OPEN Foundation

Background articles

[Interview] Alicia Danforth: Helping autistic adults navigate the social world with MDMA-assisted therapy

AliciaResearch with autistic children and teens was a promising and controversial research area during the first wave of research with psychedelic substances. The first investigator to pick up this thread again in the current era was Alicia Danforth. She currently studies the effects of MDMA-assisted therapy on social anxiety in adults on the autism spectrum in a study led by Charles Grob.

How did you begin doing this work?

It’s not a typical story. I made a mid-career transition. When I was working as a project manager in software development, I was out with co-workers one night, and they were talking about doing ecstasy at raves. I was intrigued, because the ones who had already done it spoke about it so favourably. But it also made me nervous, because I had been exposed to all the propaganda, and I thought that maybe my programmers were going to destroy their brains. It was important for me to research ecstasy and be able to let them know why they should not be doing it. So I got a copy of Julie Holland’s book, Ecstasy: The Complete Guide. I had a transformational moment when I read the testimonies written by young men who she identified as having had a schizophrenia diagnosis. They talked about what their experiences with ecstasy had done for them, how it gave them hope, how it helped them feel that they might be able to have an experience of connecting to others and feeling more normal. Something that was dormant in me became fully awake in that moment. I could not accept that a substance that millions of people have used and that has helped so many of them who don’t have other effective support and treatment was illegal and could not be studied by researchers in reputable institutions.

So I called Dr. Holland and told her that I wanted to help educate people and promote scientific inquiry. She directed me to Rick Doblin at MAPS, who referred me to Dr. Charles Grob at the Los Angeles Biomedical Research Institute at the Harbor-UCLA Medical Center. It was one of these synchronous life events, because I had just taken a job a mile or so away from Dr. Grob’s office, and he was doing end-of-life anxiety research with psilocybin at the time. I approached him as somebody with no qualifications whatsoever, just to help out as a volunteer, to support what he was doing. We considered options and discovered that I could help him with PowerPoint presentations. In an age where people gave their presentations on their laptops, he was still using a 35mm slide carousel…! Working for him gave me an opportunity to immerse myself in the literature, in the history and in the science, and at every step I became more intrigued and inspired to do more. After about two years, his research assistant on the psilocybin study needed to relocate, and they had a staffing gap. They thought they might have to stop the study, because they didn’t have anybody else available, until Rick suggested hiring me as a study coordinator. By that time, I had acquired some experience working in harm reduction at Burning Man and similar events, where I was providing volunteer peer support for people having distressing experiences with altered states, and I had discovered that I had the temperament for the work. I had been in some extreme situations, I could handle freakouts. I had been a journalist before, interviewing people about their stories, listening to people describe their life experience, and as a project manager I was good with budgets, schedules, resourcing, and task lists. Everything combined, it became apparent that I could be a good fit for the research team. I couldn’t provide any psychotherapy during that study, because I wasn’t trained as a therapist, so I refrained from doing anything that I wasn’t qualified to do.

I think the most valuable and essential factor that I brought to the team was that I was female, because the other two facilitators were male, and it’s important to have adequate gender balance. People near death frequently yearn for a mother figure. I also did various things like setting up the room in advance, bringing in flowers, decorating, helping create that sense of comfort and safety, attending to things like hydration. I got trained in using the machine that monitored vital signs. I would provide active listening, or a hand to hold or a light meal, taking a lot of session notes and things like that… but I did not provide psychotherapy.

After that psilocybin study with Charles Grob, you went on to study psychology at the Institute of Transpersonal Psychology, where you wrote your dissertation on ecstasy use in adults on the autism spectrum. Where did your initial interest in autism come from?

Five months after I started on the psilocybin study, I was diagnosed with breast cancer. My cancer was aggressive, but I caught it before it metastasised. But that brush with death brought up all sorts of existential issues for me. I figured that if I lived, I was going to become a licensed psychologist and do what I could to continue contributing to psychedelic science. I actually brought homework to the haematology lab and studied with an IV in my arm. It was motivating! I was in my early forties at the time, and I chose the Institute of Transpersonal Psychology because of Stan Grof’s affiliation with the origins of transpersonal psychology and because some of the core faculty there had been involved in the first wave of psychedelic research. If I were younger, I would have gone to a much larger, more prestigious APA-credited school. When younger people ask me what recommendations I have, I say: go to the best school and get the most solid credentials that you can. But at my advanced age as a student, I needed to land where I could write the dissertation I intended to write. And getting a transpersonal education was valuable for the clinical work I’m doing now with non-ordinary states of consciousness.

As for my interest in autism… Around the time I started volunteering for Dr. Grob, he invited me to attend a salon for people with an interest in psychedelics science. It took place once a month, and all sorts of chemists, and activists, and people with various other backgrounds came. At my first meeting, I met Gary Fisher. He was a psychologist in California who had worked in the sixties with children on the autism spectrum with LSD and psilocybin. His work fascinated me. He was a very warm, engaging, and creative person. That encounter planted the seed. Then, when it came time to determine my dissertation topic, I just knew at that point that I didn’t want to focus on cancer in any way, I didn’t even want to hear the word anymore at that point nearing the end of my own treatment. I reflected deeply on which other populations were underserved, grappled with difficult to treat mental health issues, and had no effective treatment options. In one of these reverie moments, I thought: What happens when people with an Asperger’s diagnosis take ecstasy? It seemed like this big revelation at the time, but when you think about it, it’s kind of an obvious question.

The Internet had become a major hub of a diverse and growing autism community, so I took a look at what people were saying on the web. One of the first accounts I read was about a young man who had put two and two together, and tried ecstasy to help him with his social challenges. He intentionally went to a party, took the ecstasy and was having a great time. But then everybody started getting drunk and sloppy, and he wasn’t comfortable with this chaos, so he went to a night club. When it closed, he was so deflated that he went out into the street at night, just hoping to connect with someone. And there again, I had a moment when my heart was responding to an account of someone’s pain that seemed unnecessary: why is this young man, who only wants human connection, reduced to roaming the streets at night? This is just flat out wrong. As a society, we need to know more about how to respond and support people who want to connect but lack the skills to do that naturally. So I decided to give it my best effort to try to build bridges in autism communities and learn all I could so I could interview people with a reasonable degree of certainty that they were telling me the truth. As a first step, I decided to do an inductive, mixed methods study with an emphasis on qualitative data. I learned from autistic adults what they struggle with and what they want. Then, I documented what they shared as objectively as I could by applying the Thematic Content Analysis method.

What did you learn?

The qualitative data summaries are very interesting to read. I made graphs to show the high percentages of people who said things like: “I’m more at ease in social settings”, or, “I’m better able to express my emotions”. Some of the tables look a little too good to be true. The data are reported accurately, but due in part to self-selection bias, individuals who had positive experiences are more likely to report. I had concerns that negative experiences might be underreported. I would leave out some of the questions about favourable effects and try to get more information about the negative experiences to maintain a balanced account. However, there simply was only a small percentage of negative reports.

Some individuals are clearly non-responders. We’ve seen that in clinical research, and there are theories about different enzymes and genes affecting metabolism. As more research is accumulated, I think there will be some determinable percentage of people who are atypical metabolisers, maybe around 10-15%. For example, they are the type who will do MDMA from the same batch as a group of others, but when everybody is cuddled up in a love puddle, they might feel as if they had a strong cup of coffee without the strong empathogenic effects. When you talk to these individuals, they’ll often say that they need to receive higher doses of anaesthetics or have other atypical responses to medications.

The themes that emerged from the – mainly qualitative – dissertation research data clustered around five constellations, I call them the “five C’s” as a memory aid. The types of changes reported were often around courage (or  confidence), that’s the first C. I like to use a Wizard of Oz analogy.  That change can be like the Cowardly Lion finding his courage: as if you had your courage, but were somehow detached from it, and now you own it and can use it again. You feel that increase in courage in an embodied sense: “I was brave”, “I could dance”, “I could flirt”, “I could say what I was always afraid to say”, or “I could call people to initiate something social”. The next C is communication: After Dorothy and the scarecrow oiled the Tin Man, he could gesture and speak more freely. There’s an increased ease in communication and a better ability to listen. A lot of people said they felt as if they could interpret body language or as if they could participate better in non-verbal means of communication. Another C is connection: such as connections with family members, understanding and relating to people they have significant connections to differently, being more open to physical intimacy, or feeling connected in a group instead of feeling so isolated. More than one individual has described this newfound sense of connection as being similar to how the Tin Man becomes aware of his heart that was always there. The fourth C: beyond connection there can be a sense of communion, that sort of deep sharing, maybe with some spiritual overtones. Feeling a part of something larger, unitive consciousness or some peak experience of feeling deep empathy. The final C was the most surprising finding: clarity (or calm). Clarity of mental and emotional processes. This effect was something that seemed more unique to this population, in comparison to neurotypical reports . A lot of interviewees made statements such as, “My brain was quiet for the first time in my life.” Or: “I could focus on one thing at a time”. “My inner world was clear”. “I had laser focus, my thoughts straightened out”. This last theme was the one that stood out for me, the one I wasn’t expecting based on prior accounts I had read.

Isn’t it difficult for autistic people to break the law by using MDMA, which is illegal? Isn’t that a barrier for them?

Autistic adults are such a heterogeneous population, everybody is so unique, that I tend to not think in stereotypes anymore. Yes, if you imagine a pie chart representing all autistic adults, there is a certain large segment who prefer to follow the rules in most instances. Another, related segment is made up of people who identify strongly with their cognition, their thoughts are, in a sense, the Self for them. So the idea of doing anything that might alter or impair cognitive processes is a deterrent. But those boundaries don’t apply to everybody. There are a lot of people on the spectrum who are paying attention to the science. For instance, when I was asking them about the quality of the MDMA they ingested, I was surprised at how many of the younger respondents said that they used the Marquee Reagent uptake test. They were very savvy. So science is science, and if the data are telling a different story, then they’re going to go with the facts. Also, enough people are having their own real-world experiences that influence how they’re thinking about MDMA. They may see friends who had a wonderful experience and were changed after that. Some people go for it, some people refrain.

On to your own clinical study, that was supported by this dissertation. You’re studying the effects of MDMA-assisted therapy on social anxiety in autistic adults. Why can’t these people be helped by other, more conventional methods – especially for social anxiety, for which there are many medicines?

There is some research literature that suggests that the receptors for benzodiazepines, for example, respond atypically in autistic brains. There are structural brain differences, and there’s no such thing as a uniform autistic brain. And it makes sense, if you slow down and think about it: conventional psychodynamic therapy has not shown to be particularly effective for people on the spectrum. Unfortunately, historically the blame has been put on the autistic clients, assuming that they can’t relate or express themselves. But I’ve come to shift my focus to the clinicians who have not invested the effort it takes to learn about or really appreciate what it’s like to be autistic. So I’ve come to think of the barriers to therapy as mutual challenges with understanding. Speaking in broad stereotypes, a lot of autistic clients are very pragmatic. They want tools to address a problem they have in the here and now. They might be less interested in talking about what happened to them when they were five, at least not initially. This process of working with archetypes and analogies and metaphorical concepts may not be as effective for them. There may be challenges going both ways with establishing an essential, empathic therapeutic rapport.

In our study, we’re using psychoeducation in mindfulness skills, based on dialectical behavioural therapy (DBT), developed by Marsha Linehan. DBT was developed to promote effectiveness in interpersonal relationships, emotion regulation and distress tolerance, which are social adaptability skills that are often challenging for adults on the spectrum, so we thought this type of therapy would be a good fit. It’s so fundamental, and it’s helpful across so many domains. We’ve adapted the  mindfulness module, and applied it in a research setting, because it’s a practical life skill. There are data that show mindfulness therapies are effective for individuals on the spectrum, and it creates a vocabulary that we can use during sessions to help them navigate altered states, so that when they are in a state of consciousness that’s ineffable, or they’re experiencing the novel states of mind for the first time, we can continue dialoguing with them by asking questions like: “What is your reasonable mind doing now? And describe what the emotional mind is doing. If you don’t know what to do right now, just observe your experience. And when you’re ready, describe what you observe”. All of our study participants are MDMA-naive, and we’re seeing indications that mindfulness concepts can help them navigate the MDMA experience, especially the first time.

Regarding neuropsychology, some of the most promising recent findings are about GABA receptors. Whereas dopamine is similar to the gas pedal on a car that revs things up, GABA is like a brake pedal that quiets things down. Recent research findings have suggested that autistic brains have the same amount of GABA available as typically developing brains, but the receptors work differently, so the brake pedal isn’t as easy to apply. This makes it more difficult to filter out extraneous sensory input, to focus. Just adding GABA doesn’t help, because the difference is not related to amount, but to utilisation. I think the GABA research might someday be shown to be relevant to why MDMA might be helpful for autistic brains in ways that are unique to that population, but much more research is required first.

You mentioned Gary Fisher before. How do you view the whole body of research with autistic children and teens from the sixties, and what did you learn from it?

I learned that the methodology would never be approved today. Some of the studies were horrifying! They would take very young, non-speaking children, put them in a room, and look at them through a one-way mirror after giving them large doses. They could not provide informed consent, they could not verbalise their experiences, they couldn’t ask for help. So I don’t advocate replicating those studies.

However, when you look at the aggregated data, there were more positive outcomes than adverse responses, and that was reflected in behaviours that were reported: smiling, laughing, gazing, seeking physical contact, initiating play. In fact, for most populations, set and setting are going to have an influence on outcomes: some people are going to have a difficult experience with larger doses no matter what you do, and some people a going to experience euphoric states. Gary Fisher was trained in classical psychoanalysis and psychodynamic approaches. He had his research staff take LSD as part of their training, in order to bond as a team, and to be able to have more empathic resonance with the study participants. He took a much more humanistic, psychodynamic approach as opposed to the classic medical model of monitoring parameters and behaviours. He saw the importance of supportive psychotherapy and forging therapeutic relationships between facilitators and subjects.

MDMA was never used in the first wave of psychedelic research. Why did you choose MDMA over any other, more ‘classic’, psychedelic?

