Skip to main content
Full access
Reviews and Overviews
Published Online: 20 December 2024

Benefits and Challenges of Ultra-Fast, Short-Acting Psychedelics in the Treatment of Depression

Publication: American Journal of Psychiatry

Abstract

Unlike classical antidepressants, psychedelics such as psilocybin have been shown to induce a rapid antidepressant response. In the wake of this development, interest has emerged in ultra-fast, short-acting psychedelics such as 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT) and N,N-dimethyltryptamine (DMT) with the expectation that these can produce rapid antidepressant effects following an intense but brief psychedelic intervention. The current paper reviews the clinical pharmacology of 5-MeO-DMT and DMT and their potential benefits and challenges in the treatment of depression. Both compounds display affinities for a variety of monoamine receptors and transporters, but mostly so for serotonergic (5HT) receptors, including 5HT1A and 5HT2A. Early clinical trials in small samples have shown that short interventions (15–30 min) with 5-MeO-DMT and DMT are safe and well tolerated and can induce marked improvement in symptoms of depression within 24 hours that sustain for at least 1 week. Data on long-term efficacy are currently scarce but do suggest a prolongation of the treatment response. Potential benefits of these treatments include flexible, single day dosing regimens, achievement of treatment efficacy independent from integrative therapy, and ease of clinical implementation. Future challenges include establishing the duration of the antidepressant effect and strategies on how to sustain the antidepressant response, optimization of treatment delivery parameters, and a mechanistic understanding of the clinical response. Acceptance of ultra-fast, short-acting psychedelics will depend on future randomized, placebo-controlled trials with a focus on replication, duration and maintenance of antidepressant efficacy in large patient samples.
Classic antidepressants often take weeks to months of daily dosing to achieve response and remission, while patients remain symptomatic, functionally impaired and at an increased risk of suicide during this initial treatment period (1). While full therapeutic effects eventually manifest in a large portion (i.e., 67%) of depressed patients across cumulative treatment steps, a considerable number of patients do not have satisfactory improvement despite long-term treatment (2). Importantly, a rapid antidepressant response might be critical for achieving long term clinical stability, as clinical improvement during the first few weeks of treatment with antidepressants has been associated with a reduction in neurobiological harms from recurrent, enduring depression (35). Therefore, treatments that can induce a rapid improvement of depressive symptoms, within hours or days, and whose effects can be sustained, would have an enormous impact on public health.
Psychedelics have shown promise for inducing fast and sustained symptom alleviation in depressed patients and are currently under development for the treatment of depression. A phase II, placebo controlled clinical trial in 233 participants with treatment resistant depression demonstrated significant increases in the number of responses and remissions after a single oral dose of psilocybin (25 mg) at 3 weeks, though not at 3 months (6). Similarly, treatment efficacy has been reported between 1 and 6 weeks after one or two oral doses of psilocybin in smaller trials (710), with effect sizes comparable to those observed after daily treatment with a classical antidepressant (8). The potential success of psilocybin in the treatment of depression has sparked an interest in ultra-fast and short-acting psychedelics such as 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT) and N,N-dimethyltryptamine (DMT). The latter are currently under development with the expectation that these can produce rapid antidepressant effects following an intense but brief psychedelic intervention. 5-MeO-DMT and DMT belong to the tryptamine class of psychedelics, just like psilocybin (11). Yet, where the psychedelic effects of oral psilocybin come on slow (30–60 minutes) and last long (4–6 hours) (12), those of inhaled 5-MeO-DMT and intravenous DMT occur instantly and potentially only last for 15–30 minutes (13, 14). 5-MeO-DMT and DMT are typically administered through routes that avoid the first pass-metabolism (e.g., inhalation, intravenous [IV], intramuscular, intranasal, insufflation), as oral formulations are not rendered psychoactive because of rapid degradation by monoamine oxidase (MAO) (1517). The short duration of the psychedelic experience of these formulations offers the potential of flexible dosing and increased control over the duration of treatment. For example, when administering DMT intravenously the experience can last for minutes or hours, depending on the rate of infusion (18). Short duration treatments with psychedelics might therefore offer more scalable clinical applications as compared to long duration treatments with psychedelics. The current paper will review the clinical pharmacology and the potential benefits and challenges of ultra-fast, short acting psychedelics in the treatment of depression.

Phenomenology and Pharmacology of 5-MeO-DMT and DMT

5-MeO-DMT and DMT are naturally occurring tryptamines that can be found in a range of Amazonian plant species (1921). 5-MeO-DMT has also been identified as the primary psychoactive component of the parotoid gland venom of Bufo alvarius, the Sonoran desert toad (22). The psychedelic experience induced by 5-MeO-DMT and DMT is known to differ. DMT has been reported to elicit a dynamic and rich visual experience, sometimes accompanied with sentient encounters or the sense of visiting other worlds (17, 2326). Subjective experiences with 5-MeO-DMT are often reported to be emotional and devoid of any visuals other than the experience of a “profound white light” (13, 27, 28). Both psychedelics have been associated with a feeling of “ego death” that is characterized by a dissolution of one’s sense of self and, in the case of 5-MeO-DMT, loss of memory (13, 17, 27, 28). Both 5-MeO-DMT and DMT can produce spiritual experiences with altered-self-awareness that are described as overwhelming and profound (13, 2325, 27, 28).
Both compounds display binding affinities for a wide range of monoamine receptors and transporters, but mostly so for serotonergic receptors. Receptor binding affinities of 5-MeO-DMT and DMT are listed in Table 1 and discussed below. It is suggested that the primary target of psychedelics, including 5-MeO-DMT and DMT, which underlies their psychedelic effect, is the serotonin 5HT2A receptor (11, 13, 17, 29), but contributory roles of additional receptors, such as 5HT1A, cannot be excluded (17, 3032). Receptor binding studies indeed have shown a stronger selectivity of 5-MeO-DMT for the 5HT1A receptor as compared to the 5HT2A receptor, suggesting a selectivity ratio that is substantially higher than for DMT and other psychedelics (30, 3234). Computational docking analysis further confirmed a stronger interaction of 5-MeO-DMT at the 5HT1A receptor compared to the 5HT2A receptor (32). The strong affinity of 5-MeO-DMT for 5HT1A might be very relevant as 5HT1A agonists have been implicated in the treatment of anxiety and depression (35). The functional relevance of the 5HT1A receptor for the behavioral effects of 5-MeO-DMT has been suggested in rodent studies which have shown a stronger reduction of drug discrimination effects under 5-MeO-DMT in the presence of a 5HT1A antagonist as compared to a 5HT2A antagonist (3638). Likewise, changes in cortical activity following 5-MeO-DMT in normal and 5HT2A knock-out mice were reversed by 5HT1A antagonists (39, 40). Agonism at inhibitory 5HT1A receptors is often antagonistic to agonism of 5HT2A receptors which elicit excitatory effects (35, 41, 42). However, the opposite has been shown in that 5HT1A blockade increased 5-MeO-DMT induced head twitch responses in mice, a preclinical measure of hallucinogenic effects (32). In humans, blockade of the 5HT1A receptor increased the intensity of the psychedelic experience induced by DMT (23) suggesting a modulatory interplay between the 5HT1A and 5HT2A receptors during a psychedelic experience. Yet, the functional and contributory role of the 5HT1A receptors to the phenomenology of the psychedelic experience as observed with short-acting psychedelics remains poorly understood and needs further exploration. Likewise, the contributory roles of dopaminergic, adrenergic and sigma-1 receptors (30, 33, 43) and possibly trace-amine-associated (TAAR) receptors (44) to the acute psychedelic effects of 5-MeO-DMT and DMT are not clear.
TABLE 1. Binding affinities (Ki values in nanomolar [nM]) of 5-MeO-DMT (33) and DMT (30, 33) for monoamine receptors and transporters, and sigma receptors, as determined in cloned human cell linesa
Receptor5-MeO-DMTDMT
Serotonin (5HT) receptors
 5HT1A1.9183
 5HT1B7.4129
 5HT1D6.339
 5HT1E360.2517
 5HT2A2011127
 5HT2B3884184
 5HT2C538360
 5HT5A276.62135
 5HT635.5464
 5HT73.9206
Dopamine (D) receptors
 D179.5271.1
 D2356.2>10000
 D3497.6>10000
 D43120>10000
 D5>10000>10000
Norepinephrine receptors
 α1A79.51745
 α1B356.2973.7
 α2A497.61561
 α2B3120257.7
 α2C>10000258.6
 β22032>1000
Sigma receptors
 σ1>100005209
 σ2>10000>10000
Transporters
 SERT20326000
 DAT>10000>10000
 NET2859>10000
a
SERT=serotonin transporter; DAT=Dopamine transporter; NET=norepinephrine transporter.

