• Psychedelic Medicine

PHARMACOLOGY | +80 articles

image.axd

A randomized placebo-controlled PET study of ketamine´s effect on serotonin1B
receptor binding in patients with SSRI-resistant depression.


Ketamine uses serotonin receptors to relieve depression

by Dr. Liji Thomas, MD | News Medical | 31 May 2020

Ketamine is a drug used to induce general anesthesia. It has also been found to act rapidly to relieve depression that has failed to respond to two other rounds of medication. A new study published in the journal Translational Psychiatry in May 2020 reports the mechanism of action of the drug, which was hitherto unknown. This could help find new ways to treat this debilitating and sometimes fatal condition.

The study comes from Sweden, where depression affects 10% of men and 20% of women. This makes it the most frequently diagnosed mental disorder in this country. The treatment of this condition fails after two rounds or more in about 15-30% of patients. At this point, the depression is termed difficult-to-treat.

Ketamine and depression

Ketamine is a non-competitive N-methyl D-aspartate (NMDA) receptor antagonist. It is a dissociative drug, meaning it disrupts the user's relationship with sensory perceptions.

Higher doses can cause breathing to slow dangerously, produce muscle spasms, dizziness, loss of balance, and slurring, with severe confusion. Hallucinations described as "out of body" experiences occur when emerging from the drug, which can be very distressing.

Treatment-resistant depression that has been found to respond quickly to ketamine at low doses. This has led to the approval of a nasal ketamine-containing spray in the USA and EU for this condition. Despite this knowledge, little has been known about how it acts.

Earlier research, in which Tiger and his team had collaborated, shows that the brains of people with depression presented low levels of these receptors.

In the first step, the researchers performed a randomized double-blinded controlled study on 30 patients with treatment-resistant depression. They divided them at random into two groups. One group of 20 received ketamine infusions. The other group of 10 received only saline infusions.

Neither the doctor treating them nor the patients knew who was getting what. Both before and after infusions, the patients underwent brain imaging using the PET cameras. This was repeated after 24-72 hours after the completion of the procedure.

The second part of the study involved voluntary participation. This phase included 29 individuals who got ketamine infusions twice weekly for two weeks.

Ketamine acts by bosting Serotonin 1B Receptors

When the results were assessed, the doctors found that more than 70% of the patients who had received ketamine experienced relief of depressive symptoms, as recorded using a rating scale for depression.

The researchers found that the therapeutic effects of ketamine were due to inhibition of serotonin action. Serotonin is a neurochemical which acts like a brain hormone, in part. It is best known for its significant contribution to triggering depression. Low levels of this hormone are thought to be associated with more severe depression.

Neurons have 14 serotonin receptor subtypes on their cell surfaces. However, in the current study, the researchers used a more specific molecule, a radioactive substance that binds only to serotonin 1B receptors. They thus discovered a novel mechanism of action for ketamine.

The binding of the serotonin 1B receptor causes serotonin levels to go down, while another neurochemical called dopamine, which also transmits nerve signals, is increased. Dopamine is a neurotransmitter related to the reward circuits of the brain, which means its levels cause more positive feelings or mood elevation. This feature is highly desirable in depression.

So how does ketamine do what it does? Apparently, it increases the number of serotonin 1B receptors. This is the first time this effect has been observed, and it explains the quick and favorable action of ketamine.

This discovery could trigger more research on whether the serotonin 1B receptor offers a target for newer drugs that may avoid ketamine's adverse effects even while maintaining its rapid mood-lifting capability.Another researcher, Johan Lundberg, says, "Ketamine has the advantage of being very rapid-acting, but at the same time it is a narcotic-classed drug that can lead to addiction."

The ketamine study: What was done?

In the current study, researchers used positron emission tomography (PET) to image the brains of depression patients on ketamine treatment. Calling it "the largest PET study of its kind in the world," researcher Mikael Tiger says their "focus was on finding how much of an effect ketamine has on depression, as well as trying to identify the role of serotonin 1B receptors."

 
Last edited:
shutterstock_1476864158.jpg



Therapeutic effects of N,N-Dimethyltryptamine (DMT)

by Barb Bauer | Psychedelic Science Review | 9 Jun 2020

Researchers are uncovering reasons to think there’s more to DMT than just hallucinations.

DMT (N,N-dimethyltryptamine) is a naturally occurring psychoactive molecule found in plants of several genera, including Acacia, Desmodium, Mimosa, Virola, Delosperma, and Phalaris. It is the main active compound in the beverage ayahuasca, traditionally obtained from the leaves of Psychotria viridis.

DMT has also been isolated in mammals. In 1961, Axelrod was the first to demonstrate the presence of DMT in rat and human brains. A study published in Nature in 2019 generated media coverage by finding synthesis and release of DMT in the brain of rats, leading researchers to wonder if this mechanism also occurs in human brains.

The resurging interest in the therapeutic potential of psychedelic compounds is setting the stage for more investigation into DMT. Typically, the primary area of interest for DMT research is its hallucinogenic effects, mostly in the context of ayahuasca. However, some studies within the last decade indicate DMT may have health benefits all its own.

Some possible therapeutic applications of DMT

In science, if you don’t understand how something works and what it does, it’s hard to figure out what you can do with it. Since Axelrod’s discovery in 1961, scientists have been wondering why DMT is present in humans and what it does.

In 2013, Frescka et al. published a review paper in the Journal of Neural Transmission, which suggested an answer. The authors proposed that DMT may have a role in adaptive biological processes via sigma receptors such as sigma-1. “Our main conclusion is that DMT is not only neurochemically active, but also bioactive in general. Its sigma receptor actions are not so revealing for its psychedelic effects, but rather point to a universal regulatory role in oxidative stress-induced changes at the endoplasmic reticulum–mitochondria interface.”

Building on this work, the results of a 2014 study by Szabo et al. indicated that DMT (and 5-MeO-DMT, 5-methoxy-dimethyltryptamine) modulates the inflammatory response via the sigma-1 receptor in humans. In a 2015 review article discussing psychedelics and immunomodulation, Szabo summarized, “The mentioned studies demonstrate and propose new biological roles for DMT, which may act as a systemic endogenous regulator of inflammation and immune homeostasis.”

In 2016, Carbonaro and Gatch summarized the neuropharmacology literature on DMT. They observed that the literature indicated DMT might be useful for treating anxiety, substance abuse, inflammation, and cancer. However, at the time, they cautioned, “Experimental studies have been few and it is premature to conclude that DMT may have clinically relevant uses.”

In a 2018 study using rats, researchers found that DMT (and other psychedelics) increased the number of synapses in the brain. In addition to this, the authors stated, “…serotonergic psychedelics are capable of robustly increasing neuritogenesis [growth of neurons] and/spinogenesis [growth of spines on neurons] both in vitro and in vivo.” These changes were seen in areas of the brain that regulate emotion and mood.

Lifting the veil covering DMT

Although the studies so far are intriguing, DMT has a long way to go. It faces the same stigma that has stalled research on other psychedelic compounds. However, compounds like psilocybin and LSD are being examined in a new light, hopefully laying a path for DMT to follow.

Frecska et al. eloquently summarized the overall paradigm change needed for harnessing the potential benefits of DMT: “…while DMT is a substance which produces powerful psychedelic experiences, it is better understood not as a hallucinogenic drug of abuse, but rather an agent of significant adaptive mechanisms that can also serve as a promising tool in the development of future medical therapies.”

 
Last edited:
shutterstock_1255189054.jpg



The biosynthesis of DMT

by Barb Bauer | Psychedelic Science Review | 23 Jun 2020

Understanding this is more important than ever in psychedelic research because scientists think DMT may be an endogenous neurotransmitter.

DMT is a naturally occurring psychoactive molecule found in plants - genera, including Acacia, Desmodium, Mimosa, Virola, Delosperma, Phalaris, and citrus. DMT is also in the brains of animals including mammals. Scientists have detected endogenous DMT in humans tissues, and some hypothesize that DMT may act as a neurotransmitter.

The biosynthesis of DMT

In 2018, a paper by Barker reviewed the scholarly literature on DMT, including its biosynthesis. Figure 1 below illustrates the biosynthesis steps. It begins with the amino acid tryptophan. Animals must get this amino acid in their diet (i.e., it’s an essential amino acid), whereas plants make it themselves.

- Tryptophan is converted to tryptamine by the enzyme aromatic amino acid decarboxylase (AADC).

- Tryptamine undergoes transmethylation by the enzyme indole-N-methyltransferase (INMT). The enzyme takes one methyl group from a compound called S-adenosyl-methionine (SAM) and attaches it to tryptamine (SAM is acting as a cofactor in the reaction by donating the methyl group). The reaction results in tryptamine being converted to N-methyltryptamine (NMT).

- Step 2 is repeated, with INMT taking a methyl group from another molecule of SAM, and transferring it to NMT. From this, NMT is converted into N,N-dimethyltryptamine (DMT).

3000-biosynthesis-of-DMT-e1592224149844.png

Figure 1: The biosynthesis of DMT 5. AACC = aromatic amino acid decarboxylase, INMT = indole-N-methyltransferase, SAM = S-adenosyl-methionine.

Controlling the biosynthesis of DMT

Things can get out of hand fast if there are no controls on physiological reactions like this in the body. However, it’s not entirely clear at this time how the body regulates DMT production. Figuring it out is complicated by the fact that there may be several forms of INMT (isoenzymes) within a species.8 Since the early 1970s, scientists have reported on an as yet unidentified water soluble and dialyzable inhibitor of INMT which may regulate its activity, and subsequently, DMT biosynthesis.
Continuing Research on DMT

The psychedelic science of DMT is intriguing, important, and provides many paths for research. Barker suggests that “…more research is needed on DMT’s natural role and function and interaction with other neurotransmitter systems.” To that end, he notes "the need for removing regulatory blockades that have existed for 50+ years and stymied DMT research." Noting the endogenous nature of DMT and its potential role as a neurotransmitter, he contends that DMT “deserves a special stature for future research.”

 
Last edited:
shutterstock_1084540790.jpg



Beta carbolines - Newcomers to the science of magic mushrooms

by Barb Bauer. MS | Psychedelic Science Review | 10 Jul 2020

Understanding the pharmacology of magic mushrooms is not just about tryptamines anymore.

In 2019, Blei et al. announced that they had isolated new compounds from four species of magic mushrooms, including Psilocybe cubensis. Of course, the better-known compounds like psilocybin, psilocin, and baeocystin were present in the mushrooms, but also were some compounds called beta carbolines (ß-carbolines). This groundbreaking discovery has changed the magic mushroom paradigm for psychedelic researchers.

ß-Carboline basics

ß-carbolines are naturally occurring alkaloids. They are most commonly known as components in the psychotropic beverage ayahuasca. Examples of the ß-carbolines in ayahuasca include harmine, harmane, and harmaline (note: ß-carboline is also another name for the compound norharmane).

Much of the effect of ayahuasca is due to the ability of ß-carbolines to inhibit MAO (monoamine oxidase) enzymes. This inhibition makes it possible for the psychedelic compound DMT (dimethyltryptamine, another compound in ayahuasca) to move out of the digestive system and enter the circulation.

The ß-Carbolines in magic mushrooms

Blei et al. analyzed extracts from P. cubensis, P. mexicana, P. cyanescens, and P. semilanceata. In the extracts, they identified the ß-carbolines cordysinin C, cordysinin D, harmane, harmol, norharmane, and perlolyrine. These compounds were previously isolated from fungi and plants in genera such as Cordyceps, Peganum, and Banisteriopsis, but this was a eureka moment in Psilocybe research history.

The importance of discovering ß-Carbolines in magic mushrooms

When it comes to an understanding the effects and potential therapeutic applications of magic mushroom compounds, it’s not just about tryptamines anymore. Now scientists have a new path for further discovery. Consider some of what is already known about ß-carbolines. It deepens the complexity of the already challenging magic mushroom chemical landscape.

ß-carbolines are known to play a role in the development of essential tremor (uncontrolled shaking) and have been implicated in Parkinson’s disease. In contrast, a systematic review of the literature conducted by Ferraz et al. in 2019, indicated that ß-carbolines (harmane and harmine, in particular) might have antidepressant effects. The authors summarized their findings by saying,

"In general, β-carbolines modulate 5-HT and GABA systems, promote neurogenesis, induce neuroendocrine response and restore astrocytic function, being effective when administrated acutely or chronically in different animal models, including chronic mild stress protocols."

Some of these effects of ß-carbolines come from their ability to inhibit the uptake of serotonin, dopamine, epinephrine, and norepinephrine via competitive inhibition of the receptors.

In 2018, a review article by Dai et al. focused on the occurrence, structural diversity, and biological activity of ß-carboline monomers and dimers. Their review found that some ß-carboline monomers have many effects, including antitumor, antiparasitic, and antiviral, as well as antioxidant, antidiabetic, and aphrodisiac activity. Interestingly, some ß-carboline dimers have a significantly higher level of bioactivity than their monomeric counterparts.

Further research on ß-Carbolines from magic mushrooms

It’s unknown how ß-carbolines function in magic mushrooms and the ways they may contribute to the overall psychedelic effect. The discovery of these compounds in magic mushrooms will alter the path of psychedelic drug research, perhaps leading to the discovery of even more beneficial effects.

 
Last edited:
shutterstock_1420388516.jpg



Study finds 4-HO-TMT active at the Serotonin 5-HT2A receptor

by Barb Bauer, MS | Psychedelic Science Review | 7 Jul 2020

Surprisingly, 4-HO-TMT has binding affinity similar to psilocin at 5-HT2A.

Researchers at CaaMTech, the University of Massachusetts Dartmouth, and Canopy Growth USA have published an article in ACS Omega detailing the synthesis, crystal structure, and serotonin receptor activity of 4-HO-TMT (4-hydroxy-N,N,N-trimethyltryptamine). This paper is the latest in a series from CaaMTech that have come out since early 2019.

Chadeayne et al. explained that their impetus for this study was gaining a better understanding of other potentially psychoactive molecules in magic mushrooms (aka psilocybin mushrooms) besides psilocin. They noted that the literature indicates the effects of magic mushrooms are different from pure psilocybin, suggesting a synergy such as what occurs between the compounds in cannabis. Chadeayne et al. stated,

“The extracts of ‘magic mushrooms’ demonstrate the same clinical effects as pure psilocybin at dosages that are an order of magnitude smaller, suggesting important activity by other psychoactive molecules or the presence of an entourage effect.”

4-HO-TMT is purportedly the metabolite of aeruginascin, which is found in the magic mushroom Inocybe aeruginascens. The hypothesis about the origins of this metabolite is based on an analogy to the hydrolysis of the prodrug psilocybin to the active metabolite psilocin (Figure 1). Chadeayne et al. hypothesized that aeruginascin undergoes similar hydrolysis to form 4-HO-TMT.

3000-Chadeayne-4-HO-TMT-1.png

Figure 1: Chemical structures showing the similarity that may exist between the conversion on the
prodrug psilocybin to psilocin, to that of aeruginascin to its purported metabolite 4-HO-TMT.


Testing hypotheses

4-HO-TMT has a similar chemical structure to bufotenidine, an active compound in toad venom (Figure 2). Bufotenidine is a strong selective agonist of the serotonin 5-HT3 receptor. As such, Chadeayne et al. worked from the hypothesis proposed by Jensen in 2004, that 4-HO-TMT would show similar activity at 5-HT3.

