Movatterモバイル変換


[0]ホーム

URL:


Skip to main content
NCBI home page
Search in PMCSearch
As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsement of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more:PMC Disclaimer | PMC Copyright Notice
Sage Choice logo

Microdosing psychedelics: More questions than answers? An overviewand suggestions for future research

Kim PC Kuypers1,Livia Ng2,David Erritzoe3,Gitte M Knudsen4,Charles D Nichols5,David E Nichols6,Luca Pani7,8,Anaïs Soula9,David Nutt10,
1Department of Neuropsychology andPsychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University,Maastricht, The Netherlands
2Department of Psychology, UniversityCollege London, London, UK
3Department of Psychology,Neuropsychopharmacology Unit, Imperial College London, London, UK
4Neurobiology Research Unit,Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
5Department of Pharmacology andExperimental Therapeutics, Louisiana State University Health Sciences Center, NewOrleans, LA, USA
6Purdue University College of Pharmacy,West Lafayette, LA, USA
7Department of Psychiatry and BehavioralSciences, Psychiatry University of Miami, Miami, FL, USA
8Department of Biomedical, Metabolic& Neural Sciences, University of Modena, Modena, Italy
9COMPASS Pathways, London, UK
10Neuropsychopharmacology, ImperialCollege London, London, UK

David Nutt, Neuropsychopharmacology,Imperial College London, London, UK. Email:k.kuypers@maastrichtuniversity.nl

Issue date 2019 Sep.

© The Author(s) 2019

This article is distributed under the terms of the Creative CommonsAttribution-NonCommercial 4.0 License (http://www.creativecommons.org/licenses/by-nc/4.0/) whichpermits non-commercial use, reproduction and distribution of the workwithout further permission provided the original work is attributed asspecified on the SAGE and Open Access page (https://us.sagepub.com/en-us/nam/open-access-at-sage).

PMCID: PMC6732823  PMID:31303095

Abstract

Background:

In the past few years, the issue of ‘microdosing’ psychedelics has beenopenly discussed in the public arena where claims have been made about theirpositive effect on mood state and cognitive processes such as concentration.However, there are very few scientific studies that have specificallyaddressed this issue, and there is no agreed scientific consensus on whatmicrodosing is.

Aim:

This critique paper is designed to address questions that need to be answeredby future scientific studies and to offer guidelines for these studies.

Approach:

Owing to its proximity for a possible approval in clinical use andshort-lasting pharmacokinetics, our focus is predominantly on psilocybin.Psilocybin is allegedly, next to lysergic acid diethylamide (LSD), one ofthe two most frequently used psychedelics to microdose. Where relevant andavailable, data for other psychedelic drugs are also mentioned.

Conclusion:

It is concluded that while most anecdotal reports focus on the positiveexperiences with microdosing, future research should also focus on potentialrisks of (multiple) administrations of a psychedelic in low doses. To thatend, (pre)clinical studies including biological (e.g. heart rate, receptorturnover and occupancy) as well as cognitive (e.g. memory, attention)parameters have to be conducted and will shed light on the potentialnegative consequences microdosing could have.

Keywords: Psychedelics, microdosing, psychoactive substances

Background

Psychedelics are a class of psychoactive substances that induce complex behavioural,psychological and physiological effects primarily through activation of serotonin5-HT2A receptors. In the past few years, the issue of ‘microdosing’psychedelics has been openly discussed in the public arena with several books (Cruz, 2017;Kumar, 2016;Waldman, 2017) claimingvalue to the authors who tried this concept. However, there are very few scientificstudies that have specifically addressed this issue, and there is no agreedscientific consensus on what microdosing entails (Cameron et al., 2019;Horsley et al., 2018). This paper isdesigned to address questions that need to be answered by future scientific studiesand to offer guidelines for these studies. Although a number of classic psychedelicsexist, two of them, lysergic acid diethylamide (LSD) and psilocybin, are allegedlymost frequently used to microdose. The following review focuses predominantly onpsilocybin due to its proximity for a possible approval in clinical use andshort-lasting pharmacokinetics (Passie et al., 2002) in comparison with LSD (Dolder et al., 2017). However, whererelevant and available, data for other psychedelic drugs are also mentioned.

As early as the 16th century, low doses of psilocybin, ‘teonanacatl’or sacred mushroom, were used medically (Schultes, 1940). Bernardino de Sahagún, aFranciscan friar during the period of the Spanish conquest of the Americas(1519–1521), reported that, ‘teonanacatl were … medicinal for fevers and forrheumatism. Only two or three need to be eaten. Those who eat them see visions andfeel a faintness of the heart. And they provoke lust to those who eat a number, oreven a few, of them’. However, by 1640, 94% of the Aztec population was wiped outand alongside them, the traditions involving ‘teonanacatl’. Of note, the mentioningof visions here suggests this ancient ‘low-dose’ use does not refer to what iscurrently seen as microdosing, something that will be addressed below.

Psychedelic studies underwent a significant expansion following the discovery of themind-altering properties of LSD by Albert Hofmann in 1943 (Hofmann, 1970). The subsequent growth ofpsychedelic use allegedly had a profound effect on innovation in science andtechnology. A popular example is that of Francis Crick, one of the co-discoverers ofthe double-helix structure of DNA, who used LSD, though this use was never confirmednor denied by him (Roberts,2008). Furthermore, Kary Mullis, who discovered a means to automate thepolymerase chain reaction, claimed that the idea came to him after using LSD (Doyle, 2002). Thesediscoveries greatly advanced the field of genetic research (Luke, 2006). In this atmosphere ofinnovation, Frederick Terman was appointed as Provost of Stanford, 1955–1965. Duringhis tenure, Terman ‘set out to create a community of technical scholars in SiliconValley’ (Leslie and Kargon,1996). This community developed alongside the psychedelic capital of theworld, San Francisco, and over time technology and psychedelics began to merge. By2005, the founder of Apple and one of the most influential figures in SiliconValley, Steve Jobs, highlighted that LSD had played a pivotal and transformativerole in his life (Dormehl,2012).

Although there was accumulating evidence to suggest that the intake of psychedelicsled not only to hallucinations but also to an improvement of cognition andcreativity, scientific progress in the field was prohibited by government agencieson account of the growing political concern over the recreational use ofpsychedelics (Belouin andHenningfield, 2018). Thus, the only study investigating psychedelics inproblem solving was ended by the US Food and Drug administration (FDA) in 1966(Harman et al.,1966). However, James Fadiman, a young researcher in this study, continuedhis research after the UN Convention on Psychotropic Substances of 1971 bannedpsychoactive substances and bundled his knowledge into a book, which now acts as aguide for those interested in microdosing. His bookThe PsychedelicExplorer’s Guide: Safe, Therapeutic, and Sacred Journeys (Fadiman, 2011) published in2011, is often referred to as a protocol for those practising microdosing. Of note,no study to date has revealed statistically significant effects of microdosing oncreativity under placebo-controlled circumstances (Passie, 2019).

Although microdosing became prominent due to the belief it improved cognition, agrowing number of individuals began to microdose psychedelics to improve conditionsof pain (Johnstad, 2018),cluster headache or migraine (Andersson et al., 2017). It seems that the efficacy of microdosing mayderive from its non-psychedelic dose range, which provides treatment withoutaffecting cognition. Individuals also reported relief of pain with a long-termpsychedelic microdosing regimen (Johnstad, 2018). Thus, psychedelic microdosing might constitute adifferent paradigm to single psychedelic therapeutic sessions withmacrodoses where the nature and content of the experience playsa key role in predicting therapeutic outcome (Roseman et al., 2018;Schenberg, 2018). However, many questionsremain about the definition, safety, potential mechanism and future researchinvolving microdosing.

Question 1: What does microdosing mean?

The term microdosing is not a uniquely psychedelic term. In pharmacology, microdosingis a process used in drug development (Lappin and Garner, 2008) and drug selection(Lappin et al., 2006)where a minute dose of a substance is used to assess the pharmacokinetics of a drug.A microdose, in this regulatory arena, has been defined by a position paper from theEuropean Medicines Agency 2004 (EMEA, 2003), guidelines from the U.S. Food and Drug Administration in2006 (FDA, 2006) and theMinistery of Health, Labour and Welfare in Japan in 2008 (MHLW, 2008), and the current definitiveinternational guideline in 2009 (ICH, 2009) as being a dose of drug that is 1% of the pharmacologicallyactive dose, up to a maximum of 100 µg. Thus, psychedelic microdosing (‘5–10 µg ofLSD’ (Fadiman, 2011))would be 5–10% of a usual psychoactive dose and lie between a full pharmacologicaldose (100%) and a ‘pharmacological microdose’.

Microdosing psychedelics has been described in a similar manner by differentindividuals. Fadiman describes it as a practice ‘to use sub-threshold doses ofpsychedelic drugs in an attempt to enhance cognitive tasks, to boost physical energylevels, to promote emotional balance, and to treat anxiety, depression andaddiction’ resulting in typically subtle though noticeable effects (Fadiman, 2011). Similarly,Aylet Waldman in her book (Waldman, 2017) states the same intention for microdosing but describesthe process as ‘the act of integrating sub-perceptual doses of psychedelic drugs, inyour weekly routine’. In addition, Johnstad emphasizes that ‘to microdose with apsychedelic drug means to take a dose small enough to provide no intoxication orsignificant alteration of consciousness’ (Johnstad, 2018).

Thus, the term ‘microdosing’ appears to consist of three components:

  1. The use of a low dose below the perceptual threshold that doesnotimpair ‘normal’ functioning of an individual.

  2. A procedure that includes multiple dosing sessions.

  3. The intention to improve well-being and enhance cognitive and/or emotionalprocesses.

Existing dosing categories for psychedelics when used in research areverylow dose, low dose, medium dose, andhigh dose (Table 1). A microdose hasbeen defined as approximately one-tenth to one-twentieth of a recreational dose,varying within and between substances, so it can be seen as being somewhat below avery low dose. Although microdosing of psychedelics does not have an agreedscientific definition, we have decided to continue to use the term because of itsprevalent societal use. Hopefully, this paper will help to facilitate researchtowards establishing it as a scientific construct.

Table 1.

Varying doses of psychedelic compounds used in preclinical and clinicalstudies.

SubstanceSubjects/participants (animal/human)Route of administrationMicrodoseVery low doseLow doseMedium doseHigh dose
Psilocin (Hasler et al., 2004;Wackermann et al.,2008)Human (both studies)Oral<1 mg3.15 mg8 mg15 mg22 mg
LSD (Dandiyaet al., 1969)RatsIntraperitoneally10–25 μg30– 40 μg60–110 μg150 μg200+ μg
Ibogaine HCl (Glick et al., 2000;Lotsofand Wachtel, 2002;Schechter and Gordon,1993)aRats
Humansa
Intraperitoneally
Orala
200 mg300–400 mg700 mg1400 mga2800 mg
DMT (Shulgin, 1976)HumansIntramuscular injection6 mg10 mg20 mg30 mg50–70 mgb

Per kilogram dose values have been converted to values for a 70-kgperson. These doses are approximate values.

a

Study conducted in humans using a single oral dose of 1400 mg.

b

When inhaled, 30 mg would be considered a high dose.

The most widely distributed species of psychedelic mushrooms arePsilocybecubensis and those of the genusCopelandia, whichconsists of 12 species (Guzmánet al., 1998). The psilocin (the active metabolite of psilocybin) andpsilocybin content in the whole body of these mushrooms when dried was estimated tobe in the range of 0.14–0.42% (psilocin) and 0.37–1.30% (psilocybin) forP.cubensis and 0.43–0.76% (psilocin) and 0.08–0.22% (psilocybin) forCopelandia, respectively. Thus, the former is morepsilocybin-rich than the latter, and the latter contains more psilocin compared tothe former (Tsujikawa et al.,2003). ThePsilocybe semilanceata is the most commonBritish species. This mushroom only contains psilocybin, in the range from 0.17 to1.96%, as shown by one Norwegian analysis (Christiansen et al., 1981;Rumack and Spoerke, 1994).These data show that the psilocybin concentration varies between and within speciesbut is also dependent on the time of collection, the preservation of the materialand growth conditions. User reported recreational doses depend on the species andexperience of the user (Rumackand Spoerke, 1994).

A hallucinogenic dose of driedP. cubensis, for example, is between3 and 5 g (Rumack and Spoerke,1994). These values equate to a recreational dosing range of 8.6 to 14.7mg of psilocin per dose. Thus, a microdose would range from 0.43 to 0.73 mg ofpsilocin per dose because a microdose of psilocybin is generally one-tenth of a fulldose (Fadiman, 2011).That positions a recreational dose of psilocin between a low and medium dose and amicrodose below a very low dose. However, variations in psilocin content betweendoses of dried mushroom may be seen due to variations between individual fungiwithin a species. A microdose of LSD ranges between 10 and 20 μg with 20 μg beingthe upper limit that might already produce perceptual changes in some. A microdoseof ibogaine hydrochloride is approximately 25 mg (Kroupa and Wells, 2005), and when smoked,that ofN,N-dimethyltryptamine (DMT) is approximately 6 mg (May, 2018).

Question 2: What microdosing schedules have been used?

The data presented here were collected using a search of microdosing protocols thatincluded books, online fora and surveys. The keywords of this search includedmicrodosing, microdosing protocols, microdosing approaches and psilocybin microdose.In this search, it was found that users mainly followed three approaches. The mostpopular of these was the Fadiman approach, outlined in his book (Fadiman, 2011), whichinvolves two consecutive dosing days followed by two non-dosing days. Anotherpopular approach involves ‘weekday’ dosing, i.e. from Monday to Friday and notdosing on Saturday and Sunday. Additionally, some users indicated that they followeda balanced low/microdose approach, which involved dosing every other day. Dosingperiods ranged from 1 week to 2 years. This variation in microdosing schedules wasconfirmed by a recent survey which demonstrated that half of the respondents whomicrodosed came up with their own schedule (Hutten et al., 2019).

Question 3: What controlled studies have been done so far?

The first placebo-controlled LSD microdosing study was published recently (Yanakieva et al., 2018).Findings showed a delay of time perception in the absence of self-rated effects onperception, mentation and concentration after administration of single doses of 5,10, and 20 μg LSD. To our knowledge there has been only one published study designedspecifically to measure the effects of psilocybin microdosing per se (Prochazkova et al., 2018)where the effects of psychedelic mushrooms were explored in a recreational setting.This study suffers from a number of methodological issues, particularly the lack ofa placebo control as well as uncertainty over dose taken. However, there have beenseveral more controlled studies where a low dose of psilocybin has been used as acontrol for a regular dose; these are presented below.