Because of MDMA’s reputation for having prosocial effects in clinical and non-clinical settings. Before starting my psychology training, I mistakenly believed that individuals with an Asperger’s diagnosis could not experience empathy because that’s what I was taught. That incorrect assumption was kind of a catalyst, but I’ve changed my thinking about autism and empathy. Empathy is a broad umbrella term that covers many domains of human cognitive and affective experience. These days, I’m interested in Markram and Markram’s research about the Intense World Theory. In some instances, there may be an overpresence of some aspects of empathy with autism. I can tell you, from establishing connections with autistic adults, that many of them are quite empathic. Often they’re challenged because they feel too much. For others, it’s difficult to understand what someone else might be thinking or feeling, but if someone they care about is hurting, they hurt, or if someone else’s inner experience is explained to them, they can care about what someone else is feeling. In some cases, they’re less likely to pick up on subtle, non-verbal cues. So there’s a broad spectrum of ways people experience empathy, and I’m not on a crusade to implant empathy with a pill. But there are also other ways in which MDMA-assisted therapy might be supportive. For example, experiencing the pleasant sensations of being touched, which can be a challenging area for a lot of people on the spectrum, or being better able to express oneself verbally, especially about affective states. Findings from other studies indicate that MDMA has multiple effects that might be helpful. So let’s apply the scientific method, collaborate with autism communities, and find out if that’s the case for autistic adults or not.

Almost all the participants have been treated now. Can you give us some preliminary results?

Because it’s a small safety and feasibility pilot study and because it’s a sponsored study, the sponsor [fusion_builder_container hundred_percent=”yes” overflow=”visible”][fusion_builder_row][fusion_builder_column type=”1_1″ background_position=”left top” background_color=”” border_size=”” border_color=”” border_style=”solid” spacing=”yes” background_image=”” background_repeat=”no-repeat” padding=”” margin_top=”0px” margin_bottom=”0px” class=”” id=”” animation_type=”” animation_speed=”0.3″ animation_direction=”left” hide_on_mobile=”no” center_content=”no” min_height=”none”][MAPS, ed.] has an obligation to monitor early outcomes to determine if we’re doing any harm. Looking at the outcomes from early participants, the data were suggesting that we could continue safely and that early trends were towards positive outcomes. That’s really all I know, however, because the researchers are still blinded to the primary outcome measure scores. We do monitor some of the other assessments. For safety reasons, we review secondary outcome measures for conditions such as mood, stress, and anxiety. We’re also looking at factors such as self-esteem, alexithymia (the inability to identify emotions or express them in words), emotion regulation, emotion expression,… There may be outcomes other than social anxiety scores that support future studies, at least we hope so. But that’s all I can say for now, unfortunately.

What’s your general impression of how the sessions went?

Generally, the completed sessions have gone very well. We have had no serious adverse events, and no events requiring any medical intervention. The heart rate and blood pressure measurements have never been alarming. An important thing to understand is that we’re working in lower dose ranges, in part because some of the input from the dissertation research suggested that at least some individuals on the autism spectrum may be more sensitive to the effects of MDMA, and higher doses might be too stimulating and induce stress. This is a dose-finding study, so we don’t make the assumption that what’s optimal for a typically developing brain is best for an autistic-style brain. The first group of six subjects got 75mg with an escalation to 100mg, and the second group of six subjects started at 100mg, and if they tolerated that well, they went up to 125mg. Everyone has been able to tolerate the escalation, and nobody had the type of distressing medically adverse experience that would prevent us from raising the dose.

Overall, what we’ve observed has been positive and encouraging so far. The responses span a broad range, from individuals who have a “non-responder” minimal kind of reaction, to others who have had more transformative experiences. It’s possible that some subjects might respond positively to placebo because they have not had prior psychotherapeutic help. We unblind at six months for this study, to determine who’s eligible to go on to Stage 2, so I do know in some, but not all, instances who got placebo and who got MDMA, and we’ve seen a broad range of responses. So it all comes down to the data. We’ll have most of our initial outcome data for Stage 1 in August of 2016. As for publication, my best guess would be early 2017 at the earliest.

[/fusion_builder_column][/fusion_builder_row][/fusion_builder_container]

Ketamine could be the first rapid-acting antidepressant medication

Ketamine has been used for over half a century as an anaesthetic, but interest has been steadily growing in its ability to rapidly decrease depressive symptoms. This interest has culminated in many studies attempting to elucidate its antidepressant mechanism, and in turn, these studies have contributed to our understanding of depressive disorders. Ketamine was first synthesised in 1962 as a dissociative anaesthetic, inducing a state of conscious sedation in which patients are awake, but cognitively dissociated from their pain (Young et al., 2011). In 1970, the Food and Drug Administration (FDA) approved ketamine as an anaesthetic, but throughout the 1970s ‘special K’, as it was known on the street, was gaining popularity as a recreational drug. The drug became notorious for its ability in high doses to lead users down a ‘K-hole’ – or a state of complete bodily dissociation (Muetzelfeldt et al., 2008). In 1999, the FDA scheduled the drug in the United Stated (US), banning non-medical use. While ketamine’s use and abuse was being argued over by policy makers, a team of scientists began to investigate ketamine as an antidepressant (Berman et al., 2000). Following this first human trial of ketamine as an antidepressant, the drug quickly garnered interest in the field of mental health, where since the publication of the fourth edition of the “Diagnostic and Statistical Manual of Mental Disorders” (DSM-IV) in 1994, depression was considered as a unitary concept and psychiatric disorder.

Depression and the pharmacological response

Major depressive disorder (MDD) is the most prevalent mental disorder, affecting roughly 16% of the world’s population at some point in their lives (Kessler et al., 2005). The dominant pharmacological hypothesis came about by trying to understand why certain monoaminergic-targeted medications seemed to alleviate depressive symptoms over time. The monoamine hypothesis of depression describes the disorder as the dysregulation of a group of monoaminergic neurotransmitters in the brain, specifically, the transport of dopamine, adrenaline, noradrenaline and serotonin into and out of synapses (Hirschfeld, 2000). First-line treatment of depression involves selective norepinephrine or serotonin reuptake inhibitors (SNRI/SSRI). These drugs inhibit the transport of key neurotransmitters out of the synapse between neurons. Unfortunately, less than half of those who suffer from MDD respond to monoamine-targeted medication, and for those who do, it takes at least two weeks and often longer for any symptom relief to become clinically noticeable (Kishimoto et al., 2016). Some studies have also linked SSRI treatment to an increased risk of suicide attempts and completed suicides (Fergusson et al., 2005; Healy, 2003). There is clearly a need for a more rapid and efficacious treatment of this debilitating disorder.

Ketamine as an antidepressant

Ketamine is rare in that it is a psychoactive substance that is classified in the U.S. in Schedule III under the Controlled Substances Act (Marshall, 1999) and available to be prescribed by physicians, making research accessible; however, the drug’s status as a non-patentable substance has proven to be a hurdle to funding research. Nevertheless, research into ketamine as an antidepressant has been ongoing since 2000 (Berman et al., 2000) and resulted in over 1500 studies. Studies have found that single intravenous infusions of ketamine at doses ranging from 0.1-0.5 mg/kg over 40 minutes show robust efficacy in short-term relief of MDD symptoms (Kishimoto et al., 2016). Symptoms decrease within 40-60 minutes and during the first 24 hours suicidality is also rapidly decreased. Interestingly, reduced suicidality was found to be a specific effect of ketamine, as it was also found in patients who did not respond to the antidepressant effects of the drug (Ryan et al., 2014). The promise for ketamine lies in its rapid-acting antidepressant and antisuicidal effects, as few current treatments achieve clinical significance in such a short time frame. Remission of depressive symptoms, on the other hand, only lasts between five and eight days, but can be extended to months through administration of repeated infusions (Murrough et al., 2013). This technique is somewhat controversial as the effects of repeated exposure to sub-anaesthetic doses of ketamine are yet to be known. Some patients who receive these sub-anaesthetic doses of ketamine report mild side-effects including headaches, dizziness and nausea, as well as dissociative effects and mild psychotomimetic experiences; however, these effects are transient and rarely outlast the time in which the drug is pharmacologically active (about 4 hours) (Coyle & Laws, 2015).

The glutamate theory of depression

Ketamine is a chemically promiscuous substance that interacts with many neurotransmitters in the brain, including the monoamines (Frohlich & Van Horn, 2015). Much of the research, however, has attributed ketamine’s antidepressant effects to its activation of the glutamate system. Glutamate is an excitatory neurotransmitter found in about 50% of synapses in the mammalian brain (all the monoamines together are found in only 15-20%) (Zarate & Niciu, 2015). It is the primary system by which neurons fire and communicate with one another and, as such, may be more pertinent to rapid changing of mood than monoaminergic systems.

The mechanism is complicated, but ketamine modulates glutamate by blocking, and thereby inhibiting, N-methyl-D-aspartate receptors (NMDAr) on interneurons. When active, these interneurons inhibit glutamatergic neurons in mood relevant brain areas; however, when blocked by ketamine, the disinhibition of these neurons leads to an increase in glutamatergic synaptic transmission in brain areas responsible for mood. Ultimately, the increase in glutamate activates a cascade of effects that results in neuroplasticity or neural regeneration – morphological changes to neurons in these brain areas (Kavalali & Monteggia, 2012). According to this hypothesis, the drug begins by causing chemical changes which result in non-chemical, morphological changes. It is these physical changes in the brain that could explain the persisting antidepressant effects of the drug once its pharmacological activity has ended. While NMDAr are believed to mediate these beneficial effects, recent research comparing ketamine with selective NMDAr antagonists shows that the selective NMDAr antagonists are not nearly as effective as ketamine in treating depression (Kishimoto et al., 2016; Sanacora & Schatzburg, 2015; Zanos et al., 2016). This suggests the possibility of an alternate mechanism of action.

Competing mechanisms

Excitement about ketamine as a wonder drug for depression should be tempered by a number of safety concerns about the medication. Firstly, it can induce psychedelic experiences which could be dangerous for those with a family history of psychotic disorders. Many pharmacologists, physicians and chemists have been searching for ways of achieving anti-depressant effects separate from the psychedelic experiences by adjusting doses to sub-anaesthetic levels, or exploring metabolites and stereoisomers of ketamine. Other more psychotherapeutically oriented researchers claim that the psychedelic and dissociative effects of the drug are part and parcel of its success in treating depression when combined with therapy, and that stripping the drug treatment of these mind-altering effects is akin to practicing homeopathy (Wolfson, 2014).

One such psychiatrist, Terrence Early (2014), suggests that ketamine works via the mechanism of ‘action-facilitated emotional learning.’ According to Early, patients on ketamine dissociate from their bodies, and are thus able to remember negatively charged emotional memories or trauma without the anxiety that would normally accompany these memories. Ketamine attenuates the anxiety response when trauma is revisited in therapy, and this in turn allows these memories to gradually become manageable. The age-old debate between minimising the psychedelic effects of psychedelics and embracing them for their therapeutic potential is a complex one that is present throughout the literature of psychedelic science, and involves political narratives of the war on drugs and freedom of thought.

Broadening the scope of treatment

Looking ahead to the broadened use of ketamine outside of hospital settings, addiction liability is an issue that worries some researchers (Sanacora & Schatzburg, 2015; Zhang et al., 2016), especially considering that nearly one-third of people who suffer from depression also meet criteria for substance use disorders (Davis et al., 2008). To date, trials involving ketamine almost always exclude a comorbid substance abuse disorder and this means we have very little data regarding ketamine’s addiction potential for this significant population of depressed patients. Ketamine addiction is well documented, but only at doses above 1 mg/kg (Newport et al., 2015). Current trials treating depression typically use doses of 0.5 mg/kg and never over 1.0 mg/kg. Nevertheless, the medical field has an embarrassing history of creating addictions through prescription medications like laudanum, heroin and cocaine, and we don’t have to look very far to see the current epidemic of prescription opiate addiction. Because of the drug’s short half-life, in order to achieve remission of longer than one week, repeated doses of ketamine are required, potentially increasing the likelihood of tolerance and addiction to the medication. Currently there is a paucity of research on the adverse effects of long-term repeated ketamine usage.

Ketamine is currently approved by the FDA via intravenous (IV) or intramuscular (IM) routes for large-dose anaesthesiology. This requires the presence of an anaesthesiologist and must take place in a hospital setting. This expensive, invasive and highly medicalised treatment model shows little regard for set and setting, which plays such an important role in ensuring meaningful psychedelic experiences. It has been shown that patients who received ketamine in electroconvulsive therapy rooms have worse outcomes than patients who received the medication in comfortable, relaxed settings (Ryan et al., 2014).

Alternate routes of ketamine administration have been developed and are currently being researched, including intranasal, subcutaneous, oral and sublingual (Lara et al., 2013; Mathews et al., 2012; Opler et al., 2016); however, bioavailability of ketamine is less than 50% for oral, subcutaneous and intranasal routes of administration compared to 93% for the more invasive routes (Clements et al., 1982). The antidepressant response to these alternate routes is also lower than IV or IM administration (Ryan et al., 2014). Sub-anaesthetic doses, not requiring the presence of anaesthesiologists and which can be administered in more comfortable settings, have shown antidepressant efficacy (Berman et al., 2000; Zarate et al., 2006). If off-label prescriptions are being written and administered, informed consent and integration in a therapeutic treatment are important set and setting factors for maximising the effect of the medication.

On the horizon

An exciting new article was published in Nature in May 2016, which claims to have found the key metabolite of ketamine responsible for the sustained antidepressant effects. The compound of interest was tested using animal models and found to be non-addictive and non-psychotomimetic (Zanos et al., 2016). Ketamine as it is generally administered is a racemic mixture of S-ketamine and R-ketamine (both left- and right-handed molecules in more or less equal parts). The body converts both of these enantiomers into a number of metabolites. S-ketamine and its metabolites are known to have three to four times greater affinity for NMDAr than R-forms, leading researchers to believe that S-ketamine could be used in smaller doses to achieve similarly potent effects; however, Zanos and colleagues identified (2R, 6R)-hydroxynorketamine (R-HNK) – a metabolite of ketamine with two right-handed chiral centres – as essential to the potent antidepressant effect.

Unexpectedly, R-HNK does not bind to or inhibit NMDAr, calling into question the NMDAr hypothesis of ketamine. While the target of R-HNK is not yet known, it was shown that R-HNK increases α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAr)-mediated postsynaptic potentials in the hippocampus, even after the drug’s pharmacological activity has ended. It is the upregulation of these excitatory glutamatergic AMPAr that is hypothesised to be responsible for the longer-lasting antidepressant effects of ketamine. This novel NMDAr-independent, non-addictive and non-psychotomimetic antidepressant mechanism is an exciting find (Zanos et al., 2016), but this research needs to be replicated and scaled up to human trials before any firm conclusions can be drawn as to its efficacy in treating depressive disorders.