3000-Chadeayne-4-HO-TMT-2-2048x963.png

Figure 2: The chemical structures of 4-HO-TMT and bufotenidine.

The researchers also tested their prediction that 4-HO-TMT would be inactive at receptors in the serotonin 5-HT2 family. This idea stems from what the authors called “the leading hypothesis” in the scientific community on the activity of aeruginascin at these receptors.

Synthesis of 4-HO-TMT

The synthesis of 4-HO-TMT begins with 4-AcO-DMT (aka psilacetin, 4-acetoxy-N,N-dimethyltryptamine ) fumarate, a functional analog of psilocybin. Below are the basic synthesis steps described by Chadeayne et al.

- 4-AcO-DMT fumarate is methylated in the presence of excess iodomethane, resulting in 4-AcO-TMT (4-acetoxy-N,N,N-trimethyltryptammonium) iodide. Chadeayne et al. call this compound a functional analog of Aeruginascin.

- 4-AcO-TMT iodide is hydrolyzed to 4-HO-TMT iodide in aqueous acetic acid.

- 4-HO-TMT iodide is purified by recrystallization in a methanolic solution.

- Both 4-HO-TMT and 4-AcO-TMT iodide salts were recrystallized from water to obtain their single-crystalline forms.

The authors report that,

"These are the first two quaternary tryptammonium salts ever characterized by single-crystal diffraction."

They confirmed the identity and high purity of the compounds by using NMR and elemental analysis. Further, Chadeayne et al. solved the crystal structures of the iodide salts of 4-HO-TMT and 4-AcO-TMT. In a press release, CaaMTech refers to these compounds by the names Amphoria and Prophoria, respectively.

Serotonin receptor binding affinity of 4-HO-TMT and 4-AcO-TMT

In the next part of the study, Chadeayne et al. tested the binding affinity of 4-HO-TMT and 4-AcO-TMT at four serotonin receptors, 5-HT1A, 5-HT2A, 5-HT2B, and 5-HT3. Table 1 below shows the receptor testing data. For more information on receptor binding values and what they mean, see Binding of Psilocin and Psilocybin to serotonin receptors.
Compound​
5-HT1A​
5-HT2A​
5-HT2B​
5-HT3​
4-HO-TMT​
4,400​
670.0​
120.0​
>10,000​
4-AcO-TMT​
>10,000​
>10,000​
>10,000​
>10,000​
Psilocin​
567.4​
107.2​
4.6​
>10,000​
Psilocybin​
>10,000​
>10,000​
98.7​
>10,000​
Table 1: Ki values in nM for 4-HO-TMT and 4-AcO-TMT. Values for psilocybin and psilocin are included for comparison. Ki values >10,000 imply no activity at the receptor.

4-AcO-TMT showed no binding activity at any of the receptors tested. However, 4-HO-TMT had activity at 5-HT1A, 5-HT2A, and 5-HT2B. Agonist activity at 5-HT2A is regarded by experts as the hallmark of psychedelic activity.

Although 4-AcO-TMT showed less binding affinity at 5-HT2A than psilocin, the authors concluded that the Ki values were comparable. Also, the data showed that 4-HO-TMT has a similar affinity for 5-HT2B compared to psilocybin.

Recall that one of the hypotheses Chadeayne et al. was testing was that aeruginascin’s active metabolite 4-HO-TMT would have an affinity for 5-HT3 like bufotenine. The data showed this was not the case. The Ki for 4-HO-TMT at 5-HT3 was >10,000 nM.

The authors also noted an important observation regarding the activity of 4-HO-TMT at the 5-HT2B receptor. In 2000, Rothman et al. proposed the involvement of 5-HT2B receptor activation with valvular heart disease (VHD). In the Chadeayne et al. study, the data indicated that 4-HO-TMT had significantly less binding affinity for 5-HT2B than psilocin. The comparison of Ki values is considered state of the art for estimating the safety of a compound in relation to 5-HT2B and VHD.

Despite the value gained from these tests, it is unclear why Chadeayne et al. did not test the two compounds at nicotinic acetylcholine receptors. The results may have shed some light on the possible role of aeruginascin (or its metabolite) in the phenomenon known as wood lover paralysis.

What does all this mean for psychedelic research?

This study by Chadeayne et al. makes several significant contributions to the knowledge base of psychedelic science. First, it provides a synthesis method for 4-HO-TMT, which makes the compounds available for further research. Also, the crystal structures they resolved are essential for understanding changes at the molecular level that can affect the physical properties of a compound. From this, scientists can gain more understanding of the behavior of compounds at biological receptors.

Second, the receptor binding data was surprising based on what is known about the chemistry and pharmacology of similar compounds. Despite its close structural similarity to bufotenidine, 4-HO-TMT did not show an affinity for the 5-HT3 receptor. But surprisingly, 4-HO-TMT did have an affinity for the 5-HT2A receptor, showing a magnitude comparable to psilocin.

The latter results present an intriguing puzzle about the potential psychotropic activity of 4-HO-TMT. Chadeayne et al. say that by having a quaternary ammonium group, it appears unlikely that 4-HO-TMT could cross the blood-brain barrier (BBB) and directly affect 5-HT2A receptors in the brain. However, they point to a 2014 study indicating that another quaternary ammonium compound crosses the BBB. In this study, Zacny et al. showed that the compound known as methylnaltrexone or MTNX, crossed the BBB in humans, possibly via cellular transporters. Chadeayne et al. concluded,

"...therefore, psychotropic activity [of 4-HO-TMT] remains a possibility."

Summing up their explorations in psychedelic research thus far, CaaMTech CEO Dr. Andrew Chadeayne said,

"Given the building momentum behind magic mushroom decriminalization and legalization, understanding all of the molecules in those mushrooms and their effects seems like a good idea."

 
Last edited:
soul_ship-14408f7e-c7ce-436a-a263-98789d4ae5e6.jpg



Structure of the iodide salts of DMPT and DMALT

by Barb Bauer, MS | Psychedelic Science Review | 30 Jul 2020

The catalog of compounds for use in psychedelic drug research continues to grow.

The psychedelic research company CaaMTech has published in Acta Crystallographic E, the crystal structures of the iodide salts of the quaternary tryptammonium compounds DMPT and DMALT. This study adds to the growing body of scientific work on structurally related tryptamine compounds that the company has published since early 2019.

DMPT and DMALT are structural analogs of the quaternary tryptammonium toad secretion bufotenidine and the magic mushroom compound aeruginascin. The latter two compounds have recently entered the psychedelic research arena due to their natural occurrence in living organisms alongside other psychedelic tryptamines, including bufotenin and psilocybin.

Chemically, the compounds are known as N,N-dimethyl-N-propyltryptammonium iodide (DMPT iodide), and N,N-dimethyl-N-allyltryptammonium iodide (DMALT iodide). Each crystal structure comprises one cation of the compound and one iodide anion (Figure 1).

Chadeayne-et-al.-DMPT-and-DMALT-iodide.jpg

Figure 1: The chemical structures of the salts DMPT iodide and DMALT iodide.

In the press release, CaaMTech says these are “the world’s first crystal structures of unsubstituted quaternary tryptammonium salts.”

Why solve the crystal structures of these salts?

Dr. Andrew Chadeayne, CEO of CaaMTech and lead author of the study, explained that “In nature, quaternary tryptammonium compounds are present alongside their better-known tertiary amine analogs, like psilocybin or psilocin; but their pharmacology has not been studied. Adding DMPT and DMALT to CaaMTech’s library of unique tryptamines creates new opportunities for drug development and personalized medicine.”

As part of this study, Dr. Chadeayne and his team also published the synthesis methods for making DMPT iodide and DMALT iodide. This advancement in psychedelic chemistry lets labs synthesize the compounds themselves for use in research studies.

Crystal structures in the big picture of psychedelic research

Crystal structures are essential for helping researchers ‘see’ what’s going on at a molecular level. This view gives them a better understanding of how receptors and compounds look and act in nature. Making small changes at the molecular level can translate into significant changes in the effects. Therefore, working with compounds at the molecular level is essential for understanding the mechanisms of how drugs work and how other compounds affect them.

 
Last edited:
shutterstock_226007338.jpg



Metabolizing DMT – The differences between oral administration and smoking

by Barbara Bauer, MS | Psychedelic Science Review | 11 Aug 2020

Research is indicating that a less efficient metabolic pathway plays a role in DMT’s psychoactivity.

Psychedelic Science Review
has previously written about the biosynthesis of DMT in living organisms. Understanding this three-step process is essential in psychedelic research, particularly because some researchers hypothesize that DMT may act as a neurotransmitter. DMT is also of interest to psychedelic researchers because of its role in the effects of the beverage ayahuasca.

But the biosynthesis of DMT is only one side of the coin. How the body metabolizes DMT in the brain is critical to getting the complete picture of the compound’s pharmacokinetics. Although the metabolism of DMT has been extensively studied, examining what is known reveals gaps where further research can focus its efforts.

The all-important route of administration

The metabolism, and therefore the effects of DMT, depend on how it enters the body and then the brain (the route of administration). People taking DMT orally experience virtually nothing. The absence of activity is due to monoamine oxidase enzymes (MAOs) in the body, which rapidly break down DMT into inactive metabolites before enough of it can reach the brain (another way to say DMT has low bioavailability when given orally).

However, if DMT is given orally along with a compound that inhibits MAOs, then some of it has time to pass through the digestive system and reach the brain before being metabolized. This is why ayahuasca causes effects when a person drinks it. Ayahuasca is made using other plants that contain MAO inhibitors like harmine, harmane, and harmaline.

Injecting (intravenously or intramuscularly) or smoking (vaporization and inhalation) DMT bypasses some of the first metabolism in the liver that oral administration undergoes. Therefore, the compound is pharmacologically active when administered via these routes.

The mechanisms and products of DMT metabolism

As previously mentioned, much of DMT that is orally administered is broken down by MAOs. The two primary metabolites are:
  • DMT-N-oxide (DMT-NO)​
  • Indole-3-acetic acid (IAA)​
Other metabolites include:​
  • N-methyltryptamine (NMT)​
  • 6-hydroxy-DMT (6-OH-DMT)​
  • 6-OH-DMT-N-oxide (6-OH-DMT-NO)​
In the early 1980s, Barker et al. discovered that IAA resulted from the oxidative and direct deamination of DMT by MAOs. DMT-NO is produced via N-oxidation of the 2-aminoethyl group on DMT.

Research is showing that there are other metabolic routes for the breakdown of DMT. In 2014, Gomes et al. found that DMT can also be broken down by peroxidase enzymes, resulting in other metabolites including:​
  • Hydroxy-DMT (DMT-OH)​
  • N,N-dimethyl-N-formyl-kynuramine (DMFK)​
  • N,N-dimethyl-kynuramine (DMK)​
In terms of the mechanism at work, the authors stated that “Oxidation of DMT by peroxidases apparently uses the common peroxidase cycle involving the native enzyme, compound I and compound II.”

Different routes = different mechanisms = different effects

Scientists are finding out that there are different metabolic pathways for the breakdown of DMT in the brain, depending on whether it is taken orally or smoked. And, not surprisingly, the data suggest that the subjective effects of DMT depend on the route of administration.

In 2015, Riba et al. observed differences in the metabolic pathways of DMT breakdown in volunteers, depending on whether it was smoked or taken orally. As the researchers predicted, these differences correlated with the subjective effects reported by the users.

The DMT used in the study was extracted from the root bark of the plant Mimosa tenuiflora. Isolating DMT like this for the study is essential because it removes any variables associated with the possible interactions of other compounds in the plant (the entourage effect).

The urinalysis of study participants revealed that oxidative deamination was the primary metabolic route when DMT was taken orally. These people had higher levels of IAA in their urine and no residual DMT. Specifically, IAA comprised about 97% of the compounds in the urine and DMT-NO about 3%. Notably, these subjects had no detectable DMT in their urine, demonstrating the MAO degradation pathway’s efficiency. As expected, these participants reported virtually no psychoactive effects from the oral ingestion of DMT (remember the action of MAOs in the body).

shutterstock_1356429551-1.jpg


Participants who smoked the DMT had higher levels of DMT-NO in their urine, showing more activity in the N-oxidation metabolic pathway. The levels of IAA dropped to 63%, and DMT-NO increased to 28%. Also, unmetabolized DMT accounted for about 10% of the compounds in the urine. This residual DMT suggests that this degradation pathway is less efficient than MAO. All the participants in this group reported “fully psychoactive” effects from smoking the DMT.

The authors summarized the study findings by saying, “As the highly efficient MAO-dependent first-pass metabolism is circumvented by the smoked route, DMT metabolism is directed to the less efficient N-oxidation allowing the access of larger amounts of the parent compound to the central nervous system.”

This study had another significant finding. Analysis of the data revealed a statistically significant inverse correlation between the amount of IAA in the participant’s urine and their scores on the States of Consciousness Questionnaire (SCQ).

The SCQ uses seven subscales to assess several aspects of the mystical experience. The researchers observed the correlation between IAA levels in the urine and the Internal Unity subscale of SCQ. They explained that Internal Unity assesses “the sense of pure awareness and a merging with ultimate reality.” The inverse correlation means that the lower the IAA level in a participant’s urine, the higher their rating was for Internal Unity. The authors summarized these results by saying,

Though preliminary due to the small sample size [n=6], these results suggest that psychoactivity depends on the shift from oxidative deamination to N-oxidation.

Finding more pieces of the DMT puzzle

Science continues to advance the knowledge base of how and why DMT works in the brain. The effects of orally ingesting the DMT-containing brew ayahuasca, despite naturally occurring MAOs in the body, is no longer a mystery.

Now, by isolating and testing DMT in humans, the enzymatic pathways governing its metabolism and psychoactive effects are revealing themselves. Studies comparing the metabolic and subjective effects of different routes of administration of DMT offer additional insights and further reveal the complexity of nature.

 
Last edited:
shutterstock_1476864158.jpg



New biosynthesis of psilocybin and related tryptamines

by Barb Bauer | Psychedelic Science Review | 9 April 2020

Researchers took the psilocybin-making genes from P. cubensis and put them into S. cerevisiae allowing them to produce psilocybin, several of its analogs, and a “new to nature” compound.

Psychedelic research is experiencing many scientific advances for producing psychedelic compounds. This is because trying to get meaningful and usable amounts of psilocybin and psilocin from magic mushroom flesh, for example, is not practical. The levels of these compounds in dried mushrooms are only 0.2% – 1.0% of their dry weight. Having ways to make meaningful amounts of these compounds at a reasonable cost is essential for conducting scientific studies from receptor assays to clinical trials in humans.

The Novo Nordisk Foundation Center for Biosustainability in Denmark recently published a study in the journal Metabolic Engineering describing how they bioengineered Saccharomyces cerevisiae to produce psilocybin and other related tryptamine derivatives.

PSR previously reported on several papers describing both synthetic and bioengineering methods for producing psychedelic compounds and derivatives. Both the new techniques (like the one described herein) and improvements on existing methods are critical for the advancement of psychedelic science. Two of these articles are Scientists Engineer E. Coli to Produce Psilocybin and Scientists Synthesize and Test the Magic Mushroom Compounds Baeocystin, Norbaeocystin, Norpsilocin, and Aeruginascin.

What is S. cerevisiae and why use it?

S. cerevisiae is a type of yeast. Milne et al. chose to work with it “due to its long use in industrial production as well as the fact that it naturally produces very few secondary metabolites or other tryptophan derivates, thereby facilitating simple downstream processing and purification.”