For example,Hasler andcolleagues (2004) compared four doses of psilocybin in healthy humans ina placebo-controlled experimental design and found slight physiological andpsychological differences between single administration of placebo and a very lowdose (VLD) (Hasler et al.,2004). A VLD was defined as 45 μg/kg p.o., equating to approximately 2.3mg of psilocin for an average 70-kg human. VLD was compared with a low dose (LD), amedium dose (MD) and a high dose (HD) defined as 115, 215 and 315 μg/kg p.o.,respectively. Although most physiological measures were similar between the VLD doseand placebo, a significant decrease was seen in maximum heart rate at the 6-hourpoint after VLD administration. Acute self-rated/self-reported psychologicalresponses of VLD included slight drowsiness, increased sensitivity andintensification of pre-existing mood states; an increase in introversion compared toplacebo was only shown for the MD and HD at peak drug effect, 95 minutespost-administration.

Building on that,Griffiths andcolleagues (2011) investigated the effects of psilocybin in varying doseswhere each participant received five dosing sessions, spread across 1-monthintervals (Griffiths et al.,2011). The doses used were 0, 5, 10, 20 and 30 mg/70 kg. Using a MonitorRating Questionnaire with a 5-point scale, they found that a dose of 5 mg/70 kgincreased stimulation, distance from ordinary reality and sense of peace. Intensity,somaesthesia, affect, perception, cognition and volition measured on theHallucinogen Rating Scale all increased after administration of a 5 mg/70 kg dose.In other words, they did not find a dose without psychological effects.Interestingly, when using an 11 mg/70 kg and 15 mg/70 kg dose of psilocybin,Lewis and colleagues (2017)found a significant decrease in global cerebral blood flow in the frontal, parietal,temporal, limbic, cingulate and occipital cortex, insula, caudate, putamen,pallidum, amygdala, hippocampus and thalamus (Lewis et al., 2017). This may relate to thepsychological effects seen with lower doses.

Psychological effects of microdosing have been regularly reported by users aftermultiple administrations of psilocybin. Independent accounts from online fora andsurveys (Fadiman and Korb,2019;www.thethirdwave.co;www.dmt-nexus.me, 2018;www.reddit.com, 2018) reveal thatusers report improvements in energy, mood, cognition, concentration, management ofstress, creativity, spiritual awareness, productivity, language capabilities,relationships and visual capabilities. Further, users also reported reduced anxiety,depression and addiction and pain relief. In a recent survey byAnderson and colleagues(2018), users also noted drawbacks such as illegality, stigma, physicaldiscomfort, anxiety, overstimulation, cognitive interference, emotional difficultyand uncertainty of effect (Anderson et al., 2018). All of these reports are confounded by the lackof certainty relating to the actual dose used, or indeed the provenance of theactive ingredient, and the absence of placebo conditions. For a recent review ofpast research with psychedelic microdosing, please seePassie (2019).

Question 4: Are there any relevant preclinical studies?

We found only two preclinical studies involving microdosing (Cameron et al., 2019;Horsley et al., 2018).Horsley and colleagues(2018) investigated the effect of microdosing on anxiety using anelevated plus-maze and observation of ecological behaviours. They defined amicrodose of psilocin as 0.05 mg/kg, which equates to 3.5 mg for an average 70-kghuman. Rats received three dosing sessions over 6 days with the last dosing sessionon the 6th day. Anxiety profiles were measured in Wistar rats 2 days after the finaldosing session. Ethological behaviours including rears, head dips and stretch-attendwere also measured during this period. Psilocin at 0.05 mg/kg significantly reducedentries into open arms, suggesting that microdosing may have an anxiogenic effect.This effect was not replicated in the ethological measures. Although the authorsconclude that these results might have implications for future therapeuticapplications, as they produce counter-productive behaviour, one obvious limitationis the interspecies scaling issue (Sharma and McNeill, 2009). It isquestionable whether doses administered to animals translate to humans and theauthors also acknowledged that the translational value of their results needs to bedetermined in a therapeutic context.

Cameron and colleagues(2019) tested the effect of repeated low doses of DMT in rats. They gavea dose for 2 months every third day and assessed behaviour with a broad range oftests. In a cued fear extinction learning test, they showed that animals frozesignificantly less than a control group, suggesting that DMT facilitates fearextinction memory. In the forced swim test, an antidepressant-like effect wasobserved. No change was observed in dendritic spine density in the layer V pyramidalneurons, and no changes were observed in gene expression (EGR1, EGR2, ARC, FOS, BDNFand 5HT2A). However, an impact on metabolism was observed in male rats; the weightincreased by 182%, compared to 165% with vehicle (Cameron et al., 2019). Comparable to theHorsley et al. (2018)study, the interspecies scaling is a point of discussion together with the questionof whether a short-acting substance such as DMT would show beneficial effects inhumans without administration of a monoamine oxidase (MAO) inhibitor. Lastly, itshould be emphasized that there is a need to conduct more research on long-termeffects in order to assess the long-term safety of repeat doses.

Question 5: What is the pharmacology of psychedelics when used inmicrodoses?

The pharmacology of psilocybin and psilocin is still unclear due to the rapid declinein psychedelic drug research following their being made Schedule 1 drugs in 1968(Rucker et al.,2018). This decline predated the growth of modern neuropharmacology. Thus,more research is required to build a more complete pharmacological profile ofpsilocybin and psilocin.

Psilocybin (3[2-(dimethylamino)ethyl]indol-4-ol dihydrogen phosphate ester;O-phosphoryl-4-hydroxy-N,N-dimethyltryptamine)belongs to the indolealkylamine class of psychoactive compounds (Table 2). It is an indoleprodrug characterised by a 4-substituent, a phosphate group (Repke et al., 1977), six hydrogen bondacceptors and low lipophilicity (Geiger et al., 2018). Low lipophilicity may contribute to the notionthat psilocybin does not cross the blood–brain barrier (Rautio et al., 2008).

Table 2.

The physical and chemical properties of psilocybin.

NamePsilocybin
IUPAC name3[2-(dimethylamino)ethyl]indol-4-ol dihydrogen phosphate
Other common name4-Phosphoryl-N,N-dimethyltryptamine
Chemical formulaC12H17N2O4P
Molar mass204.27 g/mol
Melting point173 to 176°C (343 to 349°F)
Physical formSolid
Soluble inWater
Saline

In vivo, however, the majority of the prodrug psilocybin is rapidly converted topsilocin by alkaline phosphatases present in the blood and tissues. Psilocin hasfewer hydrogen bond acceptors in its structure, which increases lipophilicity. Inaddition, NMR spectral studies have implicated an intramolecular hydrogen bond inpsilocin that reduces the basicity of psilocin, increases its lipophilicity and alsomay render it stable to the action of MAO (Migliaccio et al., 1981). Followingsystemic circulation, psilocin is metabolized by either phase I or phase IImetabolism (Figure 1). Theformer involves an oxidation reaction to form 4-hydroxyindole-3-acetaldehydefollowed by either an oxidation to 4-hydroxyindole-3-acetic acid or a reduction to4-hydroxytryptophole. It is believed that none of these metabolites are biologicallyactive. The latter pathway involves the formation of a psilocinO-glucuronide conjugate through small intestine and liver enzymesUGT1A10 and UGT1A9, respectively. There is evidence that to some extent,glucuronidated psilocin can be converted back to psilocin (Brown et al., 2017). Although more than 80%of psilocin undergoes phase II metabolism, both phase I and II metabolites areultimately eliminated through renal excretion.

Figure 1.

Figure 1.

The structure of (a) psilocybin and (b) psilocin.

Depending on body weight, the minimum active oral dose of psilocybin is approximately4 to 10 mg in humans (vanAmsterdam et al., 2011). Onset of action as defined by the firstappearance of acute psychological symptoms begins 20 to 60 minutes following oralingestion and 10 to 40 minutes following buccal administration (Geiger et al., 2018) andalmost immediately following i.v. injection (Carhart-Harris et al., 2012).

Psilocin begins to appear in the plasma approximately 25 minutes after oral dosage,with peak levels reached after approximately 105 ± 37 minutes (Brown et al., 2017). A typical userresponds to a full active dose for approximately 4 to 7 hours. Even a VLD canproduce responses for up to 6 hours after dose administration (Hasler et al., 2004).

Question 6: Is microdosing safe?

Preclinical studies to assess the safety of repeated doses of psilocybin in rodentshave not been conducted. That may be due to several factors, including thehistorical background of psilocybin as an ingredient in magic mushrooms that hadbeen used in many cultures without apparent harm. Evidence from these accountsdemonstrates a lack of serious adverse events resulting from psilocybinadministration. There are, however, several non-clinical investigations ofpsilocybin’s safety profile. The risk of an adverse cardiovascular event due to hERG(human ether-a-go-go-related protein) potassium channel blockade is low, with hERGassay results demonstrating minimal effect of psilocybin at concentrations up to1000 μM (nominal) and completely without effect at 100 μM. That means that unwantedcardiac chronotropic effects with microdosing are very unlikely as the maximumplasma concentration of psilocybin produced by a 25-mg dose would not reach 160 nM(Brown et al.,2017).

Other potential and serious adverse events are cardiac valvulopathies due to repeatedactivation of serotonin 5-HT2B receptors, which psilocin activates alongwith many other serotonin receptors. Several drugs have recently been pulled fromthe market due to this concern. The first example of this was the diet medicationPhen/Fen, which had an unacceptably high fatality rate due to its effects on5-HT2B receptors in the heart (Connolly et al., 1997). Another example ismethysergide, an ergot-derived prescription drug that is still being used today as aprophylaxis in difficult to treat migraine and cluster headache (MacGregor and Evers, 2017).It has a known risk of increasing cardiac valve dysfunction (Joseph et al., 2003). In early reports itwas shown that although aortic insufficiencies disappeared in most cases afterarrest of the methysergide therapy, the mitral insufficiencies remained unchanged(Graham, 1967). Itremains to be seen whether repeated low-dose psilocybin administration inpreclinical studies might produce valvular hyperplasia, and whether or not thiswould translate to the human user population. This concern is discussed more in thenext section. So far psilocybin testing in preclinical studies has not revealed anysignals of valvulopathy.

A different psychedelic that is more often used for microdosing, LSD, has beenexamined in rodents after repeated dosing schedules similar to microdosing.Comparatively low doses of LSD administered every other day for several months wereshown to produce persistent negative behavioural changes that lasted for at leastseveral weeks to months after LSD administration was discontinued (Marona-Lewicka et al.,2011). These changes included increased aggression, scruffy appearance,anhedonia and hyper-reactivity. Analysis of gene expression in key cortical regionslike the medial prefrontal cortex indicated that LSD produced alterations in genesenriched for schizophrenia and bipolar depression that lasted long after the drugwas discontinued (Martin etal., 2014). Of note, here the interspecies scaling question arises, andit is disputable whether the (low) doses used in animals are comparable to thoseused by humans (Sharma andMcNeill, 2009). Related to these preclinical findings, another primarysafety concern for 5-HT2A agonists is the potential for adversepsychological response in humans (Carhart-Harris et al., 2016;Johnston et al., 2010;Vollenweider et al.,1998).

The lethal dose of psilocybin in a single administration in 50% of animals tested(the LD50) ranges from 280 mg/kg in rats and mice to 12.5 mg/kg in rabbits (Usdin and Efron, 1972;Williams, 2013).Animals receiving a very HD of psilocybin (10 mg/kg) exhibit sympathetic systemeffects such as irregularities in heart and breathing rate as well as mydriasis,piloerection, hyperglycaemia and hypertonia (Cerletti, 1958). Similar central excitatoryeffects were seen after the administration of 2–4 mg/kg intraperitoneal psilocybinin rhesus monkeys (Horibe,1974).

Question 7: What receptors will be involved in the activity of microdosedpsilocybin?

Psilocin predominantly binds to serotonin receptors: 5-HT1A,5-HT1D, 5-HT1E, 5-HT2A, 5-HT2B,5-HT2C, 5-HT5, 5-HT6 and 5-HT7(Table 3) (McKenna et al., 1990) andthe serotonin transporter and partially to the norepinephrine transporter, similarto MDMA (Rickli et al.,2016). Hill slope values demonstrate that psilocin binds independently atall 5-HT receptors except 5-HT2B where cooperative binding is exhibited(McKenna et al.,1990).

Table 3.

An overview of 5-HT receptors that are stimulated by psilocin. Ki values arebased on displacement of an antagonist radioligand.

