authentic Human Blood and Urine. J. Anal. Toxicol 41 , 646–657. [DOI] [PubMed] [Google Scholar]
  • (119).Poklis JL, et al. (2015) Identification of Metabolite Biomarkers of the Designer Hallucinogen 25I-NBOMe in Mouse Hepatic Microsomal Preparations and Human Urine Samples Associated with Clinical Intoxication. J. Anal. Toxicol 39, 607–616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (120).Halberstadt AL (2017) Pharmacology and Toxicology of N-Benzylphenethylamine (“NBOMe”) Hallucinogens. Curr. Top. Behav. Neurosci 32, 283–311. [DOI] [PubMed] [Google Scholar]
  • (121).Andreasen MF, et al. (2015) A fatal poisoning involving 25C-NBOMe. Forensic Sci. Int 251, e1–e8. [DOI] [PubMed] [Google Scholar]
  • (122).Lowe LM, Peterson BL, and Couper FJ (2015) A Case Review of the First Analytically Confirmed 25I-NBOMe-Related Death in Washington State. J. Anal. Toxicol 39, 668–671. [DOI] [PubMed] [Google Scholar]
  • (123).Walterscheid JP, et al. (2014) Pathological Findings in 2 Cases of Fatal 25I-NBOMe Toxicity. Am. J. Forensic Med. Pathol 35, 20–25. [DOI] [PubMed] [Google Scholar]
  • (124).Umemura Y, et al. (2014) Fatal outcome of status epilepticus, hyperthermia, rhabdomyolysis, multi-organ failure, and cerebral edema after 25I-NBOMe ingestion. Neurology 82, 342. [Google Scholar]
  • (125).Kueppers VB, and Cooke CT (2015) 25I-NBOMe related death in Australia: A case report. Forensic Sci. Int 249, e15–e18. [DOI] [PubMed] [Google Scholar]
  • (126).Yoshida K, et al. (2015) A case of fatal intoxication due to the new designer drug 25B-NBOMe. Forensic Toxicol. 33, 396–401. [Google Scholar]
  • (127).Shanks KG, Sozio T, and Behonick GS (2015) Fatal Intoxications with 25B-NBOMe and 25I-NBOMe in Indiana During 2014. J. Anal. Toxicol 39, 602–606. [DOI] [PubMed] [Google Scholar]
  • (128).Laskowski LK, et al. (2015) Evolution of the NBOMes: 25C- and 25B- Sold as 25I-NBOMe. J. Med. Toxicol 11, 237–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (129).Isbister GK, Poklis A, Poklis JL, and Grice J (2015) Beware of blotting paper hallucinogens: severe toxicity with NBOMes. Med. J. Aust 203, 266–267. [DOI] [PubMed] [Google Scholar]
  • (130).Poklis JL, Nanco CR, Troendle MM, Wolf CE, and Poklis A (2014) Determination of 4-bromo-2,5-dimethoxy-N-[(2-methoxyphenyl)methyl]-benzeneethanamine (25B-NBOMe) in serum and urine by high performance liquid chromatography with tandem mass spectrometry in a case of severe intoxication. Drug Test. Anal 6, 764–769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (131).Tang MHY, Ching CK, Tsui MSH, Chu FKC, and Mak TWL (2014) Two cases of severe intoxication associated with analytically confirmed use of the novel psychoactive substances 25B-NBOMe and 25C-NBOMe. Clin. Toxicol 52, 561–565. [DOI] [PubMed] [Google Scholar]
  • (132).Gee P, Schep LJ, Jensen BP, Moore G, and Barrington S (2016) Case series: toxicity from 25B-NBOMe - a cluster of N-bomb cases. Clin. Toxicol 54, 141–146. [DOI] [PubMed] [Google Scholar]
  • (133).Zwartsen A, Hondebrink L, and Westerink RH (2018) Neurotoxicity screening of new psychoactive substances (NPS): Effects on neuronal activity in rat cortical cultures using microelectrode arrays (MEA). NeuroToxicology 66, 87–97. [DOI] [PubMed] [Google Scholar]
  • (134).Xu P, et al. (2019) 25C-NBOMe, a Novel Designer Psychedelic, Induces Neurotoxicity 50 Times More Potent Than Methamphetamine In Vitro. Neurotoxic. Res 35, 993–998. [DOI] [PubMed] [Google Scholar]
  • (135).Yoon KS, et al. (2019) 2-(2,5-Dimethoxy-4-methylphenyl)-N-(2-methoxybenzyl)ethanamine (25D-NBOMe) and N-(2-methoxybenzyl)-2,5-dimethoxy-4-chlorophenethylamine (25C-NBOMe) induce adverse cardiac effects in vitro and in vivo. Toxicol. Lett 304, 50–57. [DOI] [PubMed] [Google Scholar]

RESOURCES