They also chose this yeast species because it naturally expresses cytochrome P450 enzymes that convert tryptamine to 4-hydroxytryptamine (read more about this in the section below, Optimizing the Synthesis of Psilocybin). The presence of the enzyme is a significant cost-savings, according to the authors. It eliminates an extra synthesis step used by other yeasts, which require an expensive substrate during production.

Modifying yeast to produce psilocybin analogs

The research team inserted genetic material from magic mushrooms into S. cerevisiae. Specifically, they cut out certain psilocybin-making genes from the magic mushroom Psilocybe cubensis and put them into S. cerevisiae. They did this by using plasmids as vectors for transporting the genes into the target yeast cells. Once inside, the plasmid incorporates the new genes into the yeast’s DNA (the plasmids are “programmed” to know where to insert them). The modified yeast then starts making psilocybin according to the instructions encoded in the magic mushroom’s DNA.

The yeast also makes psilocybin derivatives

Notably, the researchers also detected several psilocybin derivatives being produced by their bioengineered yeast. The observed psilocybin analogs included psilocin, norpsilocin, baeocystin, norbaeocystin, 4-hydroxytrimethyltryptammonium (i.e., dephosphorylated aeruginascin), and the “new to nature” N-acetyl-4-hydroxytryptamine.

Why was only the dephosphorylated version found and not aeruginascin itself? The researchers noticed that the amount of psilocybin that S. cerevisiae made “was accompanied by a concomitant production of psilocin.” They theorized that the same phosphatase enzymes (of which there are several in S. cerevisiae) acting on psilocybin to produce psilocin were doing the same thing to aeruginascin. The action of these enzymes may also explain the presence of norpsilocin, which is dephosphorylated baeocystin.

Another result of this study was the creation of a biosynthetic method for producing a “new-to-nature” tryptamine derivative, N-acetyl-4-hydroxytryptamine.

The researchers made the new compound by adding the gene for the enzyme serotonin N-acetyltransferase into the DNA of a different strain of S. cerevisiae that produces 4-hydroxytryptamine. The compound is structurally similar to the neurotransmitter N-acetylserotonin (normelatonin), differing only in the position of the hydroxyl group in the 4-position instead of the 5-position.

According to the authors, this was,

"…the successful production of a novel molecule structurally similar to both psilocin and normelatonin with potentially novel pharmacological activity."

3000-N-acetylserotonin-and-N-acetyl-4-hydroxytryptamine2-2048x989.png


Optimizing the synthesis of psilocybin

Initially, the amount of psilocybin produced by the genetically engineered S. cerevisiae was low. The researchers noticed that as the yeast was making psilocybin, there was an increase in extracellular tryptamine. The accumulation of tryptamine suggested to them that its conversion to 4-hydroxytryptamine inside the yeast was not very efficient.

The researchers improved the conversion of tryptamine to 4-hydroxytryptamine by giving the yeast a more efficient gene from P. cubensis. The gene, known as cytochrome P450 reductase, catalyzes the conversion of tryptamine to 4-hydroxytryptamine. After this gene was added to the DNA of S. cerevisiae, the data showed a 29-fold increase in the production of psilocybin and psilocin and significantly reduced levels of extracellular tryptamine.

Continuing research on magic mushroom compounds is needed

This research provides another method for producing meaningful amounts of psilocybin analogs for scientific study and downstream applications. Also, synthesis of the novel compounds N-acetyl-4-hydroxytryptamine and unphosphorylated aeruginascin by strains of S. cerevisiae provides more opportunities for research into their chemistry and pharmacology.

Taking genes from one organism and splicing them into another are well-known techniques. Nevertheless, the application of this technology to create psychedelic compounds represents an exciting new area of research. Other scientists have recently noted that supply problems have limited research into the “minor” tryptamines in magic mushrooms. They also hypothesize that these compounds could have substantial therapeutic value alone or when combined with psilocybin. This recent work discussed above could offer new possibilities for supplying minor tryptamines for further research and development.

 
Last edited:
yin-yang-2346883_1280.jpg



A tale of two tryptamines: N,N-DMT and 5-MeO-DMT in combination with MAOIs

Jaywin Patel, PharmD 2021 and Benjamin Malcolm, PharmD, MPH, BCPP | Spirit Pharmacist | 26 Jun 2020
It was the best of times, it was the worst of times, it was the age of wisdom, it was the age of foolishness, it was the epoch of belief, it was the epoch of incredulity, it was the season of Light, it was the season of Darkness, it was the spring of hope, it was the winter of despair, we had everything before us, we had nothing before us, we were all going direct to Heaven, we were all going direct the other way...
Charles Dickens, ‘A Tale of Two Cities’

A tragic awakening

It was a tragic morning when family and friends awoke at their campsite to find their son and friend’s body in full rigor mortis after consuming ayahuasca and 5-MeO-DMT the night prior.

We’ll never know if the subjective experience of the 25 y/o found Caucasian male found dead at his campground was heavenly, hellish, or paradoxically both, although the headlines reported his death was due to ayahuasca.

But did ayahuasca cause his death?

The medical examiner ruled the cause of death “hallucinogenic amine intoxication” and reported a toxicologic analysis of blood:
  • High levels of 5-MeO-DMT (1.88mg/L)
  • Low levels of DMT (0.02mg/L)
  • Considerable quantities of reversible monoamine oxidase inhibitors (MAOIs) including harmala alkaloids such as harmaline, harmine, and particularly tetrahydroharmine (THH)
  • No other information was given regarding the contents of ayahuasca, although appreciable amounts of 5-MeO-DMT are not found in ayahuasca.
What exactly happened here and how? Let’s figure it out together.

Introduction to DMT, 5-MeO-DMT, and MAOIs

N,N-dimethyltryptamine (DMT) and 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT) are two awe-inspiring masterpieces of evolutionary creation in God’s palace of tryptamine psychedelics. They are both short-acting tryptamines that are rapidly and primarily metabolized by monoamine oxidase (MAO) under normal circumstances. Due to MAO being found extensively throughout the GI tract and liver, DMT and 5-MeO-DMT are heavily metabolized when taken by mouth and psychoactivity is greatly reduced. DMT is not psychoactive by mouth whereas 5-MeO-DMT has mixed reports and may retain ~⅓ of its potency (relative to insufflated).

At+a+Glance_+Tale+of+Two+Tryptamines+DMT+and+5MEO.png


For this reason, they are notoriously used via the inhalation route, resulting in incredibly intense yet short experiences, although have been used by injection (IM/IV), sublingually, or rectally. They are qualitatively quite different, with DMT users reporting interactive-relational experiences with ‘higher beings’ and kaleidoscopic hyperdimensions of incredible visual complexity. 5-MeO-DMT creates a ‘non-dual’ space in higher doses in which the subject-object relationship seems to completely collapse into an infinite totality.

MAOIs+at+a+glance.png


Metabolic breakdown of DMT or 5-MeO-DMT can be prevented by using drugs that inhibit MAO, resulting in psychoactivity when ingested orally and extended duration of effects. The practice of combining MAOIs with DMT has been done extensively over centuries in the tradition of ayahuasca and appears safe when used in ritual context. The practice of combining MAOIs with 5-MeO-DMT has little anthropologic evidence of safety or tradition and appears to be a more recent development in psychonautic exploration. It is apparently unsafe and even deadly to use 5-MeO-DMT with MAOIs. Thus our primary aims are twofold:
  1. Describe in pharmacological terms why DMT is safe to combine with MAOIs while 5-MeO-DMT is not
  2. Discuss use parameters that are likely to modulate the risk of severe adverse effects when 5-MeO-DMT is desired to be used in close temporal proximity to MAOIs
Ayahuasca, DMT, and MAOIs

DMT is found broadly in the natural world. It has been identified in numerous plants as well as an endogenous neurochemical in humans. It is famously found in the bark of acacia trees, phalaris grasses, chacruna (psychotria viridis), and chaliponga (diploterys cabrena). Chacruna and chaliponga are two plants native to the Amazon that are used in combination with banisteriopsis caapi (ayahuasca vine) to create the ritual brew known as ayahuasca. The ayahuasca vine contains harmala alkaloids (beta-carbolines) such as harmine, harmaline, and tetrahydroharmine (THH). Syrian rue (peganum harmala) seeds also contain harmine and harmaline. Collectively, harmala alkaloids are potent and reversible inhibitors of monoamine oxidase (MAOIs). The pharmacokinetic properties of DMT are significantly altered in the presence of MAOIs, leading or bioavailability when ingested by mouth and an extended duration of effect.

DMT_5-MeO-DMT+oral+bioavailability+MAOIs.png


The use of MAOIs renders the user metabolically vulnerable to potentially severe or fatal drug or dietary interactions as they are temporarily unable to metabolize neurotransmitters (serotonin, norepinephrine, dopamine) or biogenic amines (e.g. tyramine). It is well known that MAOIs can cause serotonin syndrome in combination with other serotonergic drugs such as serotonin reuptake blocking antidepressants (SSRIs) or serotonin releasing agents (MDMA). Psychostimulants (cocaine, amphetamine) as well as excessive tyramine from the diet can also lead to severe toxicities, although extreme blood pressures and brain hemorrhages occur as opposed to serotonin syndrome. Use of direct serotonin agonists such as DMT, psilocybin, and LSD has not been shown to cause severe toxicity such as serotonin syndrome with MAOIs. To be fair, there are plenty of reports of severe adverse and life-threatening reactions of ayahuasca that could be consistent with serotonin syndrome, although poison control calls illuminate that 81% are male and the median age was 21. In contrast, ritual ayahuasca drinking has made an expansion around the globe and is drunk by persons of all ages and genders, yet is not associated with these reports.


5-MeO-DMT and Bufo

5-MeO-DMT is found in the secretion of Bufo alvarius toads (Sonoran Desert Toads or Colorado River Toad). It is also found in intranasal snuffs of virola or yopo plants and can be synthetically produced. There is controversy over whether 5-MeO-DMT is endogenously produced in humans. Relative to DMT, there is much less literature available about 5-MeO-DMT and is limited primarily to preclinical experiments and self-report surveys from users. To date of this posting, there is not a single study that’s been done under clinical conditions in humans with 5-MeO-DMT.

The toad genus ‘Bufo’ relates to a commonality among the toads in that they can all produce bufotenine or 5-hydroxy-N,N-dimethyltryptamine (5-OH-DMT). Bufotenine is (much) less psychoactive than 5-MeO-DMT, although may have more vasoconstrictive or physical effects. It is solely the venom of Bufo alvarius that is known to produce 5-MeO-DMT in its venom. Bufo alvarius venom also contains bufotenine as well as bufodienolides. Furthermore, one metabolic pathway 5-MeO-DMT can take in the body includes the production of bufotenine.

Some literature describes 5-MeO-DMT as an emergent substance of abuse. 5-MeO-DMT was not officially made an illicit substance in the United States until 2010, whereas DMT has been illegal since the Drug War began ~1970. A survey of online users did not commonly report using 5-MeO-DMT recreationally and that a spiritual or ‘neoshamanic’ setting is typical for such experiences and it has not been found addictive in preclinical experiments.


Comparative Pharmacology: DMT and 5-MeO-DMT

The exact mechanism for why 5-MeO-DMT is deadly with MAOIs and DMT is not has not been discovered. However, there is enough known about the pharmacology of each substance to postulate several possibilities for why this is so. Much of what we discuss is related to serotonin pharmacology. Thus the major players are featured below for review.

Buckle up, we’re going deep into pharmacologic hyperspace!

5HT+Synapse+Normal+Logo.png


Tryptamines: Structure Activity Relationships

Both have similar chemical structures to one another. In fact, the entire and complete chemical structure of DMT is contained within 5-MeO-DMT. Other tryptamine psychedelics such as psilocybin (4-phosphoryloxy-N,N-dimethyltryptamine) also have DMT contained within it. DMT is both incredibly potent as a psychedelic on its own as well as a ‘psychedelic scaffold’ with many permutations available. For example, legendary chemist Alexander Shulgin synthesized several novel psychedelic tryptamine compounds by making modifications to DMT’s indole ring or the substitution to the (N,N-dimethyl)amine portion for larger substituents.

Tryptamine+Analogues.png


DMT is a ‘small and simple’ tryptamine psychedelic, which tends to confer pharmacologic behavior similar to the neurotransmitter serotonin itself in regards to serotonin transport and breakdown. Small structural tweaks between molecules can make large shifts in the changes in psychoactivity or physical properties of the substances. The apparent and stark difference in safety profile between DMT and 5-MeO-DMT are a good example of this and related to substitution at the 5 position. Serotonin and melatonin are also 5-substituted tryptamines, whereas psilocybin, and LSD do not share this structural overlap (4 substituted or rigidified tryptamines). In essence, 5-MeO-DMT and its metabolite 5-OH-DMT are closer in structure to serotonin and melatonin than other classic tryptamine psychedelics. It suggests substitution at the 5 position leads to small changes in potency (binding) and large changes to efficacy. We’ll explore these soon.

Tryptamine+Psychedelics+5-MeO+Tale+of+2+Tryptamines.png


5HT2A and 5HT1A receptor activation: A balancing game?

There is much to understand about how psychedelics interact with 5HT2A and other serotonin receptors. For example, it is still not understood why DMT is the only tryptamine psychedelic known to not display tolerance upon repeated dosing. It is obvious that there are differences in their interactions with serotonin receptors that likely give rise to differences in psychoactivity and safety profiles. It could be that other tryptamine psychedelics produce lowered responses relative to serotonin, while 5-MeO-DMT does the opposite. We’ll explore some speculative reasons for this now.

Both DMT and 5-MeO-DMT potently and non-selectively stimulate serotonin receptors, although 5-MeO-DMT is ~5x more potent than DMT at 5HT2A receptors. Their psychedelic effects are primarily attributed to action at 5HT2A receptors, although they bind to several other serotonin receptors including 5HT1A receptors. Substitution at the 5 position confers much higher binding affinities at 1A receptors and 5-MeO-DMT binds this receptor in a roughly equipotent manner to its binding at 5HT2A receptors and ~25x more tightly than DMT.

The 5HT2A receptor has been most heavily implicated in the development of serotonin syndrome, although the 5HT1A receptor is also thought to play a role. Interestingly, 5HT1A and 5HT2A receptors seem to have opposite roles on pyramidal neuron membrane excitability, although co-activation resulted in enhancement of response to strong excitation signals, In another experiment in mice, it was found that hyperthermia (one symptom of serotonin syndrome) in response to co-administration of 5-MeO-DMT and harmaline could be reduced by blocking either 5HT2A or 5HT1A receptors. These experiments may suggest opposing roles of 5HT1A and 5HT2A receptors in hallucinogenic action or additive physiologic responses when both receptors are strongly stimulated. In Dr. Strassman’s studies of IV DMT, use of a 5HT1A antagonist pindolol potentiated hallucinogenic effects of DMT.

Pharmacodynamics+DMT+and+5-MeO-DMT.png


Tryptamine bias: DMT and 5-MeO-DMT

5-MeO-DMT’s metabolite bufotenine is not highly psychoactive, but it does bind 5HT2A and 1A receptors with a similar affinity to its parent compound. How could this be? It is likely because (psychedelic) tryptamines display ‘bias signaling’ or functional selectivity meaning that they can preferentially activate different signaling pathways at serotonin receptors than serotonin itself. This could explain why both 5-MeO-DMT and bufotenine (5-OH-DMT) can bind to 5HT2A receptors with similar affinity, yet one is highly psychedelic while the other is weakly psychoactive.