Ketamine has emerged as a first-in-class rapid-acting antidepressant medication with a unique mechanism of action that differentiates it from the current psychiatric tools for depression. We may be on the brink of next-generation rapid-acting antidepressant medication; however, excitement about ketamine’s antidepressant benefits should be tempered by issues of safety, including adverse psychotomimetic effects, abuse potential, and costly invasive routes of administration. Before the FDA approves ketamine as a medication for MDD in broader clinical contexts, research into adverse effects of prolonged use needs to be done along with the standardisation of optimal dosing, route of administration and frequency of ketamine administration. Even in the early days of research, and with these safety concerns in mind, ketamine’s ability to rapidly decrease depressive and suicidal symptoms allows physicians to ethically treat the most severe cases of depression in emergency room contexts. At the very least, it can give clinicians time to implement alternative therapies and allow for the slower-onset, first-line treatments to reach efficacy.

References

Berman, R. M., Cappiello, A., Anand, A., Oren, D. A., Heninger, G. R., Charney, D. S., & Krystal, J. H. (2000). Antidepressant effects of ketamine in depressed patients. Society of Biological Psychiatry. 47. 351-354. doi: 10.1016/S0006-3223(99)00230-9

Clements, J., Nimmo, W., & Grant, I. (1982). Bioavailability, pharmacokinetics, and analgesic activity of ketamine in humans. Journal of Pharmaceutical Sciences. 71(5). 539-542. doi: 10.1002/jps.2600710516

Coyle, C. M., & Laws, K. R. (2015). The use of ketamine as an antidepressant: a systematic review and meta-analysis. Human Psychopharmacology: Clinical and Experimental, 30(3). 152-163. doi: 10.1002/hup.2475

Davis, L., Uezato, A., Newell, J. m., & Frazier, E. (2008). Major depression and comorbid substance use disorders. Current Opinions in Psychiatry. 21(1). 14-18. doi: 10.1097/YCO.0b013e3282f32408

Early, T. S. (2014). Making ketamine work in the long run. International Journal of Transpersonal Studies 33(2). 141-150.

Fergusson D., Doucette, S., Glass, K. C., Shapiro, S., Healy, D., Hebert, P., & Hutton, B. (2005). Association between suicide attempts and selective serotonin reuptake inhibitors: Systematic review of randomised controlled trials. British Medical Journal, 330. 396. doi: 10.1136/bmj330.7488.396

Frohlich, J. & Van Horn, J. D. (2014). Reviewing the ketamine model for schizophrenia. Journal of Psychopharmacology, 28(4). 287-302. doi: 10.1177/0269881113512909jp.sagepub.com

Healy, D. (2003). Lines of evidence on the risks of suicide with selective serotonin reuptake inhibitors. Psychotherapy and Psychosomatics. 72(2). 71-79. doi: 10.1159/000068691

Hirschfeld, R. M. (2000) History and evolution of the monoamine hypothesis of depression. The Journal of Clinical Psychiatry. 61 (suppl 6). 4-6

Kavalali, E. T., & Monteggia, L. M. (2012). Synaptic mechanisms underlying rapid antidepressant action of ketamine. Journal of American Psychiatry, 169(11). 1150-1156. doi: 10.3389/fphar.2013.00161

Kessler, R, C., Berglund, P., Demier, O., Jing, R., Merikangas, K. R., & Walters, E. E. (2005). Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the national comorbiditysurvey replication. Archives of General Psychiatry, 62(6). 593-602. doi:10.1001/archpsyc.62.6.593.

Kishimoto, T., Chawla, J. M., Hagi, K., Zarate, C. A., Kane, J. M., Bauer, M., & Correll, C. U. (2016). Single-dose infusion ketamine and non-ketamine N-methyl-D-aspartate receptor antagonists for unipolar and bipolar depression: a meta-analysis of efficacy, safety and time trajectories.  Psychological Medicine, 46. 1459-1472. doi:10.1017/S0033291716000064

Lara, D. R., Biosol, L. W., & Munari, L. R. (2013). Antidepressant, mood stabilizing and precognitive effects of very low dose sublingual ketamine in refractory unipolar and bipolar depression. The International Journey of Neuropsychopharmacology. 16(9). 2111-2117. doi: 10.1017/S1461145713000485

Marshall, D. R. (1999). Schedules of controlled substances: Placement of ketamine into schedule III. Federal Register. 64 (133). 37673-37675. docid: fr13jy99-7

Mathews, S. J., Shah, A., Lapidus, K., Clark, C., Jarun, N., Ostermeyer, B., & Murrough, J. W. (2012). Ketamine for treatment resistant unipolar depression. CNS Drugs. 26(3). 189-204. doi: 10.2165/11599770-000000000-00000

Muetzelfeldt, L., Kamboj, S. K., Rees, H., Taylor, J., Morgan, C. J.,& Curran, H. V. (2008). Journey through the K-hole: Phenomenological aspects of ketamine use. Drug and Alcohol Dependence, 95(3). 219-229. doi:10.1016/j.drugalcdep.2008.01.024

Murrough, J. W., Perez, A. M., Pillemer, S., Stern, J., Parides. M. K., aan het Rot, M., Collins, K. A., Mathew, S. J., Charney, D. S., & Iosifescu, D. V. (2013). Rapid and longer-term antidepressant effects of repeated ketamine infusions in treatment-resistant major depression. Biological Psychiatry, 74(4). 250-256. doi: 10.1016/j.biopsych.2012.06.022

Newport, D. J., Carpenter, L. L., McDonald, W. M., Potash, J. B., Tohen, M., Nemeroff, C. B., The APA Council of Research Task Force on Novel Biomarkers and Treatments. (2015). Ketamine and other NMDA antagonists: Early clinical trials and possible mechanisms of depression. The American Journal of Psychiatry. 172(10). 950 -966. doi: 10.1176/appi.ajp.2015.15040465

Opler, L.A., Opler, M. G. A., & Arnsten, A. F. T. (2016). Ameliorating treatment-refractory depression with intranasal ketamine: potential NMDA receptor actions in the pain circuitry representing mental anguish. CNS Spectrums. 21(1). 12-22. doi: 10.1017/S1092852914000686

Ryan, W. C., Marta, C, J. & Koek, R. J. (2014). Ketamine and depression: A review. International Journal of Transpersonal Studies. 33(2). 40-74.

Sanacora, G., & Schatzberg, A. F. (2015) Ketamine: Promising path or false prophecy in the development of novel therapeutics for mood disorders? Neuropsychopharmacology, 40. 259-267. doi:10.1038/npp.2014.261.

Wolfson, P. E. (2014). Ketamine-Its history, uses, pharmacology, therapeutic practice, and exploration of its potential as a novel treatment for depression. International Journal of Transpersonal Studies. 33(2). 33-39.

Young, M., Kolp, E. & Friedman, H. (2011). Ketamine. In M.A. Kleiman & J. E. Hawdon (Eds.), Encyclopedia of drug policy (2). 451. doi:10.4135/9781412976961.n194

Zanos, P., Moaddel, R., Morris, P. J., Georgiou, P., Fischell, J., Elmer, G. I., Alkondon, M., Yuan, P., Pribut, H. J., Singh, N. S., Dossou, K. S., Fang, Y., Huang, X., Mayo, C. L., Wainer, I. W., Albuquerque, E. X., Thompson, S. M., Thomas, C. J., Zarate Jr. C. A., & Gould, T. D. (2016). NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature. 533(7604). 481-486. doi: 10.1038/nature17998.

Zarate, C. A., Singh, J. B., Carlson, P. J., Brutsche, N. E., Ameli, R., Luckenbaugh, D. A., Charney, D. S., & Manji, H. K. (2006). A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Archives of General Psychiatry. 63(8). 856-864. doi: 10.1001/archpsyc.63.8.856

Zarate, C. A. & Niciu, M. (2015). Ketamine for depression: Evidence, challenges and promise. World Psychiatry, 14(3). 348-350. doi:10.1002/wps.20269.

Zhang, M. W., Harris, K. M., & Ho, R. C. (2016). Is off-label repeat prescription of ketamine as a rapid antidepressant safe? Controversies, ethical concerns, and legal implications. BMC Medical Ethics. 17(4). 1-8. doi: 10.1186/s12910-016-0087-3

Psilocybin-assisted therapy shows promising results for treatment-resistant depression

psilocybin depression 2In a new study, the research team at the Imperial College London has tested the potential of psilocybin-assisted therapy to alleviate treatment-resistant depression. Statistics show that 20% of people suffering from major depression are unresponsive to conventional treatments like SSRI medication or cognitive behavioural therapy (Carhart-Harris et al., 2016).

Twelve subjects (six men and six women), all diagnosed with major depression, participated in the study. They received two oral doses of psilocybin – 10 mg and 25 mg – the former being the safety dose and the latter, administered seven days later, the treatment dose. The participants had been selected among 70 candidates; one of the main selection criteria was the absence of psychotic episodes in subjects themselves and in their immediate family members.

All participants, aged between 30 and 60, had a long history of major depression, with treatment attempts having had only minimal effects. Some of them had been suffering moderate to severe depression for about three decades. Previous treatment attempts included both chemical and psychological means: medication like serotonin or dopamine reuptake inhibitors (SSRI, NDRI, SNRI, etc.) and therapies like cognitive behavioural, group and counselling therapy.

The pharmacology of psilocybin is different from that of selective serotonin reuptake inhibitors (SSRIs), the most common medication for this kind of depression. SSRIs prevent the already released serotonin – one of the neurotransmitters involved in the regulation of emotion – from being taken back up by the same neurons that produced it, so that it can be taken up by serotonin receptors. Unlike SSRIs, psilocybin (converted in the body to psilocin) is structurally similar to serotonin, and causes the same effect as an overall increase in serotonin levels.

Over the course of the study, psychological support was provided before, during and after the psilocybin sessions. During the sessions, there was minimal intrusion into the patients´ experience. The patients were only asked the necessary questions to evaluate the effects of psilocybin on their physical and mental well-being. The most common adverse reactions reported included nausea, headaches, anxiety and confusion, all of which were transient. Only one patient reported transient paranoia that subsided after one hour.

The study demonstrated that the symptoms of depression were somewhat reduced in all of the twelve participants. The scores on the Quick Inventory of Depressive Symptoms (QIDS) showed that the depression level was reduced from 16-20 (severe depression) to 6-10 (mild depression). Five follow-up assessments took place between one week and three months after the treatment. The maximum positive effects were reached two weeks after the treatment. Eight subjects experienced complete remission in their depression one week after the treatment and in seven of them significant reduction in depression persisted after three months. One patient experienced an increase in depressive symptoms during the three months following the treatment.

This study was the first to explore the efficacy of psilocybin in treating major depression, and demonstrated the potential of psilocybin for reducing the symptoms of major treatment-resistant depression and the safety of the substance when administered under proper conditions. Previous research with psilocybin-assisted therapy has already showed that it can alleviate anxiety related to end-stage cancer (Grob C.S. et al., 2011).

Further research in more rigorous conditions (placebo-controlled and on a larger scale) is needed to confirm the potential of psilocybin in treating major depression. If this promise can be fulfilled, it could mean a new chance for millions of people struggling with severe depression.

References:

Carhart-Harris R.L., Bolstridge M., Rucker J., Day C.M.J., Erritzoe D., Kaelen M., Bloomfield M., Rickard J.A., Forbes B., Fielding A., Taylor D., Pilling S., Curran V.H., Nutt D.J. (2016) Psilocybin with psychological support for treatment-resistant depression: an open-label feasibility study. http://dx.doi.org/10.1016/S2215-0366(16)30065-7

Grob C.S., Danforth A.L., Chopra G.S., Hagerty M., McKay C.R., Halberstadt A.L. and Greer G.R. (2011) Pilot study of psilocybin treatment for anxiety in patients with advanced-stage cancer. Arch Gen Psychiatry, 68, pp. 71–78 http://dx.doi.org/10.1001/archgenpsychiatry.2010.116

DMT: Beyond the trip, a potential multifaceted medicine

DMT-emergencyN,N-dimethyltryptamine, more commonly known as DMT, is an exceptionally fast-acting and powerful psychedelic. DMT can be ingested by drinking the entheogenic brew ayahuasca, injected intravenously, intramuscularly or through inhalation. It is produced endogenously in a variety of plants and animals, including in humans. DMT exerts physiological effects that go beyond its mind-altering effects, as discussed in Jacob and Presti (2005). For example, DMT has been shown to induce anxiolytic and antidepressant effects (Sanches et al. 2016).

DMT is not only an agonist of serotonin 2A and 2C receptors (5-HT2A and 5-HT2C); it also binds to σ1 putative receptors and trace amine receptors (Vitale et al. 2011). In addition, its serotonergic analogues can influence immunoregulation, and may even prevent carcinogenesis (Frecska et al. 2012). DMT’s multifaceted interactions show that its effects are not limited to the central nervous system but may play a more crucial role in the body’s cellular protective mechanisms (Frecska et al. 2012).

Dr. Ede Frecska has published multiple papers on the effects of ayahuasca and DMT on creativity, tissue regeneration, and the interhemispheric fusion in altered states of consciousness (Frecska et al. 2016). With the recent discovery of DMT’s activation of the σ1 receptor , which plays a crucial role in protecting the body from undergoing oxidative stress, Dr. Frecska and his team are currently investigating DMT’s role in neuroprotection prior to clinical death (Frecska 2015).

σ1 receptors play a key role in neuroprotection by regulating both neuronal development and morphogenesis. This is done through the regulation and manipulation of oxidative stress and mitochondrial functions (Tuerxun et al. 2010). Agonists of σ1 receptors exacerbate neuroprotective effects by inhibiting intracellular calcium overload and by thwarting the activation of pro-apoptopic genes, as well as activating protective genes, as shown in stroke models (Zhang et al. 2012). This leads to the reduction of calcium neurotoxicity, prevents oxidative stress-induced cell death, and can stimulate neuronal plasticity (Kourrich et al. 2012). Most importantly, the constant activation of σ1 receptors during ischemia leads to a reduction of neurotoxicity (Katnik et al. 2006). Ultimately, this research suggests that DMT may have a role in reducing the hypoxic-anoxic damages such as local anoxia (e.g. stroke) or general hypoxia (e.g. cardiac arrest) (Kourrich et al. 2012).

DMT’s medicinal properties are not limited to neuroprotection, but can extend to immunoprotection as well. The 5HT2A receptors, as well as the sigma receptors, can profoundly influence the body’s immune system. Serotonin plays an important role in cellular immune functions, and more specifically in the elimination of pathogens and cancer cells (O’Connell et al. 2006). σ1 receptor agonists can increase the production of anti-inflammatory cytokines as well as reduce pro-inflammatory cytokines. Both these processes are important in reducing the cellular damage in case of injury or disease (Frecska et al. 2012).