Dose-Finding Studies and Safety

A range of dose-finding studies in healthy volunteers has been conducted to determine safety of 5-MeO-DMT and DMT, often with the aim to determine a dosing regimen for future trials in patient populations. These trials are discussed below and summarized in Table 2. Typically, the route of administration and the dosing regimen employed in these studies has varied. In the case of 5-MeO-DMT most research so far has focused on inhaled (13, 45) and intranasal (46) formulations, but IV formulations are currently under development as well (13). The first study of its kind (45) aimed to assess the impact of a novel vaporized 5-MeO-DMT formulation (GH001) administered via inhalation as single doses of 2, 6, 12, or 18 mg, and in an individualized dose escalation regimen, on the safety, tolerability, and the psychoactive experience in five groups of healthy volunteers. The individualized dosing regimen (IDR) consisted of 6, 12, and 18 mg, where at least one and up to three doses of 5-MeO-DMT interspaced at 3 hours were given on a single day. The decision to move on to the next dose was guided by an evaluation of whether the participant achieved a peak psychedelic experience (i.e., mean subjective ratings of intensity, profoundness and loss of control equal or over 75% on a peak experience scale), at the previously administered dose. If no peak experience was achieved, the next dose was administered, after consultation with and consent from the participant. The aim of the individualized dose escalation was to maximize the intensity of the psychedelic experience across participants, as previous research with other psychedelics has indicated that the stronger the intensity, the better the therapeutic outcome (47, 48). Single doses of 5-MeO-DMT produced instant, short-acting, dose related increases in the intensity of the psychedelic experience ratings that lasted for 15–20 minutes. Up to 50% of the participants in the single dose regimens reported a peak psychedelic experience, whereas that number rose to 100% of the participants that received an individualized dose escalation of 5-MeO-DMT. The duration of the psychedelic experience as estimated by the participants was about 18 minutes. Psychedelic experiences following single, intranasal administrations (range 1–12 mg) of 5-MeO-DMT (BPL-003) appeared instantly and lasted up to 90 minutes (46). The onset, length, and intensity of the experience closely followed plasma concentration time curves of 5-MeO-DMT. Yet, the intensity of the psychedelic experience following a single intranasal dose varied considerably between individuals, as was also shown after inhalation of 5-MeO-DMT (45)
TABLE 2. Duration of the psychedelic experience, dose regimens and route of administration as reported in clinical dose findings studies with 5-MeO-DMT and DMT in healthy volunteers
ReferenceDesignRouteDose (N)Duration of Psychedelic Experience
5-MeO-DMT
 Reckweg et al., 2021 (45)Single-blind, ascending dosesInhalationSingle doses: 2, 6, 12, 18 mg (N=4–6 per dose); IDR 6+12+18 mg, spaced 3 hours apart (N=4)Up to 20 min per dose
 Rucker et al., 2024 (46)Double-blind, placebo-controlledIntranasalSingle ascending doses: 1, 2.5, 4, 6, 8, 10, 12 mg (N=4–5 per dose), placebo (N=2 per dose)Up to 90 min per dose
DMT
 Strassman et al., 1994 (54); Strassman and Qualss, 1994 (24)Double-blind, placebo-controlled, crossoverIV—infusion over 30 sec0.05, 0.1, 0.2 and 0.4 mg/kg DMT fumarate and saline placebo (N=11)20–30 min
 Strassman et al., 1996 (53)Double-blind, placebo-controlled, crossoverIV—infusion over 30 sec0.3 mg/kg DMT fumarate or saline placebo, four times, at 30 min intervals (N=17)Up to 30 min per dose
 D’Souza et al., 2022 (49)Open-label, ascending dosesIV—intravenous push over 30–60 seconds0.1 and 0.3 mg/kg spaced >48 hours apart (N=3)20–30 min per dose
 Luan et al., 2023 (52)Placebo-controlled, crossover, ascending dosesIV—bolus+continuous infusion over 30 min6 mg+0.63 mg/min (N=6); 10 mg+1.05 mg/min (N=10), 14 mg+1.46 mg/min (N=9), 18 mg+1.88 mg/min (N=6)Up to 42 min
 Vogts et al., 2023 (18)Double-blind, placebo-controlled, crossoverIV—bolus+continuous infusion over 90 minLow infusion (0.6 mg/min), high infusion (1 mg/min), low bolus+low infusion (15 mg+0.6 mg/min), and high bolus+high infusion (25 mg+1 mg/min) DMT hemifumarate (N=27)Up to 105 min
 Falchi-Carvalho et al., 2024 (55)Open-label, ascending dosesInhalation5+20 mg, 7.5+30 mg, 10+40 mg, 12.5+50 mg or 15+60 mg DMT free base (plant extract), spaced 2 hours apart (N=5–6 each)Up to 20 min per dose
 Aicher et al., 2023 (56)Double-blind, placebo-controlled, crossoverOxiOridisperable (harmine)+instranasal (DMT)100 mg harmine+10 intermittent DMT doses (plant extract) of 10 mg (N=31), spaced 15 min apart4–5 hours
 Good et al., 2023 (50); James et al., 2023 (51)Parallel, double-blind, placebo- controlledIV—two phased continuous infusion for up to 11 minutes6+3, 6+6, 6+11 or 6+15.5 mg DMT free base (N=6 each); placebo (N=8)30–40 min
In the case of DMT, most studies have focused on IV-formulations (18, 4952), the oldest dating back to 30 years ago (24, 53, 54). One study has also focused on inhaled administration (55) and another on the combination of oral DMT and a harmaline MAO inhibitor (56). The duration of the psychedelic experiences varied with the IV infusion rate and between dose formulations. In the case of IV-administration, the rate of infusions varied between 30 seconds and 90 minutes. Short duration infusions of up to 30 or 60 seconds (24, 49, 53, 54) produced a psychedelic experience that lasted up to 30 minutes, while infusion of 11 minutes (50, 51) and 90 minutes (18) extended the experience to 40 and 105 minutes, respectively. Inhalation of two subsequent doses of DMT (55) produced psychedelic effects for up to 20 minutes per dose, whereas 10 intermittent oral doses of DMT combined with a harmaline MAO inhibitor to inhibit degradation of DMT produced psychedelic effects that lasted up to 4–5 hours (56). The total dose of DMT administered in these healthy volunteer studies has also varied considerably because of differences in route and rate of administration as is shown in Table 2.
Adverse events following acute administrations of 5-MeO-DMT and DMT in clinical trials were generally mild and resolved spontaneously (18, 24, 25, 45, 4951, 53, 54, 57, 58). Both compounds produced transient increments in blood pressure and heart rate during the psychedelic experience. The most frequently reported adverse events included anxiety, nausea and headache. Subacute effects of 5-MeO-DMT, occurring in the days following dosing, included flashbacks or reactivations, i.e., brief (in the order of seconds) and benign sensory re-experiences of 5-MeO-DMT (45, 59). Reactivations following 5-MeO-DMT have also been reported in non-clinical settings (6062). These studies mostly suggest a neutral or positive valence of this phenomenon (61, 62) and a lower prevalence after intramuscular as compared to inhaled use (60). The mechanism underlying sub-acute reactivations is unknown. It has been speculated that reactivations could reflect temporary epileptoform activity related to the distinct selectivity of 5-MeO-DMT for the 5HT1A receptor (63). However, current data suggest a beneficial role of 5HT1A stimulation in most preclinical epilepsy models (64) and patients with temporal lobe epilepsy (6467). Preclinical work in cats showed that 5-MeO-DMT significantly suppressed photically induced myoclonus, suggesting antiepileptic properties (68). It might be the case however that the phenomenon of reactivations is more typical for 5-MeO-DMT than for other psychedelics.
Although psychedelic experiences as such are typically not reported as adverse events in clinical trials it should be noted that both 5-MeO-DMT and DMT can produce intense negative emotions or adverse physical effects as part of the experience (13, 26, 29, 69). Intense psychedelic experiences always carry the risk of producing psychological distress, although these are also often reported to be cathartic (70). This appears to be the case for psychedelics in general, in that intense psychedelic experiences, even when negative, are associated with enduring increases in well-being (71) or a reduction in psychopathology (48). It is noteworthy in this regard that individuals that were exposed to an intense psychedelic experience with 5-MeO-DMT or DMT in clinical trials scored relatively low on the challenging experience questionnaire (45, 56, 59). Psychological experiences with 5-MeO-DMT and DMT are however difficult to predict, as with any psychedelic, and rare psychotic reactions can occur (7274). The risk of psychosis or negative experiences should be reduced with psychiatric screening and the exclusion of patients with a predisposition towards psychotic illnesses from clinical treatment with psychedelics, by careful preparation and anticipation of the psychedelic experience, and by establishing trust and rapport between the session monitor and study participants in a safe environment (75).