Bias+Signaling+5-MeO+vs+DMT.png


It is clear that even large overdoses of classic tryptamines like psilocybin or LSD usually do not cause death. There is much less data available about DMT overdoses, although one risk assessment estimated needing to ingest ~20 cups of ayahuasca for fatality to occur. In pharmacologic terms DMT, psilocybin, and LSD are all partial agonists at 5HT2A receptors whereas it appears 5-MeO-DMT is a full agonist. Full agonists generally have more severe toxicity profiles than partial agonists, particularly at high doses. The safety profile classic psychedelic tryptamines apparently enjoy could be due to partial agonist activity or only being able to partially activate signaling associated with serotonin that is necessary for toxic responses.

This means that increasing doses of drug runs into a physiologic ceiling effect that is lesser relative to the endogenous binding substance, in this case serotonin. It may prevent or safeguard against excessive activation of serotonin signaling needed to produce lethal serotonin syndrome. It appears that 5-MeO-DMT may be different compared to classic tryptamines in this regard. Serotonin response to different tryptamine psychedelics in cortical areas of rat brains revealed that 5-MeO-DMT was able to produce a supramaximal physiologic response relative to the endogenous neurotransmitter serotonin, and was the most potent tryptamine tested.


Classic mechanisms of serotonin syndrome with MAOIs: Serotonin release or reuptake inhibition

Drugs that increase intrasynaptic serotonin cause serotonin syndrome when combined with MAOIs. Increased intrasynapic serotonin classically occurs via one of two mechanisms: blockade of serotonin reuptake or serotonin release. Selective serotonin reuptake inhibitors (SSRIs) and related antidepressants (SNRIs, select TCAs) are flagship examples of the first way intrasynaptic serotonin is increased. Several other types of drugs have the ability to block serotonin reuptake, although they are typically much less potent than antidepressants at doing so (e.g. methadone, dextromethorphan, pseudoephedrine, tramadol). Yet, they remain contraindications with some reports of serotonin syndrome associated with MAOI combinations. The other classic category of drugs that can cause fatal serotonin syndrome with MAOIs are those that release monoamines such as serotonin, dopamine, and norepinephrine (MDMA, amphetamine). Therefore, either of these properties - serotonin reuptake inhibition or release could confer high risk for serotonin toxicity.

Classic+Mechanisms+of+Serotonin+Syndrome+.png


In rat brains, 5-MeO-DMT was able to inhibit the serotonin reuptake pump, along with several other 5 substituted tryptamine compounds. This provides preliminary evidence that 5-MeO-DMT could produce serotonin syndrome through a classic mechanism (SRIs + MAOIs). DMT is known to be a substrate of the serotonin reuptake pump, but not an inhibitor. 5-MeO-DMT did not release monoamines such as serotonin however, several other 5-substituted tryptamine compounds did. DMT has been shown to be capable of resulting in carrier mediated release of serotonin, however the capability for induction of serotonin efflux is clearly much different than other serotonin releasing agents that are truly dangerous in combination with MAOIs such as MDMA. These results suggest heavy caution should be taken when using 5-substituted tryptamines with MAOIs as they typically display pharmacologic properties of drugs that are known to precipitate severe serotonin syndrome (blockade of serotonin reuptake).

There are surely other differences in these compounds relating to action at serotonin neurocircuitry as well as neurotransmitter systems beyond serotonin (e.g. σ-1 or trace amine associated receptors). To date, these other receptor interactions are not known to play a role in serotonin toxicities, although may have roles in the therapeutic properties of subjective phenomena.


Metabolism of DMT and 5-MeO-DMT

DMT and 5-MeO-DMT rely primarily on MAO for metabolism. This is the reason that they both lack or have greatly reduced oral bioavailability or are not psychoactive when taken by mouth without MAOIs. There are two big differences in the metabolism of DMT and 5-MeO-DMT that can be observed from the comparative diagram of their metabolism below.

The first is a difference in the metabolic routes. DMT is metabolized by MAO, aldehyde dehydrogenase, kyneurinase, and an N-oxidase, whereas 5-MeO-DMT is metabolized by MAO as well as CYP2D6. When 5-MeO-DMT is metabolized by CYP2D6 the metabolite formed is bufotenine (5-OH-DMT). Bufotenine lacks significant psychoactivity by most accounts, although it has vasoconstrictive properties, thus could contribute to physical toxicity . In mice, pretreatment with harmaline resulted in a 3-4x increase in systemic exposure to 5-MeO-DMT as well as a 6-9 x increase in bufotenine.

The second difference is related to the first: Harmala alkaloids inhibit both MAO and CYP2D6, so when used prior to or in conjunction with 5-MeO-DMT, they create a ‘no way out’ metabolic situation for 5-MeO-DMT. This is in contrast to DMT, which has experiments suggesting significant alternative metabolic routes. Harmala alkaloids rely on CYP2D6 metabolism themselves. When persons with different genetic predispositions for CYP2D6, predisposing them to slow and fast CYP2D6-mediated metabolism, it was found that although serum concentrations of the MAOIs doubled, the serum concentrations of DMT remained constant. This suggests alternative metabolic routes for DMT that are able to accommodate for the increased amounts of MAOIs (or that MAO is maximally inhibited at lower concentrations).

The relevance of CYP2D6 and the contributions of bufotenine to toxicity is a difficult landscape to navigate for a few reasons. The first is significant genetic variability in CYP2D6 metabolic status among human populations, with ~10% of Caucasian persons having genes encoding for ‘poor metabolizer’ status at CYP2D6. The second is CYP2D6 is primarily localized to the liver and is best positioned to metabolize drugs consumed by mouth. 5-MeO-DMT is usually consumed via an inhaled route and it’s unclear if CYP2D6 interactions and metabolism would be as relevant for inhaled use of 5-MeO-DMT compared to oral use of 5-MeO-DMT brewed with harmala alkaloids.


Preventing tragedy in awakening

The easiest and most assured way to avoid risks of combining monoamine oxidase inhibitors (MAOIs) with 5-MeO-DMT is to avoid co-ingestion. It is unlikely that this combination offers benefits superior to 5-MeO-DMT alone or DMT in combination with MAOIs such that the risks could be justified from a ‘healing and growth’ perspective. If you are seeking healing or psychospiritual growth then you may turn another direction, as there are much safer routes that offer tremendous benefit. This is not meant to say there is no merit in understanding more about the combination, only that it is better suited only for those with psychonautic intention, extensive prior experience, high risk tolerance, high levels of humility, expert knowledge, and access to immediate emergency care.

The International Center for Ethnobotanical Education and Research (ICEERS) has released a statement on the combination of ayahuasca and Bufo alvarius secretions or 5-MeO-DMT recommending individuals do not use any 5-MeO-DMT by inhalation within 24 hours of drinking ayahuasca. They recommend to avoid oral use of 5-MeO-DMT with ayahuasca. Prior to ayahuasca, 5-MeO-DMT may be used as close to 1 hour prior. They are also clear that there is no lineage of combining these sacraments in a consecutive fashion, speaking to the psychonautic nature of the exploration. Beyond physical risks, there are psychological risks that may need to be considered.

ICEERS’ recommendations are constructed based upon kinetic knowledge of time to drug elimination. For example, the harmala alkaloids have half lives of 2-3 hours for harmine and harmaline and 8-11 hours for THH. In one experiment, THH was still detected 24 hours after ayahuasca use. Due to reversibility of MAO inhibition by harmalas the metabolic capacity of the enzyme is predicted to be recovered quickly and 24 hours should be enough time for most to eliminate the majority of harmalas. However, persons who are poor metabolizers at CYP2D6 or drink ayahuasca several nights in a row may need longer time buffers prior to 5-MeO-DMT use. It should be noted that this is not the case for pharmaceutical MAOIs which should be discontinued for at least two weeks prior to 5-MeO-DMT.

Prior to use of ayahuasca or MAOIs the time for avoidance is dependent upon the contraindicated drug in question, in this case 5-MeO-DMT. Around half of 5-MeO-DMT is eliminated in ~15 mins, thus 1 hours is predicted to be enough time to eliminate almost all of the drug from the system and be ‘safe’ to ingest MAOIs.

Other factors that are likely to contribute significantly to risk of fatal serotonin syndrome are the dose of 5-MeO-DMT or MAOIs administered as well as the route of administration. The reader is referred to Jonathan Ott’s ‘pharmauasca’ bioassays for more information on ‘what’s been done.’ Readers are cautioned that one person’s experience is insufficient to think such combinations are ‘safe.’


Case summary

In the case report that began our tale ‘A Tragic Awakening’ the decedent had more THH in their system than other harmala alkaloids and low levels of DMT, suggesting he drank ayahuasca earlier the evening prior. The high levels of 5-MeO-DMT suggested use at a time period approximately four hours after original use of ayahuasca. Notably, diphenhydramine (Benadryl) was also found in the decedent’s system, which could have also contributed to the toxicity experienced. As a 25 y/o male, he was consistent with both demographics reported in poison control data related to ayhausca and epidemiology of persons that use 5-MeO-DMT. Due to being Caucasian, there was a greater chance he had a CYP2D6 genetic profile causing him to be a poor metabolizer at CYP2D6. Ultimately, ruling the cause of death as hallucinogenic amine intoxication is fair, however ruling the cause of death due to ayahuasca is not accurate.
“I wish you to know that you have been the last dream of my soul.”
― Charles Dickens, A Tale of Two Cities
This death is one example of a tragically preventable occurrence. It is with the last piece of my soul that the pharmacology in this post is helpful in guiding safe decision making in beneficial use and psychonautic exploration of these two incredible tryptamines. I believe the decedent would agree. Younger males discovering the world of DMT, ayahuasca, and 5-MeO-DMT are a high-risk demographic of users and psychedelic communities may consider efforts to target drug safety education towards these persons as they begin their journey with such psychologically and physically powerful substances.

Tryptamine tale take aways

DMT and 5-MeO-DMT are two short acting tryptamines that have gained popularity in use via the inhalation route. Simultaneously, ayahuasca has spread worldwide and use of various tryptamine and MAOI combinations has increased. Careful pharmacologic analysis of effects at SERT, VMAT2, MAO, and 5HT2A/1A receptors can shed light on observed differences in safety profiles of psychedelic tryptamines with MAOIs. 5-MeO-DMT is potentially unsafe with MAOIs for many reasons including full 5HT2A receptor agonism, full 5HT1A receptor agonism, SERT pump inhibition, active metabolites, and lack of alternate metabolic routes when used with harmala MAOIs. Persons should not use 5-MeO-DMT containing products within 24 hours of ingesting MAO inhibiting harmala alkaloids.

 
Last edited:
mdma+neurotoxicity+cover.jpg



Is MDMA Neurotoxic?

by Aimee Sarmiento, PharmD 2021 and Benjamin Malcolm, PharmD, MPH, BCPP | Spirit Pharmacist | 20 Jun 2020

Neurotoxicity resulting in low mood, anxiety, insomnia and problems with cognition has been reported and well documented in literature relating to use of ‘Ecstasy’, but what about pure MDMA (3,4-methylenedioxymethamphetamine)?

MDMA-induced neurotoxicity use has been the subject of intense controversy and even scandal within the medical community over decades. As MDMA assisted psychotherapy (MDMA-AP) progresses toward approval as a legal therapeutic entity, the questions regarding neurotoxicity of MDMA will be clinically relevant as patients and providers consider risks and benefits of use. Moreover, clinical trials are shedding some light on thresholds for neurotoxic effects and providing high quality data to help guide safe use.

In this post we’ll give some background information on what neurotoxicity is, how MDMA may act as a neurotoxin, summarize research findings on MDMA-induced neurotoxicity, and explain use parameters that are likely to prevent development of significant neurotoxic effects.


First of all, what IS neurotoxicity?

Neurotoxicity involves damage to neurons of the central or peripheral nervous system. It can be caused by several things, including drugs. One common misconception is that neurotoxicity exists as an all or nothing event - that is your neurons are either healthy or destroyed, although it is best understood as a symptom spectrum that can be reversible in some cases and irreversible in others. It can begin with small functional deficits, which gradually progress to a larger-scale functional decline or symptoms.

In the case of neurotoxicity as it relates to MDMA, this progression may present itself in an insidious manner, whether it be gradual loss of memory, behavioral and mood issues, or decreased cognitive function over repeated exposures. Of course, large overdoses can produce severe neurotoxicity rapidly. Drugs can produce neurotoxicity through many different types of mechanisms, too many to recount here, although it can be noted that a drug may have a dosing window in which no neurotoxicity is observed due to the dose being modest enough to not push a biological system to such extreme points that toxic responses occur. Even substances we consider completely benign and healthful, such as water, can become neurotoxic under particular circumstances (e.g. drinking 2 gallons in a single hour).

Therefore, our question of MDMA-induced neurotoxicity needs more nuance than ‘is it neurotoxic or not?’ The answer is “yes it is”, but it’s misleading. This is because it inevitably depends on the circumstances of administration. Instead, the better question to ask is ‘under what conditions does MDMA cause neurotoxicity?’


How does MDMA lead to neurotoxicity?

Before discussing how MDMA leads to neurotoxicity, it may be helpful to review how MDMA works. MDMA acts on several neurotransmitter systems, although primarily releases serotonin from presynaptic nerve terminals, resulting in acute depletion of serotonin stores and synaptic flooding of serotonin. This has led researchers to believe neurotoxicity is a consequence of damage to serotonergic neurocircuitry. Subsequently, evidence of neurotoxicity for MDMA is mainly quantified through 5-HT (serotonin) concentrations, activity or presence of enzymes involved with serotonin synthesis or transport (TH and SERT), and visualization of axons immunoreactive for 5-HT or SERT through imaging.

5HT+Synapse+MDMA+MOA.png


Mechanisms of MDMA-induced neurotoxicity

Sometimes it may be as simple as the dose determining if poisonous or not. With higher doses there is a greater chance of profound serotonin depletion. Frequency of use is also probably consequential as dosing on a weekly basis (as some may be if frequenting raves often) is not enough time to allow for recovery from prior use, even in some taking doses in clinical environments. This is rather simple in that your serotonin neuron simply has a capacity for how high the concentrations can be without damage and how fast it can return to balance after use. The two may go hand in hand as frequent use can produce tolerance and result in increased doses when used.

Other explanations are more complicated. For example, MDMA is metabolized to reactive metabolites such as HHMA or HHA, which can cause cellular damage. The serotonin excess may also lead to lasting reduction in gene expression resulting in lowered expressed of 5HT or SERT. The latter hypothesis has challenges the ‘neurodegenerative’ theory of MDMA induce neurotoxicity which argues for direct damage to serotonin neurocircuits and argues instead for a genetic reason for changes in serotonin function.