Receptor typeG-proteinDistributionPhysiological responseBehavioural responseAgonistAntagonistDrug classes that act on this receptorPsilocin binding affinity: Ki (nM) (Halberstadtand Geyer, 2011)
5-HT1AGiCerebral cortex, hippocampus, septum, amygdala and raphe nucleusin high densities. Low amounts also exist in the basal gangliaand thalamus (Beliveau et al., 2017)I. Hypotension
II. Increase DA release in the medialprefrontal cortex, striatum and hippocampus
I. Decreased anxiety and depression (Campos and Guimarães,2008)
II. Impairment of declarative and non-declarativememory
III. Decreased aggression andimpulsivity
IV. Inhibition of drug-seeking behaviour
8-OH DPAT, buspirone,
5-CT (Barnes and Sharp, 1999),psilocybin (Mckenna et al., 1990)
WAY 100135 (methiothepin nonselective) (Barnes and Sharp,1999)I Analgesics (agonists)
II Antidepressants (post-synapticreceptor agonists and pre-synaptic autoreceptorantagonists)
III Anxiolytics (antagonists)
567.4
5-HT1DGiTrigeminal sensory neurones including peripheral and centralprojections to dural blood vessels and to the medulla (Longmore etal., 1997)Inhibition of adenylyl cyclaseI. Modulates locomotion and anxiety
II. Migraineprophylaxis (Longmore et al., 1997)
Dextromethorphan, sumatriptan, L694247, 5-CT (Barnes andSharp, 1999), psilocybin (McKenna et al.,1990)Sumatriptan, GR 127935 (metergoline, methiothepin nonselective)(Barnesand Sharp, 1999)I. Triptans (agonists used for migraine)36.4
5-HT1EGiHigh levels in olfactory bulb glomeruli and molecular layer ofdentate gyrus. Low amounts in the adventitial layer of cerebralarteries (Klein and Teitler, 2012)Inhibition of adenylyl cyclase5-HT (Barnesand Sharp, 1999), psilocybin (McKenna et al.,1990)None (methiothepin weak) (Barnes and Sharp,1999)N/A
5-HT2AGq/11High concentrations on the apical dendrites of pyramidal cellsin layer V, neocortex (mainly prefrontal, parietal andsomatosensory cortex) and the olfactory tubercle, as well ascardiovascular system (Beliveau et al.,2017)I. Vasoconstriction
II. Plateletaggregation
III. Bronchoconstriction
IV. Anti-inflammatory
I. Modulates addiction
II. Increasedanxiety
III. Increased appetite
IV. Improvedcognition (learning and memory)
V. Decreasedsleep
VI. Modulates sexual behaviour
Alpha-methyl-5-HT, DOI (Barnes and Sharp, 1999),psilocybin (McKenna et al., 1990)Ketanserin, pimvanserin, pirenperone (Barnes and Sharp,1999)I. Atypical antipsychotics(antagonists)
II. Antidepressants and anxiolytics(antagonists)
107.2
5-HT2BGq/11Predominantly peripheral, widespread tissue distributionincluding liver and kidneys (Julius et al.,1990)VasoconstrictionI. Regulates sleep (Qian et al.,2017)
II. Increased GI motility, especially smallintestine
III. Increased cardiac hypertrophy in mice
Alpha-methyl-5-HT, DOI (Barnes and Sharp, 1999),psilocybin (Mckenna et al., 1990)SB 200646 (also 5-HT2C antagonist)4.6
5-HT2CGq/11Mainly in choroid plexus, high concentrations in hippocampus,anterior olfactory nucleus, substantia nigra, amygdala,subthalamic nucleus and lateral habenula (Julius et al.,1990)I. Vasoconstriction
II. Increase phosphoinositideturnover
I. Increased anxiety
II. Increased GImotility
III. Modulates locomotion
IV. Modulatesmood and sexual behaviour
Alpha-methyl-5-HT, DOI, psilocybin (McKenna et al.,1990)Mesulergine (also 5-HT2A antagonist)I. Antidepressant (antagonists)
II. Orexigenic(antagonists)
III. Anorectic(agonists)
IV. Antipsychotic (agonists)
97.3
5HT5AGi/GoHigh concentrations in olfactory bulb and medial habenula ofwild-type mice. Lower densities in neocortex, hippocampus andtrigeminal nucleusN/AI. Modulates locomotion
II. Increases sleep
5-CT, valerenic acid (partial agonist)Methiothepin, ritanserin, asenapine, psilocybin (McKenna etal., 1990)N/A83.7
5-HT6GsPredominantly in the caudate nucleus, with lower concentrationsin hippocampus and amygdala. Very low levels of expression inthe thalamus, subthalamic nucleus and substantial nigra (Yoshioka etal., 1998)Activation of adenylyl cyclase (HEK 293 cells)I. Increased anxiety
II. Reduced cognition andmemory
III. Negative effect on mood
EDMT, EMD-386,088Amitriptyline aripiprazole, MS-245, psilocybin (McKenna etal., 1990)I. Antidepressants (antagonists)
II. Anxiolytics(antagonists)
III. Nootropics(antagonists)
IV. Anorectics (antagonists)
57.0
5-HT7GsPredominantly the caudate and putamen nuclei, the pyramidallayer of the CA2 field of the hippocampus, the centromedialthalamic
nucleus and the dorsal raphe nucleus (Ruat et al.,1993)
I. Activation of adenylyl cyclase (HeLa cells and COScells)
II. Vasoconstriction
I. Increased anxiety
II. Decreasedmood
III. Reduced working and reference memory
5-CT, 8-OH-DPAT, aripiprazole, AS-19, psilocybin (Glennon,2003)Methiothepin, mianserin, SB-269,270I. Antidepressants (antagonists)
II. Anxiolytics(antagonists)
III. Nootropics (antagonists)
3.5

Cerebral 5-HT receptors that can be stimulated by psilocin are highly distributedamong different regions (Table3). Many behavioural and neuropsychological effects claimed to beelicited by microdosing are known to be modulated by these receptors (Anderson et al., 2018).

Psilocin acts as a partial agonist at the 5-HT2A receptor with 46%(+/−2.4) response compared with the response produced by serotonin for signallingthrough the phospholipase C (PLC) pathway (Kurrasch-Orbaugh et al., 2003). It has alower binding affinity to the 5-HT2A receptor compared to LSD (Rickli et al., 2016).Currently there is only one study of the in vivo cerebral 5-HT2A receptoroccupancy produced by the psilocybin metabolite psilocin in humans. That was done bythe Copenhagen group led by Knudsen who used the PET tracer[11C]Cimbi-36. This tracer is an agonist of the 5-HT2Areceptor and therefore particularly sensitive to displacement by another agonist,psilocin. Having performed a dose-finding study of psilocybin that ranged from 3 to30 mg p.o. per person, they found that the plasma concentration that produced a 50%occupancy of the 5-HT2A receptor was 1.95 (range 1.16–3.15) μg psilocin/L(Madsen et al.,2019). They also found that plasma psilocin was positively correlated withsubjective intensity ratings and that doses producing less than 20% occupancy (i.e.probably less than 0.028 mg/kg body weight) were not detectable either bypsychological or physiological measurements (Madsen et al., 2019), suggesting that thisconcentration might represent the threshold for microdosing, based on brain5-HT2A receptor occupancy.

At this dose level, several 5-HT receptors other than the 5-HT2A receptormay also be affected. This could include antagonist activity at the 5-HT6and 5-HT7 receptors that may improve mood and cognition (Ballaz et al., 2007;Mnie-Filali et al., 2009).The 5-HT7 receptor is also implicated in the regulation of circadianrhythms (Lovenberg et al.,1993). Upon assessing binding affinity of LSD and DMT at 5-HT7receptors, similarly high Ki values of 9.5 nM (Ruat et al., 1993) were found.Additionally, it has been found that 5-HT7 receptor activation reducessecondary hypersensitization in response to capsaicin in mice (Brenchat et al., 2009). Thus, psilocybinagonist activity at 5-HT7 may relate to the ancient use of ‘teonanacatl’to ease rheumatism.

Further, psilocin also binds with relatively high affinities to 5-HT1D(Ki = 36.4 nM) and 5-HT2B (Ki = 4.6 nM)receptors. 5-HT1D is predominantly expressed in the trigeminal system,which may account for the recent reports of self-medication using microdoses ofpsychedelics to produce migraine prophylaxis (May, 2018). With regard to the5-HT2B there are concerns of the development of cardiac valvulopathyassociated with agonism at 5-HT2B (Elangbam et al., 2005). It is mostly theuse of intermittent high-dose psilocybin intake that has been discredited. However,even with repeated microdosing there is a possibility that 5-HT2Breceptors might be stimulated enough to lead to tissue overgrowth. A potentialmitigation against this risk is the suggestion that the efficacy of psilocin (EC50> 20 μM) (May, 2018)is lower than that of 5-HT. Nonetheless, because psilocin also has a higher affinityfor the 5-HT2B receptor than 5-HT, further investigation is needed tounderstand better the risks associated with microdosing.

Some stimulation at 5-HT1A receptors may also occur. Such activity hasbeen implicated in the mechanism of action of antidepressant medications includingSSRIs (Celeda et al., 2013). Activation of these receptors by psilocin couldconceivably be involved in reduction in anxiety and increased mood swings (Carhart-Harris and Nutt,2017) due to dense distribution of the receptor in the midbrain, limbicand cortical regions that regulate stress and emotion.

Question 8: Are the claims of the benefits of microdosing biologicallyplausible?

There have been only a few studies on the basic neurobiology of psychedelics at the5-HT2A receptor. Recent work has shown that psychedelics like DOI andLSD directly produce transcriptional activation of Immediate Early Genes (IEGs) likecfos in only about 5% of neurons within key brain structures,and that these activated Trigger Population neurons express significantly higherlevels of receptor than the non-activated neurons (Martin and Nichols, 2016). Transcriptionalactivation of IEGs within neurons is generally accepted to be a reliable marker forneural activity (Joo et al., 2016). Further, psychedelics also act on subsets ofinhibitory neurons, and non-neuronal cells like glia and astrocytes (Martin and Nichols, 2016).Together, these data indicate that within specific brain regions, psychedelicstrigger complex patterns of excitatory and inhibitory neurons in small subsets ofcells, and that how these cells are activated differs between brain regions (Martin and Nichols,2016).

Genes acutely activated by LSD in the brain are predominantly involved in synapticplasticity (Nichols andSanders-Bush, 2002;Nichols and Sanders-Bush, 2004). Accordingly, activation of5-HT2A receptors in brain slice culture modulates aspects oflong-term plasticity, and expression of brain-derived neurotrophic factor (BDNF)(Vollenweider and Kometer,2010). BDNF expression is also observed to increase in primary culturesof cortical neurons 24 hours following the application of psychedelics (DOI, DMT,LSD) (Ly et al., 2018).Blockade of the receptor for BDNF, Trk-B, prevents increased spinogenesis andsynaptogenesis in cortical neurons that have been treated with psychedelics.Interestingly, the mammalian target of the rapamycin (mTOR) pathway is activateddownstream of psychedelics in cortical neuron cultures similarly to ketamine, andlikely mechanistically underlies the synaptogenesis (Ly et al., 2018). None of these preclinicalstudies, however, utilized psilocybin, and it remains to be seen if it produces thesame effects, and if so, at what dose?

Psychedelics are known to induce behavioural tolerance, an absence of behaviouraleffects after repeated intake of a substance. Previously it was shown thatbehavioural effects were for example diminished after repeated doses of LSD (Abramson et al., 1956); inaddition, another study showed these effects to be associated with reduced cortical5-HT2A receptor binding (Gresch et al., 2005). Serotoninsyndrome-related symptoms, skin jerks, shaking behaviour and hyperthermia, inducedby a single dose of the 5-HT2A agonist DOI in rats were absent afterrepeated low dosing, suggesting behavioural tolerance (Pranzatelli and Pluchino, 1991). Althoughspeculative, this downregulation of the 5-HT2A receptor might be amechanism of action underlying some of its putative therapeutic effects. An exampleis obsessive-compulsive disorder (OCD), a psychiatric condition that ischaracterized by increased 5-HT2A binding (Adams et al., 2005). Preliminary data haveshown that administration of low to high doses of psilocybin lead to symptomreduction in patients with OCD (Moreno et al., 2006). It was previously suggested that a re-balancebetween 5-HT1A and 5-HT2A receptors might be responsible forobserved therapeutic actions (Buchborn et al., 2014), but this remains to be investigated.

In peripheral tissues, very low doses of psychedelics have profound anti-inflammatoryeffects (Yu et al.,2008). In general, psychedelics in the phenethylamine class such as DOI havemore potency than those in the ergoline class such as LSD. In a rodent model ofasthma, for example, levels of the R stereoisomer (R)-DOI that are30 times lower than the behavioural threshold can have profound effects to preventinflammation, T-Helper Cell Type 2 (Th2) cell recruitment, eosinophilia and mucusproduction in the lung, resulting in animals that can breathe normally afterexposure to an allergen (Nau etal., 2015). In another animal model of inflammatory bowel disease, levelsof the psychedelic (R)-DOI 30 times lower than the behaviouralthreshold nearly completely prevented intestinal inflammation (Nau et al., 2013). It remains to be seenwhether very low levels of psychedelics are also anti-inflammatory in humans, and ifthe anti-inflammatory activity also occurs in the brain, but if these findings dotranslate then levels typically used in microdosing regimes for some psychedeliccompounds would be predicted to have significant and beneficial anti-inflammatoryeffects. Interestingly, although LSD is one of the most powerful and potentmind-altering psychedelics, it is comparatively among the least potentanti-inflammatories tested (Yuet al., 2008).

Question 9: What is the legal position of microdosing?

The answer to this question is complex due to differences in national regulations. Ingeneral, under the UN Conventions, LSD and related compounds, psilocybin and DMT arecontrolled as Schedule 1 drugs – i.e. are defined as being the most harmful and ashaving no medicinal value. In other words, they are subject to the most extremerestrictions and penalties for unapproved possession. These constraints apply to anydose of the drug, even a sub-psychoactive or microdose level. Research can becarried out with the right ethical regulatory and institutional approvals, butdosing would have to be conducted in a secure environment like a hospital orresearch ward. For repeated microdosing, this adds significant costs and complexityto any study, which is likely why none have yet been reported.

However, this situation is easing for psilocybin as a result of several successfulclinical trials in recent years, and both the European and US regulators have givenapproval for studies (NIH,2018) with psychedelic doses of synthetic psilocybin made to GMPstandards. That means that microdosing trials of similarly sourced product forclinical therapy are likely also to be approved, though as yet we do not believe anyhave been submitted.

Question 10: What are the regulatory issues?

Unfortunately, due to their long history of anecdotal use in recreational settings,none of the psychedelics has ever followed the conventional drug research anddevelopment path expected by contemporary standards. Thus, at best, doses have beenselected based on published data in a variety of indications but mostly to providean indication for an upper safety limit. In a pooled analysis of psilocybinStuderus et al. (2011)classified active oral doses within a vast range of about one order of magnitudedifference, between 0.045 and 0.315 mg/kg, which translates into 3.15 to 22.05 mgfor a 70-kg human (Studerus etal., 2011). Such a range is quite surprising for active principles withthe pharmacological potency of psychedelics. Given such underdetermination,regulatory standards will most likely require dedicated dose-finding studies (morethan one) to provide a rationale explaining the known individual differences thathave been reported in the clinical response to treatment and most importantly, thedose chosen for late development. In this context, the information provided fromoral dosing, resulting plasma psilocin levels and corresponding brain5-HT2A receptor occupancy will turn out to be informative.

Parallel fixed dose designs are usually recommended (ICH, 1994). In some cases, four arm rangestudies could be necessary; under these circumstances microdoses could be used totest pseudo-placebo properties or alternatively a peculiar pharmacologicalactivity.