Indications of Therapeutic Efficacy

Preliminary indications for the therapeutic potential of 5-MeO-DMT and DMT mostly come from prospective observational studies on the naturalistic use of smoked 5-MeO-DMT (synthetic or toad venom) and DMT containing brews such as ayahuasca. These studies demonstrated immediate (within 24 hours) and lasting improvements (i.e., up to 4 weeks) relative to baseline, in self-reported ratings of depression, anxiety, stress, mindfulness-related capacities, and satisfaction with life, after a single administration of the substance in healthy volunteers (21, 7681) and depressed patients (82). Naturalistic studies also come with notable limitations, such as the lack of dose information, lack of control groups, and high drop-out rates. Yet, persisting improvements in mood and attitude towards life have also been observed in healthy volunteers at 1 and 4 months after participation in a placebo-controlled trial with intermittent intranasal dosing of DMT (100 mg) combined with harmine (56). Likewise, a placebo-controlled, prospective study in healthy volunteers reported improvements in ratings of depression at 1–2 weeks following IV DMT doses (7–20 mg) (83).
To date, a limited number of early clinical trials have assessed the potential for antidepressant efficacy of 5-MeO-DMT and DMT in small samples of depressed patient populations. These are discussed below and summarized in Table 3. The first placebo controlled trial was conducted in 29 patients suffering from treatment resistant depression (TRD) and demonstrated the clinical utility of ayahuasca to rapidly decrease symptoms of depression by more than 50% at 1 and 7 days after treatment (84) as rated with the Hamilton Depression Rating Scale (HAM-D). On day 7, 43% of the patients were in remission. It should be noted however that therapeutic indications observed for ayahuasca brews do not exclusively generalize to DMT. Ayahuasca brews also contain β-carboline alkaloids such as harmine, harmaline, and tetrahydroharmine (85). The latter are monoamine oxidase inhibitors (MAOI) allowing DMT to reach the central nervous system for a prolonged period of time (86) and that have an accepted clinical utility as antidepressants by themselves (87, 88).
TABLE 3. Summary of study designs, support models, and primary outcomes in early clinical trials with 5-MeO-DMT and DMT in depressed patient populations, as compared to a representative psilocybin trial in depressiona
ReferenceDisorderDesignPreparationDoseDose Session DurationIntegrationTherapy SupportModel(Primary) Outcome
Psilocybin
 Goodwin et al., 2022 (6)TRD (N=233)Randomized, double-blind, placebo controlledTwo sessions of 1 hour10 or 25 mg (oral)6–8 hours2.5 hours2 therapistsNondirective psychological support (manualized)MADRS −12 (25 mg), 29% in remission; −7.9 (10 mg), 9% in remission, at week 3
5-MeO-DMT
 Reckweg et al., 2023 (59)TRD (N=16)Open-label, single doses and IDRInhaled single doses 12, 18 mg (N=4 each); IDR 6+12+18 mg, spaced 3 hours apart (N=8)20 min per doseStandard medical careMADRS −24.4, 87.5% in remission (IDR) at day 7
DMT
 Palhano-Fontes et al., 2019 (84)TRD (N=29)Randomized, double-blind, placebo controlledAyahuasca brew 0.36 mg/kg DMT+1.86 mg/kg harmine+0.24 mg/kg harmaline+1.2 mg/kg tetrahydroharmine5–6 hoursNondirective support as neededHAM-D −14.4, 43% in remission at day 7
 D’Souza et al., 2022 (49)MDD (N=7)Open-label, ascending doses45 min0.1 and 0.3 mg/kg IV spaced >48 hours apart15–30 min2 hours debriefingNondirective psychological supportHAM-D −4.5 at day 1
 Falchi-carvalho et al., 2024 (89)TRD (N=6)Open-label, ascending dosesPsychological preparation session15+60 mg (free base) inhaled spaced 90 min apart2 × 15 minIntegration session after each dose and at day 1 and day 7Nondirective psychological supportMADRS −22, 66.67% in remission at day 7
a
HAM-D=Hamilton Depression Rating Scale; MADRS=Montgomery–Åsberg Depression Rating Scale; MDD=major depressive disorder; TRD=treatment resistant depression.
The first exploratory study (49) to assess the therapeutic potential of DMT in patients (N=7) with major depression disorder (MDD), reported a significant mean decrement (−4.5 HAM-D score) in symptoms as compared to baseline, the day after an IV DMT administration of 0.3 mg/kg (about 21 mg for a person of 70 kg). The antidepressant effect was relatively small, only a 19% symptom reduction from baseline, possibly because the doses were relatively low. A larger antidepressant effect was reported in a Brazilian trial in which patients suffering from TRD (N=6) where treated with two subsequent doses of inhaled DMT (15 mg and 60 mg) (89). Response rates (i.e., a reduction of 50% or more from baseline) at 1 and 7 days after treatment were 83.33% as assessed with the Montgomery–Åsberg Depression Rating Scale (MADRS). At 1 month after treatment the response rate was 66.67%. Remission rates at 7 days and at 1 month after treatment were 66.67% and 50% respectively. None of these studies provided pharmacokinetics of DMT, which makes it difficult to compare the bioavailability of IV and inhaled formulations. Subjective ratings of the psychedelic experiences in both DMT studies suggest however that the intensity was higher after the inhaled dose formulation (84.15%) as compared to the IV formulation (55.83%) as assessed on visual analogue scales.
Potent and rapid antidepressant effects of a vaporized 5-MeO-DMT formulation (GH001) were demonstrated in a clinical phase 1/2 trial involving 16 patients with treatment resistant depression (59). The trial investigated two single dose levels (12 and 18 mg) of 5-MeO-DMT and an individualized dosing regimen with up to three increasing doses (6, 12, and 18 mg) of 5-MeO-DMT within a single day, with a primary endpoint of efficacy in the IDR group, as assessed by the proportion of patients in remission (i.e., MADRS ≤10) 7 days after treatment. The proportion of patients in remission at day 7 were 50% and 25% in the 12 mg and 18 mg groups respectively, and 87.5% in the IDR group. In this group, seven out of eight patients achieved a peak experience, six after the second administration (6 mg and 12 mg doses), and one after the third administration (6 mg, 12 mg, and 18 mg doses). All remissions were observed from day 1, and 60% of remissions were already observed 2 hours after treatment. The subjective, mean intensity of the psychedelic effect in the IDR group was around 90% as assessed with a peak experience scale.
Clinical studies in depressed patients thus have suggested that 5-MeO-DMT and DMT induce a therapeutic response within 24 hours that persists for at least 1 week (59) and 1 month (89) respectively. It is unknown at present how long the therapeutic effects of 5-MeO-DMT and DMT will last. It is to be expected that therapeutic efficacy of ultra-fast, short-acting psychedelics will not last forever, and that patient relapse rates will start to increase over weeks and months, just as has been previously observed after treatments with traditional antidepressants (90), ketamine (91), psilocybin (6), and ECT (92). Future trials with 5-MeO-DMT and DMT should therefore be designed to treat depressed patients over longer time intervals of 6–12 months, to establish the duration of the therapeutic response, and to probe the feasibility and efficacy of treatment re-dosing in case of patient relapse. Such studies can provide insights into how to prolong and extend the rapid acting benefits of ultra-fast, short-acting psychedelics in the treatment of depression.

Dose Escalation and Efficacy

A principal advantage of ultra-fast, short acting psychedelics is that they allow for repeated dosing in a single session in order to achieve dose optimization for individual patients in a clinical setting. Individualized dosing regimens might allow for a reduction in inter-individual variations in subjective (93, 94) and therapeutic responses (47, 95) to a psychedelic that have been observed after a fixed dose. Inter-individual variations in the response to a psychedelic might result from biological variations in 5HT2A receptor binding (96), drug metabolism (97), as well as from variations in subjective expectations (98, 99). Yet, the intensity and quality of the psychedelic experience may be relevant, because positive associations between the intensity of psychedelic experiences and therapeutic effects in patients with depression have been reported in a range of clinical trials (47, 48, 84, 95, 100, 101), though not all (9).
These data therefore suggest that individualized dose escalation of ultra-fast, short acting psychedelics may be best suited for clinical applications that aim to maximize the psychedelic experience to elicit a strong therapeutic response. So far, individualized dose escalation regimens have only been considered as part of treatment protocols for 5-MeO-DMT. In these protocols, successive, dose escalation steps were offered to healthy volunteers and patients with treatment resistant depression with the aim to increase dosing up until an individual achieved a maximal “peak” psychedelic experience (45, 59). In healthy volunteers, individualized dose escalations of a vaporized formulation of 5-MeO-DMT (GH001) consistently produced peak psychedelic experiences while the intensity of the subjective response to a single dose administration was more variable (45). Similar associations between dose regimens of 5-MeO-DMT (GH001) and the intensity of the psychedelic experience have been reported in patients with treatment resistant depression (59). More importantly, a larger fraction (87.5%) of patients that received an individualized dose escalation of vaporized 5-MeO-DMT to elicit a peak experience achieved remission as compared to the fraction of patients (25%–50%) who achieved remission after a single dose (59). These data support the notion that intensity of the psychedelic experience is predictive of the therapeutic response and can be employed as a subjective marker to guide individualized dosing regimens.
Two DMT trials in depressed patient populations also employed dose escalations that were either spaced 48 hours (49) or 90 minutes (89) apart. Dose escalations were fixed and did not allow for flexible, personalized treatment. In the first study (49), dose escalation only generated a moderate intensity of the psychedelic experience, possibly because the doses were relatively low and widely spaced apart. Yet, response (83.33%) and remission (66.67%) rates in the second study (89) at 7 days after dosing were not so much different from response (100%) and remission (87.5%) rates observed with IDR treatments with 5-MeO-DMT at 7 days after dosing (59). Likewise, as previously indicated, the mean intensity of the psychedelic experience achieved in both studies appears similar. This suggests that fixed incremental dose regimen might also beneficially affect treatment outcome. The gradual transition from low to high doses in dose escalation regimens possibly enables patients to mitigate their fear and increase their confidence to proceed to the next dose level, and thereby provide a smoother entry into a full dose psychedelic experience. The importance of a positive mindset for the quality of a psychedelic experience has been well documented and might add to the antidepressant effects of psychedelics (99, 102). Large patient studies that allow comparisons between single and incremental dose regimens and assess mindset and quality of the psychedelic experience at each dose step will be needed to disentangle potential pharmacological and non-pharmacological aspects of dose escalation regimens.