It is crucial to note that MDMA’s neurotoxic potential is increased by other substances such as alcohol or amphetamine, which are commonly taken in conjunction with MDMA at nightlife events. Concurrent substances can be ingested either advertently or inadvertently as many Ecstasy tablets contain adulterants or misrepresented substances. For example, one analysis found almost half of ecstasy tablets continued < 67% MDMA and that caffeine and amphetamine were common adulterants. Other common adulterants include novel psychoactive substances such as other phenethylamine (amphetamine) based drugs. Many events sell alcohol or may limit water availability. In combination, both alcohol and MDMA exert neurotoxic effects by impairing the survival of neuronal precursors in the hippocampal dentate gyrus, an area of the brain important for neuronal generation, learning, and memory. Beyond alcohol or amphetamine(s), rave scenes can expose users to myriad other substances, such as GHB (gamma-hydroxybutyrate) or ketamine, which may also increase risk. Recently and tragically, other adulterants from diverse drug classes have began to be found on ecstasy tablets such as fentanyl. Ecstasy is recommended to be tested for presence of desired agent (MDMA) and absence of deadly adulterants (fentanyl) by harm reduction organizations. Further information and testing kits for purchase can be found at dancesafe.org

Other aspects of MDMA use that may contribute to neurotoxicity is the recreational environment and timing of administration. Users may stay up all night on MDMA and related stimulants, which could trigger a sleep deprived state. Sleep deprivation is well known to impair cognition and lower mood, thus plausibly leads to some of the observed week after effects. This issue may be minimized by timing MDMA administration such that the user can fall asleep at a regular time or with minimal disruption to habitual circadian patterns. Another factor that may play a role in toxicity is the bioenergetic stress (namely thermal stress) secondary to MDMA being used in hot environments, raising core body temperatures, and being associated with high output physical activity.

Though the jury is still out on exactly how MDMA exerts its neurotoxic effects, the bottom line is that MDMA has several underlying explanations of why it could be neurotoxic as well as evidence from recreational use environments that it can be neurotoxic under certain circumstances. There circumstances tend to work in conjunction within subcultures that use Ecstasy heavily.


MDMA+Neurotoxicity+Harm+Reduction+Graphic+.png


At what point does MDMA become neurotoxic?

The potential for MDMA to act as a neurotoxin is determined by the dose, frequency of exposure, timeline of exposure, concurrent substance use, and overall setting or context in which it is administered.

Doses of 1.7mg/kg (~125mg in a ~160lb adult) have been evaluated in clinical laboratory conditions without evidence of damage to serotonin neurocircuits or functional deficits. In the six phase II trials of MDMA-AP for PTSD conducted thus far, doses have ranged between 75-187.5mg and participants have undergone 2-3 experimental sessions spaced 3-5 weeks apart. In phase III trials, the maximum dose in any session will be 120mg as an initial dose and 60mg as a booster dose (180mg total) and all participants will undergo 3 sessions. In phase II, there were no detected changes in neurocognitive function over a 2-month period following the second and third experimental sessions. During MDMA-AP sessions participants reported a range of short-lived side effects characteristic of amphetamine use and mild serotonin excess. In the week after use fatigue peaked on day 1 and decreased towards day 6, the need for more sleep peaked on day 2 and decreased towards day 6, and low mood peaked between days 2-4 and decreased towards day 6. Notably, in 4 of the 6 phase II studies, depression scores were measured and showed greater improvements than placebo over the course of the trial (supplementary material online e-tables 4-6). In summary, it’s clear that a course of 2-3 MDMA sessions spaced at least a month apart utilizing doses of 75-125mg followed by a booster dose of 37.5mg-75mg 2-3 hours later does not produce persistent neurotoxic effects.

In the previously mentioned study of MDMA samples, the ones that contained >67% MDMA averaged 205mg per dose, higher than the initial and booster doses in MDMA-AP combined (however weights include fillers thus is likely an overestimate of MDMA per tablet). Consumption of multiple tablets (stacking) can bring doses up to 0.5g or more throughout an evening for some users; similar to amounts consumed in all 3 MDMA-AP sessions combined. Stacking was investigated in laboratory conditions by giving 100mg of MDMA followed by another 100mg of MDMA 4 hours later. Results demonstrated that blood concentrations, cardiovascular stress, and temperature were all increased, however subjective intensity of effect was similar due to development of rapid tolerance to pleasurable effects. This may cause users to underestimate the physical stress they are placing on their body with repeated dosing.


So, what does the research tell us so far?

There are two converging streams of evidence here. One from recreation literature and Ecstasy use suggesting potential for neurotoxicity. The other from clinical environments in which MDMA is predominantly therapeutic, has a promising safety profile, and lacks evidence of clinically significant neurotoxicity despite using measures that could reasonably detect such problems. Therefore, models of dosing from clinical trials could be extrapolated and generalized into a harm reduction framework that could be applied broadly to persons who use MDMA.

While phase II trials did detect several cases of persons experiencing side effects of MDMA in the week after their session, the changes were transient and resolved after a week in almost all cases. This suggests that transient changes to mood, sleep, or cognition in the week after use is a side effect that persons should be educated about, while neurotoxicity is an adverse drug reaction associated with ‘incorrect’ or harmful administration patterns.


How can MDMA-induced neurotoxicity be avoided?

The good news is that avoiding or severely limiting the potential for significant neurotoxicity occurring with MDMA is straight-forward, not difficult, and preserves the ability for profound benefits.
  • Limit MDMA doses to the moderate range 75-125mg initially
  • Limit dosing in a single session to a single booster of 50% the original dose
  • Space MDMA sessions at least one month apart
  • Limit dosing to 3-4 times per calendar year
  • Avoid using other drugs and alcohol with MDMA
  • Test ‘ecstasy’ tablets for presence of MDMA prior to use (as well as absence of other drugs such as fentanyl)
  • Limit sleep disruption due to MDMA use
  • Plan regular breaks and sip water if exposed to hot environments (do not drink excessive amounts of water)
  • If you notice a pattern of low mood or cognitive problems in the days after MDMA use that seems to worsen upon repeated administration or is persistent beyond a few days post-use then take an extended break from using MDMA or similar substances
Do antidepressants or 5-HTP prevent MDMA-induced neurotoxicity?

Serotonin blocking antidepressants such as Selective Serotonin Reuptake Inhibitors (SSRIs e.g. Prozac or fluoxetine) can diminish neurotoxic potential of MDMA. However, this is a poor strategy overall because serotonin blocking antidepressants also greatly diminish the subjective experience of MDMA.

Supplements such as L-tryptophan or 5-HTP are able to boost serotonin synthesis. It may not be wise to take large doses prior to MDMA use as it could increase the risk of physical side effects of serotonin excess such as nausea, vomiting, or diarrhea. Conversely, it may be reasonable to use L-tryptophan or 5-HTP for a few days after MDMA use to aid in restoring serotonin levels.

Neither antidepressants nor serotonin precursor supplementation has adequate data suggesting benefits to recommend use. While further research and testing of interventions to reduce either week after side effects or prevent neurotoxic effects is encouraged, current clinical trial data do not support such measures as necessary.


Conclusion

Neurotoxicity can manifest in many forms and severity levels and MDMA appears to be neurotoxic under certain circumstances. High doses or several doses throughout the period of ingestion, frequent use (e.g. weekly use at nightlife events), aduleration, and substance mixing all are implicated in the adverse event of MDMA neurotoxicity. Data from phase II trials of MDMA-AP do not support risks of clinically significant neurotoxicity from MDMA use as side effects were observed to be mild and limited to the week after use, while participants also saw profound improvements in clinical symptoms for PTSD. In summary, MDMA induced neurotoxicity is easy to avoid with moderate doses, adequate time between use, and pure substances without combination.

 
Last edited:
a1c7bc54-f850-4aaf-bbe2-07592b6c3835



MSP-1014 - A Next Generation Psychedelic Medicine

Mindset Pharma | GlobeNewsWire | 3 Jun 2021​
  • MSP-1014 selected based on head-to-head comparison to psilocybin and its active metabolite psilocin​
  • MSP-1014 showed greater safety and efficacy in in vitro and in vivo preclinical models​
  • Cost effective for cGMP manufacturing and IND-enabling studies​
Mindset Pharma Inc., a drug discovery and development company focused on creating optimized and patentable next-generation psychedelic medicines to treat neurological and psychiatric disorders with unmet medical needs, today announced that it has selected its lead clinical candidate, MSP-1014, a differentiated psilocybin-based compound from its Family 1 of novel, patent-pending psychedelic compounds to move forward into current good manufacturing practice (cGMP) compliant manufacturing and investigational new drug (IND)-enabling studies.

“The selection of Mindset’s first lead candidate, MSP-1014, is a significant milestone in our clinical journey. We are excited to enter the final preclinical development step prior to commencing first-in-human clinical trials. Our next generation psychedelic compounds represent the flourishing evolution of therapeutics to effectively address neurological and neuropsychiatric disorders,” said James Lanthier, CEO of Mindset.

“MSP-1014 demonstrates superior preclinical characteristics in head-to-head comparison with psilocybin and its active metabolite psilocin, including increased safety and efficacy, which we believe will potentially result in lower clinical doses thereby indirectly decreasing safety concerns, and manufacturing advantages. MSP-1014 has the potential to be a safer, more efficacious analog to psilocybin, with reduced potential side effects. Given its chemical profile, we anticipate that MSP-1014 will have the potential to treat mood disorders, including major depressive disorder, substance misuse disorders and end-of-life angst associated with terminal illnesses, including cancer. Therefore, we believe that MSP-1014 has the potential to be a first-in-class psychedelic drug candidate. With our first lead candidate selected, Mindset continues to steadily advance its position in developing innovative next-generation psychedelic therapeutics that couple the life-changing potential of psychedelic drugs with novel and patentable new chemical entities that are more predictable, convenient, and safer than first-generation psychedelics."

Mindset’s Family 1 compounds leverage state-of-the art modern drug design using conjugated and deuterated psilocybin and psilocin design strategies to improve on psilocybin’s potential toxicity and pharmacokinetic profiles. Rodent preclinical studies showed that MSP-1014 displayed superior in vivo and safety profiles in mice compared to psilocybin at a range of doses, and 5-HT2A subtype activation in rats. The superiority to psilocybin is due in part to the incorporation of a conjugated amplification moiety (CAM) that enhances 5-HT2A specific effects while reducing non-specific effects. In addition, the manufacturing process of MSP-1014 precludes the phosphorylation step, one of the most challenging chemical synthesis steps in psilocybin manufacturing.

The Company’s Family 1 includes patent-pending compounds comprising two patent applications with priority dates of February 2020. The Company has run a comprehensive range of specialized in vitro and in vivo tests on its patent-pending novel compounds to select the optimal psychedelic drug candidate for progressing towards human clinical trials. These studies have revealed a high rate of success among many of its compounds (see Mindset's news release of April 29, 2021), giving Mindset an exceptional pool of new drug candidates from which it will continue to identify additional suitable compounds. The Company intends to further develop candidates from Mindset Families 2-4 with the goal of selecting additional lead candidates in the near future.

For additional detail on our Family 1 lead candidate MSP-1014, please watch the following interview from Mindset’s management team:



About Mindset Pharma Inc.

Mindset Pharma Inc. is a drug discovery and development company focused on creating optimized and patentable next-generation psychedelic medicines to treat neurological and psychiatric disorders with unmet needs. Mindset was established in order to develop next generation pharmaceutical assets that leverage the breakthrough therapeutic potential of psychedelic drugs. Mindset is developing several novel families of next generation psychedelic compounds, as well as an innovative process to chemically synthesize psilocybin as well as its own proprietary compounds.

For further information on Mindset, please visit our website at www.mindsetpharma.com.

 
Last edited:
Psychedelic-trees.jpg



The crystal structure of LSD bound to the activated 5-HT2A serotonin receptor (5-HT2AR)

by Lily Aleksandrova, MSc, PhD | Psychedelic Science Review | 29 Sep 2020

Scientists have solved the long-awaited crystal structure of LSD bound to the activated 5-HT2A serotonin receptor (5-HT2AR). The discovery reported by Kim at al. in the journal Cell sheds light on how compounds interact with and activate this canonical receptor. 5-HT2AR binding is the key molecular event thought to trigger the vast psychedelic experience and possibly holds broad therapeutic potential. That is why these findings are ground-breaking for understanding how psychedelics work.


The 5-HT2A receptor and its role in psychedelic drug action

The hallucinogenic effects of psychedelics, such as LSD and psilocybin, are related to their ability to activate 5-HT2AR. Simply stated this causes neurons in the human cortex to fire in disorganized patterns, inducing an altered state of consciousness referred to as the “psychedelic trip.”

Serotonin receptors belong to an important family of cell surface G-protein coupled receptors (GPCRs). The binding of an agonist causes GPCR activation. This activation releases the intracellular G protein partner (hence the name “GPCR”), which then launches a specific and coordinated biological response inside the cell.

Importantly, small differences in the drug-receptor interaction can translate into significant changes in the observed clinical effect. Therefore, it is essential to precisely visualize and map out how a compound interacts with its receptor at the molecular level.


Discovery in the making: Scientific stepping stones

To achieve this critical visualization, structural biologists used cutting-edge, high-resolution imaging techniques, earning several recent Nobel Prizes in chemistry for their work in 2012 and 2017. X-ray crystallography measures how X-ray beams reflect off the surface of a drug or protein. Complete crystal structures are the ‘gold standard,’ but they can take years to solve depending on the molecular target’s ease of handling and complexity. Recently, cryogenic electron microscopy (cryo-EM), which literally freezes a molecular event in time, has become a popular and powerful alternative.

Crucial to the Kim et al. work is that the crystal structure of LSD bound to the human serotonin 5-HT2B receptor was reported in 2017. Both studies were done at the lab of Dr. Bryan Roth, a molecular pharmacologist and psychiatrist at The University of North Carolina School of Medicine. Because 5-HT2AR and 5-HT2BR are similar in how they bind to psychedelics, the LSD-2B crystal structure provided important clues as to how LSD might interact with the 2A receptor.

However, until now, scientists had been unable to precisely visualize a psychedelic compound bound to the 5-HT2A receptor. Figure 1 shows the crystal structure of LSD bound to 5-HT2AR from the Kim et al. study.


New study adds missing puzzle piece

Building on Roth’s previous work, Kim et al. and Roth joined forces with Georgios Skiniotis, a structural biologist at Stanford University’s School of Medicine. Using X-ray crystallography and cryo-EM, the two teams created detailed 3D images of how LSD and other compounds bind to and activate 5-HT2AR.

Molecular maps such as these reveal unique and shared mechanisms of interaction for each drug-receptor complex. Such insights add another piece to the puzzle, bringing scientists closer to understanding how psychedelics work on a molecular level. These discoveries also provide additional clues regarding strategies for harnessing the therapeutic power of psychedelics.


Binding details and future psychedelic research paths

Capping off decades of work, Dr. Kuglae Kim of the Department of Pharmacology at the University of North Carolina Chapel Hill (UNCCH) School of Medicine and colleagues have determined the crystal structure of the psychedelic drug LSD bound to the activated 5-HT2A serotonin receptor (5-HT2AR). The discovery reported in the journal Cell sheds light on how compounds interact with and activate this canonical mind-altering receptor. The research team was led by Dr. Bryan Roth, a molecular pharmacologist and psychiatrist at the (UNCCH) School of Medicine and director of the National Institute of Mental Health (NIMH) Psychoactive Drug Screening Program (PDSP).

This article takes a deeper dive into what scientists learned from these molecular maps created using X-ray crystallography and cryo-EM, discussed above. Kim et al.’s findings are ground-breaking, not only for understanding how psychedelics work but also for designing the next generation of neuropsychiatric drugs.


Receptor binding – LSD’s unique interactions with 5-HT2AR

LSD and 25CN-NBOH* act as partial and full agonists for the 5-HT2A receptor (5-HT2AR), respectively, distinguished by the extent of receptor activation. The high-resolution structures published by Kim et al. show how each agonist binds to an overlapping pocket within 5-HT2AR. The study reports subtle but critical differences in binding between the two hallucinogens, supporting previous research.