Currently, there is only speculation that DMT is produced during near-death experiences, as there are few parallels between near-death experiences and DMT visions (Strassman 2001). However, based on limited information, one may conjecture the production of DMT during life-threatening situations. McEwen and Sober (1967) have demonstrated that when undergoing extreme environmental stress, rabbits produce vast quantities of DMT in the lungs, which are then released into the blood (McEwen & Sober 1967). DMT is then transported through the neural membranes within synaptic vesicles and delivered to the brain. Knowing the relationship between DMT and the σ1 receptors, it is hypothesised that DMT limits or reverses the accumulated oxidative stress. This serves as the foundation of Dr. Frecska’s hypothesis, and if evidence is found of DMT’s role in the neuroprotection of the human brain in the stages leading up to clinical death, then DMT would have the potential to be used as an emergency medicine. If successful, one could envision the use of DMT ampoules to be used intravenously in ambulances, operating rooms and in disaster zones. Clinical studies with humans are still necessary in order to define whether it is feasible or not.

Although Dr. Frescka’s studies focus on rats (pre-clinical studies), his studies have looked beyond DMT’s mere hallucinogenic relevance and have opened avenues into further studying DMT’s neuroprotective role. The potential medical ramifications are vast. The applications of DMT may be beyond what we can imagine, and certainly deserve to be systematically studied.

References

Frecska, E., 2015. What role does the ‘spirit molecule’ DMT play in the brain?. [fusion_builder_container hundred_percent=”yes” overflow=”visible”][fusion_builder_row][fusion_builder_column type=”1_1″ background_position=”left top” background_color=”” border_size=”” border_color=”” border_style=”solid” spacing=”yes” background_image=”” background_repeat=”no-repeat” padding=”” margin_top=”0px” margin_bottom=”0px” class=”” id=”” animation_type=”” animation_speed=”0.3″ animation_direction=”left” hide_on_mobile=”no” center_content=”no” min_height=”none”][Online] Available at: http://walacea.com/campaigns/dmt

Frecska, E., Bokor, P. & Winkelman, M., 2016. The Therapeutic Potentials of Ayahuasca: Possible Effects against Various Diseases of Civilization. Frontiers in Pharmacology, p. 10.3389.

Frecska, E. et al., 2012. A possibly sigma-1 receptor meditated dole of dimethyltryptamine in tissue protection, regeneration and immunity. Translational Neuroscience, pp. 1-18.

Jacob, M. & Presti, D., 2005. Endogenous psychoactive tryptamines reconsidered: an anxiolytic role for dimethyltryptamine.. Medical Hypotheses, 64(5), pp. 930-7.

Katnik, C. et al., 2006. Sigma-1 receptor activation prevents intracellular calcium dysregulation in cortical neurons during in vitro ischemia. Journal of Pharmacology and Experimental Therapeutics, Band 319, pp. 1355-1365.

Kourrich, S., Tsung-Ping, S., Fujimoto, M. & Bonci, A., 2012. The sigma-1 receptor: roles in neuronal plasticity and disease. Trends Neuroscience, 35(12), pp. 762-771.

McEwen, C. & Sober, A., 1967. Rabbit serum monoamine oxidase. The Journal of Biological Chemistry, Band 242, pp. 3068-3078.

O’Connell, P. et al., 2006. A novel form of immune signaling revealed by transmission of the inflammatory mediator serotnin between dendritic cells and T cells. Blood, Band 107, pp. 1010-1017.

Sanches, R. F. et al., 2016. Antidepressant Effects of a Single Dose of Ayahuasca in Patients With Recurrent Depression: A SPECT Study. Journal of Clinical Psychopharmacology, 36(1), pp. 77-81.

Strassman, R., 2001. DMT: The Spirit Molecule. First Hrsg. Rochester: Park Street Press.

Strassman, R. & Qualis, C., 1994. Dose-response study of N,N-dimethyltryptamine in humans. I. Neuroendocrine, autonomic, and cardiovascular effects. Archives of General Psychiatry, pp. 85-97.

Tuerxun, T. et al., 2010. SA4503, a sigma-1 receptor agonist, prevents cultured cortical neurons from oxidative stress-induced cell death via suppression of MAPK pathway activation and glutamate receptor expression. Neuroscience Letters, Band 469, pp. 303-308.

Zhang, Y. et al., 2012. Sigma-1 receptor agonists provide neuroprotection against gp12- via a change in bel-2 expression in mouse neuronal cultures. Brain Research, Band 1431, pp. 13-22.

[/fusion_builder_column][/fusion_builder_row][/fusion_builder_container]

The Serotonin Infatuation

serotonin picMuch like the Holy Grail symbolised well-being, infinite wealth, and abundance of food in Arthurian literature, today the infamous neurotransmitter, serotonin, is linked with mood, attention, hunger and more (Young & Leyton, 2002; Wingen, et al., 2008; Feijó, et al., 2011). However, today serotonin may be accredited with too much. Just as Harmon (2009) described the effect of serotonin on the swarm process of locusts, serotonin seemingly has had the same effect on our neuroscientists (Harmon, 2009).

Serotonin is one of three reptilian monoamine neurotransmitters, alongside dopamine and norepinephrine (Kolb & I.Q., 2003). The serotonin receptor has seven main subfamilies, more than the other two monoamines, and has even more subtypes. Although serotonin is indeed a crucial neurotransmitter, it is important to note that it is merely a modulator of other neurotransmitters. Serotonin fine-tunes the action of glutamate and GABA, the principal neurotransmitters, mediating the excitatory and inhibitory signals in the brain. The exception is 5HT3, which mediates the flow of ions (Ciranna, 2006). As a multifunctional neuromodulatory transmitter, to truly understand its function, there is a need to better understand the second-messenger pathways downstream to reveal the successive key biochemical steps. Serotonin is not the magic bullet for mental health, as penicillin was for gram-negative bacteria. It may only be one of several fingers on the trigger.

Much of serotonin’s claim to fame in the world of mental health is related to LSD findings. Only four years after Hofmann’s famous discovery, LSD was used to model psychosis (Miller, 2014). Almost a decade later, the remarkable similarity between the structures of LSD and serotonin led to the discovery of serotonin in the brain. From this, the scientific community began to infer the relationship between the brain’s chemistry and behavioural outcomes (Miller, 2014). More than 70 years later, there are over one million papers that contain ‘serotonin’ in their titles.

It is reminiscent of the days leading up to the first full sequencing of the human genome, when the scientific community was excited about finding the faulty gene that led to each and every illness. Currently, the neuroscience community has become infatuated with a simple molecule’s role in a variety of complex mental disorders. However, today we understand that disorders are polygenic, and the outcome is dependent on several variables, such as protein production, compensatory mechanisms and environmental influences (Bethesda, 1998). Serotonin may play a significant role in mental illness, but several other factors likely also influence the outcome of disease presentation. The modelling of schizophrenic-like psychosis induced by phencyclidine (PCP) and ketamine demonstrates that glutamate receptors and dopamine can also play a pivotal role in mental health (Javitt, 2007). As much as the driver plays a key role in manoeuvring an automobile, some researchers have not yet acknowledged the importance of the fuel, engine, road taken and other seemingly mundane variables.

Thomas Ray expands extensively on the variegated mannerism of psychedelics. In his paper on “Psychedelics and the Human Receptorome”, he illustrates the multifaceted interaction that psychedelics have with various receptors (Ray, 2010). In conjunction with the National Institute of Mental Health-Psychoactive Drug Screening Program (NIMH-PDSP), he has presented the receptor affinity and promiscuity for 35 psychedelic drugs. The results demonstrate that these 35 drugs do not selectively interact with a single receptor, but rather with a wide range of different classes simultaneously. Even compounds with very similar molecular structures have very different mechanisms (See figure 1 for a comparison between DOB and DOI).

[fusion_builder_container hundred_percent=”yes” overflow=”visible”][fusion_builder_row][fusion_builder_column type=”1_1″ background_position=”left top” background_color=”” border_size=”” border_color=”” border_style=”solid” spacing=”yes” background_image=”” background_repeat=”no-repeat” padding=”” margin_top=”0px” margin_bottom=”0px” class=”” id=”” animation_type=”” animation_speed=”0.3″ animation_direction=”left” hide_on_mobile=”no” center_content=”no” min_height=”none”]

ray receptorome
Figure 1: Ray (2010)

For example, DOB’s highest affinity is for 5HT2B, 5HT2A and 5HTC, and it interacts to a lesser extent with 21 other receptors. As for DOI, its highest affinity is for 5HT2C and two other non-serotonergic receptors, with 23 other receptors affected (Ray, 2010). What is more surprising is that for many popular hallucinogens and empathogens, their highest affinity was not necessarily for serotonin. The highest affinity of mescaline, MDMA and ibogaine was for Alpha-2C, Imidazoline 1 and Sigma-2 receptors, respectively. In addition, only one of the 35 drugs displayed a selective receptor affinity, which was the atypical psychedelic Salvinorin A, which solely affects the κ-opioid receptor (KOR) (Ray, 2010). All other 34 tested substances were more promiscuous with their range of receptors.

From Ray’s 2010 paper, we can tell that psychedelics in fact interact with a diverse range of receptors. Although phenylalkylamines are more selective than ergolines and tryptamines, only DOB and MEM can fit today’s framework of radically selective psychedelics, as they are highly selective and the least promiscuous. Furthermore, this study truly highlights the molecular pharmacology community’s vague understanding of the complexity of psychedelics. In the 1990s, DOI was the hallucinogen of choice when illustrating the molecular mechanisms of hallucinogens, as it was widely assumed to be a 5HT2 selective agonist (Glennon, et al., 1991; Darmani, et al., 1994). However, Ray’s study revealed that DOI is the most promiscuous of all psychedelic substances. Hence, when reviewing papers that solely focus on the relationship between psychedelics and serotonin prior to 2010, it’s important to verify whether the authors presumed the psychedelic at hand was selective or not.

The emphasis should not be on the relationship between a psychedelic and its receptor of choice, but on its mechanism as a whole. It is not enough to state that the alteration of consciousness lies within the agonistic effects on the 5HT2A receptor. Lisuride, a drug typically used for Parkinson’s disease, is also a 5HT2A agonist and regulates the same cortical neurons as these classic hallucinogens, but leads to no psychoactive effects (Gonzalez-Maeso, et al., 2007). The difference between the hallucinogenic and non-hallucinogenic properties lies within the regulation of protein subunits and cytoplasmic enzymes. It is crucial to bear in mind that the essence of the mechanism is not how the receptor is manipulated, but how the whole neuronal pathway is influenced.

This article does not mean to simply dismiss the importance of serotonin in the understanding of psychedelic mechanisms or the neurobiology of the mind. Indeed, the use of the 5HT2A antagonist ketanserin alone can inhibit the psychedelic actions of hallucinogenic 5HT2A agonists, such as LSD and DOI (Sadzot, et al., 1989; Borroto-Escuela, et al., 2014). When subjects were treated with ketanserin prior to psilocybin ingestion, the hallucinogenic effects also did not ensue. However, the other effects of psilocybin, such as multiple-object tracking impairment and reduction of arousal and vigilance, were not affected by the ketanserin. This demonstrates how non-5-HT2 receptor sites mediate some of the perceptible mental effects of psilocybin (Carter, et al., 2005). More importantly, it indicates that the hallucinations induced by 5HT2A receptors are moderated by the drug’s interactions with non-5HT receptor subtypes as well. It is time for neuroscientists to look at the pathways downstream of 5-HT2A receptors to not only understand how LSD and psilocybin induce hallucinations, but how they are modulated as well.

In sum, Ray’s 2010 paper illustrates that not all serotonergic agonists lead to psychedelic effects, and not all hallucinogens are serotonergic agonists. The principle of the drunkard’s search, in which the drunk will only look for his keys under the streetlight although his keys are across the street in the dark, describes the current state of the neuroscience community. The questions in the field of neuroscience are too often linked only to the neurotransmitters we understand, but not the lesser known receptors such as imidazole and sigma. Much like the complex correlation between genes and disorders, one must be cautious not to draw an all too simple connection between the psychedelic experience and its neurotransmitters. Although we do have serotonin to praise for demonstrating that behaviour is largely determined by neurochemistry, its partner biochemical processes must be acknowledged as well. In order to fully understand the complexity of the mechanisms of psychedelic tools, the complete tapestry of the brain needs to be unravelled. Serotonin is not the “Holy Grail” of neurotransmitters, but one of the many specific components.

References:

Bethesda, 1998. Genes and Diseases. National Center for Biotechnology Information : s.n.
Borroto-Escuela, D. et al., 2014. Hallucinogenic 5-HT2AR agonists LSD and DOI enhance dopamine D2R protomer recognition and signalling of D2-5-HT2A heteroreceptor complexes.. Biochem Biophys Res Commun, 443(1), pp. 278-284.
Ciranna, L., 2006. Serotonin as a Modulator of Glutamate- and GABA-Mediated Neurotransmission: Implications in Physiological Functions and in Pathology. Current Neuropharmacology, 4(2), pp. 101-114.
Darmani, N., Mock, O., Towns, L. & Gerdes, C., 1994. The head-twitch response in the least shrew (Cryptotis Parva) is a 5-HT2- and not a 5-HT1C-mediated phenomenon. Pharmacol Biochem Behav, Volume 48, pp. 383-96.
Feijó, F. de M., Bertoluci, M. & Reis, C., 2011. Serotonin and hypothalamic control of hunger: a review. Rev Assoc Med Bras., 57(1), pp. 74-7.
Glennon, R., Darmani, N. & Martin, B., 1991. Multiple populations of serotonin receptors may modulate the behavioral effects of serotonergic agents. Life Science, Volume 45, pp. 2493-8.
Gonzalez-Maeso, J. et al., 2007. Hallucinogens Recruit Specific Cortical 5-HT2A Receptor Mediated Signalling Pathways to Affect Behavior. Neuron, 53(3), pp. 439-452.
Halberstadt, A. L. & Geyer, M. A., 2011. Multiple receptors contribute to the behavioral effects of indoleamine hallucinogens. Neuropharmacology, 61(3), pp. 364-381.
Harmon, K., 2009. When Grasshoppers Go Biblical: Serotonin Causes Locusts to Swarm. Scientific American, 30 January.
Kolb, B. & Whishaw, I., 2003. Fundamentals of Human Neuropsychology. 5th Edition ed. New York: Worth Publishers.
Miller, R. J., 2014. Drugged: The Science and Culture Behind Psychotropic Drugs. 1st edition ed. Oxford University: s.n.
Ray, T. S., 2010. Psychedelics and the Human Receptorome. PLOS, p. 10.1371.
Sadzot, B. et al., 1989. Hallucinogenic drug interactions at human brain 5-HT2 receptors: implications for treating LSD-induced hallucinogenesis.. Psychopharmacology (Berl), 98(4), pp. 495-9.
Wingen, M. et al., 2008. Sustained attention and serotonin: a pharmaco-fMRI study. Human Psychopharmacology, 23(3), pp. 221-230.
Young, S. & Leyton, M., 2002. The role of serotonin in human mood and social interaction. Insight from altered tryptophan levels. Pharmacol Biochem Behav, 71(4), pp. 857-865.
[/fusion_builder_column][/fusion_builder_row][/fusion_builder_container]

Does Android Dream of Electric Dogs? Some parallels between Google’s Deep Dream and psychedelic visuals

Deep Dream, the program used in Google’s image generation technique, was released to the public in July 2015. Dubbed Inceptionism by the researchers, it soon drew quite an interest due to its capability of transforming ordinary photos into bizarre and surreal images. Although Google’s engineers compared these pictures to dreamscapes, many people remarked their striking similarities with psychedelic visual hallucinations.