Tolerance and Sensitization

Tolerance and sensitization are neuroadaptive processes characterized by a rapid decrease and increase respectively in responsiveness after repeated exposure to a given drug. Rapid tolerance (also called tachyphylaxis) to the autonomic and psychological effects of a psychedelic has been shown to occur in humans after a few moderate daily doses of LSD, but sensitivity to LSD also quickly reversed after 1 or 2 days of discontinuation (103). Tolerance and cross-tolerance have also been demonstrated with other classical psychedelics such as psilocybin and mescaline in animal models of psychedelic action (104), and several studies have shown that rapid tolerance to psychedelics correlates with downregulation of 5HT2A receptors (11) or glutamate binding sites (105). However, tolerance to the behavioral effects of DMT appears less pronounced in humans. The intensity of the subjective psychedelic experience of intravenous 0.3 mg/kg DMT in healthy volunteers did not change over four successive administrations on a single day, whereas physiological reactivity decreased (53). An indication of moderate acute tolerance was observed in a study with continuous DMT administration in healthy volunteers, where subjective effects of DMT remained stable during 90 minutes of infusion, despite increasing plasma concentration (18). In animals, chronic administration of 5-MeO-DMT (2 mg/kg i.p., every 30 minutes for 4 hours) produced a rapid tolerance to the behavioral effects of the drug and reduced the duration of the effect from 14.9 minutes to 1.2 minutes (106). Tolerance, however, totally disappeared within 4 hours following termination of 5-MeO-DMT treatment (106). In humans, 12 mg and 18 mg of 5-MeO-DMT preceded by 6 mg or by 6 mg and 12 mg, respectively (all interspaced at 3 hours), consistently produced more intense psychedelic peak experiences as compared to single doses of 12 mg and 18 mg (45, 59). These differences may simply be accounted for by inter-individual variations in reactivity to 5-MeO-DMT but may also hint in the direction of increased sensitization. The latter may arise from experience learning and decreased anxiety after exposure to the first dose of 5-MeO-DMT and increased “openness” for a subsequent dose. In theory, sensitization may also result from 5HT2A receptor upregulation, but no study so far has evaluated 5HT2A receptor density in humans after exposure to repeated doses of 5-MeO-DMT. Sensitization to the psychedelic action of 5-MeO-DMT was demonstrated however in mice (i.e., enhanced head-twitch responses) with prior exposure to electroconvulsive shocks that caused 5HT2A receptor upregulation (107). Overall, human studies designed to assess tolerance and sensitization following closely interspaced doses of 5-MeO-DMT and DMT are scarce and need further investigation, but the clinical data so far seems to suggest that a solid psychedelic response can be maintained or achieved following close repetitions of ultra-fast, short-acting psychedelics.

Therapeutic Mechanisms

Several neurobiological and psychological mechanisms have been implicated in the antidepressant response to treatments with classic psychedelics that are also relevant to 5-MeO-DMT and DMT.
Neuroimaging studies in major depressive disorder have revealed distinct pattern of hyperconnectivity and hypoconnectivity within the default mode network (DMN) and the frontal parietal network (FPN) respectively, as well as compromised cross-network connectivity of the salience network (SN) (108110). A recent psilocybin trial in patients with major depression demonstrated post-treatment symptom relief to be associated with increased functional connectivity between these networks as defined by diminished network modularity (101). Studies in healthy volunteers also demonstrated a global increase in cross-network connectivity after treatment with classic psychedelics (111), including ayahuasca (112) and DMT (58). Ayahuasca increased DMN-SN connectivity and within network connectivity at 24 hours after dosing, while DMT acutely increased global functional connectivity (58). These data suggest that psychedelics may exert some of their antidepressant effects by increasing cross-connectivity of brain networks that are deficient in major depression. Whether specific actions of 5-MeO-DMT and DMT on brain function differ from long-acting psychedelics, however, is difficult to determine given that direct comparator studies are lacking.
Other mechanisms of action that have been implicated to contribute to the antidepressant response of psychedelics include the enduring increases in neuroplasticity (113) and immunomodulation (114) that can be observed post-treatment. A large number of reports suggest that decreased neuroplasticity, and neural atrophy in the hippocampus and the prefrontal cortex (PFC) play an essential role in the general pathophysiology of depression (115118) and that structural repair can be stimulated with compounds that promote structural and functional neural plasticity (115, 119, 120). The neuroplasticity-promoting effects of several serotonergic psychedelics, including 5-MeO-DMT and DMT, have been reported in vitro (121, 122). In vivo, single dose injections of 5-MeO-DMT rapidly increased the proliferation of neural progenitors and their dendritic complexity in the dentate gyrus of mice (123). Likewise, activation of the innate immune system, as indicated by elevated blood levels of pro-inflammatory cytokines and acute phase proteins, is associated with major depressive disorder (124). In vivo studies have demonstrated that 5-MeO-DMT and DMT inhibit the production of pro-inflammatory cytokines (i.e., IL-1 beta, IL-6, TNF-alpha, and the chemokine IL-8), while increasing the secretion of the anti-inflammatory cytokine IL-10 (125). Anti-inflammatory action of 5-MeO-DMT was also shown in humans as indicated by a rapid decrease in salivary IL-6 levels after a single dose (78). In TRD patients, reductions in MADRS rating following treatment with ayahuasca were associated with increased levels of a potential marker of neuroplasticity, i.e., brain-derived neurotropic factor (BDNF) (126), and reductions in an inflammatory marker, i.e., C-reactive protein (127). Yet, definitive evidence supporting the contribution of increased neuroplasticity and immunomodulation to the antidepressant response induced by ultra-fast, short-acting psychedelics is currently lacking and should come from future clinical trials.
The antidepressant response to 5-MeO-DMT and DMT might however also be psychologically driven. Psychedelic experiences with 5-MeO-DMT and DMT, depending on the dose and formulation, have been reported to be very intense (18, 25, 45, 58, 59), potentially inducing an “ontological shock” that may lead to radical shifts in self-perception and increased psychological flexibility when coping with day-to-day stressors (79, 80). Psychedelic alterations in consciousness are characterized by a perturbation of self-referential processing, giving rise to subjective states where self-other boundaries disappear (128, 129). Anecdotal as well as clinical reports have suggested that the experience of intense subjective psychedelic effects can facilitate psychological shifts and insights in behavioral patterns and it has been argued that these are fundamentally important to an enduring therapeutic response with a psychedelic (130132). Others have argued that psychedelic experiences are just epiphenomena of underlying neurobiological mechanisms that are truly driving the therapeutic response (133). Both perspectives are not necessarily mutually exclusive when explaining the therapeutic effects of 5-MeO-DMT and DMT.

Clinical Implementation

Advanced results from clinical trials of psilocybin for depression suggest that regulatory approval of psychedelic therapy is within reach, which is why questions on the societal impact of psychedelics as they are scaled up to clinical practice have started to be raised. While economic analyses of cost-effectiveness and cost-benefits of psychedelic treatment versus traditional antidepressant treatment are lagging behind (134), high costs of current treatment models with psilocybin (610) that involve the presence of multiple therapists during lengthy dosing sessions and integrated (psycho)therapy (see Table 3 for a prototype example of a psilocybin treatment model), are anticipated to impede their accessibility. This has fueled a fundamental discussion on the need to combine psychedelic treatment with integrated therapy as their interactive contribution to the therapeutic outcome is primarily hypothesized (135, 136), but has not been rigorously tested (137). Inclusion of assisted therapy in clinical trials with psychedelics furthermore provides a challenging mixture for regulatory bodies that are traditionally asked to review efficacy of drugs rather than psychotherapy (138). It has been suggested therefore that psychological support during psychedelic treatment should primarily focus on safety rather than efficacy (139).
Challenges of scaling psychedelic treatment for clinical practice will also affect potential treatment programs with 5-MeO-DMT and DMT. Early patient studies, as shown in Table 3, have explored the option to deliver psychedelic treatment in the absence of specialized therapists or therapy, but in the presence of standard medical care and psychological support to provide guidance and safety during psychedelic treatment. In DMT studies, preparation and integration/debriefing sessions were reported to be an explicit part of the treatment protocol (49, 89), but without much specification and without a need for therapists integrated into the trial. In patient studies with ayahuasca (84) and 5-MeO-DMT (59), no preparation or integration sessions were scheduled, but medical and psychological support was present during treatments and provided on an as needed basis. Psychedelic treatment models that do not capitalize on integrative therapy as part of the psychedelic experience might be easier and less costly to implement in clinical settings, provided that antidepressant efficacy is maintained. Yet, even treatment models that focus on safety and care should incorporate directions on how to create a trusted environment in which patients feel safeguarded and prepared for an experience with 5-MeO-DMT and DMT. Standardization is recommended for such guidelines.
Short-acting formulations of psychedelics may also reduce the time and effort that both patients and staff must invest in treatments, which might be preferable over long-acting formulations. Even with short-acting formulations, total treatment duration might still be considerable depending on a number of parameters such as rate of IV infusion, the presence of MAO inhibitors, or the number of administrations. As can be observed in Tables 2 and 3, IV infusion rates that have been explored varied from 60 seconds to 90 minutes, with psychedelic experiences lasting between 15 and 105 minutes. Intranasal DMT experiences even extended to 4–5 hours when combined with harmaline. While inhaled formulations of DMT and 5-MeO-DMT always produced short lasting experiences, the total treatment duration could still last somewhere between 3 and 9 hours depending on the number of dose increments and the duration of dosing intervals. It is to be expected that some parameters of 5-MeO-DMT and DMT treatment regimens will be adapted in future drug development trials in order to find the optimal balance between treatment delivery and efficacy. Such insights will contribute to future consensus on definitional terms of short-acting formulations and their applications.