Another issue pertains to the limited pharmacokinetic data available in order toevaluate a dose-concentration–response relationship for psychedelics. Updated ADMEstudies are not available for psychedelics, although the characterization of theirmetabolites and their role in the active principle efficacy or safety profile mightprove relevant to interpret and predict their clinical effect. Once pharmacokineticsof the parent compound and its metabolite(s) are established, variation of clearanceif any, its prediction by body weight and the concentration–response relationshipfor the claimed clinical effect must be presented. If possible, biomarker(s) (e.g.single-nucleotide polymorphisms (SNPs)) at the 5-HT receptor subtypes where theyhave affinity should be linked to the risk/benefit profile and thus to thetherapeutic effect, and they could be used to enrich/stratify the population ofinterest. Because psychedelics have been reported by some to possess a largeinter-individual sensitivity, the definition of a precise concentration–responserelationship may be difficult to demonstrate, especially once a microdose range isreached.

Question 11: What are the future research needs?

Microdosing is generally accepted as the use of a functionally low dose of apsychedelic compound over multiple dosing sessions with the intention of improvingmental and physical well-being, cognition or creativity (Fadiman, 2011;Johnstad, 2018). A systematic study ofmicrodosing psychedelics investigated by means of observation changes inpsychological variables of microdosers. Small changes in a sub-set of variables werefound, i.e. decreased depression and stress, decreased mind wandering, increasedabsorption and increased neuroticism. Interestingly, these variables were not thosethat participants most expected to change, suggesting that long-term changes may bedue to biological changes and not only expectations (Polito and Stevenson, 2019). Nonetheless,the possible effects and implications of microdosing remain largely unknown.Although there is a large database of reported effects of ‘microdosing’ on onlinefora, the true amount of active substance in these is unknown as are the peak plasmapsilocin concentrations achieved during ‘intoxication’. Further, while in theseanecdotal reports the user deliberately ingests a substance for a reason, expectingpositive effects, it is difficult to distinguish between expectation ‘placebo’effects and the effect of a microdose. These non-pharmacological effects, describedas set and setting, are also known to be of influence when taking a full dose of apsychedelic (Hartogsohn,2017). Another unknown is whether effects are noticeable after only onemicrodose or that a certain ‘build-up’ is needed, supported by underlyingneurobiological changes, before effects occur.

Therefore, rigorous placebo-controlled clinical studies need to be conducted withdifferent low doses of the drug to determine whether there is any evidence for theclaims being made by microdosers. The types of cognitive testing performed shouldinclude several different validated psychological instruments and preferably coverthe concepts mentioned in the Research Domain Criteria (Cuthbert and Kozak, 2013), and not simplyrely on anecdotal accounts or simple tests. Generated knowledge in healthyvolunteers will provide clear information on which cognitive aspects can be enhancedwith microdosing. This knowledge will provide a first hint as to whether microdosingcan be of value in the treatment of specific symptoms in psychiatric populations.Anecdotal reports suggest, for example, that microdosing might help in combattingattention deficit hyperactivity disorder (ADHD) symptoms; studies including measuresrelated to symptom domains like executive functioning, attention and temporalprocessing will help to decode the potential of microdosing as a therapeutic agent.In terms of biological mechanism of action, more (pre)clinical work needs to beperformed to understand fully the complex interaction of different cell types, andtheir responses to psychedelics at the molecular level such as elucidatingperipheral or central signalling pathways involved, if any, in the process ofmicrodosing (Kuypers,2019). Resulting findings can provide theoretical grounds for whymicrodosing could work in alleviating cluster headache in patients suffering from it(Anderson et al.,2018;Johnstad,2018).

Whereas most anecdotal reports focus on the positive experiences with microdosing,future research should investigate the molecular mechanisms behind low-dosepsilocybin behavioural effects as well as address potential risks of (multiple)administrations of a psychedelic in low doses. Although extensive toxicology hasbeen conducted on a single active dose of psilocybin and has been proven to be safe(Brown et al., 2017;Johnson et al.,2018), further research is required to understand better the possible healthrisks incurred by microdosing, especially in relation to cardiac and lung tissue.These studies would involve (pre)clinical safety and tolerability tests of multiplelow/microdoses of psilocybin over an extended period of time. To that end,continuous monitoring of physiological parameters including heart functioning inaddition to assessment of receptor turnover at low/microdoses as well as receptoroccupancy will shed light on the potential negative consequences microdosing couldhave.

Acknowledgments

Livia Ng was a paid intern and Anaïs Soula an employee for COMPASS Pathways.

Footnotes

Declaration of conflicting interest: The author(s) declared the following potential conflicts of interest with respectto the research, authorship, and/or publication of this article: David Nutt is ascientific adviser and Luca Pani is a consultant for COMPASSPathways. The otherauthors declare that there is no conflict of interest.

Funding: The author(s) received no financial support for the research, authorship, and/orpublication of this article.