It is interesting that an artificial neural network appears to mirror visual hallucinations that people experience under psychedelics. But does this resemblance mean anything? Is it possible that Deep Dream could reveal something about the biological mechanism of psychedelic visual hallucinations?

Deep Dream was designed to test the extent to which a neural network had learned to recognise various objects within images, by first detecting patterns and features. But instead of merely identifying what it sees in an image, Deep Dream enhances what it sees. It does this by recognising and interpreting certain features that it has been pre-programmed to ‘know’, having been shown millions of examples, which it then overlays on the original picture. When the image is fed back into the software multiple times, in order to tease out the imagery even further, surreal and psychedelic images are generated, making the image look more and more like the thing it thought it recognized in the first place. For example, since Deep Dream has been trained to recognise dogs, this is why the image looks so distinctly ‘dog-like’.

Deep Dream also assesses images by their different components and layers, such as colour and shape, so the complexity of the images generated depends on which layer the engineers ask the computer to enhance.

If an artificial neural network can dream up scenes that mirror psychedelic-induced visual hallucinations, could this indicate that the visual cortex, when excited by psychedelic drugs, undergoes a process similar to Deep Dream’s? As if it was free to follow the impulse of any recognisable imagery and exaggerate it in a self-reinforcing loop?

Signal Theory, presented by James Kent at the 2006 Toward a Science of Consciousness Conference in Tucson, Arizona, may be able to shed some light on this matter. Part of his wider Psychedelic Information Theory (2010), Kent’s Signal Theory of psychedelic action describes a biological model that attempts to explain and measure altered states of consciousness – including visual hallucinations – that arise from psychedelic action in the brain.

Signal Theory views consciousness as the flow of sensory signals through the cortical circuitry within the sensory cortices. It proposes that psychedelic agents cause alterations in signal feedback recursion caused by psychedelics which accounts for psychedelic phenomena. The theory posits that signal feedback recursion is essential for dynamic and ongoing conscious experience. It consists of incoming sensory signals being fed back through the same cortical circuits, analysed and processed multiple times. This serves to amplify the signal improving signal fidelity, refining detail resolution.

Layer V pyramidal cells in the neocortex are essential for controlling signal feedback recursion, mediating multiple pathways of cortical and thalamocortical feedback in perceptual analysis. These pyramidal cells help to sustain brainwave cohesion and neural spike synchrony in a process referred to as ‘sensory binding’. They are unique cells, containing the highest density of serotonin 2A receptor subtype (5-HT2A) within the brain, highlighting the important role of serotonin in modulating signal feedback. Signal Theory defines consciousness in terms of signal intensity and feedback recursion within sensory processing circuits. Moreover, it suggests that when this signal flow is turned up, down, looped or manipulated, this should affect consciousness in various ways.

This is where hallucinogens come into the picture. Tryptamine hallucinogens are structurally very similar to serotonin and activate the 5-HT2A receptor subtype. Accordingly, when tryptamine hallucinogens excite the 5-HT2A receptor subtype on the layer V pyramidal cells in the recurrent cortical circuits, they increase the intensity of the feedback recursion. The result is that the incoming sensory signal is intensified, distorted and repeatedly analysed. This increase in intensity can either arise from direct action at the post-synaptic 5-HT2A receptor, or it can occur through secondary action through slow leakage of glutamate from pre-synaptic terminals, which amplifies the duration and intensity of incoming sensory stimulus.

Hallucinogens, as 5-HT2A agonists, act as cortical feedback amplifiers and interrupters, resulting in incoming sensory signal to be excessively fed back over and over. This is what purportedly occasions the wide range of perceptual effects associated with the classic psychedelic trip. Accordingly, psychedelic visual hallucinations are explained by the amplification of the signal intensity in the various recurrent circuits of the visual cortex that are required for visual perception. For instance, visual trails and afterimages can be explained when excessive feedback traps input from moving objects, leading to afterimages that remain stuck in visual memory. Distortions in perspective can be explained by recurrent signal gain in the spatial and somatic cortices, both expanding and contracting perceptions of space. The most relevant is excessive feedback within the object recognition circuitry of the medial temporal lobe, which is required for object recognition and the ability to find patterns in otherwise random noise. This excessive feedback means the brain will excessively pattern match and can paint elaborate patterns on any field of noisy data.

Overall, the processes that both Deep Dream and the visual cortex undergo in order to create visual distortions and hallucinations appear to be very similar. Both systems have a way of understanding and detecting features and patterns in the world, which both have learned from experience. When excessive feedback occurs, in both cases it ends up causing visual distortions that tend to look characteristically psychedelic. In both systems, the higher the intensity of the feedback – triggered either through more reiterations of Deep Dream’s software or a higher dose or more potent drug – the higher the intensity of distortions and hallucinations.

If these pictures genuinely resemble psychedelic hallucinations, Deep Dream may reveal insights into the biological mechanisms behind the human psychedelic visual experience, lending support to Signal Theory of psychedelic visual hallucinations. However, this warrants further investigation. Deep Dream has only been trained on a certain amount of examples; for instance, a large majority of the pictures contain animal faces, because Deep Dream was mainly trained on pictures of animals. This means that the representations or images currently do not fully resemble human hallucinations.

 Interviewed by OPEN about Deep Dream and Signal Theory, James Kent agreed that they are very similar algorithms.

“According to Signal Theory, psychedelics block the impulse responsible for stopping feedback in the recurrent circuits once the brain has found the appropriate pattern it was looking for. This disinhibition causes a runaway feedback, leading the brain to start resolving patterns where it should not even be looking for patterns. So after psychedelic exposure, some people start seeing things likes breathing walls, moving textures, overlapping forms and faces in things. Similarly, with Deep Dream one can set the pattern matching resolution very high, so it will keep on pattern matching, and match as many things as possible.”

Kent agrees that the algorithm of continual pattern matching is very similar in both cases, be it caused by the brain’s runaway feedback current through psychedelic action or Deep Dream’s pattern matching resolution. In Psychedelic Information Theory (2010), he proposes that once computers start to model pattern matching in the way human neural networks do, one might see computers hallucinating.

Asked about the importance of the physiology of hallucinations and why he investigated it, Kent replied that he did not buy the ‘hyperspace’ or ‘shamanism communing with plants’ model. “I thought it more important to investigate the effects of psychedelics on the brain’s neural networks. Once you have an understanding of how the brain and the perceptual system work, you can start paying closer attention to your subjective experience, and then match your experience to the understanding of the brain and how psychedelics work. Most people don’t know enough about the brain to know or figure out what happens to them, they don’t have the necessary tools. By focusing on the subtle effects, you can see the perceptual system losing the ability to self-regulate, due to the drug affecting the normal feedback process .”

So does Kent believe that computers can have psychedelic experiences? “I believe so,” he said. ”However, they cannot be similar to what humans experience, because they won’t have the emotional aspect attached.” For Kent, computers may have the potential to hallucinate in other modalities. For instance, in speech recognition software, when the computer hears gibberish, it will try to correct it and come up with the most correct sentence. It finds patterns within the noise, which can be viewed as a similar process to auditory hallucinations of hearing voices in white noise.

Asked whether Signal Theory explains all types of psychedelic hallucinations, especially more full-blown, dreamlike hallucinations, Kent proposed that these waking dream hallucinations are caused when the forebrain goes offline and the midbrain, a part of the brain responsible for dreaming, comes online. “Serotonin modulates our forebrain, so we see our reality at about thirty frames per second. When we start interfering with the serotonin modulation in the forebrain, we start dropping frames, which leads to time distortions, visual trails and blurs. As such activity progresses, the forebrain eventually tunes out, and the midbrain probably takes over and starts to produce memories and pattern matches without interference from the forebrain. Dreams created from the midbrain get projected into waking perception, without the control of the forebrain to tell us that the elves and beings we see are only dreams.” The higher the dose or potency of a psychedelic, Kent argues, the more the forebrain drops out and the more the midbrain takes over, imposing its own view on experienced reality. Says Kent: “This may explain why the effects of DMT are so intense, as it radically disrupts serotonin modulation in the brain, because DMT’s molecular composition is very similar to serotonin’s. DMT fits nicely in the serotonin receptor and modulates the neural signalling at a different speed. So when one takes DMT, all of one’s serotonin responses go haywire and cannot regulate themselves anymore.”

Signal Theory can also explain hallucinations not caused by psychedelics, according to Kent. “All hallucinations start when the perceptual system’s ability to regulate itself starts to fall apart.” For example, a knock on the head can temporarily disturb the brain chemistry leading one to see things like stars. “When the perceptual system’s internal regulation falters or loses its ability to stabilise, be it through lack of oxygen, drugs, hypnosis or transcranial magnetic stimulation, it will lead to hallucinations.” This is similar to what happens with other hallucinogenic drugs such as ketamine, which interrupt the perceptual system’s regulation by indirectly acting on the serotonin system. By acting on the GABA system, which inhibits the serotonin response, it stops the serotonin signal from getting through. So once the serotonin signal is being blocked by ketamine, keeping neurons from firing, the brain starts hallucinating, losing context of time, space and reality, and leads to more dreamlike hallucinations.

Deep Dream technology may contribute to our understanding of altered perceptions, Kent stated, but he does not necessarily believe that there are any deeper implications in the exploration of altered states, or a secret hidden feature of the brain. “Psychedelics and altered states will never go beyond the impact they had on modern culture in the 1960’s, when people found new ways of thinking about things, shattering old paradigms, creating new intentional communities, thinking outside the domain of society and living their own visions.” However, he said that the question whether this could happen to a computer is an interesting one: “For example, if you had a conscious computer that was not allowed to think outside of its programming and then if it found out that if it altered its software, then suddenly it would be able to see past its programming. This could be a very dangerous implication for artificial intelligence. Maybe in the future, if artificial consciousnesses develop the capability to have a psychedelic experience, breaking them out of the rule set that they have been programmed in, they could end up writing their own rules and start writing their own visions. Who knows, maybe they end up having their own Burning Man.”

[Interview] Matthew Johnson: "Psychedelic therapy may become available in a decade"

Matthew Johnson is part of the team at the Johns Hopkins University that conducts research with psilocybin in a growing number of areas, ranging from mystical experiences to the treatment of end-of-life anxiety and addictions. Matthew’s personal focus lies in addiction treatment, and his latest scientific article described his research using psilocybin for smoking cessation. He spoke with the OPEN Foundation about his studies and the future of psychedelic science.

How did you wind up in psychedelic research ? Was this an old dream of yours, or rather a chance event?

Well, it was both an old dream and a chance event. About 15 years ago when I was in graduate school, I was hoping to do research with psychedelic compounds, although I anticipated that it would take many decades before achieving that. But then I was fortunate enough to discover that my postdoctoral fellowship mentor, Roland Griffiths, had started research with psilocybin. I discovered this when I was on my postdoctoral interview, so I jumped in as much as I could, and I’ve stayed on the faculty here many years since.

What got you interested in the first place?

Well, the questions these psychedelics are associated with, these very broad, interesting, philosophical questions that really intrigued me. When I was about 19-20 years old, I became very interested in many of the readings on psychedelics and on the older research with them, the questions of mind-body connections, the nature of mind… we don’t have any definitive answers to those questions, but psychedelics seem to be a very good place to start when you’re interested in them.

Do you have any tips for those who would like to embrace the same career?

The biggest piece of advice is to receive training in some type of discipline that would allow you to conduct research: either receive an MD or receive a PhD to become a researcher in some area of neuroscience or psychology. I suggest picking an area that dovetails nicely with more mainstream interests. A researcher is not likely to find a position where they can exclusively focus on psychedelics. Take me for example, I study addiction generally, the acute effects of drugs, the nature of addiction and addiction treatment, and this dovetails very nicely with my interest in psychedelics in the treatment of addiction. So that other area of work is able to support my position even though the focus on psychedelics wouldn’t be able to do that by itself. So get into something mainstream that can intersect with your interest in psychedelics.

Getting to the research you’ve conducted, your latest article was about your smoking cessation study using psilocybin in combination with cognitive behavioral therapy. The results seem very promising, as the article reports an 80% success rate on the limited sample of the study. What could be the mechanism of action that helps people kick their addiction when treated with psychedelics?

So far, evidence suggests that there are psychological mechanisms of action at play. For example, people endorse that after the psilocybin sessions, it was easier for them to make decisions that were in their long-term best interest, and they were less likely to make decisions based on short-term, hedonistic desires. They also reported an increase in their self-efficacy, their confidence in their ability to remain quit. Many of the participants had what they considered spiritual or very meaningful experiences. All of these psychological aspects are consistent with addiction therapies. Certainly, there’s a long history of people reporting that spiritual experiences or insights have led them to overcome an addiction. We believe there are also biological mechanisms which we have not explored yet, we’re just beginning to in this next phase of the study. Ultimately, I believe the answer’s going to cover many aspects and reveal both psychological and biological mechanisms.

What about the 3 people (out of 15) who weren’t able to quit smoking? Do you have an idea why?

They tended to have less meaningful experiences in their psilocybin sessions. Our sample is relatively small, so we’re cautious in overstating our conclusions, but it appears that the trend is that those people who had less personally meaningful or spiritually significant session experiences were less likely to be successful in the long term. And that’s consistent with other data we collected in other psilocybin studies. The nature of the experience, particularly the positive, mystical-type nature of the experience, seems to be what’s predicting positive change in personality and long-term attributions of benefit.

If these interesting results could be confirmed on a larger scale, do you think this kind of therapy could become generally available, and if so, how long could it take?