Discussion

Although findings from early trials with 5-MeO-DMT and DMT in the treatment of depression are encouraging, there are also a number of limitations, the most important ones being the small sample size and the open label design of most studies. Studies with a small sample size are prone to overestimation of treatment effect sizes, particularly in the absence of a proper control group. Open-label trials with psychedelics might be particularly sensitive to extra-pharmacological factors such as expectancy effects that are known to moderate treatment responses (140). Patients who apply for participation in trials with psychedelics may enter with high treatment expectations that have been shaped and hyped though many media channels over recent years. Such expectations might drive strong placebo or nocebo effects that can add to or bias the actual treatment effects (99, 102, 140). That is, clinical trial participants who realize that they are in the active treatment group may show a strong clinical response because of increased expectations, whereas those who find out that they are in the placebo group may feel worse because of decreased expectations. Future trials with 5-MeO-DMT and DMT will therefore need to include large patient samples and add placebo and active control groups to control for non-pharmacological confounders and to properly mask allocation to psychedelic treatments. The latter is an acknowledged challenge for any trial with psychedelics given that these produce unique psychedelic experiences that can be easily recognized (140). Unfortunately, active treatment controls which can fully blind allocation to a psychedelic arm in a clinical trial are presently not available, which leaves the alternative design aimed at showing a dose dependent response, as a second best to control for positive expectations. In addition, nocebo effects can be minimized by offering patients in the placebo group the perspective of entering an open-label extension with the psychedelic treatment, after completion of the double-blind phase of the trial.
There are also unexplored grounds. Current trials with psychedelics have primarily focused on the onset and duration of a treatment response after one or two dosing days, without paying attention to strategies to sustain or reintroduce a treatment response in case patients relapse. Unlike clinical trials with classical antidepressants where a demonstration of sustained treatment efficacy over a couple of weeks was sufficient to assume a closed-loop treatment model based on daily dosing, clinical trials with single dose administrations of psychedelics have provided open-loop models in which continuation of treatment efficacy is not accounted for. Future trials with psychedelics, including 5-MeO-DMT and DMT, would therefore have to incorporate strategies aimed at continuation of the antidepressant response to provide a closed-loop model of psychedelic treatment. Such strategies could for example include re-dosing sessions for patients who relapse in order to assess whether renewed exposure to the psychedelic treatment produces a similar antidepressant response as observed after first exposure. Alternative to re-dosing, other approaches may be explored to sustain the antidepressant response, such as concurrent treatment with traditional antidepressants, neurostimulation, and/or cognitive training programs which potentially may leverage enduring and adaptive learning and coping behavior during critical, post-treatment windows of neuroplasticity (141).
In sum, clinical studies on 5-MeO-DMT and DMT have highlighted a number of potential benefits in the treatment of depression as well as challenges for future clinical implementations, as listed in Table 4. Ultra-fast, short-acting psychedelics may acquire a unique position amongst the medical armamentarium of antidepressants; one that capitalizes on instant and durable relief, with only brief interventions. Acceptance of ultra-fast, short-acting psychedelics by regulatory authorities will depend on future clinical trials with a focus on replication, duration, and maintenance of antidepressant efficacy in large patient samples.
TABLE 4. A summary of potential benefits and challenges of ultra-fast, short-acting psychedelics (5-MeO-DMT and DMT) as treatment for depression
Potential benefitsFuture challenges
Fast and prolonged antidepressant responseReplication of early findings in placebo-controlled trials in large patient samples
Therapeutic response independent of integrative therapyDetermination of the duration of the antidepressant response
Brief interventionStrategies to maintain the antidepressant response
Well toleratedOptimization of treatment regimen parameters (infusion rate, number dose escalation, duration dose interval)
Single day treatmentsDetermination of tolerance and sensitization during repeated or steady dosing
Flexibility of dosingFunctional contributions of 5HT1A and 5HT2A receptors to phenomenology
Experimental control over duration of psychedelic experience (IV)Determination of neurobiological and/or psychological mechanisms that underlie the antidepressant response
Clinical implementation less labor intensiveGuidelines for building trusted and safeguarded environment to deliver treatment

Footnote

Dr. Ramaekers drafted the manuscript which was edited and commented on by Dr. Reckweg and Dr. Mason. All authors contributed to the final version.