References

  1. Abramson HA, Jarvik ME, Gorin MH, et al. (1956) Lysergicacid diethylamide (LSD-25): XVII. tolerance development and its relationshipto a theory of psychosis. J Psychol41: 81–105. [Google Scholar]
  2. Adams KH, Hansen ES, Pinborg LH, et al. (2005) Patientswith obsessive-compulsive disorder have increased 5-HT2A receptor binding inthe caudate nuclei. Int JNeuropsychopharmacol8: 391–401. [DOI] [PubMed] [Google Scholar]
  3. Anderson T, Petranker R, Dinh-Williams LA. (2018) Demography of microdosingcommunity survey. Available at:osf.io/g5cwy(accessed 25 June 2019). [Google Scholar]
  4. Andersson M, Persson M, Kjellgren A. (2017) Psychoactive substances as alast resort: A qualitative study of self-treatment of migraine and clusterheadaches. Harm Reduct J14: 60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Ballaz SJ, Akil H, Watson SJ. (2007) Analysis of 5-HT6 and 5-HT7receptor gene expression in rats showing differences in novelty-seekingbehavior. Neuroscience147: 428–438. [DOI] [PubMed] [Google Scholar]
  6. Barnes NM, Sharp T. (1999) A review of central 5-HTreceptors and their function.Neuropharmacology38: 1083–1152. [DOI] [PubMed] [Google Scholar]
  7. Beliveau V, Ganz M, Feng L, et al. (2017) Ahigh-resolution in vivo atlas of the human brain’s serotoninsystem. J Neurosci37: 120–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Belouin SJ, Henningfield JE. (2018) Psychedelics: Where we arenow, why we got here, what we must do.Neuropharmacology142: 7–19. [DOI] [PubMed] [Google Scholar]
  9. Brenchat A, Romero L, Garcia M, et al. (2009) 5-HT7receptor activation inhibits mechanical hypersensitivity secondary tocapsaicin sensitization in mice. Pain141: 239–247. [DOI] [PubMed] [Google Scholar]
  10. Brown RT, Nicholas CR, Cozzi NV, et al. (2017)Pharmacokinetics of escalating doses of oral psilocybin inhealthy adults. Clin Pharmacokinet56: 1543–1554. [DOI] [PubMed] [Google Scholar]
  11. Buchborn T, Schröder H, Höllt V, et al. (2014) Repeatedlysergic acid diethylamide in an animal model of depression: Normalisationof learning behaviour and hippocampal serotonin 5-HT2signalling. J Psychopharmacol28: 545–552. [DOI] [PubMed] [Google Scholar]
  12. Cameron LP, Benson CJ, Defelice BC, et al. (2019) Chronic,intermittent microdoses of the psychedelic N,N-Dimethyltryptamine (DMT)produce positive effects on mood and anxiety in rodents.ACS Chem Neurosci. Available at: 10.1021/acschemneuro.8b00692 (accessed25 June 2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Carhart-Harris RL, Erritzoe D, Williams T, et al. (2012) Neuralcorrelates of the psychedelic state as determined by fMRI studies withpsilocybin. Proc Natl Acad Sci109: 2138–2143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Carhart-Harris RL, Kaelen M, Bolstridge M, et al. (2016) Theparadoxical psychological effects of lysergic acid diethylamide(LSD). Psychol Med46: 1379–1390. [DOI] [PubMed] [Google Scholar]
  15. Carhart-Harris RL, Nutt DJ. (2017) Serotonin and brain function:A tale of two receptors. J Psychopharmacol31: 1091–1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Cerletti A. (1958) Etude Pharmacologique de laPsilocybine. In: Heim R, Wasson RG. (eds) Les Champignons Hallucinogenes duMexique. Paris: Musée dehistorie naturelle. [Google Scholar]
  17. Christiansen A, Rasmussen K, Tønnesen F. (1981) Determination of psilocybinin Psilocybe semilanceata using high-performance liquid chromatography on asilica column. J Chromatogr A,210:163–167. [Google Scholar]
  18. Connolly HM, Crary JL, McGoon MD, et al. (1997) Valvularheart disease associated with fenfluramine–phentermine.N Engl J Med337: 581–588. [DOI] [PubMed] [Google Scholar]
  19. Cruz RW. (2017) Microdosing LSD: The DefinitiveGuide to Increased Creativity and Productivity.Independently Published. [Google Scholar]
  20. Cuthbert BN, Kozak MJ. (2013) Constructing constructs forpsychopathology: The NIMH research domain criteria.J Abnorm Psychol122: 928–937. [DOI] [PubMed] [Google Scholar]
  21. Dandiya PC, Gupta BD, Gupta ML, et al. (1969) Effectsof LSD on open field performance in rats.Psychopharmacologia15: 333–340. [DOI] [PubMed] [Google Scholar]
  22. Dolder PC, Schmid Y, Steuer AE, et al. (2017)Pharmacokinetics and pharmacodynamics of lysergic aciddiethylamide in healthy subjects. ClinPharmacokinet56: 1219–1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Dormehl L. (2012) The Apple Revolution: Steve Jobs,the Counter Culture and how the Crazy Ones Took Over the World.London: VirginBooks. [Google Scholar]
  24. Doyle R. (2002) LSDNA: Rhetoric,consciousness expansion, and the emergence of biotechnology.Philosop Rhetoric35: 153–174. [Google Scholar]
  25. Elangbam CS, Lightfoot RM, Yoon LW, et al. (2005)5-Hydroxytryptamine (5HT) receptors in the heart valves ofcynomolgus monkeys and Sprague-Dawley rats. JHistochem Cytochem53: 671–677. [DOI] [PubMed] [Google Scholar]
  26. EMEA (2003) EMEA Position Paper onNon-clinical Safety Studies to Support Clinical Trials with a SingleMicrodose. London:European Agency for the Evaluation of MedicinalProducts. [Google Scholar]
  27. Fadiman J. (2011) The Psychedelic Explorer’s Guide:Safe, Therapeutic, and Sacred Journeys. South Paris,ME: Park StreetPress. [Google Scholar]
  28. Fadiman J, Korb S. (2019) Might microdosingpsychedelics be safe and beneficial? An initial exploration.J Psychoactive Drugs51: 118–122. [DOI] [PubMed] [Google Scholar]
  29. FDA (2006) Guidance for Industry,Investigators, and Reviewers Exploratory IND Studies.Rockville, MD: U.S.Department of Health and Human Services Food and Drug Administration Centerfor Drug Evaluation and Research (CDER). [Google Scholar]
  30. Geiger HA, Wurst MG, Daniels RN. (2018) DARK classics in chemicalneuroscience: Psilocybin. ACS Chem Neurosci9: 2438–2447. [DOI] [PubMed] [Google Scholar]
  31. Glennon RA. (2003) Higher-end serotoninreceptors: 5-HT5, 5-HT6, and 5-HT7. J MedChem46: 2795–2812. [DOI] [PubMed] [Google Scholar]
  32. Glick SD, Maisonneuve IM, Szumlinski KK. (2000) 18-Methoxycoronaridine(18-MC) and ibogaine: Comparison of antiaddictive efficacy, toxicity, andmechanisms of action. Ann N Y Acad Sci914: 369–386. [DOI] [PubMed] [Google Scholar]
  33. Graham JR. (1967) Cardiac and pulmonaryfibrosis during methysergide therapy for headache.Trans Am Clin Climatol Assoc78: 79–92. [PMC free article] [PubMed] [Google Scholar]
  34. Gresch PJ, Smith RL, Barrett RJ, et al. (2005)Behavioral tolerance to lysergic acid diethylamide isassociated with reduced serotonin-2A receptor signaling in ratcortex. Neuropsychopharmacology30: 1693. [DOI] [PubMed] [Google Scholar]
  35. Griffiths RR, Johnson MW, Richards WA, et al. (2011)Psilocybin occasioned mystical-type experiences: Immediateand persisting dose-related effects.Psychopharmacology218: 649–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Guzmán G, Allen JW, Gartz J. (1998) A worldwide geographicaldistribution of the neurotropic fungi, an analysis anddiscussion. Annali del Museo Civico diRovereto14: 189–280. [Google Scholar]
  37. Halberstadt AL, Geyer MA. (2011) Multiple receptors contributeto the behavioral effects of indoleamine hallucinogens.Neuropharmacology61: 364–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Harman WW, Mckim RH, Mogar RE, et al. (1966)Psychedelic agents in creative problem-solving: A pilotstudy. Psychol Rep19: 211–227. [DOI] [PubMed] [Google Scholar]
  39. Hartogsohn I. (2017) Constructing drug effects: Ahistory of set and setting. Drug Sci PolLaw3: 2050324516683325. [Google Scholar]
  40. Hasler F, Grimberg U, Benz MA, et al. (2004) Acutepsychological and physiological effects of psilocybin in healthy humans: Adouble-blind, placebo-controlled dose-effect study.Psychopharmacology (Berl)172: 145–156. [DOI] [PubMed] [Google Scholar]
  41. Hofmann A. (1970) The discovery of LSD andsubsequent investigations on naturally occurringhallucinogens. In: Ayd FJ, Blackwell B. (eds) Discoveries in Biological Psychiatry.Philadelphia:Lippincott, pp. 91–106. [Google Scholar]
  42. Horibe M. (1974) The effects of psilocybin onEEG and behaviour in monkeys. Act Nerv Super(Praha)16: 40–42. [PubMed] [Google Scholar]
  43. Horsley RR, Palenicek T, Kolin J, et al. (2018) Psilocinand ketamine microdosing: Effects of subchronic intermittent microdoses inthe elevated plus-maze in male Wistar rats. BehavPharmacol29: 530–536. [DOI] [PubMed] [Google Scholar]
  44. Hutten NRPW, Mason NL, Dolder PC, et al. (2019) Motivesand side-effects of microdosing with psychedelics amongusers. Int J Neuropsychopharmacol22: 5426–5434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. ICH (1994) Topic E4 Dose ResponseInformation to Support Drug Registration.London: European Agency forthe Evaluation of Medicinal Products. [Google Scholar]
  46. ICH (2009) Note for Guidance onNon-Clinical Safety Pharmacology Studies for Human PharmaceuticalsM3(R2). London:European Medicines Agency. [Google Scholar]
  47. Johnson MW, Griffiths RR, Hendricks PS, et al. (2018) Theabuse potential of medical psilocybin according to the 8 factors of theControlled Substances Act.Neuropharmacology142: 143–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Johnstad PG. (2018) Powerful substances in tinyamounts: An interview study of psychedelic microdosing.Nord Stud Alcohol Dr35: 39–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Johnston LD, O’Malley PM, Bachman JG, et al. (2010) Monitoring theFuture: National Survey Results on Drug Use, 1975–2009. Volume I: SecondarySchool Students. NIH Publication No. 10–7584.National Institute on Drug Abuse(NIDA). [Google Scholar]
  50. Joseph T, Tam SKC, Kamat BR, et al. (2003)Successful repair of aortic and mitral incompetence inducedby methylsergide maleate: Confirmation by intraoperative transesophagealechocardiography. Echocardiography20: 283–287. [DOI] [PubMed] [Google Scholar]
  51. Julius D, Huang KN, Livelli TJ, et al. (1990) The 5HT2receptor defines a family of structurally distinct but functionallyconserved serotonin receptors. Proc Natl Acad Sci US A87: 928–932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Klein MT, Teitler M. (2012) Distribution of 5-HT(1E)receptors in the mammalian brain and cerebral vasculature: Animmunohistochemical and pharmacological study. Br JPharmacol166: 1290–1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Kroupa PK, Wells H. (2005) Ibogaine in the 21st century:Boosters, Tune-ups and Maintenance.Multidisciplinary Association for Psychedelic Studies(MAPS)BulletinXV: 21–24. [Google Scholar]
  54. Kumar N. (2016) Microdosing: Enhance Your Vitality,Temperament and Efficiency with Hallucinogenic.Scotts Valley, CA:CreateSpace Independent PublishingPlatform. [Google Scholar]
  55. Kurrasch-Orbaugh DM, Watts VJ, Barker EL, et al. (2003)Serotonin 5-hydroxytryptamine 2A receptor-coupledphospholipase C and phospholipase A2 signaling pathways have differentreceptor reserves. J Pharmacol Exp Ther304: 229–237. [DOI] [PubMed] [Google Scholar]
  56. Kuypers KPC. (2019) Psychedelic medicine: Thebiology underlying the persisting psychedelic effects.Med Hypotheses125: 21–24. [DOI] [PubMed] [Google Scholar]
  57. Lappin G, Garner RC. (2008) The utility of microdosingover the past 5 years. Expert Opin Drug MetabToxicol4: 1499–1506. [DOI] [PubMed] [Google Scholar]
  58. Lappin G, Kuhnz W, Jochemsen R, et al. (2006) Use ofmicrodosing to predict pharmacokinetics at the therapeutic dose: Experiencewith 5 drugs. Clin Pharmacol Ther80: 203–215. [DOI] [PubMed] [Google Scholar]
  59. Leslie SW, Kargon RH. (1996) Selling Silicon Valley:Frederick Terman’s model for regional advantage. BusHist Rev70: 435–472. [Google Scholar]
  60. Lewis CR, Preller KH, Kraehenmann R, et al. (2017) Two doseinvestigation of the 5-HT-agonist psilocybin on relative and global cerebralblood flow. Neuroimage159: 70–78. [DOI] [PubMed] [Google Scholar]
  61. Longmore J, Shaw D, Smith D, et al. (1997)Differential distribution of 5Ht1D-and 5HT1B-immunoreactivitywithin the human trigemino-cerebrovascular system: Implications for thediscovery of new antimigraine drugs.Cephalalgia17: 833–842. [DOI] [PubMed] [Google Scholar]
  62. Lotsof HS, Wachtel B. (2002) Manual for Ibogaine Therapy:Screening, Safety, Monitoring & Aftercare.Ibogaine Dossier. [Google Scholar]
  63. Lovenberg TW, Baron BM, De Lecea L, et al. (1993) A noveladenylyl cyclase-activating serotonin receptor (5-HT7) implicated in theregulation of mammalian circadian rhythms.Neuron11: 449–458. [DOI] [PubMed] [Google Scholar]
  64. Luke D. (2006) A Tribute to Albert Hofmannon his 100th Birthday: The Mysterious Discovery of LSD and the Impact ofPsychedelics on Parapsychology. The ParanormalReview37: 1–8. [Google Scholar]
  65. Ly C, Greb AC, Cameron LP, et al. (2018)Psychedelics promote structural and functional neuralplasticity. Cell Rep23: 3170–3182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. MacGregor EA, Evers S. (2017) The role of methysergide inmigraine and cluster headache treatment worldwide: A survey in members ofthe International Headache Society.Cephalalgia37: 1106–1108. [DOI] [PubMed] [Google Scholar]
  67. Madsen MK, Fisher PM, Burmester D, et al. (2019)Psychedelic effects of psilocybin correlate with serotonin 2Areceptor occupancy and plasma psilocin levels.Neuropsychopharmacology44: 1328–1334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Marona-Lewicka D, Nichols CD, Nichols DE. (2011) An animal model ofschizophrenia based on chronic LSD administration: Old idea, newresults. Neuropharmacology61: 503–512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Martin DA, Marona-Lewicka D, Nichols DE. (2014) Chronic LSD alters geneexpression profiles in the mPFC relevant to schizophrenia.Neuropharmacology83: 1–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Martin DA, Nichols CD. (2016) Psychedelics recruit multiplecellular types and produce complex transcriptional responses within thebrain. EBioMedicine11: 262–277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. May J. (2018) The Ibogaine Conversation Part 9:How Safe is Ibogaine? [Online]. Available at:https://www.psymposia.com/magazine/how-safe-is-ibogaine-we-asked-clare-wilkins-who-has-facilitated-over-700-treatments/.
  72. McKenna DJ, Repke DB, Lo L, et al. (1990)Differential interactions of indolealkylamines with5-hydroxytryptamine receptor subtypes.Neuropharmacology29: 193–198. [DOI] [PubMed] [Google Scholar]
  73. Migliaccio GP, Shieh TL, Byrn SR, et al. (1981)Comparison of solution conformational preferences for thehallucinogens bufotenin and psilocin using 360-MHz proton NMRspectroscopy. J Med Chem24: 206–209. [DOI] [PubMed] [Google Scholar]
  74. Ministery of Health, Labour and Welfare (2008)Microdose clinical studies. In:Pharmaceutical and Medical Safety Bureau, M. O. H. L. A. W.(ed.) English Regulatory Information Task Force Japan PharmaceuticalManufacturers Association. Tokyo,Japan:MHLW. [Google Scholar]
  75. Mnie-Filali O, Lambas-Senas L, Scarna H, et al. (2009)Therapeutic potential of 5-HT7 receptors in mooddisorders. Curr Drug Targets10: 1109–1117. [DOI] [PubMed] [Google Scholar]
  76. Moreno FA, Wiegand CB, Taitano EK, et al. (2006) Safety,tolerability, and efficacy of psilocybin in 9 patients withobsessive-compulsive disorder. J ClinPsychiatry67: 1735–1740. [DOI] [PubMed] [Google Scholar]
  77. Nau F, Jr., Miller J, Saravia J, et al. (2015)Serotonin 5-HT(2) receptor activation prevents allergicasthma in a mouse model. Am J Physiol Lung Cell MolPhysiol308: L191–L198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Nau F, Jr, Yu B, Martin D, et al. (2013)Serotonin 5-HT2A receptor activation blocks TNF-alphamediated inflammation in vivo. PLoS One8: e75426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Nichols CD, Sanders-Bush E. (2002) A single dose of lysergicacid diethylamide influences gene expression patterns within the mammalianbrain. Neuropsychopharmacology26: 634–642. [DOI] [PubMed] [Google Scholar]
  80. Nichols CD, Sanders-Bush E. (2004) Molecular genetic responsesto lysergic acid diethylamide include transcriptional activation of MAPkinase phosphatase-1, C/EBP-beta and ILAD-1, a novel gene with homology toarrestins. J Neurochem90: 576–584. [DOI] [PubMed] [Google Scholar]
  81. NIH (2018) Trials with‘Psilocybin’. NIH US National Libary of Medicine.ClinicalTrials.gov.
  82. Passie T. (2019) Science of MicrodosingPsychedelics. London:Psychedelic Press. [Google Scholar]
  83. Passie T, Seifert J, Schneider U, et al. (2002) Thepharmacology of psilocybin. Addict Biol7: 357–364. [DOI] [PubMed] [Google Scholar]
  84. Polito V, Stevenson RJ. (2019) A systematic study ofmicrodosing psychedelics. PLoS One14: e0211023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Pranzatelli MR, Pluchino RS. (1991) The relation of central5-HT1A and 5-HT2 receptors: Low dose agonist-induced selective tolerance inthe rat. Pharmacol Biochem Behav39: 407–413. [DOI] [PubMed] [Google Scholar]
  86. Prochazkova L, Lippelt DP, Colzato LS, et al. (2018)Exploring the effect of microdosing psychedelics oncreativity in an open-label natural setting.Psychopharmacology235: 3401–3413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Qian Y, Cao Y, Deng B, et al. (2017) Sleephomeostasis regulated by 5HT2b receptor in a small subset of neurons in thedorsal fan-shaped body of Drosophila. Elife6: e26519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Rautio J, Laine K, Gynther M, et al. (2008) Prodrugapproaches for CNS delivery. AAPS J10: 92–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Repke DB, Leslie DT, Mandell DM, et al. (1977) GLC-massspectral analysis of psilocin and psilocybin. JPharm Sci66: 743–744. [DOI] [PubMed] [Google Scholar]
  90. Rickli A, Moning OD, Hoener MC, et al. (2016) Receptorinteraction profiles of novel psychoactive tryptamines compared with classichallucinogens. Eur Neuropsychopharmacol26: 1327–1337. [DOI] [PubMed] [Google Scholar]
  91. Roberts A. (2008) Albion Dreaming: A Popular Historyof LSD in Britain (Revised Edition with a New Foreword by Dr. SueBlackmore). Singapore: MarshallCavendish International (Asia) PtdLimited. [Google Scholar]
  92. Roseman L, Nutt DJ, Carhart-Harris RL. (2018) Quality of acute psychedelicexperience predicts therapeutic efficacy of psilocybin fortreatment-resistant depression. FrontPharmacol8: 974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Ruat M, Traiffort E, Leurs R, et al. (1993)Molecular cloning, characterization, and localization of ahigh-affinity serotonin receptor (5-HT7) activating cAMPformation. Proc Natl Acad Sci U S A90: 8547–8551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Rucker JJH, J Iliff, DJ Nutt. (2018) Psychiatry & thepsychedelic drugs. Past, present & future.Neuropharmacology142: 200–218. [DOI] [PubMed] [Google Scholar]
  95. Rumack BH, Spoerke DG. (1994) Handbook of Mushroom Poisoning:Diagnosis and Treatment. Boca Raton,FL: Taylor &Francis. [Google Scholar]
  96. Schechter MD, Gordon TL. (1993) Comparison of the behavioraleffects of ibogaine from three sources: Mediation of discriminativeactivity. Eur J Pharmacol249: 79–84. [DOI] [PubMed] [Google Scholar]
  97. Schenberg EE. (2018) Psychedelic-assistedpsychotherapy: A paradigm shift in psychiatric research anddevelopment. Front Pharmacol9: 733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Schultes RE. (1940) Teonanacatl: The narcoticmushroom of the Aztecs. Am Anthropol42: 429–443. [Google Scholar]
  99. Sharma V, McNeill JH. (2009) To scale or not to scale: Theprinciples of dose extrapolation. Br JPharmacol157: 907–921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Shulgin AT. (1976) DMT &TMA-2. J Psychedelic Drugs8: 167–169. [Google Scholar]
  101. Studerus E, Kometer M, Hasler F, et al. (2011) Acute,subacute and long-term subjective effects of psilocybin in healthy humans: Apooled analysis of experimental studies. JPsychopharmacol25: 1434–1452. [DOI] [PubMed] [Google Scholar]
  102. Tsujikawa K, Kanamori T, Iwata Y, et al. (2003)Morphological and chemical analysis of magic mushrooms inJapan. Forensic Sci Int138: 85–90. [DOI] [PubMed] [Google Scholar]
  103. Usdin E, Efron DH. (1972) Psychotropic Drugs and RelatedCompounds. Washington, DC:National Institute of MentalHealth. [Google Scholar]
  104. van Amsterdam J, Opperhuizen A, Van Den Brink W. (2011) Harm potential of magicmushroom use: A review. Regul ToxicolPharmacol59: 423–429. [DOI] [PubMed] [Google Scholar]
  105. Vollenweider FX, Kometer M. (2010) The neurobiology ofpsychedelic drugs: Implications for the treatment of mooddisorders. Nat Rev Neurosci11: 642–651. [DOI] [PubMed] [Google Scholar]
  106. Vollenweider FX, Vollenweider-Scherpenhuyzen MF, Babler A, et al. (1998)Psilocybin induces schizophrenia-like psychosis in humans viaa serotonin-2 agonist action. Neuroreport9: 3897–3902. [DOI] [PubMed] [Google Scholar]
  107. Wackermann J, Wittmann M, Hasler F, et al. (2008) Effectsof varied doses of psilocybin on time interval reproduction in humansubjects. Neurosci Lett435: 51–55. [DOI] [PubMed] [Google Scholar]
  108. Waldman A. (2017) A Really Good Day: How MicrodosingMade a Mega Difference in My Mood, My Marriage, and My Life.New York:Knopf. [Google Scholar]
  109. Williams M. (ed.) (2013) The Merck Index: AnEncyclopedia of Chemicals, Drugs, and Biologicals. 15thed.Cambridge: Royal Society ofChemistry. [Google Scholar]
  110. www.dmt-nexus.me(2018) Microdosing [Online] (accessed28 August 2018).
  111. www.reddit.com(2018) Psilocybin, Microdose Providing more Energy andMotivation: Microdosing [Online] (accessed 28 August2018).
  112. Yanakieva S, Polychroni N, Family N, et al. (2018) Theeffects of microdose LSD on time perception: A randomised, double-blind,placebo-controlled trial. Psychopharmacology(Berl). Epub ahead of print 26 November 2018. DOI: 10.1007/s00213-018-5119-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Yoshioka M, Matsumoto M, Togashi H, et al. (1998) Centraldistribution and function of 5-HT6 receptor subtype in the ratbrain. Life Sci62: 1473–1477. [DOI] [PubMed] [Google Scholar]
  114. Yu B, Becnel J, Zerfaoui M, et al. (2008)Serotonin 5-hydroxytryptamine(2A) receptor activationsuppresses tumor necrosis factor-alpha-induced inflammation withextraordinary potency. J Pharmacol Exp Ther327: 316–323. [DOI] [PubMed] [Google Scholar]
J Psychopharmacol. 2019 Jul 14;33(9):1039–1057.