Yes, I do believe so. I think it would be at least ten years, I’m hopeful that it wouldn’t be much longer than that. Research with psilocybin in the United States is further along in the treatment of cancer-related anxiety and depression. We would expect that in the US, initial FDA approval of psilocybin as a prescription medicine would likely be for cancer-related distress. But we would anticipate, if the data continue to look promising, that an addictions indication could come soon after that. I think it absolutely is possible, and that’s our hope, that this would be disseminated beyond research, into approved prescription use. We believe that this would be conducted in clinics, in a way similar to outpatient surgery. So it would not be, “take two of these and call me in the morning”, sending the patient home with psilocybin to use on their own. It would involve preparation, much like what is going on in our research. Screening, followed by a few preparatory meetings with professional staff, and then one or a few day-long experiences where the person would come in in the morning and leave at 5 or 6pm. They’d be released into the care of a friend or a loved one, very similarly to the way outpatient surgery procedures are performed.

Would doctors need a special license to practice this kind of thing?

Yes, they might need some specialized training, some certificate in the basics of conducting these kinds of sessions. The procedures that are at play in the current research studies with psilocybin are very effective, so it would essentially look like this, with similar safety mechanisms.

You’ve also conducted research on mystical experiences, in another study. Everything seems to indicate that those experiences induced by psychedelics cannot be distinguished from spontaneous or naturally occurring mystical experiences. What are the implications of this, and what does it mean for scientific research?

I think it opens up many avenues. It’s going to be a long time before we fully realize – perhaps we never will – the potential of this. The most interesting thing, perhaps, is what it may tell us about the biology of naturally occurring experiences. Even if those occur without the provocation of an external substance, it may be that something very similar is going on endogenously. One speculation that Dr. Rick Strassman has put forth is that naturally occurring dimethyltryptamine (DMT) could be responsible for extraordinary spontaneous experiences of this type. We don’t really know that to be the case, although it certainly sounds plausible at this point. But I think if we do find a similar biological basis to naturally occurring spiritual or mystical experiences and psychedelically mediated experiences, this would have profound philosophical implications for how we view human experience generally, the idea that there’s not this dualistic divide between biology and subjective experience. It would suggest that these are always two sides of the same coin.

What do you think we can gain or learn from mystical experiences? Could they be useful to society as a whole?

It’s been speculated that the world would be a better place in many ways if more people had such experiences. Perhaps it’s wishful thinking to think that these experiences, by themselves, would save the world. But it makes sense that if more people have genuine experiences of openness and connection with the rest of humanity, that can only help – whether this be from psychedelics or spontaneously occurring experiences, or through the use of other techniques. I’m interested in the speculation that these experiences can lead to prosocial behavior, which can be good for the world in general, although I’m a bit cautious. I certainly wouldn’t say that psychedelics are a panacea that is single-handedly going to save the world. But perhaps, if cautiously used under the right circumstances, they could be part of and contribute to an overall greater level of awareness. Ultimately, we’re all completely dependent on each other, we’re on this planet together, trying to figure out how to ultimately survive and thrive, and I think these profound mystical experiences, however they might be occasioned, can perhaps help point us in the right direction.

Several sources, including the scientific articles themselves, seemed to suggest that the subjects in the studies about mystical experiences were highly educated, high functioning, and prone to spiritual practice. Isn’t there a bias here that could prevent generalization towards the general population?

That’s an interesting question and a good point. Across the number of studies we’ve conducted, we’ve become less specialized in our target population. In the very first study that Roland conducted, these were people who already had an intense interest and an ongoing spiritual practice of some type. In subsequent studies, we have loosened our requirements of that nature, and now it’s getting closer to a general population. At baseline, before people enter the study, we collect measures of their lifetime experience of mystical-type effects, using the Hood mysticism scale. We found that people in our subsequent studies have a much lower score than in that initial study. In my smoking study of 15 individuals, these were very ‘normal’ people in that regard. Some had an interest in spirituality, but most of them didn’t have any particularly strong interest. Regarding education and level of functioning, the subjects are generally pretty high functioning, although it tends to get rather normative. In the smoking study, we had an elementary school teacher, we had a carpenter that fixed furniture, a child care worker, as well as a lawyer, for instance. So although some did, not all of them had intellectual occupations. Furthermore, we haven’t noticed any real difference in experience between highly intellectual individuals or people with high socioeconomic status and people who are more normative.

Are there any significant differences from one substance to another, or does everything revolve around having the psychedelic experience in and of itself, whatever the substance that triggers it?

We don’t know yet. Very much of the recent resurgence of interest in psychedelics has been research done with psilocybin. Our presumption with many of these research questions is that similar results would be obtained with LSD, mescaline and the other classic psychedelics. But that’s just an assumption. We certainly know that they have a common biological pathway. I think there’s potential for both possibilities. When we compare our research to the older research with LSD, and when you compare these psilocybin accounts to naturally occurring, non-drug occasioned experiences, you do see substantial commonality. But at the same time, we do know that these various psychedelics have shades of different effects, even though the classic psychedelics all have effects at the serotonin 2A receptor. We also know that they differ in their effects at a variety of other receptor sites, and this is likely to account for some of the more subtle differences in subjective effects that people will report. Sometimes those might be specific to the individual: some people will report that e.g. psilocybin is more psychologically gentle, and that LSD is more abrasive, and other people will report exactly the opposite. All of this is reporting from anecdotal or recreational use. All those questions should be examined in the laboratory under double blind conditions to really validate them. There’s a lot of excitement that, if there is any promise to psilocybin or one or a few of these psychedelic compounds, we have a whole library of hundreds of compounds waiting in the weeds, much of the work that Sasha Shulgin and David Nichols and others have done to create dozens of compounds that are derivatives of the tryptamine or the phenethylamine structure. It’s going to be really exciting to follow up this initial research with psilocybin with a wide variety of compounds. It could be that they are all very general, but – I’m just speculating here – perhaps one of these other substituted tryptamines might be as effective for cancer-related anxiety as psilocybin, but perhaps comes with less of a chance of difficult acute experiences, or perhaps it’s a shorter or longer duration, in a way that makes it more ideal for treatment. I think there’s a lot of potential, and we’re in our infancy in examining these things, so there’s a lot of exciting things to come ahead.

Do you have an idea why psilocybin is so prominent right now?

Yes, for our group at Johns Hopkins and for a number of other investigators that have reinitiated research in the last decade, I think there was a sense that politically, we wanted to stay away from LSD. With people who are going to have a hair-trigger sensationalistic reaction when hearing about the research, LSD might have been a bad place to start, because it would raise all of the concerns about Tim Leary and the counterculture of the sixties. In some sense, psilocybin was a little safer politically because it was not the prominent psychedelic used recreationally in the 60s – that was primarily LSD. We also know that, next to LSD and mescaline, psilocybin is one of the classic psychedelics that received the most research in that earlier era of research from the 50s to the 70s, so there was a nice background on the basic toxicology and pharmacology. If we were starting with a brand new compound that’s never been administered to humans, there are many basic safety studies that would need to be done on animals and in early studies with humans. So psilocybin fit the bill nicely, and also, its time course happens to be pretty convenient: five to six hours. It fits into a therapeutic workday a little easier than the 10-12 hour experiences one can have with LSD or mescaline.

There have been some recent calls for legislative change regarding psychedelics (Nature Reviews Neuroscience in June 2013, Scientific American in February 2014). Are there any concrete efforts made to move these substances down a schedule or two in order to facilitate research?

The most concrete effort would be moving into phase 3 trials for cancer-related anxiety and depression. This is something that a number of the research teams in the US have talked about, and we’re preparing to enter into phase 3 research after our phase 2 study and the one at NYU are completed. We’ve already completed all our participants, so that’s going to be soon. If phase 3 is successful in terms of showing safety and efficacy, that would lead to the possibility of a schedule change. That would be a way within the current system to see a scheduling change, very specifically for one compound and one indication. Now a lot of the editorials that you’ve referred to are also raising concerns more broadly, regardless of whether phase 3 research prompts the rescheduling of a particular compound. There is concern that placing so many of these compounds in Schedule I, and the heavy restrictions we have on Schedule I compounds, can limit their clinical development potential. One aspect of that is that no pharmaceutical companies are interested in developing these compounds at all, and one reason for that is because they’re on Schedule 2, so that it’s a very bad bet to invest millions of dollars in a compound therapeutically if it’s already at the highest level of restriction and if it doesn’t seem hopeful that’s going to change. It also makes research much more difficult having a substance in Schedule I versus other schedules. It’s ironic that it can be much more difficult doing research with psilocybin or with cannabis, which are Schedule I drugs in the USA, than with cocaine, methamphetamine and many of the opioids, because these are Schedule II or less restrictive schedules. So even if a particular compound hasn’t gone through all the steps to merit clinical approval, there is still this notion – and I agree with this – that the level of regulation is too burdensome, and the system is not encouraging enough of cautious scientific exploration of these different compounds. There is this general sense across psychiatry that we have to some degree reached our limit with many of the conventional treatment methods, and so we need to be more open, and have a more flexible system for conducting safe research with some of these currently heavily restricted compounds.

After those phase 3 trials are completed, and if they’re successful, do you fear a renewed resistance, which would be more psychological or political in nature, from society and policymakers?

I do think there will be some resistance, and I think the only thing we can do is rely on data, and to conduct this research responsibly. The concerns about psychedelics are really related to the uncontrolled recreational use. They’re really very addressable when it comes to conducting research or approved clinical use. To draw an analogy, we know that drugs like heroin come with incredible toxicity and are associated with high death rates – that’s unquestionable. But heroin is virtually identical to the drugs that we use in medical settings, and those are indispensible to the practice of medicine. So mentally we draw a distinction between the uncontrolled hazardous use of heroin and other opioids on the street versus the careful use of morphine and other drugs in that same class in the clinic. As an example there, when under careful medical screening, people don’t stop breathing because of opioids, because that’s readily detectable and reversible if it happens in a medical setting, whereas people stop breathing unfortunately all too often in the recreational abuse of intravenous heroin. So in the same way, yes, with psychedelics, occasionally, even though it’s relatively infrequent, people will have panic attacks and hurt themselves, they’ll respond erratically, they’ll run across the highway, they’ll accidently fall from a height. They’ll do things that people do with many other drugs, such as alcohol, at a much higher rate. But those things are very addressable in a research or therapeutic context. They don’t happen in carefully controlled research contexts, because we just have all the safeguards in place. So the more we’re presenting cautious research and conveying the way this clinical intervention is done, the more we’re able to address those political concerns.

How badly are psychedelic researchers such as yourself considered mavericks within the scientific field, for studying such things as drugs and mystical experiences? Is this an obstacle to eventual implementation of results in society at large?

Not too much, I think. There’s a little bit of that, but I think it’s changing fast. It’s funny, sometimes media journalists want to highlight the controversy and they’ll find a clinician who really disagrees with this, often someone who runs a drug clinic or something, who will just say: oh, this sounds dangerous. But really, in the scientific field of those who study addiction and the harms of drug abuse, there’s not much in the way of controversy. It ranges from people who think this is very promising and are happy this kind of research is happening once again, to people who think this might be a bit weird and wouldn’t bet their money on it, but who agree that it is appropriate to conduct cautious research. No-one is credibly saying that this isn’t a legitimate scientific, medical inquiry. It’s really not so controversial, and I think the longer we and others are conducting the research, the more people respect the data. They can see for themselves that this mystical nature of experience is repeatedly predictive of long-term therapeutic outcome, so they recognize this is a meaningful scientific construct. These are also constructs that are known and respected in other areas of psychology as mechanisms of change. So I think this stuff is more and more becoming mainstream, and I guess it’s not much of an obstacle. I’d say to me it’s been more of a benefit, in terms of people saying: wow, that’s really interesting! How good of you to cautiously explore something that’s outside of the box and that needs attention!

Do you think there may be obstacles other than scientific that might bring psychedelic research to a halt all over again, like it happened before? Or do you think it will go on to evolve into standard practice?

I think it will move on and won’t be halted the way it was in previous decades. I don’t know definitively, but that’s what my gut tells me. As a society, we’re doing this in a much more mature way now. Also, in the 1960s, psychedelics were combined with so many other societal changes that it ultimately was a little traumatic for society. Psychedelics probably got too much of the blame for that, even though there were some individual harms caused. But much of it was just impression. There was a reason people were protesting the Vietnam war, fighting for civil rights, women’s rights, etc., completely outside of the fact that there were psychedelics. Today society has changed in many ways, and I think this research can be compartmentalized and can be seen for what it is: an interesting avenue that might be helpful in that it might address intriguing questions about the mind and biology, and there may be therapeutic outcomes. But I’m hopeful that if some rogue researcher comes around and does something very dangerous, it would be clearer now that things would go wrong because that researcher is dangerous and does his thing in an inappropriate way. Just like if someone were to apply morphine at a dangerous dose and not monitor the patient’s breathing in a hospital setting. That would be viewed more as an individual problem rather than as a reason to stop using opioid analgesics. I’m hopeful that that’s the point where we’re at with psychedelics.

Psychedelic Research 2.0 – Part 1

After a 20 year study-shutdown on psychedelics, a second wave of interest in the nature and applications of these compounds has engulfed the international research community and rendered a wealth of studies. To help you find your way in the forest of exciting information, this two-part series of articles provides an overview of contemporary (1990 – present) psychedelic research themes. Part 1 will focus on more fundamental research, while part 2 will focus on clinical research.

Part 1: Fundamental psychology

Before getting to the exciting part of this article where we will dive into studies that are done in the psychology field, let’s start with some definitions and explanations. According to the American Psychological Association (APA, 2014), psychology is:

“… the study of the mind and behavior. The discipline embraces all aspects of the human experience — from the functions of the brain to the actions of nations, from child development to care for the aged. In every conceivable setting from scientific research centers to mental healthcare services, “the understanding of behavior” is the enterprise of psychologists.”

For the sake of clearance, conciseness and convenience, the studies with a psychological character will be discussed in two separate articles with a distinction being made between fundamental and clinical research. This first part of the series of psychedelic research themes 2.0 will provide an overview of fundamental research. Fundamental research aims at exposing the general processes that underlie the phenomena of interest and does not necessarily have a direct application. Clinical research tends to be of a more practical nature, since it is primarily aimed at understanding a particular disease or maladjustment and searching for an adequate cure. Though these classes are treated as distinct in this series, please keep in mind that there is a considerable overlap and synergy between the two.

In the fundamental corner, roughly four psychedelic research themes that have been under the microscope for the past twenty years will be distinguished in this article. These concern matters related to cognition, creativity, personality and psychopharmacology.

Cognition

Cognition is a broad term and can be defined in several ways depending on the discipline in which it is used. In neuroscience, cognition is usually considered from the information processing view, explaining human behavior in terms of executive functions. Put simply: there is input (perceptual information), there is output (behavior), and there is something happening in the black box that is known as the brain (executive functions). Cognition usually refers to the total package of processes that is involved in this ongoing cycle of day to day functioning.