References

1.
Machado-Vieira R, Salvadore G, Luckenbaugh DA, et al: Rapid onset of antidepressant action: a new paradigm in the research and treatment of major depressive disorder. J Clin Psychiatry 2008; 69:946–958
2.
Rush AJ, Trivedi MH, Wisniewski SR, et al: Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry 2006; 163:1905–1917
3.
Sheline YI, Liston C, McEwen BS: Parsing the hippocampus in depression: chronic stress, hippocampal volume, and major depressive disorder. Biol Psychiatry 2019; 85:436–438
4.
Sheline YI, Sanghavi M, Mintun MA, et al: Depression duration but not age predicts hippocampal volume loss in medically healthy women with recurrent major depression. J Neurosci 1999; 19:5034–5043
5.
Sheline YI, Wang PW, Gado MH, et al: Hippocampal atrophy in recurrent major depression. Proc Natl Acad Sci U S A 1996; 93:3908–3913
6.
Goodwin GM, Aaronson ST, Alvarez O, et al: Single-dose psilocybin for a treatment-resistant episode of major depression. N Engl J Med 2022; 387:1637–1648
7.
Davis AK, Barrett FS, May DG, et al: Effects of psilocybin-assisted therapy on major depressive disorder: a randomized clinical trial. JAMA Psychiatry 2021; 78:481–489
8.
Carhart-Harris R, Giribaldi B, Watts R, et al: Trial of psilocybin versus escitalopram for depression. N Engl J Med 2021; 384:1402–1411
9.
von Rotz R, Schindowski EM, Jungwirth J, et al: Single-dose psilocybin-assisted therapy in major depressive disorder: a placebo-controlled, double-blind, randomised clinical trial. EClinicalMedicine 2022; 56:101809
10.
Raison CL, Sanacora G, Woolley J, et al: Single-dose psilocybin treatment for major depressive disorder: a randomized clinical trial. JAMA 2023; 330:843–853
11.
Nichols DE: Psychedelics. Pharmacol Rev 2016; 68:264–355
12.
Holze F, Becker AM, Kolaczynska KE, et al: Pharmacokinetics and pharmacodynamics of oral psilocybin administration in healthy participants. Clin Pharmacol Ther 2023; 113:822–831
13.
Reckweg JT, Uthaug MV, Szabo A, et al: The clinical pharmacology and potential therapeutic applications of 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT). J Neurochem 2022; 162:128–146
14.
James E, Keppler J, L Robertshaw T, et al: N,N-dimethyltryptamine and amazonian ayahuasca plant medicine. Hum Psychopharmacol 2022; 37:e2835
15.
Riba J, McIlhenny EH, Valle M, et al: Metabolism and disposition of N,N-dimethyltryptamine and harmala alkaloids after oral administration of ayahuasca. Drug Test Anal 2012; 4:610–616
16.
Shen HW, Jiang XL, Winter JC, et al: Psychedelic 5-methoxy-N,N-dimethyltryptamine: metabolism, pharmacokinetics, drug interactions, and pharmacological actions. Curr Drug Metab 2010; 11:659–666
17.
Barker SA: N,N-dimethyltryptamine (DMT), an endogenous hallucinogen: past, present, and future research to determine its role and function. Front Neurosci 2018; 12:536
18.
Vogt SB, Ley L, Erne L, et al: Acute effects of intravenous DMT in a randomized placebo-controlled study in healthy participants. Transl Psychiatry 2023; 13:172
19.
Pachter IJ, Zacharias DE, Ribeiro O: Indole alkaloids of acer saccharinum (the silver maple), dictyoloma incanescens, piptadenia colubrina, and mimosa hostilis. J Org Chem 1959; 24:1285–1287
20.
Agurell S, Holmstedt B, Lindgren JE, et al: Alkaloids in certain species of virola and other South American plants of ethnopharmacologic interest. Acta Chem Scand 1969; 23:903–916
21.
Domínguez-Clavé E, Soler J, Elices M, et al: Ayahuasca: pharmacology, neuroscience and therapeutic potential. Brain Res Bull 2016; 126:89–101
22.
Weil AT, Davis W: Bufo alvarius: a potent hallucinogen of animal origin. J Ethnopharmacol 1994; 41:1–8
23.
Strassman RJ: Human psychopharmacology of N,N-dimethyltryptamine. Behav Brain Res 1996; 73:121–124
24.
Strassman RJ, Qualls CR: Dose-response study of N,N-dimethyltryptamine in humans. I. Neuroendocrine, autonomic, and cardiovascular effects. Arch Gen Psychiatry 1994; 51:85–97
25.
Timmermann C, Roseman L, Williams L, et al: DMT models the near-death experience. Front Psychol 2018; 9:1424
26.
Lawrence DW, Carhart-Harris R, Griffiths R, et al: Phenomenology and content of the inhaled N, N-dimethyltryptamine (N, N-DMT) experience. Sci Rep 2022; 12:8562
27.
Davis AK, Barsuglia JP, Lancelotta R, et al: The epidemiology of 5-methoxy- N, N-dimethyltryptamine (5-MeO-DMT) use: benefits, consequences, patterns of use, subjective effects, and reasons for consumption. J Psychopharmacol 2018; 32:779–792
28.
Michael P, Luke D, Robinson O: This is your brain on death: a comparative analysis of a near-death experience and subsequent 5-Methoxy-DMT experience. Front Psychol 2023; 14:1083361
29.
Ermakova AO, Dunbar F, Rucker J, et al: A narrative synthesis of research with 5-MeO-DMT. J Psychopharmacol 2022; 36:273–294
30.
Halberstadt AL, Geyer MA: Multiple receptors contribute to the behavioral effects of indoleamine hallucinogens. Neuropharmacology 2011; 61:364–381
31.
Carhart-Harris RL, Nutt DJ: Serotonin and brain function: a tale of two receptors. J Psychopharmacol 2017; 31:1091–1120
32.
Puigseslloses P, Nadal-Gratacós N, Ketsela G, et al: Structure-activity relationships of serotonergic 5-MeO-DMT derivatives: insights into psychoactive and thermoregulatory properties. Mol Psychiatry 2024
33.
Ray TS: Psychedelics and the human receptorome. PLoS One 2010; 5:e9019
34.
Besnard J, Ruda GF, Setola V, et al: Automated design of ligands to polypharmacological profiles. Nature 2012; 492:215–220
35.
Celada P, Bortolozzi A, Artigas F: Serotonin 5-HT1A receptors as targets for agents to treat psychiatric disorders: rationale and current status of research. CNS Drugs 2013; 27:703–716
36.
Spencer DG, Jr., Glaser T, Traber J: Serotonin receptor subtype mediation of the interoceptive discriminative stimuli induced by 5-methoxy-N,N-dimethyltryptamine. Psychopharmacology (Berl) 1987; 93:158–166
37.
Schreiber R, de Vry J: Studies on the neuronal circuits involved in the discriminative stimulus effects of 5-hydroxytryptamine1A receptor agonists in the rat. J Pharmacol Exp Ther 1993; 265:572–579
38.
Winter JC, Filipink RA, Timineri D, et al: The paradox of 5-methoxy-N,N-dimethyltryptamine: an indoleamine hallucinogen that induces stimulus control via 5-HT1A receptors. Pharmacol Biochem Behav 2000; 65:75–82
39.
Riga MS, Bortolozzi A, Campa L, et al: The serotonergic hallucinogen 5-methoxy-N,N-dimethyltryptamine disrupts cortical activity in a regionally-selective manner via 5-HT(1A) and 5-HT(2A) receptors. Neuropharmacology 2016; 101:370–378
40.
Riga MS, Soria G, Tudela R, et al: The natural hallucinogen 5-MeO-DMT, component of ayahuasca, disrupts cortical function in rats: reversal by antipsychotic drugs. Int J Neuropsychopharmacol 2014; 17:1269–1282
41.
Araneda R, Andrade R: 5-Hydroxytryptamine2 and 5-hydroxytryptamine 1A receptors mediate opposing responses on membrane excitability in rat association cortex. Neuroscience 1991; 40:399–412
42.
Carli M, Baviera M, Invernizzi RW, et al: Dissociable contribution of 5-HT1A and 5-HT2A receptors in the medial prefrontal cortex to different aspects of executive control such as impulsivity and compulsive perseveration in rats. Neuropsychopharmacology 2006; 31:757–767
43.
Fontanilla D, Johannessen M, Hajipour AR, et al: The hallucinogen N,N-dimethyltryptamine (DMT) is an endogenous sigma-1 receptor regulator. Science 2009; 323:934–937
44.
Simmler LD, Buchy D, Chaboz S, et al: In vitro characterization of psychoactive substances at rat, mouse, and human trace amine-associated receptor 1. J Pharmacol Exp Ther 2016; 357:134–144
45.
Reckweg J, Mason NL, van Leeuwen C, et al: A phase 1, dose-ranging study to assess safety and psychoactive effects of a vaporized 5-methoxy-N, N-dimethyltryptamine formulation (GH001) in healthy volunteers. Front Pharmacol 2021; 12:760671
46.
Rucker JJ, Roberts C, Seynaeve M, et al: Phase 1, placebo-controlled, single ascending dose trial to evaluate the safety, pharmacokinetics and effect on altered states of consciousness of intranasal BPL-003 (5-methoxy-N,N-dimethyltryptamine benzoate) in healthy participants. J Psychopharmacol 2024:2698811241246857. Online ahead of print
47.
Roseman L, Nutt DJ, Carhart-Harris RL: Quality of acute psychedelic experience predicts therapeutic efficacy of psilocybin for treatment-resistant depression. Front Pharmacol 2017; 8:974
48.
Romeo B, Hermand M, Pétillion A, et al: Clinical and biological predictors of psychedelic response in the treatment of psychiatric and addictive disorders: a systematic review. J Psychiatr Res. 2021; 137:273–282
49.
D’Souza DC, Syed SA, Flynn LT, et al: Exploratory study of the dose-related safety, tolerability, and efficacy of dimethyltryptamine (DMT) in healthy volunteers and major depressive disorder. Neuropsychopharmacology 2022; 47:1854–1862
50.
Good M, Joel Z, Benway T, et al: Pharmacokinetics of N,N-dimethyltryptamine in humans. Eur J Drug Metab Pharmacokinet 2023; 48:311–327
51.
James E, Erritzoe D, Benway T, et al: Safety, tolerability, pharmacodynamic and wellbeing effects of SPL026 (dimethyltryptamine fumarate) in healthy participants: a randomized, placebo-controlled phase 1 trial. Front Psychiatry. 2023; 14:1305796
52.
Luan LX, Eckernas E, Ashton M, et al: Psychological and physiological effects of extended DMT. J Psychopharmacol 2024; 38:56–67
53.
Strassman RJ, Qualls CR, Berg LM: Differential tolerance to biological and subjective effects of four closely spaced doses of N,N-dimethyltryptamine in humans. Biol Psychiatry 1996; 39:784–795
54.
Strassman RJ, Qualls CR, Uhlenhuth EH, et al: Dose-response study of N,N-dimethyltryptamine in humans. II. Subjective effects and preliminary results of a new rating scale. Arch Gen Psychiatry 1994; 51:98–108
55.
Falchi-Carvalho M, Wießner I, Silva SRB, et al: Safety and tolerability of inhaled N,N-Dimethyltryptamine (BMND01 candidate): a phase I clinical trial. Eur Neuropsychopharmacol 2024; 80:27–35
56.
Aicher HD, Mueller MJ, Dornbierer DA, et al: Potential therapeutic effects of an ayahuasca-inspired N,N-DMT and harmine formulation: a controlled trial in healthy subjects. Front Psychiatry 2023; 14:1302559
57.
Gouzoulis-Mayfrank E, Heekeren K, Neukirch A, et al: Psychological effects of (S)-ketamine and N,N-dimethyltryptamine (DMT): a double-blind, cross-over study in healthy volunteers. Pharmacopsychiatry 2005; 38:301–311,
58.
Timmermann C, Roseman L, Haridas S, et al: Human brain effects of DMT assessed via EEG-fMRI. Proc Natl Acad Sci U S A 2023; 120:e2218949120
59.
Reckweg JT, van Leeuwen CJ, Henquet C, et al: A phase 1/2 trial to assess safety and efficacy of a vaporized 5-methoxy-N,N-dimethyltryptamine formulation (GH001) in patients with treatment-resistant depression. Front Psychiatry 2023; 14:1133414
60.
Uthaug MV, Lancelotta R, Ortiz Bernal AM, et al: Comparison of reactivation experiences following vaporization and intramuscular injection (IM) of synthetic 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT) in a naturalistic setting. J Psychedelic Stud 2020; 2:1–10
61.
Ortiz Bernal AM, Raison CL, Lancelotta RL, et al: Reactivations after 5-methoxy-N,N-dimethyltryptamine use in naturalistic settings: an initial exploratory analysis of the phenomenon’s predictors and its emotional valence. Front Psychiatry 2022; 13:1049643
62.
Davis AK: The potential for healing and harm with 5-MeO-DMT; in Horizons Conference, 2019
63.
Dourron HM, Nichols CD, Simonsson O, et al: 5-MeO-DMT: an atypical psychedelic with unique pharmacology, phenomenology & risk? Psychopharmacology (Berl) 2023. Online ahead of print
64.
Gharedaghi MH, Seyedabadi M, Ghia JE, et al: The role of different serotonin receptor subtypes in seizure susceptibility. Exp Brain Res 2014; 232:347–367
65.
Toczek MT, Carson RE, Lang L, et al: PET imaging of 5-HT1A receptor binding in patients with temporal lobe epilepsy. Neurology 2003; 60:749–756
66.
Sourbron J, Lagae L: Serotonin receptors in epilepsy: novel treatment targets? Epilepsia Open. 2022; 7:231–246,
67.
Martínez-Aguirre C, Carmona-Cruz F, Velasco AL, et al: Cannabidiol acts at 5-HT1A receptors in the human brain: relevance for treating temporal lobe epilepsy. Front Behav Neurosci 2020; 14:611278
68.
Wada Y, Hasegawa H, Nakamura M, et al: Behavioral and electroencephalographic effects of a serotonin receptor agonist (5-methoxy-N,N-dimethyltryptamine) in a feline model of photosensitive epilepsy. Neurosci Lett 1992; 138:115–118
69.
Barker SA: Administration of N,N-dimethyltryptamine (DMT) in psychedelic therapeutics and research and the study of endogenous DMT. Psychopharmacology (Berl) 2022; 239:1749–1763
70.