In 2017, Wacker et al., working with 5-HT2BR, observed that a key amino acid residue (L229ECL2) forms a ‘‘lid’’ over LSD, locking it in place.2 This prolongs LSD’s binding time and, in turn, its biological effects. Now, Kim et al. have found a single residue unique to 5-HT2AR, which is also necessary for the unusually long action of LSD.

For comparison, Kim et al. also studied methiothepin, an inverse agonist with opposing receptor effects and vastly different biological activity. Somewhat surprisingly, this compound had a very similar binding position, only slightly expanding the receptor pocket. Interestingly, one key amino acid was necessary for binding all the compounds to 5-HT2AR.


Receptor activation – LSD biases downstream 5-HT2AR signalling

The structures also revealed important conformational rearrangements that happen during receptor activation. For example, agonist binding displaces a particular 5-HT2AR amino acid, tryptophan W3366.48, which acts as a “molecular switch,’’ turning on the receptor. However, activating the receptor is more complex than just turning it on or off.

Importantly, LSD is also unique in that it is a “biased” agonist. Instead of signalling primarily through the classical G protein-dependent pathway, it preferentially recruits other signalling partners called β-arrestins, with their own set of unique biological effects.

In this study, the drug-receptor complex is seen linked to its G protein (called Gαq) or to β-arrestin. Interestingly, Kim et al. observed that several amino acids within 5-HT2AR are necessary for each of these receptor interactions. For example, a few key amino acid residues underlie 5-HT2AR’s ability to activate Gαq. Curiously, when one is mutated, the receptor binding switches from Gαq to β-arrestin. Such structure-function relationships can begin to explain why an agonist such as LSD might exhibit biased signalling, i.e., a drug stabilized a specific 5-HT2AR signalling complex, ultimately determining its clinical effects.


Implications for neuropsychiatric drug discovery

Psychedelics are emerging as potential therapeutics for various neuropsychiatric conditions, including depression, anxiety, and substance abuse. Although crystal structures can never fully explain how a drug works, they improve our mechanistic understanding of its therapeutic and side effects. Such findings help scientists understand the complex relationship between a drug’s molecular structure, its receptor effects and its clinical action.

The study by Kim et al. opens the door to eliminating the psychedelic effects of these compounds while retaining their therapeutic effects. IF that’s at all possible. In any case, other researchers are already designing drugs with the anti-inflammatory properties of psychedelics without their psychedelic effects. And, Dr. Roth is leading a $27 million project for developing better psychiatric medications without major side effects, announced earlier this year.

In their paper, Kim et al. say their work will facilitate “a structure-guided search to identify more selective and efficacious 5-HT2AR agonists as potential innovative neuropsychiatric therapeutics.” Next, the team is developing tools to screen new drug candidates for their ability to activate the receptor without causing psychedelic side effects.

In a recent interview with Inverse, Dr. Roth exclaimed, “Now we know how psychedelic drugs work – finally!” He added,
"By taking apart the signalling processes involved with an LSD experience piece by piece, we might be able to switch some effects on while keeping others off."

 
Last edited:
5fa3dab4dde5f.jpeg



Jordi Riba and the Challenges of Studying Ayahuasca

by Mario de la Fuente Revenga, PharmD, PhD | Psychedelic Science Review | 9 Nov 2020

Ayahuasca, the quintessential Amazonian psychedelic brew, harbors a complex pharmacology that Jordi Riba helped disentangle.

The popularized term ayahuasca is just one of several used to designate a South-American psychoactive beverage found all over the Amazonian basin. Very different mixtures of plants can be found in ayahuasca, each contributing with different pharmacologically active chemicals to this 'psychedelic soup.' From the lab bench point of view, the “spirit” of the plants comes in various arrangements of carbon, nitrogen, and oxygen一in other words, the different alkaloids they bring to the mixture.

Breaking down the actions of the constellation of chemicals present in the different forms of ayahuasca is a pharmacological nightmare. That was not a deterrent to the late psychedelic researcher Dr. Jordi Riba, who focused most of his research on understanding how these chemicals come together to produce the unique effects of the archetypal Amazonian psychotropic brew.


The challenges of bringing ayahuasca into a clinical setting

Making a sacramental psychedelic brew from the Amazonian rainforest speak into the scientific vernacular is a daunting task. The preparation needs to be standardized following secular protocols to both control the presence of active components and warrant the safety of the participant volunteers. Regarding the latter, very detailed protocols need to be in place to pass the filter of risk-aversive ethics committees.

Out of the most popular psychedelics, ayahuasca is likely the one with the greatest mystical aura associated with it. Preconceived and romanticized ideas relative to its traditional use can greatly affect the manifestation of subjective effects一and even account for a good share of them. Discounting such confounding factors from the effects of actual active material is not an easy task; this is a great challenge that Dr. Riba had to face throughout his career.

Thousands of years of traditional use cannot do away with the gold standard of clinical testing, the placebo-controlled evaluation. To mask the treatment to the eyes of the participants一and facilitate the process of standardization and dosing一Jordi Riba and his collaborators designed a capsule formulation containing freeze-dried ayahuasca procured by the Santo Daime church. This preparation, paired with placebo containing capsules, was crucial to establish unequivocally the psychedelic nature of the effects of ayahuasca.

Jordi Riba, an all-terrain scientist, did not stop there though一he explored thoroughly the multiple dimensions of the pharmacology of ayahuasca and the interaction of its components with human physiology and psychology.


The complex pharmacology of ayahuasca and its manifestations

While most psychedelics can single-handedly produce their effects, the mixture of plants present in ayahuasca is crucial to its mechanism of action. The vine Banisteriopsis sp. contributes in a very special way to make the N,N-dimethyltryptamine (DMT) from Psychotria viridis一or other sources一active for hours upon ingestion. This orally active form of DMT is possible thanks to the inhibition of metabolic enzymes that would otherwise quickly destroy DMT. These crucial actors belong to a group of chemicals known as beta-carbolines (sometimes referred to as harmala alkaloids). Aware of their importance, Jordi Riba thoroughly explored these co-adjuvants to better understand how their presence in the organism could contribute to modulating the psychedelic experience elicited by DMT.

While the fundamental psychedelic effect can be attributed to the interaction of DMT with serotonin receptors, dissecting the whole breadth of the individual contributions of each active component is a challenge of its own.

Over the past few years, Dr. Riba dived one level deeper to study the molecular effects of the main components of ayahuasca. Some of his recent collaborative efforts showed how the different components present in ayahuasca partake in the stimulation of neurogenesis (i.e., the birth and growth of new neurons) in vitro and in the mouse brain. Intriguingly, such effects appear to involve the sigma receptor, an underdog in the pharmacology of psychedelics. The translational relevance of such findings will have to be validated, yet they open intriguing questions worth addressing in the future.

Mounting scientific literature points to the possibility that psychedelics produce lasting effects of potential therapeutic value for mental health. Jordi Riba and collaborators performed both observational and experimental studies that addressed the potential occurrence of enduring effects following ayahuasca use.

A longitudinal study that compared frequent users of ayahuasca within a religious context in a Brazillian population found that regular users scored lower in measures related to psychopathology and higher in scales related to positive life attitude. While a number of confounding factors might affect these measures (i.e., religiosity on either group), some of their most recent reports describe lingering improvements in different domains of mindfulness in the aftermath of an ayahuasca session. Such changes could ultimately be relevant to emotional processing.


Concluding remarks

The rainforest is inhospitable to the man, but so was the lab bench for the most paradigmatic psychedelic brew native to the Amazon. Jordi Riba devoted most of his research career to disentangling the pharmacology of ayahuasca and understanding the nature of its effects in humans. His contributions remain as a living example of the challenges he took on and overcame, making room for ayahuasca in contemporary psychedelic science.

 
Last edited:
dims



Aeruginascin identified in psilocybe cubensis magic mushrooms

by Barbara Bauer, MS | Psychedelic Science Review | 13 Nov 2020

This compound may be more ubiquitous than scientists originally thought. But no one has really gone looking for it yet.

Scientists in Prague, Czechoslovakia, recently identified the compound aeruginascin in Psilocybe cubensis magic mushrooms. In their paper published in Drug Testing and Analysis, Klára Gotvaldová and her research team reported: “an extraction procedure and a UHPLC-MS/MS analytical method for the analysis of psilocybin, psilocin, baeocystin, norbaeocystin, and aeruginascin in fungal biomass.” According to the paper, all these analytes were present in the P. cubensis samples studied. The authors noted that “concentrations of aeruginascin are low and have not been reported before.” This paper is the first report of the compound aeruginascin in any mushroom aside from Inocybe aeruginascens or Pholiotina cyanopus.

Coming up to speed with Aeruginascin

Aeruginascin was first discovered by Jochen Gartz in 1987 in Inocybe aeruginascens, a psilocybin-containing mushroom. Gartz observed that this species had a relatively high concentration of what he hypothesized at the time was aeruginascin. He made another interesting observation in a subsequent 1989 paper. Gartz compared and analyzed the experiences from 24 cases of people who accidentally ingested I. aeruginascens to those who accidentally consumed magic mushroom species with high levels of psilocybin and psilocin.

The people ingesting I. aeruginascens reported only euphoric experiences. Gartz described the effects on those consuming mushrooms with high levels of psilocybin and psilocin as an “often slight and in some cases deep dysphoric mood” accompanied by psychosis, panic, and anxiety. Gartz also found in this study that the levels of aeruginascin in the mushrooms were comparable to those of psilocybin and baeocystin.

Over the years, Gartz’s observations have raised questions about whether aeruginascin modifies the clinical effects of psilocybin, psilocin, or both to increase the likelihood of a positive outcome. Gartz himself stated in his 1989 paper the following hypothesis,​
"Aeruginascin seems to modify the pharmacological action of psilocybin to give an always euphoric mood during the ingestion of mushrooms."

Early this year, Alexander Sherwood and his colleagues synthesized aeruginascin, and a research team led by CaaMTech’s Andrew Chadeayne synthesized 4-OH-TMT, the hydrolysis product of aeruginascin and its putative active metabolite. In their study, Chadeayne et al. also synthesized 4-AcO-TMT, which serves as a prodrug of 4-OH-TMT and provides an alternative to aeruginascin. Although Sherwood did not study the pharmacology of aeruginascin, Chadeayne et al. demonstrated that 4-OH-TMT binds to 5-HT2A receptors with a greater than expected affinity.

In addition, as Sherwood et al. reviewed in their 2020 paper, there exist anecdotal reports of muscle paralysis, weakness, or both in people after eating some species of psychedelic mushrooms containing aeruginascin. Sherwood et al. state, “The speculations on these activities are largely unsubstantiated, though the assumption is tempting given the structural similarity of aeruginascin to bufotenedine, a toad toxin known to be a potent peripherally active 5-HT3 agonist.” The authors added, “…the pharmacological activity of aeruginascin remains unexplored.”

The impact of this new study on psychedelic mushroom research

New discoveries add to and change scientific theories all the time. Recall that the magic mushroom compound norpsilocin wasn’t discovered until 2017 and has since been shown to have potency exceeding psilocin’s. This study from Gotvaldová et al. highlights the importance of understanding the chemical composition of magic mushrooms–including all the active molecules, not just psilocybin.

Their work indicates that aeruginascin is more prevalent in mushrooms than scientists initially thought. And its presence makes sense given scientists’ understanding of the biosynthetic pathway for making psilocybin and other tryptamine compounds in mushrooms.

From a structural standpoint, aeruginascin is simply psilocybin with one additional methyl group (see illustration above). Theoretically, another methyl group could be added to psilocybin via the methyltransferase enzymes that mushrooms use to make psilocybin. From this perspective, it’s somewhat surprising that aeruginascin isn’t present in more species and varieties of magic mushrooms. Unless it is, and it’s just that no one has gone looking for it yet.

 
Last edited:
giphy.gif



Dose-response study of LSD confirms critical role of Serotonin 2A Receptor*

by Lily Aleksandrova, MSc, PhD | Psychedelic Science Reveiw | 26 Nov 2020

Escalating LSD doses lead to more pronounced subjective and autonomic effects, while pre-administration of ketanserin prevents the effects of a high LSD dose.

The therapeutic potential of LSD for a variety of conditions, including addiction, anxiety, and depression, has been highlighted by several recent placebo-controlled trials. Although such studies support LSD’s safety over a wide range of therapeutically relevant doses, they generally only include a single high LSD dose. This underscores the need to develop optimal dosing protocols in order to maximize the therapeutic benefits against the potential risks of compounds such as LSD. Importantly, the first modern study to investigate the subjective and adverse effects of LSD over a range of well-defined doses in healthy subjects was published last month in the journal Neuropsychopharmacology.

Holze et al. administered a range of pharmaceutically well-defined LSD doses to healthy subjects

The small double-blind, randomized, placebo-controlled trial was conducted by Holze et al. at the Liechti lab, University of Basel, Switzerland. It included 16 healthy volunteers (8 male and 8 female, mean age: 29 ± 6.4 years) who underwent six 25 hour-long experimental test sessions, separated by at least 10 days. In each session (given in random order), subjects received either placebo, LSD (25, 50, 100, and 200µg), or 200µg LSD 1h after administration of ketanserin, a Serotonin 2A (5-HT2A) Receptor antagonist (blocker).

It is becoming increasingly important that psychedelic compounds used in clinical research are standardized and developed according to pharmaceutical standards. Importantly, unlike other groups, Holze et al. confirmed the LSD content and pharmaceutical stability of each dose. Specifically, LSD of >99% purity was obtained and prepared as oral solutions of 25 and 100µg according to a set of regulatory standards called “good manufacturing practice.” These doses were then analytically confirmed using techniques previously used at the Liechti lab.

Dose-response Relationships in Terms of LSD’s Subjective and Physiological Effects

As expected, escalating doses administered to healthy subjects led to proportional increases in plasma LSD concentrations (Figure 1). The rate of LSD elimination was proportional to the amount of drug in the body.

Subjective effects over the first 24h after dosing were measured using numerous validated self-rating scales reported useful in predicting therapeutic long-term responses, particularly Oceanic Boundlessness and anxiety measures (Figure 1). LSD subjective effects were dose-dependent and started at the lowest 25µg dose, with a ceiling for “good drug effects” generally seen at 100µg (Figure 1). The 200µg dose did produce higher ratings of blissful state, insightfulness, and changed meaning, but also higher anxiety and ego dissolution.

The average duration of self-reported effects increased from 6.7h at 25µg to 11h at 200µg LSD, due to a faster onset and longer duration with the higher doses. Although participants were blinded to the treatment condition in each session, most of them retrospectively identified the LSD dose they had received correctly. Generally, the 100 and 200µg doses were indistinguishable, while 25µg was readily distinguished from the placebo. Holze et al. also evaluated the so-called autonomic effects of LSD, and found moderate increases in blood pressure and heart rate at the high doses, as previously seen.

In the current study, plasma levels of brain-derived neurotrophic factor (BDNF), a key biomarker for neuroplasticity, tended to be higher post-LSD compared to placebo. However, contrary to a recent Hutten et al. study,5 only the 200µg dose caused a significant increase in BDNF, reaching a peak at 6h.