Commentary

Microdosing psychedelics: Too much hype, almost no rigorous research

A call to explore microdosing’s psychological effects and therapeuticpotential within psychiatry

Stephen Ross

NYU Addictive Disorders and Experimental Therapeutics Research Laboratory, New YorkUniversity, New York, NY, USA

Corresponding author:

Stephen Ross, NYU Addictive Disorders and Experimental Therapeutics ResearchLaboratory, 462 First Ave, New York, NY 10016, USA. Email:Stephen.Ross@nyulangone.org;Stephen.ross@nyumc.org

This timely article (‘Microdosing psychedelics: More questions than answers? Anoverview and suggestions for future research’) provides an excellent overview on thetopic of microdosing psychedelics. The review focuses predominantly on psilocybinwith some mention of other psychedelics (i.e. LSD, ibogaine and DMT). This articleis organized around a series of questions and answers that includes the followingtopics related to microdosing: basic definitions, varying dosing regimens, relevantpreclinical data, pharmacology, safety issues, potential biological mechanisms ofaction, scientific evidence base (or relative lack thereof) from publishedpeer-reviewed research, legal and regulatory issues, and a call for rigorousplacebo-controlled trials to test the various beneficial claims of microdosing madeso far by anecdotal reports.

The therapeutic application of the classical psychedelics within psychiatry has madea historic come back within the last two decades and is at inflection point with agrowing body of rigorously conducted research pointing to the promising clinicalutility of psychedelic-assisted psychotherapies to treat a range of psychiatricdisorders, with the data most robust for: cancer-related psychological andexistential distress (Gasser etal., 2014;Griffithset al., 2016;Grobet al., 2011;Rosset al., 2016), alcohol and tobacco addiction (Bogenschutz et al., 2015;Johnson et al., 2014), andmajor depression (Carhart-Harriset al., 2016;Osorioet al., 2015). The doses used in all of these clinical trials are of the‘macro’ variety, typically consisting of moderate to high-dose psilocybin,administered as part of the psychedelic therapy model in combination withpsychotherapy with the goal of inducing significant alterations of consciousness,including mystico-mimetic states (Bogenschutz and Ross, 2018).

In contrast, and as defined in this article, microdosing is a practice of consumingvery low doses of a psychedelic substance (that produce acute drug effects that areeither not perceptible or minimally identifiable), utilizing a repeated dosingregimen, and with the goal of improving well-being, enhancing productivity orincreasing creativity. It is distinct from both psychedelic therapy (i.e. using highdoses of psychedelics to occasion mystical-type experiences, and used historicallyto treat conditions such as alcoholism and terminal cancer-related existentialdistress) and psycholytic therapy (i.e. using lower doses of psychedelics to produceperceptible and significant shifts in cognition and affect to enhance psychotherapy,historically used in combination with psychoanalytic psychotherapy to treat variouspsychiatric conditions such as anxiety spectrum disorders) (Ross and Bogenschutz, 2017). Structuredmicrodosing regimens employed so far (typically by lay individuals with theintention of receiving some positive benefit) range from daily use to dosing every 3days (Fadiman, 2011), andoccurring over varying time-frames from monthly to ongoing. Microdosing has surgedin popularity over the last several years and has been associated with the use ofserotoninergic psychedelics (i.e. LSD, psilocybin, ayahuasca, iboga), cannabinoids(i.e. CBD), and dissociative anaesthetics (i.e. ketamine) (Kitchens, 2018). Even though microdosingdoes not cause significant alterations in perception with acute administration, ithas led to numerous anecdotal reports (featured in a rapidly growing number of mediaarticles) of claimed benefits in a variety of domains including: Psychological (i.e.improved mood, energy, emotional balance, empathy, openness, introspection;decreased pain and alcohol/drug use or craving); Cognitive (i.e. improved focus,concentration, mental clarity); Creativity (i.e. improved idea generation anddivergent thought processes); Spirituality (i.e. improved meaning in life);Interpersonal (i.e. improved connectedness, sensitivity to others, relationalskills); and General well-being and quality of life (i.e. improved sleep quality,healthy eating habits, sexual function) (Austin, 2016;Kitchens, 2018).

Despite all of the growing interest and purported claims of benefits of microdosing,basic knowledge about microdosing, including therapeutic effects, is virtuallyabsent due to almost no peer-reviewed publications stemming from rigorouslyconducted research. Some of the most significant and important gaps in the knowledgebase of microdosing include the absence or paucity of: rigorous design methodologyincluding randomization, placebo control, adequate blinding integrity, and the useof appropriate inclusion/exclusion criteria to minimize potential harm; priorapproval with appropriate ethical and human subjects review boards; the use of anddispensation of an exact known dose of pharmaceutical grade psychedelic in acontrolled setting; understanding basic effects on psychological, cognitive andaffective domains; adequate safety monitoring and knowledge of acute or long-termsafety issues; understanding of basic neurobiological or potential mechanisms ofaction, such as neuroimaging (i.e. PET, functional MRI), physiological data (i.e.EEG, MEG) and biomarkers (i.e. BDNF).

This article touches on most of these gaps. The review article points out that therehas only been one published trial that specifically examined the effects ofpsilocybin microdosing. This was an open-label, non-controlled trial, with uncertaindoses of psilocybin ingested, that was conducted without ethical approval. Itreported on improvements in two creativity-related problem-solving tasks followingrecreational psilocybin microdosing (Prochazkova et al., 2018). A limitation ofthe present article is the focus on psilocybin even though online fora suggest thatrecreational LSD microdosing may be as common (if not more so) than psilocybinmicrodosing (www.reddit.com,2019). By not focusing equally on LSD microdosing, the article does notcapture the most rigorously conducted and peer-reviewed published trial ofmicrodosing to date. Unlike all of the prior publications on microdosing, this trialwas approved by an independent ethics committee, included random assignment,double-blind methodology, and the use of a placebo condition (Yanakieva, 2019). Further, appropriateinclusion/exclusion criteria were used to enhance safety, the trial was conducted ina monitored inpatient setting to optimize safety and exact doses of pharmaceuticalgrade cGMP LSD were administered. In comparing three microdoses of LSD (5, 10 and 20µg) to placebo, the trial reported that LSD microdosing produced temporal dilationin the absence of significant consciousness alteration (Yanakieva et al., 2019).

The review article also focuses on some theoretical safety concerns such as the linkbetween 5-HT2B agonism and cardiac valvulopathies (Cavera and Guillon,2014). The article would have been strengthened by a call to monitor broadly forpotential adverse medical and psychiatric effects in future microdosing research tobe able to identify all of the unknown risks that may be associated withmicrodosing. For example, we do not know the risks associated with the interactionbetween microdosing and underlying psychiatric illnesses. All of the moderntherapeutic trials of psychedelic-assisted psychotherapies exclude participants withpsychotic spectrum illnesses because of the known negative association betweenpsychedelic use and psychotic exacerbation in those with underlying psychoticillness (Ross and Peselow,2012). In the early stages of microdosing research, it would be importantto begin cautiously by excluding individuals with significant psychiatric, medicalor neurological illnesses.

One area not covered by the review article is speculation on the potentialtherapeutic utility of microdosing within psychiatry. If some of the claimedpsychological or cognitive benefits (i.e. improved mood and attention; decreasedsubstance craving and pain perception) of microdosing are real (i.e. not simply dueto placebo or expectancy effects), the next logical step would be to test thepotential efficacy of microdosing in various clinical populations (i.e. majordepression, bipolar depression, ADHD, addictive disorders, pain disorders) throughRCTs, and beyond through the drug development process. Funding sources would have tobe considered, whether that would come through private philanthropy, pharma orgovernmental funding agencies (i.e. NIHR in Europe or NIH in the USA). Finally, ifmicrodosing proves to be effective for the many claimed effects reported in the laypublic (i.e. enhanced creativity, work productivity, learning, memory, empathy,connection to others, spirituality), it could potentially be used to improvefunction in ‘normals’ without specific disease states, although it is unclear howdrug development would proceed for non-clinical entities by using mostly illegalsubstances.

Footnotes

Declaration of conflicting interests: The author(s) declared no potential conflicts of interest with respect to theresearch, authorship, and/or publication of this article.

Funding: The author(s) received no financial support for the research, authorship,and/or publication of this article.

References

  1. Austin P. (2016) Everything You Want to KnowAbout Microdosing. Available at:https://thethirdwave.co(2019) Microdosing mushrooms [Online] (accessed 24 June2019).
  2. Bogenschutz MP, Ross S. (2018) Therapeutic applicationsof classic hallucinogens. Curr Top BehavNeurosci36: 361–391. [DOI] [PubMed] [Google Scholar]
  3. Bogenschutz MP, Forcehimes AA, Pommy JA, et al. (2015)Psilocybin-assisted treatment for alcohol dependence: Aproof-of-concept study. JPsychopharmacol29: 289–299. [DOI] [PubMed] [Google Scholar]
  4. Carhart-Harris RL, Bolstridge M, Rucker J, et al. (2016)Psilocybin with psychological support fortreatment-resistant depression: An open-label feasibilitystudy. Lancet Psychiatry3: 619–627. [DOI] [PubMed] [Google Scholar]
  5. Fadiman J. (2011) The Psychedelic Explorer’sGuide: Safe, Therapeutic, and Sacred Journeys.Rochester, VT: ParkStreet Press. [Google Scholar]
  6. Gasser P, Holstein D, Michel Y, et al. (2014)Safety and efficacy of lysergic aciddiethylamide-assisted psychotherapy for anxiety associated withlife-threatening diseases. J Nerv MentDis202:513–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Griffiths RR, Johnson MW, Carducci MA, et al. (2016)Psilocybin produces substantial and sustained decrease indepression and anxiety in patients with life-threatening cancer: Arandomized double-blind trial. JPsychopharmacol30:1181–1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Grob CS, Danforth AL, Chopra GS, et al. (2011)Pilot study of psilocybin treatment for anxiety inpatients with advanced-stage cancer. Arch GenPsychiatry68: 71–78. [DOI] [PubMed] [Google Scholar]
  9. Johnson MW, Garcia-Romeu A, Cosimano MP, et al. (2014)Pilot study of the 5-HT2AR agonist psilocybin in thetreatment of tobacco addiction. JPsychopharmacol28: 983–992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Kitchens S. (2018) Microdosing’sMicromoment. New York Magazine.https://www.thecut.com/2018/05/microdosing-guide-and-explainer.html(accessed 15 January 2019).
  11. Osorio Fde L, Sanches RF, Macedo LR, et al. (2015)Antidepressant effects of a single dose of ayahuasca inpatients with recurrent depression: A preliminaryreport. Braz J Psychiatry37: 13–20. [DOI] [PubMed] [Google Scholar]
  12. Prochazkova L, Lippelt DP, Colzato LS, et al. (2018)Exploring the effect of microdosing psychedelics oncreativity in an open-label natural setting.Psychopharmacology (Berl)235:3401–3413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Ross S, Peselow E. (2012) Co-occurring psychoticand addictive disorders: Neurobiology and diagnosis.Clin Neuropharmacol35: 235–243. [DOI] [PubMed] [Google Scholar]
  14. Ross S, Bossis A, Guss J, et al. (2016)Rapid and sustained symptom reduction followingpsilocybin treatment for anxiety and depression in patients withlife-threatening cancer: A randomized controlled trial.J Psychopharmacol30:1165–1180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Ross S, Bogenschutz M. (2017) Hallucinogen-assistedpsychotherapy. In: Sadock B, Sadock V, Ruiz P. (eds) Kaplan and Sadock’s Comprehensive Textbook ofPsychiatry. 10th ed.Philadelphia, PA: WoltersKluwer. [Google Scholar]
  16. www.reddit.com (2019)https://www.reddit.com/r/microdosing/wiki/index(accessed 15 January 2019).
  17. Yanakieva S, Polychroni N, Family N, et al. (2019) Theeffects of microdose LSD on time perception: A randomized, double-blind,placebo-controlled trial. Psychopharmacology(Berl)236:1159–1170. [DOI] [PMC free article] [PubMed] [Google Scholar]
J Psychopharmacol. 2019 Jul 14;33(9):1039–1057.

Commentary

Psychedelics: What are low doses and ‘microdoses’?

Matthias E Liechti

Division of Clinical Pharmacology and Toxicology, Department of Biomedicine andDepartment of Clinical Research, University Hospital Basel and University of Basel,Basel, Switzerland

Corresponding author:

Matthias E Liechti, Division of Clinical Pharmacology and Toxicology, UniversityHospital Basel, Hebelstrasse 2, Basel, CH-4031, Switzerland. Email:Matthias.liechti@usb.ch

In clinical pharmacology, ‘microdosing’ refers to the use of a dose of a substancewith <1% pharmacological activity to study pharmacokinetics (PK), includingabsorption, distribution, metabolism and excretion, potentially usingnon-good-manufacturing practice (GMP) material and/or radiolabelled substances (e.g.Muehlan et al.,2018). ‘Microdosing’ with lysergic acid diethylamide (LSD) or psilocybinrefers to the use of low to very low doses (i.e. 5–20% of fully active doses;Table 1). I would ratherlabel such use as ‘low to very low doses’.