Davis AK, Lancelotta R: The healing potential of 5-MeO-DMT: results from two survey studies. Midwest Psychedelic Therapy Symposium, Madison, Wisconsin, 2018
71.
Carbonaro TM, Bradstreet MP, Barrett FS, et al: Survey study of challenging experiences after ingesting psilocybin mushrooms: acute and enduring positive and negative consequences. J Psychopharmacol 2016; 30:1268–1278
72.
Dos Santos RG, Bouso JC, Hallak JEC: Ayahuasca, dimethyltryptamine, and psychosis: a systematic review of human studies. Ther Adv Psychopharmacol 2017; 7:141–157
73.
Metzner R: The Toad and the Jaguar: A Field Report of Underground Research on a Visionary Medicine: Bufo alvarius and 5-Methoxy-Dimethyltryptamine. Regent press, 2013
74.
Shulgin AT, Shulgin A: TIHKAL: The Continuation. Berkeley, Transform Press, 1997
75.
Johnson M, Richards W, Griffiths R: Human hallucinogen research: guidelines for safety. J Psychopharmacol. 2008; 22:603–620
76.
Soler J, Elices M, Franquesa A, et al: Exploring the therapeutic potential of ayahuasca: acute intake increases mindfulness-related capacities. Psychopharmacology (Berl) 2016; 233:823–829
77.
Sampedro F, de la Fuente Revenga M, Valle M, et al: Assessing the psychedelic “After-Glow” in ayahuasca users: post-acute neurometabolic and functional connectivity changes are associated with enhanced mindfulness capacities. Int J Neuropsychopharmacol 2017; 20:698–711
78.
Uthaug MV, Lancelotta R, Szabo A, et al: Prospective examination of synthetic 5-methoxy-N,N-dimethyltryptamine inhalation: effects on salivary IL-6, cortisol levels, affect, and non-judgment. Psychopharmacology (Berl) 2020; 237:773–785
79.
Uthaug MV, Lancelotta R, van Oorsouw K, et al: A single inhalation of vapor from dried toad secretion containing 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT) in a naturalistic setting is related to sustained enhancement of satisfaction with life, mindfulness-related capacities, and a decrement of psychopathological symptoms. Psychopharmacology (Berl) 2019; 236:2653–2666
80.
Uthaug MV, van Oorsouw K, Kuypers KPC, et al: Sub-acute and long-term effects of ayahuasca on affect and cognitive thinking style and their association with ego dissolution. Psychopharmacology 2018; 235:2979–2989
81.
Kiraga MK, Mason NL, Uthaug MV, et al: Persisting effects of ayahuasca on empathy, creative thinking, decentering, personality, and well-being. Front Pharmacol 2021; 12:721537
82.
van Oorsouw K, Toennes SW, Ramaekers JG: Therapeutic effect of an ayahuasca analogue in clinically depressed patients: a longitudinal observational study. Psychopharmacology (Berl) 2022; 239:1839–1852
83.
Timmermann C, Zeifman RJ, Erritzoe D, et al: Effects of DMT on mental health outcomes in healthy volunteers. Sci Rep 2024; 14:3097
84.
Palhano-Fontes F, Barreto D, Onias H, et al: Rapid antidepressant effects of the psychedelic ayahuasca in treatment-resistant depression: a randomized placebo-controlled trial. Psychol Med 2019; 49:655–663
85.
Labate BC, Feeney K: Ayahuasca and the process of regulation in Brazil and internationally: implications and challenges. Int J Drug Pol 2012; 23:154–161
86.
Riba J, Valle M, Urbano G, et al: Human pharmacology of ayahuasca: subjective and cardiovascular effects, monoamine metabolite excretion, and pharmacokinetics. J Pharmacol Exp Ther 2003; 306:73–83
87.
Amrein R, Hetzel W, Stabl M, et al: RIMA: a safe concept in the treatment of depression with moclobemide. Can J Psychiatry 1992; 37:7–11
88.
Lum CT, Stahl SM: Opportunities for reversible inhibitors of monoamine oxidase-A (RIMAs) in the treatment of depression. CNS Spectr 2012; 17:107–120
89.
Falchi-Carvalho M, Barros H, Bolcont R, et al: The antidepressant effects of vaporized N,N-Dimethyltryptamine: a preliminary report in treatment-resistant depression. medRxiv 2024
90.
Lewis G, Marston L, Duffy L, et al: Maintenance or discontinuation of antidepressants in primary care. N Engl J Med. 2021; 385:1257–1267
91.
Smith-Apeldoorn SY, Veraart JK, Spijker J, et al: Maintenance ketamine treatment for depression: a systematic review of efficacy, safety, and tolerability. Lancet Psychiatry 2022; 9:907–921
92.
Rowland T, Mann R, Azeem S: The efficacy and tolerability of continuation and maintenance electroconvulsive therapy for depression: a systematic review of randomized and observational studies. J ECT 2023; 39:141–150
93.
Studerus E, Gamma A, Vollenweider FX: Psychometric evaluation of the altered states of consciousness rating scale (OAV). PLoS One 2010; 5:e12412
94.
Hutten NRPW, Mason NL, Dolder PC, et al: Mood and cognition after administration of low LSD doses in healthy volunteers: a placebo controlled dose-effect finding study. Eur Neuropsychopharmacol 2020; 41:81–91
95.
Griffiths RR, Johnson MW, Carducci MA, et al: Psilocybin produces substantial and sustained decreases in depression and anxiety in patients with life-threatening cancer: a randomized double-blind trial. J Psychopharmacol 2016; 30:1181–1197
96.
Stenbæk DS, Madsen MK, Ozenne B, et al: Brain serotonin 2A receptor binding predicts subjective temporal and mystical effects of psilocybin in healthy humans. J Psychopharmacol 2021; 35:459–468
97.
Vizeli P, Straumann I, Holze F, et al: Genetic influence of CYP2D6 on pharmacokinetics and acute subjective effects of LSD in a pooled analysis. Sci Rep 2021; 11:10851
98.
Haijen ECHM, Kaelen M, Roseman L, et al: Predicting responses to psychedelics: a prospective study. Front Pharmacol 2018; 9:897
99.
Hartogsohn I: Set and setting, psychedelics and the placebo response: an extra-pharmacological perspective on psychopharmacology. J Psychopharmacol 2016; 30:1259–1267
100.
Ross S, Bossis A, Guss J, et al: Rapid and sustained symptom reduction following psilocybin treatment for anxiety and depression in patients with life-threatening cancer: a randomized controlled trial. J Psychopharmacol 2016; 30:1165–1180
101.
Daws RE, Timmermann C, Giribaldi B, et al: Increased global integration in the brain after psilocybin therapy for depression. Nat Med 2022; 28:844–851
102.
Pronovost-Morgan C, Hartogsohn I, Ramaekers JG: Harnessing placebo: lessons from psychedelic science. J Psychopharmacol 2023; 37:866–875
103.
Passie T, Halpern JH, Stichtenoth DO, et al: The pharmacology of lysergic acid diethylamide: a review. CNS Neurosci Ther 2008; 14:295–314
104.
de la Fuente Revenga M, Jaster AM, McGinn J, et al: Tolerance and cross-tolerance among psychedelic and nonpsychedelic 5-HT2A receptor agonists in mice. ACS Chem Neurosci 2022; 13:2436–2448
105.
Buchborn T, Schröder H, Dieterich DC, et al: Tolerance to LSD and DOB induced shaking behaviour: differential adaptations of frontocortical 5-HT(2A) and glutamate receptor binding sites. Behav Brain Res 2015; 281:62–68
106.
Trulson ME, Keltch GF: Development of tolerance to repeated administration of 5-methoxy-N,N-dimethyltryptamine in rats. Eur J Pharmacol 1985; 108:33–37
107.
Metz A, Heal DJ: In mice repeated administration of electroconvulsive shock or desmethylimipramine produces rapid alterations in 5-HT2-mediated head-twitch responses and cortical 5-HT2 receptor number. Eur J Pharmacol 1986; 126:159–162
108.
Manoliu A, Meng C, Brandl F, et al: Insular dysfunction within the salience network is associated with severity of symptoms and aberrant inter-network connectivity in major depressive disorder. Front Hum Neurosci 2014; 7:930
109.
Kaiser RH, Andrews-Hanna JR, Wager TD, et al: Large-scale network dysfunction in major depressive disorder: a meta-analysis of resting-state functional connectivity. JAMA Psychiatry 2015; 72:603–611
110.
Li H, Lin X, Liu L, et al: Disruption of the structural and functional connectivity of the frontoparietal network underlies symptomatic anxiety in late-life depression. Neuroimage Clin 2020; 28:102398
111.
Kwan AC, Olson DE, Preller KH, et al: The neural basis of psychedelic action. Nat Neurosci 2022; 25:1407–1419
112.
Palhano-Fontes F, Andrade KC, Tofoli LF, et al: The psychedelic state induced by ayahuasca modulates the activity and connectivity of the default mode network. PLoS One 2015; 10:e0118143
113.
de Vos CMH, Mason NL, Kuypers KPC: Psychedelics and neuroplasticity: a systematic review unraveling the biological underpinnings of psychedelics. Front Psychiatry 2021; 12:724606
114.
Szabo A: Psychedelics and immunomodulation: novel approaches and therapeutic opportunities. Front Immunol 2015; 6:358
115.
Duman RS: Synaptic plasticity and mood disorders. Mol Psychiatry 2002; 7:S29–S34
116.
Videbech P, Ravnkilde B: Hippocampal volume and depression: a meta-analysis of MRI studies. Am J Psychiatry 2004; 161:1957–1966
117.
Duman RS, Aghajanian GK: Synaptic dysfunction in depression: potential therapeutic targets. Science 2012; 338:68–72
118.
Duman RS, Aghajanian GK, Sanacora G, et al: Synaptic plasticity and depression: new insights from stress and rapid-acting antidepressants. Nat Med 2016; 22:238–249
119.
Krystal JH, Tolin DF, Sanacora G, et al: Neuroplasticity as a target for the pharmacotherapy of anxiety disorders, mood disorders, and schizophrenia. Drug Discov Today 2009; 14:690–697
120.
Castrén E, Antila H: Neuronal plasticity and neurotrophic factors in drug responses. Mol Psychiatry 2017; 22:1085–1095
121.
Ly C, Greb AC, Cameron LP, et al: Psychedelics promote structural and functional neural plasticity. Cell Rep 2018; 23:3170–3182
122.
Dakic V, Minardi Nascimento J, Costa Sartore R, et al: Short term changes in the proteome of human cerebral organoids induced by 5-MeO-DMT. Sci Rep 2017; 7:12863
123.
Lima da Cruz RV, Moulin TC, Petiz LL, et al: A single dose of 5-MeO-DMT stimulates cell proliferation, neuronal survivability, morphological and functional changes in adult mice ventral dentate gyrus. Front Mol Neurosci 2018; 11:312
124.
Wittenberg GM, Stylianou A, Zhang Y, et al: Effects of immunomodulatory drugs on depressive symptoms: a mega-analysis of randomized, placebo-controlled clinical trials in inflammatory disorders. Mol Psychiatry 2020; 25:1275–1285
125.
Szabo A, Kovacs A, Frecska E, et al: Psychedelic N,N-dimethyltryptamine and 5-methoxy-N,N-dimethyltryptamine modulate innate and adaptive inflammatory responses through the sigma-1 receptor of human monocyte-derived dendritic cells. PLoS One 2014; 9:e106533
126.
de Almeida RN, Galvão ACM, da Silva FS, et al: Modulation of serum brain-derived neurotrophic factor by a single dose of ayahuasca: observation from a randomized controlled trial. Front Psychol 2019; 10:1234
127.
Galvão-Coelho NL, de Menezes Galvão AC, de Almeida RN, et al: Changes in inflammatory biomarkers are related to the antidepressant effects of Ayahuasca. J Psychopharmacol 2020; 34:1125–1133
128.
Nour MM, Evans L, Nutt D, et al: Ego-dissolution and psychedelics: validation of the ego-dissolution inventory (EDI). Front Hum Neurosci 2016; 10:269
129.
Preller KH, Vollenweider FX: Phenomenology, structure, and dynamic of psychedelic states. Curr Top Behav Neurosci 2018; 36:221–256
130.
Carhart-Harris RL, Goodwin GM: The therapeutic potential of psychedelic drugs: past, present, and future. Neuropsychopharmacology 2017; 42:2105–2113
131.
Yaden DB, Griffiths RR: The subjective effects of psychedelics are necessary for their enduring therapeutic effects. ACS Pharmacol Transl Sci 2021; 4:568–572
132.
Nayak SM, Singh M, Yaden DB, et al: Belief changes associated with psychedelic use. J Psychopharmacol 2023; 37:80–92
133.
Olson DE: The subjective effects of psychedelics may not be necessary for their enduring therapeutic effects. ACS Pharmacol Transl Sci 2021; 4:563–567
134.
Marseille E, Bertozzi S, Kahn JG: The economics of psychedelic-assisted therapies: a research agenda. Front Psychiatry 2022; 13:1025726
135.
Yaden DB, Earp D, Graziosi M, et al: Psychedelics and psychotherapy: cognitive-behavioral approaches as default. Front Psychol 2022; 13:873279
136.
Cavarra M, Falzone A, Ramaekers JG, et al: Psychedelic-assisted psychotherapy-A systematic review of associated psychological interventions. Front Psychol 2022; 13:887255
137.
Heifets BD, Olson DE: Therapeutic mechanisms of psychedelics and entactogens. Neuropsychopharmacology 2024; 49:104–118
138.
FDA: Psychedelic Drugs Considerations for Clinical Investigations, 2023
139.
Goodwin GM, Malievskaia E, Fonzo GA, et al: Must psilocybin always “assist psychotherapy”? Am J Psychiatry 2024; 181:20–25
140.
Aday JS, Heifets BD, Pratscher SD, et al: Great expectations: recommendations for improving the methodological rigor of psychedelic clinical trials. Psychopharmacology (Berl) 2022; 239:1989–2010
141.
Nardou R, Sawyer E, Song YJ, et al: Psychedelics reopen the social reward learning critical period. Nature 2023; 618:790–798