The critical role of the Serotonin 5-HT2A Receptor in the positive and adverse effects of LSD

Importantly, pre-administration of a 5-HT2A receptor antagonist effectively prevented the normal response of participants to high-dose LSD, as previously reported.6 Specifically, ketanserin (40 mg) reduced the subjective effects of the 200µg dose to levels observed following 25µg LSD. Consistent with this, the LSD + ketanserin combination was identified either correctly or as a low dose by participants after the test session. In addition, the 2A blocker appeared to prevent both the autonomic effects and the increases in plasma BDNF normally observed following 200µg LSD.

This pilot study confirms that LSD’s subjective and autonomic effects in healthy subjects seem to be primarily mediated by serotonin 5-HT2A receptor activation. Although encouraging, this study has several limitations, including the small number of healthy volunteers, which likely have positive expectations and some substance experiences, and the highly controlled research (and not therapeutic) setting. Finally, since ketanserin is a high-affinity but non-selective 5-HT2A antagonist, non-specific actions at other receptors cannot be excluded. Nonetheless, ketanserin effectively blocks the effects of high-dose LSD and maybe at the heart of the LSD “neutralizer technology” announced earlier this year.

Study may assist with dose-finding for future LSD research

Based on these results, Holze et al. propose that an analytically confirmed 100µg dose of LSD may optimize the positive drug effects against the risk of acute adverse reactions, making it most suitable for the treatment of depression and anxiety. Indeed, acutely higher Oceanic Boundlessness and lower anxiety ratings seem to predict a better psychedelic treatment response. Finally, Holze et al. hypothesize that although the 25µg “minidose” used here was clearly acutely psychoactive in most subjects, the authors state that LSD “microdoses” (1–20 µg) are unlikely to impair cognition or produce adverse effects.

After the completion of this study, the Liechti lab announced a research collaboration and licensing agreement with the psychedelic company MindMed. MindMed’s co-founder and co-CEO, J.R. Rahn, commented in a press release: “We see this now completed study as an important stepping stone with highly relevant data to support Project Lucy as the team identifies optimal dose levels of LSD to test in the intended Phase 2b trial of an anxiety disorder.”


*From the article here :
 
Last edited:
ShulCov.jpg.1200x400_q85.jpg



How to change your ability to change

by Abigail Calder, MSc | Psychedelic Science Review | 7 Dec 2020

The monoamine hypothesis of depression leaves many patients without effective help. It’s time for a paradigm shift, and psychedelics may be leading the way.

Until recently, the story of depression went something like this: Due to a complex interplay of factors, including stress and genetics and other imperfectly understood things, the brain lacks enough monoamine neurotransmitters – particularly serotonin. Treatment, therefore, means replacing the missing neurotransmitters with specialized drugs that increase the brain’s serotonin levels.

That works persistently well – for about 12% of patients. And for some doubly unlucky people, antidepressants may be even worse than a placebo. Sometimes, scientific stories are missing a few chapters.

Disorders of rigidity

Rather than a simple serotonin deficiency, depression can also be seen as a disorder of rigidity.

It traps people in negative thinking. Depressed patients tend to interpret ambiguous information negatively, and because life is often ambiguous, this has disastrous results. Patients develop a decidedly negative self-image, pessimistic expectations for the future, and a sense of hopelessness that is difficult to shake.

Although it is difficult to directly relate mental states with biological events, scientists see a potential connection between cognitive rigidity and a kind of biological rigidity, specifically reductions in neuroplasticity. Neuroplasticity, roughly speaking, is the brain’s ability to form new connections and adapt to the constant changes in the world around it. Depressed brains suffer from a pronounced lack of neuroplasticity, particularly in regions relevant for mood regulation and self image. Neurons atrophy and send out fewer dendritic spines to connect with their neighbors, and the brain produces fewer growth factors that stimulate neuroplasticity. This sometimes begins with chronic stress, although other triggers certainly exist. The resulting lack of biological flexibility may be one reason depressive disorders are so hard to break free from.

How to change your ability to change

If impaired neuroplasticity is an important part of depression’s pathophysiology, it stands to reason that stimulating plasticity could reduce symptoms. Antidepressants seem to stimulate neuroplasticity to some extent, but they must be taken chronically, cause objectionable side effects, and take two weeks to work at all. Thus, some scientists are looking to other plasticity-stimulating molecules: those that work well, work quickly, and work after being taken only once. Since 2018, they have a name: the psychoplastogens.

Many psychoplastogens are psychedelics, and indeed, most psychedelics stimulate neuroplasticity. Ketamine, which also rapidly reduces depressive symptoms, is also in this class, as are several molecules with no psychoactive effects at all.8 Psychoplastogens are all characterized by their ability to cross into the brain and rapidly stimulate neuroplasticity, and those that are the most useful do this only in particular circuits and brain regions. Most importantly, changes in neuroplasticity seem to stick around: Ketamine’s antidepressant effects last for 1-2 weeks, while in clinical trials with psilocybin, patients seem to need treatment anywhere between once a month and just once, period.

This theory also has an essential second chapter: psychoplastogens are best combined with psychotherapy. There is still no magic pill that can cure depression. Instead, these drugs may open up a “window of plasticity” during which the brain – the patient, really – is more responsive to therapy. Enhanced neuroplasticity alone is not automatically good; it has to happen in the right neural circuits and, ideally, be paired with therapeutically helpful experiences. Therapy and other supportive activities give the newly plastic brain a positive direction in which to change.

Theory means “a work in progress”

The neuroplasticity theory may explain a lot, but all theories are works in progress. So far, scientists have found evidence that psychedelics stimulate several important elements of neuroplasticity in rodents, and they have reason to conclude that this could also happen in humans. But just as depression isn’t reducible to “too little serotonin,” it is also not simply caused by “too little plasticity.” Psychiatry is moving away from such reductionist views.

It also isn’t clear yet whether impaired neuroplasticity in certain neural circuits directly causes depression, or whether stimulating plasticity is truly a reason that psychedelic therapy works in humans, as opposed to other aspects of the subjective psychedelic experience and its underlying biology. And human research still suffers from “technical difficulties”: measuring neuroplasticity in the lab involves slicing brains up or putting them through what is essentially a professional blender. Although scientists can measure proxies of neuroplasticity without turning anyone’s brain into a smoothie, the resulting data can be hard to interpret. This means that we have little direct evidence of psychedelics stimulating neuroplasticity in humans.

But although the unknown can disappoint, it is also promising. Psychedelics also seem to treat anxiety, addictions, and PTSD in addition to depression, and researchers are also interested in obsessive-compulsive disorder, eating disorders, and even personality disorders. Could these also be “disorders of rigidity?” Psychologists have long wondered if a general pathology underlies seemingly distinct mental health problems, especially because many of these disorders tend to occur together. While one could rightly criticize this idea as too simple, pursuing it in research may nevertheless bring science closer to the more complex truth. Perhaps impaired neuroplasticity isn’t the cause of all of these disorders, but it may be standing in the way when people try to get better.

The take-home message

A new story of depression goes like this: at some point in a patient’s life, the brain’s natural ability to adapt begins to atrophy. This loss of neuroplasticity may happen in circuits that regulate mood, and once they are impaired, the brain ends up “stuck” in a depressive disorder. Restoring neuroplasticity could reopen the highway to health.

As with many theories, this one is attractive because it explains an initially mysterious observation. Ketamine, psychedelics, and other psychoplastogens seem to be astonishingly effective at treating depression, and likely other disorders as well. The neuroplasticity theory of drugs and depression elegantly explains why. If it stands the test of time and science, this theory could lead to more breakthroughs in research on mental health and beyond.

 
Last edited:
Salvia-divinorum-plant.jpg

Salvia Divinorum

A first look at how Salvinorin A works in the brain

Johns Hopkins Medicine | Neuroscience News | 3 Jan 2021

As with other psychedelics, Salvinorin A increased activity across the brain. However, Salvinorin A use resulted in more random and disconnected signaling in the default mode network, a brain network associated with relaxing and daydreaming.

Recent interest in developing therapies from psychedelic drugs has scientists exploring how these chemicals work in the brain. Recently, Johns Hopkins Medicine researchers imaged the brains of people who had vaped Salvinorin A, a drug used in Native Mexican rituals, and found that, like other psychedelic drugs, it increased the communication across parts of the brain.

However, the primary effects of the drug that they observed — as reported in their paper Scientific Reports—suggest that Salvinorin A results in more random or disconnected signaling within the default mode network, which is the part of the brain most active when a person is sitting still, relaxing, daydreaming or otherwise not engaged in externally directed mental exercise.

Salvinorin A is a chemical found in the plant Salvia divinorum that produces powerful out-of-body and amnesia-like experiences over a short time frame similar to the effects of nitrous oxide (“laughing gas”), which is used at dental offices.

“Salvinorin A seems to do all the things that researchers believe happen when other psychedelics (such as LSD and psilocybin) act on the brain,” says Manoj Doss, Ph.D.,postdoctoral research fellow at the Center for Psychedelic and Consciousness Research at the Johns Hopkins University School of Medicine.

“The funny thing is that some researchers have selectively focused on the default mode network when that’s not even where they find the strongest effects of classic psychedelics. In contrast, that is, in fact, where we found the strongest effects of Salvinorin A.”

In the study, 12 men vaped Salvinorin A crystals and then were scanned by functional magnetic resonance imaging (fMRI) for the next 15 minutes to measure brain activity across networks in the brain. The researchers evaluated the activity within and among eight brain networks: three involved in vision and five others responsible for moving and touch, processing information from the outside world, paying attention, reward evaluation and activating the default mode network when the mind is at rest.

Most of the fMRI-scanned networks seemed to communicate more with one another when influenced by Salvinorin A than when the drug was not present, but they communicated less within themselves. The researchers observed particularly decreased synchronization of the default mode network, meaning that the brain’s electrical signals became more random and unpredictable.

In future experiments, the research team plans to compare these findings about Salvinorin A’s impact on the brain with data from imaging done on brains under the influence of other psychedelic drugs.

 
Last edited:
51bc885a87910a362d0ddd6212007c0ae79c2566-1000x668.jpg



Assessing synthetic cannabinoids and cathinones in neuropsychiatric toxicological emergencies*

Sameer Hassamal, MD and Sunita Hassamal, MD | Psychiatric Times | 15 Jan 2021

Synthetic cannabinoids and cathinones are novel psychoactive substances (NPS) that have surreptitiously emerged on the market to mimic the effects of established drugs. These designer substances are gaining popularity due to their widespread availability and safety misperceptions. Temporal trend epidemiological data analyzed by the Centers for Disease Control indicate that acute poisonings related to synthetic cannabinoids and cathinones are increasing in the United States.

The rise in synthetic cathinone and cannabinoid use among patients susceptible to adverse neuropsychiatric reactions has been linked to a wide range of unpredictable effects, including agitation, psychosis, and delirium.3 Emergency department (ED) psychiatrists are on the frontlines of care, and must provide the initial management and stabilization of the complex and challenging toxic effects of these drugs. Prompt recognition and management is essential to reduce morbidity and mortality.


Table 1. Common Adverse Neuropsychiatric Effects of Synthetic Cannabinoid and Cathinone Intoxication.


Synthetic cannabinoids

Colloquially known as spice, these designer synthetic cannabinoid receptor agonists have potent intrinsic activity at the cannabinoid (CB) 1 and 2 receptors. The compounds were originally developed to study the endocannabinoid system, but they emerged as a drug of abuse in 2008.5 Initially designed to mimic the effects of delta 9-tetrahydrocannabidiol, synthetic cannabinoids are 2 to 100 times more potent than marijuana at the CB1 and CB2 receptor, resulting in a wide range of adverse neuropsychiatric symptoms. (Table 1)

Furthermore, synthetic cannabinoids are often adulterated with additional ingredients to either enhance or attenuate the drug high, resulting in clinically unpredictable toxicological profiles. As a consequence, there is no specific toxidrome associated with synthetic cannabinoid intoxication, and patients will typically present with a constellation of sympathomimetic, anticholinergic, and marijuana toxic effects. (Table 2). The most common sympathetic nervous system cardiovascular effects are tachycardia (46 percent) and hypertension (21 percent). The anticholinergic effects closely overlap with signs and symptoms of a sympathomimetic toxidrome.

Distinguishing symptoms specific to anticholinergic toxicity include: dry skin, cotton mouth, absent bowel sounds, blurred vision, and urinary retention. Moreover, patients may additionally present with marijuana intoxication symptoms (ataxia, delayed reaction time, and conjunctival injection). Laboratory studies are indicated in cases of severe intoxication with agitation or seizures due to the resulting life-threatening metabolic derangements.


Table 2. Common Signs and Symptoms of Synthetic Cannabinoid and Cathinone Toxicity


Rates of synthetic cannabinoid use are increasing in the United States, with the lifetime prevalence estimated to be between 0.2% and 4%. Moreover, between January 2016 and September 2019, ED visits related to synthetic cannabinoid toxicity significantly increased in the country’s Midwestern and Northeastern regions. Overall, synthetic cannabinoids harms are increasing, and psychiatrists need to be cognizant about the clinical presentations and evidence based treatments.

Synthetic cathinones

Colloquially known as bath salts, these agents are synthetic psychostimulants structurally related to cathinone, a monoamine alkaloid found in the Khat plant. As substituted phenethylamines, synthetic cathinones are structurally and pharmacologically similar to amphetamine and 3,4-methylenedioxymethamphetamine (MDMA). These agents are commonly abused for their euphoric, empathogenic, and stimulating effects. The most commonly abused synthetic cathinones are 4-methylmethcathinone (mephedrone), 3,4-methylenedioxy-N-methylcathinone (methylone), and 4-methylenedioxypyrovalerone (MDPV). Synthetic cathinones produce major effects on the Central Nervous System (CNS), and are linked to adverse neuropsychiatric complications (Table 1). Synthetic cathinones stimulate the release of dopamine and inhibit the reuptake of epinephrine, norepinephrine, and serotonin inducing a broad range of sympathomimetic, serotonergic, and hallucinogenic adverse toxidrome effects (Table 2). The most frequent sympathetic nervous system cardiovascular effects are tachycardia (46 percent) and hypertension (21 percent). The hallucinogenic effects (perceptual distortions, depersonalization, synesthesia) can be highly distressing, which often prompts emergency psychiatric care. Ancillary lab studies should be ordered in cases of severe agitation because of the increased risk for acute kidney injury, rhabdomyolysis, lactic acidosis, stroke, and intracranial hemorrhage.

Self-reported prevalence of synthetic cathinone use is an estimated 0.05%. Current estimates are imprecise, however, as established surveys and standard immunoassay testing are not sensitive for detecting cathinones. The most recent data from the Drug Abuse Warning Network report indicates 22,904 ED visits were related to synthetic cathinone toxicity in 2014. The frequency of ED utilization is likely increasing, as synthetic cathinones are steadily becoming more popular despite the potential harms.

General management principles

The diagnosis of intoxication is based on a history of exposure and presence of characteristic signs and symptoms. Patients may unknowingly or inadvertently administer synthetic cannabinoids and cathinones, as these compounds are oftentimes adulterated with other illicit drugs such as MDMA. Although there is no specific toxidrome, patients will typically present with signs and symptoms of adrenergic neurohumoral activation. The neurotoxic symptoms (including aggression, confusion, and psychosis) may overlap with a primary mental illness, and psychiatrists must rely on the clinical history and physical exam findings to accurately diagnose toxicity. Urine immunoassay testing is the most common method to screen for substance use. In the setting of acute intoxication, however, urine immunoassay testing has a low positive predictive value for synthetic cannabinoids and cathinones. While gas chromatography/mass spectrometry (GC/MS) is considered the gold standard for detecting substances, the molecular geometry of synthetic cannabinoids and cathinones is everchanging, making it increasingly difficult to directly identify these compounds. Moreover, GC/MS is not readily available in all emergency hospitals, and the results may not return in a timely manner to aid in the diagnosis process. An atypical onset of psychosis and agitation alongside hyper-sympathetic activation and a mixed toxidrome is strongly suggestive of intoxication.