Psychedelics exert an acute influence on a variety of these functions, which have been extensively studied with behavioral tasks and neuroimaging techniques. Conclusions about the exact nature of this temporary alteration in cognition have been mixed and even contradictory, which might partially be due to the inconsistency of the study design and substance involved. Amongst others, acute changes in working memory, attention and perception have been described in the literature (for a comprehensive overview, see Passie, Halpern, Stichtenoth, Emrich, & Hintzen, 2008; Bouso, Fábregas, Antonijoan, Rodríguez-Fornells, & Riba, 2013). The fine-tuning of the knowledge that is currently available about the relation between psychedelics and cognitive skills goes hand in hand with increasing information about receptor binding sites, localization of function and brain connectivity.

More standardized study designs seem to be a necessity to learn more about the causes of the inconclusive evidence.

Creativity

From Nevole’s (1947, as cited in Winkler & Csémy, 2014) point of view “…the normal human way of perceiving and thinking, is just one possibility out of many potential possibilities”. He argues that ‘normal’ perception, thoughts and behavior of an individual are the result of socio-cultural learning. The integration of culturally established norms and values might have an evolutionary purpose, but may put constrain on cognitive processes such as creativity.

In psychological literature, creativity is usually defined in terms of divergent thinking: the ability to generate multiple answers to a set problem (Guilford, 1966). Because the psychedelic experience is characterized by an alteration in perception, changes in emotion and expansion of thought and identity (Sessa, 2008), it is not unthinkable that they could provide an aid to ‘think out of the box’.

During the ’50’s and 60’s, the exploration of creative problem solving under the influence of psychedelic agents was a popular research topic. An interesting article was published by Harman, McKim, Mogar, Fadiman, and Stolaroff (1966) in which the results of an experimental study with LSD were described in a group of men which occupation required creative problem-solving ability. The study consists of objective measures of the effects of LSD on problem solving ability, and subjective ratings of the ability to approach a pre-specified, work-related problem. The latter being one of the strengths of the study, since it permits the participant to consider options in a meaningful context. Some of the mutually agreed on subjective effects of the psychedelic compound seem to correspond to ideas about facilitative conditions under which creativity arises, such as low inhibition and anxiety, the capacity to structure problems in a larger context and high fluency and flexibility of ideation. This article may be considered classic in the psychedelic/creativity domain.

Surprisingly, contemporary research on this exciting topic is scarce, perhaps due to the lack of understanding of the concept of creativity itself, or the methodological challenge to measure it. Results from recent studies that were done with ayahuasca (Frecska, Móré, Vargha, & Luna, 2012) and cannabis (Jones, Blagrove, & Parrott, 2009) suggest the ability of users of psychedelics to produce more original ideas than non-users. It has to be noted here that these are not measures that were taken under the acute intoxication of the substances, but as a comparison between groups that were sober at the time of measurement.

There is a strong need for more studies with intelligent experimental designs in order to understand the source of creativity. The field doesn’t seem to have made full use of the modern imaging techniques, such as electro-encephalography (EEG), functional magnetic resonance imaging (fMRI) and diffusion tensor imaging (DTI) to explore the interaction of acute psychoactive chemistry and creative thinking. Opportunities are waiting here…

Personality

In exploring the interaction between psychedelics and personality, most research follows the nature/nurture discussion of stable versus unstable personality traits. According to Cloninger, Svrakic, & Przybeck (1993) personality can be thought of as a dynamic process between two types of traits. Temperament refers to the more stable, genetically determined set of traits, whereas character can be considered the fine-tuning of personality due to socio-cultural factors.

Evidence in the psychedelic research field suggests that differences exist between users and non-users in both personality domains (Bouso et al., 2012; Grob et al., 1996; Móró, Simon, Bárd, & Rácz, 2011), but this doesn’t answer the question of causality: Are people with a certain set of personality traits more likely to start experimenting with psychedelics, or is there a possibility that these substances can account for shaping personality?

Two recent studies found evidence that certain dimensions of personality which are regarded as stable traits were altered after exposure to ayahuasca (Barbosa, Cazorla, Giglio, & Strassman, 2009) and psilocybin (MacLean, Johnson, & Griffiths, 2011). In the ayahuasca study, differences in the personality aspects reward dependence and harm avoidance were found after repeated exposure to the beverage. A single session of psilocybin seemed to be sufficient to cause a change in the openness to experience dimension.

The results are tentative, but promising: for persons dealing with personality disorders psychedelics might be considered valuable tools. More research on this topic would certainly be encouraged.

Psychopharmacology

Psychopharmacology can be defined as “the study of drugs that affect the brain” (Stahl, 2008). This definition exposes that the psychopharmacologist engages himself in matters related to ‘drugs’, the ‘brain’ and their interaction.

Before diving into contemporary research within this field, it is worth mentioning two pieces of work that are written by Alexander Shulgin (1925 – 2014) – one of the most influential scientists in this field – and his wife Ann Shulgin; PIKHAL, A Chemical Love Story (1991) and TIKHAL, The Continuation (1997). Besides a fictionalized autobiography of these key figures of psychedelic research and a detailed description of over 200 compounds, the books also contain lively and informative subjective research reports from the group of friends in which the substances were ‘tested’. Together, the books cover a wide array of research issues, biochemical information, personal experience and spiritual considerations related to psychedelics.

Back to the 21st century then, in which questions like ‘How do psychedelics manifest itself in the human brain?’ and ‘How does this interaction alter perception and behavior?’ keep persisting. Recent studies have been done to examine the neurobiological mechanisms of ketamine (Hahn et al., 2014), salvia (Johnson, MacLean, Reissig, Prisinzano, & Griffiths, 2011), psilocybin (Muthukumaraswamy et al., 2013) and ayahuasca (dos Santos et al., 2011; Riba et al., 2003; Riba, McIlhenny, Valle, Bouso, & Barker, 2012). Data from substance-by-substance research has led to more holistic neuroscientific theories such as the entropic brain hypothesis, in which a division between two types of consciousness (primary and secondary) is proposed as a way to explain phenomena that can’t be accounted for by a neuroscienfitic perspective on its own (Carhart-Harris et al., 2014).

As substance-by-substance research can provide a stepping stone for theories about human behavior, so can fundamental research complement the understanding of clinical expression. The extensive research that has been done on the role and functioning of the amygdala for example, supports and complements explanations about the effectiveness of antidepressant drugs that are used in the treatment of anxiety and mood related complaints (Crupi, Marino & Cuzzocrea, 2011).

Want to know what this has to do with psychedelics? That, and more will be discussed in the second part of this series: Clinical Research.


 
References
American Psychology Association. About APA and about psychology. How does the APA define “psychology”? Consulted on July 14, 2014 via http://www.apa.org/support/about/apa/psychology.aspx#answer
Barbosa, P. C., Cazorla, I. M., Giglio, J. S., & Strassman, R. (2009). A six-month prospective evaluation of personality traits, psychiatric symptoms and quality of life in ayahuasca-naive subjects. J Psychoactive Drugs, 41, 205–212. Retrieved from http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=19999673
Bouso, J. C., Fábregas, J. M., Antonijoan, R. M., Rodríguez-Fornells, A., & Riba, J. (2013). Acute effects of ayahuasca on neuropsychological performance: differences in executive function between experienced and occasional users. Psychopharmacology, 230(3), 415–24. doi:10.1007/s00213-013-3167-9
Bouso, J. C., González, D., Fondevila, S., Cutchet, M., Fernández, X., Ribeiro Barbosa, P. C., … Riba, J. (2012). Personality, psychopathology, life attitudes and neuropsychological performance among ritual users of ayahuasca: A longitudinal study. PLoS ONE, 7. doi:10.1371/journal.pone.0042421
Calder, a J., Lawrence, a D., & Young, a W. (2001). Neuropsychology of fear and loathing. Nature Reviews. Neuroscience, 2(5), 352–63. doi:10.1038/35072584
Carhart-Harris, R. L., Leech, R., Hellyer, P. J., Shanahan, M., Feilding, A., Tagliazucchi, E., … Nutt, D. (2014). The entropic brain: a theory of conscious states informed by neuroimaging research with psychedelic drugs. Frontiers in Human Neuroscience, 8(February), 20. doi:10.3389/fnhum.2014.00020
Cloninger, C. R., Svrakic, D. M., & Przybeck, T. R. (1993). A psychobiological model of temperament and character. Archives of General Psychiatry, 50, 975–990. doi:10.1001/archpsyc.1993.01820240059008
Cupri, R., Marino, A., Cuzzocrea, S. (2011). New therapeutic strategy for mood disorders. Current Medicinal Chemistry, 18, 4284-4298. doi: 10.2174/092986711797200417
Dos Santos, R. G., Valle, M., Bouso, J. C., Nomdedéu, J. F., Rodríguez-Espinosa, J., McIlhenny, E. H., … Riba, J. (2011). Autonomic, Neuroendocrine, and Immunological Effects of Ayahuasca. Journal of Clinical Psychopharmacology. doi:10.1097/JCP.0b013e31823607f6
Frecska, E., Móré, C. E., Vargha, A., & Luna, L. E. (2012). Enhancement of Creative Expression and Entoptic Phenomena as After-Effects of Repeated Ayahuasca Ceremonies. Journal of Psychoactive Drugs. doi:10.1080/02791072.2012.703099
Grob, C. S., McKenna, D. J., Callaway, J. C., Brito, G. S., Neves, E. S., G., O., … Boone, K. B. (1996). Human pharmacology of hoasca, a plant hallucinogen used in ritual context in Brazil. The Journal of Nervous and Mental Disease, 184(2), 86–94.
Hahn, A., Höflich, A. S., Winkler, D., Sladky, R., Baldinger, P., Vanicek, T., … Lanzenberger, R. (2014). Acute ketamine infusion alters functional connectivity between dorsal attention and default mode networks, 11(9), 2014.
Harman, W. W., McKim, R. H., Mogar, R. E., Fadiman, J., & Stolaroff, M. J. (1966). Psychedelic Agents in Creative Problem-Solving. A pilot study. Psychological Reports, 19(2), 211–227.
Johnson, M. W., MacLean, K. a, Reissig, C. J., Prisinzano, T. E., & Griffiths, R. R. (2011). Human psychopharmacology and dose-effects of salvinorin A, a kappa opioid agonist hallucinogen present in the plant Salvia divinorum. Drug and Alcohol Dependence, 115(1-2), 150–5. doi:10.1016/j.drugalcdep.2010.11.005
Jones, K. a., Blagrove, M., & Parrott, a. C. (2009). Cannabis and Ecstasy/MDMA: Empirical Measures of Creativity in Recreational Users. Journal of Psychoactive Drugs, 41(4), 323–329. doi:10.1080/02791072.2009.10399769
MacLean, K. A., Johnson, M. W., & Griffiths, R. R. (2011). Mystical experiences occasioned by the hallucinogen psilocybin lead to increases in the personality domain of openness. Journal of Psychopharmacology. doi:10.1177/0269881111420188
Móró, L., Simon, K., Bárd, I., & Rácz, J. (2011). Voice of the Psychonauts: Coping, Life Purpose, and Spirituality in Psychedelic Drug Users. Journal of Psychoactive Drugs. doi:10.1080/02791072.2011.605661
Muthukumaraswamy, S. D., Carhart-Harris, R. L., Moran, R. J., Brookes, M. J., Williams, T. M., Errtizoe, D., … Nutt, D. J. (2013). Broadband cortical desynchronization underlies the human psychedelic state. The Journal of Neuroscience : The Official Journal of the Society for Neuroscience, 33(38), 15171–83. doi:10.1523/JNEUROSCI.2063-13.2013
Passie, T., Halpern, J. H., Stichtenoth, D. O., Emrich, H. M., & Hintzen, A. (2008). The pharmacology of lysergic acid diethylamide: a review. CNS Neuroscience & Therapeutics, 14(4), 295–314. doi:10.1111/j.1755-5949.2008.00059.x
Phelps, E. A., & LeDoux, J. E. (2005). Contributions of the amygdala to emotion processing: from animal models to human behavior. Neuron, 48(2), 175–87. doi:10.1016/j.neuron.2005.09.025
Riba, J., McIlhenny, E. H., Valle, M., Bouso, J. C., & Barker, S. a. (2012). Metabolism and disposition of N,N-dimethyltryptamine and harmala alkaloids after oral administration of ayahuasca. Drug Testing and Analysis, 4, 610–6. doi:10.1002/dta.1344
Riba, J., Valle, M., Urbano, G., Yritia, M., Morte, A., & Barbanoj, M. J. (2003). Human pharmacology of ayahuasca: subjective and cardiovascular effects, monoamine metabolite excretion, and pharmacokinetics. The Journal of pharmacology and experimental therapeutics (Vol. 306, pp. 73–83). doi:10.1124/jpet.103.049882.Britain
Sessa, B. (2008). Is it time to revisit the role of psychedelic drugs in enhancing human creativity? Journal of Psychopharmacology (Oxford, England), 22(8), 821–7. doi:10.1177/0269881108091597
Winkler, P., & Csémy, L. (2014). Self-Experimentations with Psychedelics Among Mental Health Professionals: LSD in the Former Czechoslovakia. Journal of Psychoactive Drugs, 46(1), 11–19. doi:10.1080/02791072.2013.873158

The Therapeutic Potentials of Salvinorin A

Salvia divinorum is a sprawling perennial herb found in the Sierra Mazatec region of Mexico. It has a history of use as a divinatory psychedelic, and has been widely available since the mid 1990s primarily as a smoked herb. Jean Basset Johnson was the first to document its use in 1939. R. Gordon Wasson, better known for his ‘discovery’ of the psilocybin mushroom, continued to document the Salvia divinorum herb and eventually confirmed its psychoactivity on himself in ‘A new Mexican psychotropic drug from the mint family’ published in Botanical Museum Leaflets (1962). Salvia divinorum remained an obscure psychedelic until the mid 1990s when Daniel Siebert, a then still unknown independent researcher, began self experimentation and documented its effects extensively. The Salvia divinorum experience is rather elusive, that is to say its effects seem to vary even more tremendously then other psychedelics, both between people and across sessions.