Information & Authors

Information

Published In

Go to American Journal of Psychiatry
Go to American Journal of Psychiatry
American Journal of Psychiatry
Pages: 33 - 46
PubMed: 39741439

History

Received: 3 November 2023
Revision received: 3 April 2024
Revision received: 23 April 2024
Published online: 20 December 2024
Published in print: January 01, 2025

Keywords

  1. 5-MeO-DMT
  2. DMT
  3. depression
  4. psychedelics

Authors

Details

Johannes G. Ramaekers, Ph.D. [email protected]
Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, the Netherlands.
Johannes T. Reckweg, Ph.D.
Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, the Netherlands.
Natasha L. Mason, Ph.D.
Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, the Netherlands.

Notes

Send correspondence to Dr. Ramaekers ([email protected]).

Competing Interests

Dr. Ramaekers and Dr. Reckweg are scientific consultants to GH Research. GH Research was not involved in this manuscript. Dr. Mason reports no financial relationships with commercial interests.

Metrics & Citations

Metrics

Citations

Export Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click Download.

For more information or tips please see 'Downloading to a citation manager' in the Help menu.

Format
Citation style
Style
Copy to clipboard

View Options

View options

PDF/EPUB

View PDF/EPUB

Login options

Already a subscriber? Access your subscription through your login credentials or your institution for full access to this article.

Personal login Institutional Login Open Athens login

Not a subscriber?

Subscribe Now / Learn More

PsychiatryOnline subscription options offer access to the DSM-5-TR® library, books, journals, CME, and patient resources. This all-in-one virtual library provides psychiatrists and mental health professionals with key resources for diagnosis, treatment, research, and professional development.

Need more help? PsychiatryOnline Customer Service may be reached by emailing [email protected] or by calling 800-368-5777 (in the U.S.) or 703-907-7322 (outside the U.S.).

Media

Figures

Other

Tables

Share

Share

Share article link

Share