While I mostly agree with the views in the article, I would like to add someadditional thoughts on dosing with a focus on LSD. What is actually meant by themicrograms of LSD doses? Correct doses are obviously unknown in the case of illicitsources, and the correct reporting of doses is also an issue in scientificpublications. Research in the 1950s to 1970s used LSD tartrate, whereas modernresearch commonly uses LSD base (Liechti, 2017), with the exception of atleast one study (Yanakieva etal., 2019). A dose of 100 µg LSD base corresponds to 123 µg LSD tartrate(when being free of crystal water or methanol solvate;Mesley and Evans, 1969) – this dose maynot be considered low (Table1). Thus, researchers need to indicate whether they are reporting base orsalt doses. Additionally, LSD is mostly not a pharmaceutical product and notmanufactured according to GMP with defined content and stability. Researchersproduce their own formulations. It is often not reported where the substance wasobtained from and the way in which it was formulated as the final study drug. Thisraises many questions. Are the reported doses salt or base? What is the purity? Whatis the content of crystal water, solvate and/or residual solvents? How was thisfactored into the reported dose? What is the content uniformity and stability of theproduct throughout the study duration? What is the amount of inactive iso-LSD in thepharmaceutical formulation?

LSD is inactivated to iso-LSD depending on temperature, solvent and pH and thus maybe unstable in certain formulations. Other stress factors such as light, oxygen ortap water chlorine may also lead to decomposition of the LSD molecule. In fact,amounts of iso-LSD were detected in plasma in research subjects, indicating thatapproximately 30% of the LSD that was administered likely isomerized to inactiveiso-LSD possibly within the LSD capsules that were used (Steuer et al., 2017). Novel PK studies(Holze et al. 2019)that use validly defined doses of a novel LSD formulation indicate that previousstudies used 70 and 140 µg of LSD base (equivalent to 86 and 172 µg LSD tartrate)rather than the reported oral doses of 100 and 200 µg LSD base (Dolder et al., 2017;Liechti, 2017;Preller et al., 2017). Inaddition to correctly assessing and reporting the study drug, PK data are needed,thus providing the basis of the present discussion and any drug development. Plasmaconcentrations provide an objective measure of the substance that actually arrivesin the body by accounting for dosing, bioavailability and inter-individualdifferences in absorption, distribution, metabolism and excretion. Plasmaconcentrations are also needed to compare formulations and substance exposuresbetween different studies and different research groups and should be generated foreach novel formulation.

With regard to ‘microdosing with LSD’, researchers cannot unequivocally conclude that5–20 µg LSD base has no acute subjective effects or does not impair normalfunctioning as noted in the definition of ‘microdosing’. I would argue that thesedoses can produce similar acute subjective effects but to a lesser degree comparedwith clearly psychoactive doses (> 25 µg LSD base). A recent placebo-controlledstudy that used single doses of 5–20 µg LSD tartrate (GMP-formulated, 4-16 µg LSDbase) reported statistically significant acute subjective drug effects (Yanakieva et al., 2019).Studies that used PK-pharmacodynamic modelling (Dolder et al., 2015,2017;Holze et al., 2019) allow the evaluationof plasma concentration/dose–effect relationships of LSD in humans, in whichsubjective effects parallel plasma LSD concentration–time curves. Plasmaconcentrations of LSD at the EC50 for ‘good subjective drug ratings’ arevery low (1 ng/mL). Subjective mean good drug effects of 25% of these full doseshave been reported at an average plasma LSD concentration of 0.4 ng/mL (Dolder et al., 2017;Holze et al., 2019). TheCmax after 100 µg LSD base administration was 1.7 ng/mL (Holze et al., 2019). Basedon the linear PK of LSD, Cmax values of 0.4 ng/mL are likely reached witha 25-µg dose. Based on the available data and ongoing studies, base doses of LSD of25, 10 or 5 µg can be postulated to produce approximately 25%, 10% and 5%,respectively, of the effects of a fully active dose of 100 µg LSD base.Additionally, there is no indication that the acute subjective effects of these verylow to low doses of LSD are qualitatively different from a full dose. Thus, there isno apparent evidence that LSD, which impairs cognition at active doses, wouldmagically enhance concentration when used at lower doses as described by users. Infact, decreases in self-reported concentration and task-measured cognitiveperformance have been reported at high and low plasma LSD concentrations (Dolder et al., 2016;Schmid et al., 2015). Verylow to low doses of 5–20 µg LSD tartrate (‘microdoses’) did not significantly alterself-ratings of perceptual distortion or subjective concentration, but trendstowards impairments were observed (Yanakieva et al., 2019). These effectswould likely become significant in well-powered studies with larger samplesizes.

This raises another important issue. Very low doses of substances will produce verysmall effects. To validly assess small treatment effects, study sample sizes need tobe larger than those that are common in experimental psychiatric research.Additionally, sensitive measures need to be used. Many validated psychiatric scalesmay not be sufficiently sensitive to detect small drug effects, and simpledrug-effect visual analogue scale ratings are likely better. Furthermore, vitalsigns could serve as a simple objective measure of reactivity. However, psychiatricstudies typically have shortcomings with regard to performing vital signassessments. For example, very low to low doses of psilocybin did not alter bloodpressure in one study (Hasler etal., 2004). However, for this finding to be valid, vital signs need to beassessed according to standard operating procedures. This includes the use of scaledinstruments, exactly determined time points after drug administration, time-matchedplacebo control measures and so on. For example, a safety and efficacy studyreported that LSD did not significantly alter blood pressure or heart rate at a doseof 200 µg LSD (Gasser et al.,2014). However, this same dose and lower doses were subsequently shown tosignificantly and relevantly increase blood pressure, heart rate, body temperatureand pupil size in studies that used standardized measures (Schmid et al., 2015;Dolder et al., 2016).

Altogether, we are still in the incipient stage of modern research on psychedelics.Even for relatively high doses of these substances, the data are still scant andinconsistent. Thus, it is too early to make valid conclusions about the effects ofvery low doses of these substances. Currently (Passie, 2019), I see no valid data thatindicate that LSD or psilocybin has either beneficial or adverse effects on healthwhen used repeatedly at low to very low doses (Passie, 2019). This simply needs to bestudied further.

Acknowledgments

I thank Daniel Trachsel, Felix Müller, Patrick Vizeli, Yasmin Schmid andFriederike Holze for comments on the manuscript and Michael Arends formanuscript editing.

Footnotes

Declaration of conflicting interests: The author(s) declared no potential conflicts of interest with respect to theresearch, authorship, and/or publication of this article.

Funding: The author(s) received no financial support for the research, authorship,and/or publication of this article.

References

  1. Dolder PC, Schmid Y, Haschke M, et al. (2015)Pharmacokinetics and concentration-effect relationship oforal LSD in humans. Int JNeuropsychopharmacol19: pii: pyv072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Dolder PC, Schmid Y, Mueller F, et al. (2016) LSDacutely impairs fear recognition and enhances emotional empathy andsociality. Neuropsychopharmacology41:2638–2646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Dolder PC, Schmid Y, Steuer AE, et al. (2017)Pharmacokinetics and pharmacodynamics of lysergic aciddiethylamide in healthy subjects. ClinPharmacokinetics56:1219–1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Gasser P, Holstein D, Michel Y, et al. (2014)Safety and efficacy of lysergic aciddiethylamide-assisted psychotherapy for anxiety associated withlife-threatening diseases. J Nerv MentDis202:513–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Hasler F, Grimberg U, Benz MA, et al. (2004)Acute psychological and physiological effects ofpsilocybin in healthy humans: A double-blind, placebo-controlleddose-effect study. Psychopharmacology172:145–156. [DOI] [PubMed] [Google Scholar]
  6. Holze F, Duthaler U, Vizeli P, et al. (2019)Pharmacokinetics and subjective effects of a novel oralLSD formulation in healthy subjects. Br J ClinPharmacol85:1474–1483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Liechti ME. (2017) Modern clinical researchon LSD. Neuropsychopharmacology42:2114–2127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Mesley RJ, Evans WH. (1969) Infrared identificationof lysergide (LSD). J Pharm Pharmacol21: 713–720. [DOI] [PubMed] [Google Scholar]
  9. Muehlan C, Heuberger J, Juif PE, et al. (2018)Accelerated development of the dual orexin receptorantagonist ACT-541468: Integration of a microtracer in a first-in-humanstudy. Clin Pharmacol Ther104:1022–1029. [DOI] [PubMed] [Google Scholar]
  10. Passie T. (2019) The Science of MicrodosingPsychedelics. London, UK:Psychedelic Press. [Google Scholar]
  11. Preller KH, Herdener M, Pokorny T, et al. (2017) Thefabric of meaning and subjective effects in LSD-induced states depend onserotonin 2A receptor activation. CurrBiol27: 451–457. [DOI] [PubMed] [Google Scholar]
  12. Schmid Y, Enzler F, Gasser P, et al. (2015)Acute effects of lysergic acid diethylamide in healthysubjects. Biol Psychiatry78: 544–553. [DOI] [PubMed] [Google Scholar]
  13. Steuer AE, Poetzsch M, Stock L, et al. (2017)Development and validation of an ultra-fast and sensitivemicroflow liquid chromatography-tandem mass spectrometry (MFLC-MS/MS)method for quantification of LSD and its metabolites in plasma andapplication to a controlled LSD administration study inhumans. Drug Test Anal9: 788–797. [DOI] [PubMed] [Google Scholar]
  14. Yanakieva S, Polychroni N, Family N, et al. (2019) Theeffects of microdose LSD on time perception: a randomised, double-blind,placebo-controlled trial. Psychopharmacology(Berl)236:1159–1170. [DOI] [PMC free article] [PubMed] [Google Scholar]
J Psychopharmacol. 2019 Jul 14;33(9):1039–1057.

Commentary

James Fadiman and Sophia Korb

Corresponding author:

James Fadiman, 1070 Colby Ave, Menlo Park, CA 94025, USA. Email:jfadiman@gmail.com

As microdoses are being used worldwide, this is a timely article. Prudently, theauthors have focused on synthesized psilocybin, as it may soon become moreavailable. As our own research has been entirely anecdotal, and although it includesreports from 51 countries and thousands of individuals, it does not answer any ofthe questions raised here. What our exploratory findings may have done is help raisethe level of interest about the reported negative or positive effects and mechanismsof action. While we have added suggestions and noted a few concerns here, theinvestigations proposed in the article are all necessary and fundamental.

Early on, it could be important to determine if the same weight of psilocybin in amushroom with its other alkaloids (found in over 100 mushroom species) has a similarbehavioural profile to the synthetic. Equally useful, and perhaps eventually asnecessary, would be to replicate the same study with LSD-25 and 1P-LSD. The reasonfor suggesting these equivalence studies is that of the several hundred thousandpeople known to have microdosed, less than 1% of them actually used the GMP gradepsilocybin. If their experiences differ from those using the synthetic substance, agreat deal of otherwise correlative data would need to be put aside.

It seems to us that the worry about cardiac valvulopathy is excessive, given theoverall safety profiles of all of the classic psychedelics described in several ofDr. Nutt’s publications.

The Fen-Phen experiences of heart valve disease development in the 1980s and 1990sinspired new research in identifying the specific 5-HT receptor subtype involved indrug-induced heart valve disease. In the cases of cardiotoxicity and Fen-Phen, both5-HTP2A and 5-HTP2B are implicated. In fact, ‘norfenfluramine was found to be twoorders of magnitude more potent at 5-HT2B and 5-HT2C receptorscompared with 5-HT2A receptors’ (Hutcheson, 2011). While we have someinformation about the affinity of LSD towards different receptors, we have littleinformation about how its unique ‘crystal structure’ may result in different hearthealth outcomes (Wacker et al.,2017).

Affinity does not tell the whole story. The doses of Fen-Phen used in the 1980s and1990s far exceed the doses used in microdosing, seemingly resulting in severalorders of magnitude moreactivity at the receptors. Additionally,in the cases of heart valve disease in Fen-Phen, all of the patients weresymptomatic. Of the thousands of people who microdosed, no one has reported anyheart valve trouble during their period of microdosing, and many people have beenmicrodosing for over a year. All the people we have surveyed with heart problems hadthem before they started microdosing.

The problem, and it is a very real one, is that this article will be reviewed andpopularized over the many different psychedelic and general media sites with varyingdegrees of accuracy. Since it is highly unlikely that large-scale long-term researchnecessary to investigate this possibility will ever be funded, the concern willnever be validated or disproved. There were a number of frightening scenarios raisedabout psychedelics during the earlier research era, about LSD in particular, none ofwhich were ultimately verified. However, their wide circulation led to considerableand unnecessary fears among millions of individuals using these substances. We needbe careful not to create such fears before we have evidence. Given the serious andmultiple warnings given out with most prescription medicine, that there might beunknown side effects to microdoses is to belabour the obvious.

We would look for an expansion of the receptor research (Question 7). It would be agreat gift to all psychedelic research if studies could begin to go beyond measuring5-HTP2A receptors and include, at least, the mTOR and TrkB signalling pathways aswell (Ly et al.,2018).

A question to investigate is how the well-described accelerated neural plasticity ofa number of psychedelics at high doses is diminished or intensified through periodicmicrodosing. Early speculation by Kornfeld (Kornfeld and Fadiman, 2013) has now beenartfully demonstrated by the work of Ly’s group (Ly, et al. 2018). This seems to be anespecially fruitful area, given the growing body of research linking neuralplasticity with both mental illness and recovery.

We are encouraged that in Question 8, the authors went beyond 5-HTP2A receptors andlooked at peripheral tissues with doses well below behavioural thresholds as well.We hope the number and kinds of physical systems evaluated for effects continue toexpand. For example, although it is now generally accepted that the number ofneurons that exist outside of the brain exceeds the number within it, psychedelicresearchers have not yet developed research methods to measure changes in gutneurons due to the effects of psychedelics or how those changes affect human biologyand behaviour.

Finally, the issues of dose and schedule remain critical. While many pharmaceuticalshave a given activity and that more or less of a dose leads to more or less of thesame activity, this is not true for psychedelics at higher doses and far less so formicrodoses. One size does not fit all, so that the identical dose, howevercalculated, will not yield the same results across individuals. This may be a hardproblem, especially given the few research models popular in pharmacology ingeneral. As for the effects of multiple doses over time, there has never been asuggested protocol that did not include days without dosing, in contrast to almostall psychiatric medications that warn of potential serious health issues if even asingle dose is missed. For this and other reasons, psychedelics do not fit neatlyinto much of current psychopharmacology and thus need to be researched.