Because of the variability in clinical presentations and lack of diagnostic testing, psychiatrists should strongly consider the possibility of intoxication in any patient with no known psychiatric history, a negative urine drug screen, and no known contributing medical mimickers (presenting with a mixed toxidrome). Patients will not necessarily fit neatly into a particular toxidrome, but rather present with overlapping signs and symptoms from multiple toxidrome classes. Consequently, differentiating synthetic cannabinoid and synthetic cathinone toxicity can be challenging. Regardless, supportive care and symptom management is the mainstay of the treatment. Benzodiazepines are considered the first line of treatment to reduce the sympathomimetic neural stimulation (including hypertension, agitation, and seizures).

Agitation

Agitation is the most common neuropsychiatric complication associated with synthetic cannabinoid and cathinone ingestion. Katz and colleagues reported that 40% of intoxicated patients evaluated in an ED will experience severe agitation. The spectrum of psychomotor agitation can be objectively measured and identified with a standardized rating scale such as the Behavioral Activity Rating Scale (BARS). For patients with mild-moderate agitation, the patient may initially be calmed with verbal de-escalation techniques, and environmental modification. For patients with moderate-severe agitation, benzodiazepines remain the first line treatment for reducing moderate to severe agitation. Among the benzodiazepines, midazolam and lorazepam are considered the initial drugs of choice for decreasing agitation because of their rapid onset of action, short half-life, and parenteral route of administration availability. Antipsychotics are commonly administered with benzodiazepines to manage the psychotomimetic effects. However, risks associated with administering antipsychotics in acute poisonings settings must be carefully considered. Specifically, antipsychotics: may increase the risk of seizure induction; have anticholinergic effects; impair thermoregulation mechanisms; and prolong the QTc interval. Serotonergic sedatives such as fentanyl should be avoided because of the increased risk of serotonin toxicity associated with synthetic cannabinoids and cathinones.

Excited delirium

A subset of patients will develop excited delirium following ingestion of psychostimulants. A systematic review reported that 25% to 94% of patients diagnosed with excited delirium were recently exposed to synthetic cannabinoids and/or cathinones. The American College of Emergency Physicians has provided guidance on evaluating excited delirium: a minimum of 6 out of 10 clinical criteria are required to make a diagnosis. If undertreated, the case-fatality rate is estimated to be 20%, partly due to catecholaminergic cardio-respiratory failure. The cornerstone of treatment is rapid sedation to reduce the sympathomimetic outflow and facilitate lifesaving treatments.

To date, no large scale randomized controlled trials have evaluated pharmacological treatments for excited delirium. Nonetheless standard of care is to administer benzodiazepines and antipsychotics to decrease psychomotor agitation. In the emergency setting, IM ketamine (4-5 mg/kg) is increasingly used for sedation of agitated patients with excited delirium. Initial studies in the pre-hospital setting (emergency medical services) reported that intubation rates following ketamine administration were as high as 60%. Emerging literature supports that IM ketamine administration in the hospital setting (emergency department) is a safe and effective treatment for excited delirium while not significantly affecting the cardiopulmonary system. Mankowitz and colleagues reported that more patients required intubation after ketamine administration in the pre-hospital setting compared to the hospital setting. Prospective studies conducted in the emergency department have demonstrated that there is no significant difference in incidence of complications between IM ketamine and IM haloperidol. The side effects of single doses of IM ketamine for agitation are generally transient and self-limiting: emesis, hypersalivation, recovery agitation (also known as emergence reaction), hypertension, and tachycardia. Moreover, ketamine has other potential benefits, including a faster onset to sedation compared with benzodiazepines and antipsychotics. There are observational reports of ketamine exacerbating psychosis in susceptible patients. Larger studies, however, have concluded that acute- and short-term administration of ketamine for agitation does not increase psychiatric inpatient admissions or ED psychiatric evaluations. Physical restraints should be used as a last resort, primarily as a bridge to pharmacologic treatment, as prolonged use of physical restraints is likely to worsen delirium and rhabdomyolysis, leading to renal failure, electrolyte abnormalities, and arrhythmias.

Psychosis

Synthetic cannabinoids have been linked to psychotic reactions, such as paranoia, catatonia, dissociation, auditory, and/or visual hallucinations. Longitudinal studies have reported that consumption of potent cannabinoids quintuples the risk of a psychotic episode. The psychotomimetic effects are usually transient and resolve within 5 to 8 days; however, up to one-third of patients will transition to schizophrenia. Although there is no established antidote, preliminary reports suggest that cannabinol, an allosteric modulator at the CB1 receptor, may reverse the neurotoxic effects.

Similarly, synthetic cathinones have pro-psychotic effects, potentially triggering a psychotic state in vulnerable groups of patients. An observational study reported that synthetic stimulants were implicated in approximately 25% of ED visits involving drug-related psychosis. Specifically, mephedrone was the most common synthetic cathinone involved in stimulant psychosis, accounting for approximately 20% of stimulant related psychotic cases. Synthetic cathinone induced psychosis is typically brief and resolves within days to weeks; however, 25% of patients will transition to a schizophrenia spectrum disorder.

The psychotomimetic symptoms can be controlled with first- and second-generation antipsychotic medications with careful consideration of side effect profiles. Among the first-generation antipsychotics, the butyrophenone neuroleptics IM haloperidol and IM droperidol are reasonable pharmacotherapeutic options. In comparison to IM haloperidol, IM droperidol has a faster onset of action, rapidly preventing and treating emergent psychotic agitation within 5 to 10 minutes of intramuscular administration.36 Some expert panels have recommended avoiding butyrophenone neuroleptics in cases of substance-induced psychosis due to the pro-convulsant properties, cardiotoxic effects, and extrapyramidal symptoms. Among the second-generation antipsychotics, p.o. olanzapine has a track record of treating NPS-induced psychosis, and pro-drug websites have touted olanzapine as the ideal medication to terminate so-called “bad trips.” In spite of its efficacy, olanzapine has anticholinergic and central thermoregulatory effects impairing core temperature regulation. In theory, p.o. or IM aripiprazole and ziprasidone are probably the safest antipsychotics to administer because of the relatively favorable safety profile, and negligible anticholinergic and seizurogenic effects.

Despite the infamous concerns about QTc prolongation, epidemiological studies do not support a correlation between ziprasidone and torsade de pointes. In many instances, detoxification with benzodiazepines and rest will often quell the substance-induced symptoms without the added need for antipsychotics. Given the risks associated with neuroleptics, especially in antipsychotic naïve patients, it may be best to consider a benzodiazepine as the first line agent to reduce the psychotic agitation. Antipsychotics can be initiated if the psychosis continues to persist after the detoxification period, or if the patient has a underlying primary psychotic illness.

Concluding summary

Synthetic cannabinoid and cathinone harms are increasing, and psychiatrists need to be cognizant of the clinical presentations and evidence-based treatments. Synthetic cannabinoids and cathinones have pro-psychotic and agitative effects, and the toxic reactions can mimic a primary mental illness. There is no definitive diagnostic test to differentiate primary versus secondary drug induced mental illness, and ED psychiatrists must rely on the history and physical examination. In a subset of cases, the overstimulation of catecholamines may result in an excited delirium. Benzodiazepines are considered the first line treatment to reduce the sympathomimetic neural stimulation.

*From the article here :
 
Last edited:
brain-neuron-receptor.jpg



New psychedelic compound synthesized for clinical use: 5-MeO-DMT Succinate

by Barbara E. Bauer, MS | Psychedelic Sciebnce Review | 18 Jan 2021

Leading scientists continue to remove roadblocks by developing methods for synthesizing psychedelic compounds for further research.

In a March 2020 Psychedelic Science Review article, Dr. Alexander Sherwood of the Usona Institute noted that psychedelic research had been impeded by a lack of access to pure, well-characterized research chemicals. The article also covered his addressing this unmet need by publishing synthetic methods for making several psilocybin analogs.

Dr. Sherwood is the lead author of a recently published paper in ACS Omega titled “Synthesis and Characterization of 5-MeO-DMT Succinate for Clinical Use.” The paper details a synthetic route for making the succinate salt of 5-MeO-DMT, a psychotropic compound found in the venom of some toad species. This effort is another step forward in providing pure, well-characterized research chemicals for use in psychedelic research.

In the paper, Dr. Sherwood explains the critical nature of the work. Currently, there are only a handful of psychedelic compounds in clinical development, and improving access to pure compounds would enable the needed research studies and clinical trials.

Why make the succinate salt of 5-MeO-DMT?

The paper explains in detail the rationale behind the research team’s decision to focus on 5-MeO-DMT succinate. First, they examined the available evidence to determine the best route of administration for the compound. They found chemical, physiological, and practical drawbacks to methods such as smoking, vaporizing, transdermal, and intravenous administration.

They followed the research to a 2020 study published by Uthaug et al. This study reported that the intramuscular (IM) injection of 5-MeO-DMT in a naturalistic setting was “…associated with lower and less doses, lower frequencies of reporting reactivation, a higher frequency of physical tension release, and longer onset of acute effects” compared to vaporization. Sherwood et al. noted that IM injection also allows for precise metering of doses.

Working from a 2018 study by Gupta et al. the team stated that “…a range of pharmaceutically acceptable salt forms were considered from acids with sufficient pKa difference to fully protonate 6 [5-MeO-DMT], including the counterions chloride, sulfate, fumarate, succinate, maleate, lysate, oxalate, benzoate, tartrate, mesylate, or acetate.” Testing narrowed the field down to four salt forms: hydrochloride, sulfate, fumarate, and succinate.

Various issues with water solubility and chemical stability were identified with three of the salts. The succinate form was chosen due to its high water solubility and having less potential for chemical reactivity. The latter is especially important during autoclave sterilization, which may be required for preparing sterile solutions.

5-MeO-DMT Succinate synthesis overview

As part of this study, Sherwood et al. developed an improved method for synthesizing 5-MeO-DMT freebase, the starting material for making the succinate salt. They explained, “The key features of the developed process were an optimized Fischer indole reaction with advantageous inclusion of acetonitrile cosolvent to provide crude freebase.” Also noted were “…greener solvent choices with an intermediate purification via filtration through a silica pad.”

Their new synthesis method for making the 5-MeO-DMT succinate API (active pharmaceutical ingredient) resulted in a 49% overall yield and 99.86% purity of the final product. The method includes the use of methanol, activated charcoal decolorizing, and purification using an acetone slurry.

Along with the succinate acid salt, the team identified a degradation product, the N-oxide form of 5-MeO-DMT. However, research by Shen et al. in 2010 indicates that the N-oxide form is also a 5-MeO-DMT metabolite. As such, Sherwood et al. explained that this allows some flexibility in how much N-oxide can be in the API.

In concluding, Sherwood et al. stated, “The first production run has provided sufficient API to meet current clinical and nonclinical needs to enable first-in-human clinical trials with 6 [5-MeO-DMT].”

Understanding psychedelic drug pharmacology is critical to psychedelic therapies

Interestingly, Dr. Sherwood notes that psychedelic drugs are generally non-selective, “simultaneously interacting” with several serotonin receptors in a “synergistic way,” resulting in “variable degrees of synergistic polypharmacology.” Given this breadth and variability of psychedelic pharmacology, it is feasible that various psychedelic compounds could be used within different contexts or for treating different mental and physical conditions.

Until psychedelic science reaches the level of understanding necessary for making precise formulations, it seems that one course of action is making and studying a wide range of psychedelic compounds. This recent paper by Sherwood et al. serves the goal of providing access to another compound (5-MeO-DMT) with significant clinical potential.

*From the article here :
 
Last edited:
pcblsd_withskull.jpg



The structure-activity relationship of psilocybin analogs

by Barbara E. Bauer, MS | Psychedelic Science Review | 12 Feb 2021

Seventeen 4-acetoxy and 4-hydroxy substituted derivatives “have psilocybin-like pharmacological properties.”

Structure-activity relationships (SARs) are essential in medical and pharmaceutical research because they show how the chemical structure of a compound relates to its biological activity. When looking at the big picture, SARs underscore the importance of researching and understanding the chemical variability of all the naturally occurring psychedelic compounds in an organism.

In December 2020, a team of scientists led by Dr. Adam Klein published the results of their work studying the SAR of 17 structurally related tryptamine compounds, all analogs of the magic mushroom compound psilocbyin.1 According to the authors, “there has been little systematic investigation of the structure-activity relationships of 4-substituted tryptamine derivatives,” and in the case of the 4-substituted derivatives, “almost nothing is known about their pharmacological effects.”

The tryptamines used in the study were all 4-substituted N,N-di-alkyltryptamines. These compounds can be further divided into two categories, 4-acetoxy (4-AcO) substituted and, 4-hydroxy (4-HO) substituted.

The salt forms of the compounds in the study were:​
  • 4-AcO-DMT fumarate​
  • 4-HO-DMT (psilocin)​
  • 4-HO-DET hydrochloride​
  • 4-AcO-DET fumarate​
  • 4-HO-MET hemifumarate​
  • 4-AcO-MET fumarate​
  • 4-HO-MPT fumarate​
  • 4-AcO-MPT fumarate​
  • 4-HO-EPT 3:2 fumarate​
  • 4-AcO-EPT fumarate​
  • 4-HO-DPT hemifumarate​
  • 4-AcO-DPT fumarate​
  • 4-HO-MIPT hemifumarate​
  • 4-AcO-MIPT fumarate​
  • 4-HO-DIPT hydrochloride​
  • 4-AcO-DIPT acetate​
  • 4-HO-MALT 3:2 fumarate​
The team measured the in vitro and in vivo activity of these compounds. They conducted calcium mobilization assays for assessing functional activity at several serotonin receptor subtypes, particularly 5-HT2A. They also used the head twitch response (HTR) test in mice as a behavioral model for LSD-like psychedelic activity.

Below is an overview of the study findings.​
  • All compounds behaved as full or partial agonists at the serotonin receptors, displaying similar potencies at 5-HT2A and 5-HT2B.​
  • Some compounds with bulkier N-alkyl groups (e.g., N,N-diisopropyl) had lower potency at 5-HT2C and higher 5-HT2B receptor efficacy.​
  • Compared to their 4-HO analogs, the 4-AcO compounds exhibited reduced in vitro 5-HT2A potency by about 10- to 20-fold.​
  • In contrast, the 4-AcO and 4-HO compounds exhibited similar agonist efficacy in vivo, as shown in the HTR experiments, “suggesting that O-acetylated tryptamines may be deacetylated in vivo, acting as prodrugs.”​
Klein et al. summarized the results by saying,​
…the tryptamine derivatives have psilocybin-like pharmacological properties, supporting their classification as psychedelic drugs.

Conclusion

SARs have proven to be valuable tools in other recent studies on psychedelic compounds. These include kratom,2 tabernanthalog,3 and the discovery of an anti-inflammatory pharmacophore in rats, 2C-H.4 Klein et al.’s use of SARs has added essential information to the growing knowledge base of the pharmacology of psychedelics. This fundamental knowledge forms a base from which a myriad of other investigations can be conducted.

 
Last edited:
Top