Our few concerns aside, these research proposals are a major step forward forpsychedelics in general and microdoses in particular.

References

  1. Hutcheson JD, Setola V, Roth BL, et al. (2011)Serotonin receptors and heart valve disease: It was meant2B. Pharmacol Ther132:146–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Ly C, Greb AC, Cameron LP, et al. (2018)Psychedelics promote structural and functional neuralplasticity. Cell Rep23:3170–3182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Kornfeld A, Fadiman J. (2013) Psychedelic inducedexperiences. In: Friedman HL, Hartelius G. (eds).The Wiley-Blackwell Handbook of TranspersonalPsychology. New York:Wiley/Blackwell, pp.352–366. [Google Scholar]
  4. Wacker D, Wang S, McCorvy JD, et al. (2017)Crystal structure of an LSD-bound human serotoninreceptor. Cell168:377–389.e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
J Psychopharmacol. 2019 Jul 14;33(9):1039–1057.

Commentary

Torsten Passie

Corresponding author:

Torsten Passie, Hannover Medical School, Psychiatry, Böttcherstr. 11, Hannover,D-30419, Germany. Email:dr.passie@gmx.de

Psychedelics and creativity

I would recommend not mentioning just these very few anecdotal cases in respectto creativity and psychedelics. There are some good studies and reviews aboutthe subject (Hartmann,1969) on 40 prominent painters at the German Max Planck Institute ofPsychiatry in Munich and the review on the subject byKrippner (1985).

When it comes to the very few studies looking for low doses of psychedelics andcreativity (review inPassie, 2019), no study revealed any significant/relevant effectsunder controlled scientific conditions (e.g.McGlothlin et al., 1967). The study ofProchazkova et al.(2018) claimed (under weakly controlled conditions in respect todosing and environment) increased lateral thinking, which has been discussed asa marker of creativity. This study employed doses of psilocybin that were abovethe perceptible level (4–8 mg p.o.). However, increased lateral thinking doesnot mean that the drugged subjects have shown increased creativity in a validsense, e.g. creating more original painting.

Definitions: Microdosing and minidosing

I do not agree with the authors’ narrow definition, since it does not reflectfully what is used in the literature and the appropriate Internet entries. Ithas been proposed that the term ‘mini-dosing’ could be used to separate theapproach of taking small perceptible doses. It is also clear that many authorsand Internet entries suggest the practice of just taking one dose at a timerather than a few ones consecutively as, for example, seen with the Fadimanscheme. This is also valid for taking a tenth or a twentieth of a usual dose.However, the issues related to definitions point towards the question/definitionof what is considered a ‘full dose’. In the case of LSD, some authors reasonablyargue that 150 µg is a full dose (especially in females), whereas othersconsider 250 µg a full dose. This is a significant issue, because 15 µg isusually not perceptible by most subjects, but a dose of 25 µg is for mostsubjects (as shown in some scientific studies). Therefore, the definition has tobe sharpened before scientific consensus can be reached and the evidence fromso-called microdosers disseminated on the Internet as well as studies ofanecdotal evidence (e.g.Johnson, 2018), which suffers from such inaccuracies, can be takenseriously.

Dosing of dried mushrooms

Plant/fungal material is generally quite unreliable for calculating a dose. I donot agree with the author’s statement that 3.5 gP. cubensis is‘a usual recreational dose’. Most recreational users take 1 to 2.5 g as arecreational dose, which is also recommended in most books in the field. From myexperience, and the research studies ofAbramson and Rolo (1967), I wouldstate that a dose of psilocybin below 3 mg is below the perceptible range.Usually, doses above this level can become apparent. For example,Prochazkova et al.(2018) used 4–8 mg psilocybin, i.e. more than a microdose, thus, moreconsistent with the definition of what might be considered a minidose.

The most used dosing regime and effects of micro- and minidosing

It can be easily seen in Internet entries that most subjects who take microdosesrecreationally for ‘bettering performance’ take doses that give them someperceptible effects. Even a microdosing proponent like Paul Austin recommendsdoses where you can feel/perceive some alterations to some extent. How would youbetter your performance if nothing can be felt from a dose?

Following my comprehensive research into this topic (Passie, 2019), I have never come acrossanything about a ‘workaholic approach’ (dosing during weekdays, but not onweekends) as suggested by the authors. This also does not make much sense form apharmacological point of view, because tolerance to LSD develops very quickly.Be reminded, that the US military has dosed soldiers with increasing daily dosesto try to make them ‘immune’ to LSD’s effects (Ketchum, 2006).

I think that a minidose (e.g. 20 to 50 µg LSD), in contrast to a microdose (whichI define as something below 20 µg LSD, e.g. 5–15 µg), makes a significantdifference in terms of recreational as well as scientific studies as itdefinitely alters psychological functioning and the cognitive system.

However, this alteration is not in any way equivalent to stimulants like Ritalinor amphetamine as is sometimes reported anecdotally. It is more a dissociationfrom the environment and the person itself. Cognitive abilities have been provento be compromised in many studies with LSD and psilocybin employing a very lowdose range (Passie,2019). It is also important to register these (potentially)distracting effects. There are also a few scientific reports of people who havebeen given very low doses of LSD for treating depressive mood. These hadnegative results with very few patients experiencing a small improvement.

What is plausible (and has been experienced by this author) is that a minimalsympathomimetic effect (for sure not compatible with any sort of usualstimulant!), which might be still there up to 20 hours after a 10- to 15-µg doseof LSD, can cause problems with falling asleep, especially in sensitive persons.It is of interest how long it takes following intake for effects to occur. Onecould think of a train of effects induced in the organism, which is pushed onand may influence the organism even after virtually all of the substance hasleft the organism.

I definitely do not see the Fadiman protocol (5–15 µg LSD every third day) as themost used approach. It might be viewed as the most widely known, but by far (!)the most ‘microdosers’ use one occasional dose, not a regular intake. This alsomakes it very questionable what effects can be/are felt or not, especially whenit comes to taking 10 µg just a few times per year (which is apparently whatmost users do). As Fadiman’s coworker on his more or less systematic Internetsurveys, Sophia Korb has mentioned in a lecture (conference ‘BeyondPsychedelics’, Prague 2018) that they know of just three persons who have dosedregularly (according to the Fadiman protocol) for more than 3 weeks. These threesubjects were terminal cancer patients and felt quite normal up to day 50.Between days 50 and 60 they all became much more psychologically labile, i.e.having larger mood changes (in the positive as well negative direction, withdaily fluctuations), as measured using the PANAS scale.

To my mind, the study published byHorsley et al. (2018) does not have anyseriously calculable implications for humans. Its limitations should bediscussed.

Possible alterations of gene expression and receptor proteins

There are serious doubts that the repeated doses of LSD, which have been used inrodents, are comparable to microdoses in humans. I am not an expert oninterspecies scaling, but, for example, (just by simplified arithmetic) thestudies byMartin et al.(2014) have used doses which are 12,000 times higher than a microdosein humans. According to a recent review (Sharma et al., 2009), it appears notto be congruent with scientific data to state that the dose used byMartin et al. (2014)is in any way comparable to a microdose in humans. Therefore, to date we knownothing about possible changes in gene expression induced by regular LSD intakein humans.

I doubt that the gene/BDNF changes which were found with very high daily doses inanimals can be scaled up to humans using microdoses every few days. Issues ofadaption and tolerance should be discussed in this respect.

Receptors are proteins. These proteins and others might be altered by repeatedintake of, e.g. LSD, even in very low doses (Buchborn et al., 2016). Even if thisis somewhat speculative, it seems probable.

On the possible induction of cardiovascular valvopathy

In respect to a possible induction of cardiovascular valvulopathy by chronic2-HT2R activation, it is worth mentioning that the studies ofBender and Sankar(1968) in the 1960s involved doses of 100 µg LSD for up to 35 monthson a daily basis without any observable damage. However, their methods ofinvestigation might not have been sensitive enough to detect damage. It is alsotrue that just a very small part of the patient population taking ergotcompounds (e.g. methysergide) do in fact develop valvulopathy. It is also worthmentioning that if a valvulopathy is detected in a patient, in all cases itdisappears within a short time after stopping the medication. There is just onecase documented in the literature where surgery was necessary (Graham, 1967).

Footnotes

Declaration of conflicting interests: The author(s) declared no potential conflicts of interest with respect to theresearch, authorship, and/or publication of this article.

Funding: The author(s) received no financial support for the research, authorship,and/or publication of this article.

References

  1. Abramson H, Rolo A. (1967) Comparison of LSD withmethysergide and psilocybin on test subjects. JAsthma Res3: 81-96 [DOI] [PubMed] [Google Scholar]
  2. Bender L, Sankar DVS. (1968) Chromosome damage notfound in leukocytes of children treated with LSD-25.Science159: 749. [DOI] [PubMed] [Google Scholar]
  3. Buchborn T, Grecksch G, Dieterich DC, et al. (2016)Tolerance to lysergic acid diethylamide: overview,correlates, and clinical implications. In: Preedy VR. (ed.) Neuropathology of Drug Addictions andSubstance Misuse, Volume 2NewYork: Academic Press,pp. 846–858. [Google Scholar]
  4. Graham JR. (1967) Cardiac and pulmonaryfibrosis during methysergide therapy for headache.Am J Med Sci254: 1–12. [DOI] [PubMed] [Google Scholar]
  5. Hartmann RP. (1969) Malerei aus Bereichen desUnbewußten. Künstler experimentieren unter LSD.Köln:Dumont. [Google Scholar]
  6. Horsley RR, Palenicek T, Kolin J, et al. (2018)Psilocin and ketamine microdosing: Effects of subchronicintermittent microdoses in the elevated plus-maze in male Wistarrats. Behav Pharmacol29: 530–536. [DOI] [PubMed] [Google Scholar]
  7. Johnson MW, Griffiths RR, Hendricks PS. (2018) The abuse potential ofmedical psilocybin according to the 8 factors of the ControlledSubstances Act. Neuropharmacology142: 143–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Krippner S. (1985) Psychedelic drugs andcreativity. J Psychoactive Drugs17: 235–246. [DOI] [PubMed] [Google Scholar]
  9. McGlothlin WH, Cohen S, McGlothlin MS. (1967) Long lasting effects ofLSD on normals. Arch Gen Psychiatry17: 521–532. [DOI] [PubMed] [Google Scholar]
  10. Martin DA, Marona-Lewicka D, Nichols DE, et al. (2014)Chronic LSD alters gene expression profiles in the mPFCrelevant to schizophrenia.Neuropharmacology83: 1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Passie T. (2019) The Science of MicrodosingPsychedelics. London:Psychedelic Press. [Google Scholar]
  12. Prochazkova L, Lippelt DP, Colzato LS, et al. (2018)Exploring the effect of microdosing psychedelics oncreativity in an open-label natural setting.Psychopharmacology235:3401–3413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Sharma V, McNeill JH. (2009) To scale or not to scale:the principles of dose extrapolation. Br JPharmacol157:907–921. [DOI] [PMC free article] [PubMed] [Google Scholar]
J Psychopharmacol. 2019 Jul 14;33(9):1039–1057.

Responses to the microdosing critique reviews

David Nutt1 and Kim PC Kuypers2

1Neuropsychopharmacology, Imperial College London, London, UK

2Department of Neuropsychology and Psychopharmacology, Faculty ofPsychology and Neuroscience, Maastricht University, Maastricht, The Netherlands

Corresponding author:

Kim PC Kuypers, Department of Neuropsychology and Psychopharmacology, Faculty ofPsychology and Neuroscience, Maastricht University, Maastricht, The Netherlands.Email:k.kuypers@maastrichtuniversity.nl

We would like to thank the four respondents for their thoughtful and insightfulcontributions to this work. We should also like to make it clear that in addition totheir commentaries they also made significant observations on our critique pointingout some errors and omissions that we have taken account of to improve themanuscript. Please read the critiques with this in mind as some of their commentshave now been dealt with in our text.

We were pleased that all felt it a significant contribution to the field. They alsohave significantly enlarged the scope of our review with their very helpfulcontributions to aspects such as current practice, impact on creativity, value andrelevance of rodent studies to mention but a few. Where necessary we mention theseissues below though much of their comments and additional insights and referencesreflect their extensive expertise in this field, and we are very grateful for theseas they significantly improve the scientific scope and value of this critique.

A couple of their points need addressing, starting with terminology. We like thedistinction made between minidose and microdose and would be happy to see this inuse. A minidose being one that has a detectable effect whereas a microdose does not.Of course what the actual drug amount is for each is uncertain, and to what extentthe psychoactive effect of a minidose might be ‘allowed’ to have and still be called‘mini’ would need more consideration, but until then, Passie’s suggestion of 5–15 µgLSD makes sense, though Liechti’s observations need also to be taken intoaccount.

We accept that our focus was on psilocybin and the reason for that was simple – it isalmost certainly going to be the first serotonergic psychedelic made available as amedicine. For this reason, it is also currently the only psychedelic made to GMPstandards and approved for medical trials with patients. This means that psilocybinis the best choice for microdosing research, certainly in patients and also forvolunteer studies in countries where GMP production is required for healthyvolunteers. But we fully agree that LSD should also be studied given its widespreaduse by the microdosing community. For those who chose to use this psychedelic thenFadiman’s and Passie’s approaches would make a good starting point.

The problem with all psychedelic microdosing studies is how to do it legally andethically, and this is the big question that needs answering. Maybe a change in theregulations to exclude microdoses from the list of controlled drugs could be sought?After all, when used singly they are below the threshold for subjective effects andso are not psychoactive.

Overall, we are pleased with the results of our efforts and those of the reviewers.Microdosing is a current phenomenon whose value and safety are uncertain. Muchresearch is needed to properly evaluate the personal psychological and healthclaims. We hope that this set of papers will give impetus to this research and alsoset it in a solid framework.


Articles from Journal of Psychopharmacology (Oxford, England) are provided here courtesy ofSAGE Publications

ACTIONS

RESOURCES


[8]ページ先頭

©2009-2025 Movatter.jp