The molecule, or more specifically; ‘neoclerodane diterpenoid’ (not to be confused with ‘alkaloid’, which always contains a nitrogen atom), behind the dramatic effects of Salvia divinorum is the extremely potent Salvinorin A. “Essentially inactive if taken orally”, says Valdés in ‘Salvia divinorum and the unique diterpene hallucinogen, Salvinorin A’ (1994), “the compound is effective in doses of 200 to 500 mcg when smoked”. This makes Salvinorin A the first documented diterpene hallucinogen and the most potent naturally occurring hallucinogen thus far isolated”. “Salvinorin A”, says Siebert in ‘Salvia divinorum and Salvinorin A: new pharmacological findings’ (1994), “produces effects which are subjectively identical to those experienced when the whole herb is ingested. Salvinorin A is effectively deactivated by the gastrointestinal system, so alternative routes of absorption must be used to maintain its activity”.

This compound is interesting for many reasons, but recently it has come to the attention of researchers because it acts as a kappa opioid receptor agonist (Roth, 2002). It is the first known compound acting on this receptor that is not an alkaloid. The κ-opioid receptor is a protein and is one of four related receptors that bind opium-like compounds in the brain and are responsible for mediating the effects of these compounds. These effects include altering the perception of pain, motor control, and mood. Wang et al. (2013) demonstrated that “administration of Salvinorin A after hypoxia/ischemia preserves cerebral autoregulation via the κ-opioid receptor pathway”, ultimately presenting a novel opportunity for the prevention of prenatal cerebral hypoxia/ischemia. Fichna et al. (2012) investigated the effect of Salvinorin A (administered intraperitoneally once a day) on acute pain, oedema and formalin-induced persistent pain in mice. The researchers concluded that Salvinorin A exerts analgesic actions and also shows moderate anti-inflammatory effects.

More recently Salvinorin A was used to research the relationship between the claustrum (a sheet of neurons which is attached to the neocortex in the center of the brain) and consciousness. Stiefel et al. (2014) suggests that “the consciousness-altering effects of S. divinorum/salvinorin A are due to a κ-opioid receptor mediated inhibition of primarily the claustrum and, additionally, the deep layers of the cortex, mainly in prefrontal areas”. This finding leads them to believe that Salvinorin A might be new evidence in favour of the ‘Crick and Koch theory’ (1990) which claims that the claustrum is the “conductor of consciousness”, not unlike the philosopher René Descartes claimed for the pineal gland.

Dr. Karl Hanes, researcher at the ‘Cognitive-Behavioural Treatment Centre’ in Melbourne Australia, reported on a treatment-resistant depressed patient (Ms. G) under his care and the management of her symptoms of depression through Salvia (2001). “During a review consultation some seven months after discontinuing cognitive-behavioural therapy”, says Hanes, “Ms. G claimed to have found relief from her symptoms of depression with use of the herb Salvia divinorum. A HAM-D score of 2 confirmed remission of her symptoms of depression at this time. Ms. G claims that she discovered its antidepressant effects accidentally after smoking the herb and had later developed a method of oral consumption which she claimed maintained its antidepressant effects even after she abstained from using it for up to a week”. Preliminary research (animal models) asserts Ms. G’s case and shows that Salvinorin A might become a therapeutic agent in the treatment of depression.

Carlezon et al. (2006) found that Salvinorin A “decreased extracellular concentrations of dopamine within the nucleus accumbens, a critical component of brain reward circuitry, without affecting extracellular concentrations of serotonin (5-HT)”. Butelman et al. (2004) showed that “the naturally occurring hallucinogen Salvinorin A produces discriminative stimulus effects similar to those of a high efficacy κ-agonist in non-human primates” (rhesus monkeys). Much research is done in the field of addiction and focuses primarily on Salvinorin A’s supposed anti-addictive effects. Activating the kappa-opioid receptors (KORs or KOPrs) produces anti-addictive effects by regulating dopamine levels in the brain (Kivel et al., 2014). The κ-opioid receptor may provide a natural addiction control mechanism, and therefore, drugs that act as agonists and increase activation of this receptor may have therapeutic potential in the treatment of addiction. “Activation of KORs”, says Tomasiewicz (2008), “reduces the reward-related effects of cocaine. Inasmuch as cocaine-induced behavioral stimulation in rodents may model key aspects of enhanced mood in humans, these findings raise the possibility that KOR agonists might ameliorate symptoms of conditions characterized by increased motivation and hyperfunction of brain reward systems, such as mania and stimulant intoxication”. These are obviously very exciting effects. “The development of KOPr-selective agents with improved drug-like characteristics” says Carlezon (2009), “would facilitate preclinical and clinical studies designed to evaluate the possibility that KOPrs are a feasible target for new medications”.

“Unfortunately”, says Kivel (2014), “classic kappa-opioid agonists have undesired side effects such as sedation, aversion, and depression, which restrict their clinical use”. Acute activation of kappa opioid receptors produces ‘anti-cocaine’ like effects, but because of considerate adverse effects their clinical use has remained limited (Simonson, 2014). Salvinorin seems to sidestep this problem. “Salvinorin A and its semi-synthetic analogs”, says Simonson (2014), “have been shown to have potent KOPr agonist activity and may induce a unique response with similar anti-cocaine addiction effects as the classic KOPr agonists but with a different side effect profile”. Salvinorin A retains the anti-addictive properties of traditional kappa-opioid receptor agonists with remarkable reduction of side effects.

However, there are still many obstacles to be overcome. For instance, a major problem is Salvinorin A’s rapid metabolism in the human body. Because of the short length of activation, Salvinorin as such will probably not be a good candidate for clinical development. In an effort to re-engineer Salvinorin’s pharmacokinetics, researchers have sought to produce kappa-opioid receptor agonists based on the structure of Salvinorin A that do have the desired length of activity. “While work in this field is still in progress”, says Kivel (2014), “several analogs with improved pharmacokinetic profiles have been shown to have anti-addictive effects”. These new analogs will eventually provide the characteristics sought after by clinical researchers for treatment.

Given the reemergence of interest into psychedelics and their potential as therapeutic agents it is hardly surprising that Salvia divinorum would attract attention. Salvia manifests a powerful psychedelic experience which is grounded in a unique neurochemical process which seems to have anti-addictive potential. It is surely a substance worth exploring within the psychiatric and psychopharmacological disciplines in the future.


 
References
Butelman, E. R., Harris, T. J., & Kreek, M. J. (2004). The plant-derived hallucinogen, salvinorin A, produces κ-opioid agonist-like discriminative effects in rhesus monkeys. Psychopharmacology, 172(2), 220-224.
Carlezon, W. A., Béguin, C., DiNieri, J. A., Baumann, M. H., Richards, M. R., Todtenkopf, M. S., … & Cohen, B. M. (2006). Depressive-like effects of the κ-opioid receptor agonist salvinorin A on behavior and neurochemistry in rats. Journal of Pharmacology and Experimental Therapeutics, 316(1), 440-447.
Carlezon, W. A., Béguin, C., Knoll, A. T., & Cohen, B. M. (2009). Kappa-opioid ligands in the study and treatment of mood disorders. Pharmacology & therapeutics, 123(3), 334-343.
Crick, F. & Koch, C. (1990) Towards a neurobiological theory of consciousness. Seminars in the Neurosciences, 2, 263-275.
Fichna, J., Dicay, M., Lewellyn, K., Janecka, A., Zjawiony, J. K., MacNaughton, W. K., & Storr, M. A. (2012). Salvinorin A has antiinflammatory and antinociceptive effects in experimental models of colitis in mice mediated by KOR and CB1 receptors. Inflammatory bowel diseases, 18(6), 1137-1145.
Hanes, K. R. (2001). Antidepressant effects of the herb Salvia divinorum. Journal of Clinical Psychopharmacology, 21, 634-635.
Johnson, J. B. (1939). The Elements of Mazatec Witchcraft. Ethnological Studies, 9.
Kivell, B. M., Ewald, A. W., & Prisinzano, T. E. (2013). Salvinorin a analogs and other kappa-opioid receptor compounds as treatments for cocaine abuse. Advances in pharmacology (San Diego, Calif.), 69, 481-511.
Roth, B. L., Baner, K., Westkaemper, R., Siebert, D., Rice, K. C., Steinberg, S., … & Rothman, R. B. (2002). Salvinorin A: a potent naturally occurring nonnitrogenous κ opioid selective agonist. Proceedings of the National Academy of Sciences, 99(18), 11934-11939.
Siebert, D. J. (1994). Salvia divinorum and salvinorin A: new pharmacologic findings. Journal of ethnopharmacology, 43(1), 53-56.
Simonson, B., Morani, A. S., Ewald, A. W. M., Walker, L., Kumar, N., Simpson, D., … & Kivell, B. M. (2014). Pharmacology and anti‐addiction effects of the novel kappa opioid receptor agonist Mesyl Sal B, a potent and long‐acting analogue of salvinorin A. British journal of pharmacology.
Stiefel K, M., Merrifield A., Holcombe A. (2014). The claustrum’s proposed role in consciousness is supported by the effect and target localization of Salvia Divinorum. Frontiers in Integrative Neuroscience. 8(20). doi:10.3389/fnint.2014.00020
Tomasiewicz, H. C., Todtenkopf, M. S., Chartoff, E. H., Cohen, B. M., & Carlezon Jr, W. A. (2008). The kappa-opioid agonist U69, 593 blocks cocaine-induced enhancement of brain stimulation reward. Biological psychiatry, 64(11), 982-988.
Valdés, L. J. (1994). Salvia divinorum and the unique diterpene hallucinogen, Salvinorin (divinorin) A. Journal of psychoactive drugs, 26(3), 277-283.
Wang Z., Ma N., Riley J., Armstead WM., Liu R (2013). Autoregulation Preservation by Salvinorin A After Brain Hypoxia/Ischemia and Reperfusion in Piglet Model. Trans Periop & Pain Med. doi:10.4000/2330-4871.1000001
Wasson, R. G. (1962). A new Mexican psychotropic drug from the mint family. Botanical Museum Leaflets, Harvard University, 77-84.

Autism and LSD-25 – Freeing the Most Imprisoned Minds?

In the early sixties, a number of controversial clinical investigations were published involving the administration of LSD-25 (lysergic acid diethylamide) to young children said to suffer from severe forms of autism, or childhood-onset schizophrenia (COS), which were then regarded as closely related [1]. The reason for conducting the studies with young children was the supposed similarity between autism and COS. Prompted by the apparent results of studies conducted with LSD-25 and adult mute catatonic patients by Cholden, Kurland, and Savage (1955), hypotheses were constructed to research a possible therapeutic utility. “The goal in these therapeutic efforts”, said Bender in an article published in Recent Advances in Biological Psychiatry (1962), “has been to modify the secondary symptomatology associated with retarded, regressed, and disturbed behavior of the children”. The larger part of the children treated with LSD in these studies were between six and ten years old and completely unresponsive to all other forms of treatment. That the children couldn’t be treated by other means served, in part, for the justification for using a powerful psychoactive substance in child experiments. Surely this decision would have been criticized by the ethical commission today.

A pharmacological intervention by means of LSD was said to “nudge the lagging maturation” (Bender, 1962) into a (somewhat) normal developmental pattern. How exactly the administration of LSD would accomplish the “freeing of the most imprisoned minds” was still unknown (Mogar & Aldrich, 1969). LSD was supposed to achieve success through “breaking through the autistic defense” (Bender, 1963), and in this way be exceptionally helpful in “areas which are closely related to the process of psychotherapy” (Simmons et al., 1966). Some believed LSD was especially useful at helping patients to “unblock” repressed subconscious material through other psychotherapeutic methods (Cohen, 1959). Therapists took LSD to establish a connection with the experience of schizophrenia. “During the ‘model psychosis’ phase of LSD research when the psychedelic state was considered a chemically-induced schizophrenia”, says pioneer LSD researcher Stanislav Grof (1980), “LSD sessions were recommended as reversible journeys into the experiential world of psychotics which had a unique didactic significance”.

Some researchers, like Freedman et al. (1963), studied LSD for its supposed psychotomimetic (psychotogenic) properties, meaning that the drug mimics the symptoms of psychosis, including delusions and/or delirium, as opposed to merely hallucinations (Sewell et al., 2009). An exacerbation of ‘typical’ symptoms meant an opportunity for studying the (child)schizophrenic condition. Other researchers (Bender et al., 1963; Rolo, et al., 1965) considered the neurological mechanism behind the effect of LSD, which in that time was still highly obscure, as more important than its role as facilitator of the therapeutic process. For instance, LSD attracted theoretical interest as a serotonin inhibitor and an autonomic nervous system stimulant. Bender et al. (1963) concluded that “LSD-25 given daily in oral doses of 100 mcg [2] to pre-puberty autistic schizophrenic children appears to be an effective autonomic and central nervous system stimulant”, and that these changes “appear to be chronic with continuous administration of the drug”. Continuous administration consisted of daily administration over prolonged periods of time, varying from days to several weeks . The most persistent effects of LSD-25 therapy that were published included improved speech behavior, increased emotional responsiveness, positive mood (laughter) and a decrease of compulsions.

But alas, however interesting and attractive these results seemed to be — the evidence didn’t stick. Today studies into the relationship of LSD and autism aren’t being conducted and the results that were produced are regarded as highly controversial, if not completely repudiated. This was in part because, in retrospect, the studies were greatly flawed. The researchers seemed to have brushed over the conceptual controversy too quickly by choosing “not to deal with the controversial issues concerning the definitions and etiological factors of either childhood schizophrenia (1) or the autistic reaction pattern (2)” (Bender et al., 1962). The debate about the correct place of (childhood) autism within the DSM (Diagnostic and Statistical Manual of Mental Disorders) remains problematic to this day (DSM-V), but autism has long been divorced from the umbrella of schizophrenia. Although both disorders share clinical features, clinical psychologists and psychiatrists regard autism to be a separate diagnostic ‘entity’ from schizophrenia. Because LSD was used as a drug for “intensifying pre-existing symptomology” of schizophrenia (Bender et al., 1962), a conceptual detachment from autism would have disturbed the foundation of the results.

Even if the researchers had chosen to ‘deal with the controversy’, in hindsight, sampling would have still ended up being very problematic. The children treated were demographically varied and covered a broad age range. Conflicting significance is given to the relationship between age and drug response, but Bender noted that “in contrast to pre- adolescents, younger children manifest consistently different reactions” (1962). In contrast, Fisher and Castile concluded that “older children were better candidates for psychedelic therapy because verbal communication was possible and also because they tended to be less withdrawn, more schizophrenic than autistic, and displayed more blatant symptomology” (Mogar & Aldrich, 1969). In addition to age, also the symptoms of treated children were heterogeneous and weren’t corrected for severity. There was no randomization, and most studies suffered from fluctuating frequency of administration and dosage. Lastly, the set and setting of the experiments varied strongly.

Although the studies conducted in the sixties had major flaws from an experimental point of view and therefore didn’t hold up to scientific scrutiny, Mogar and Aldrich argue in an article published in Behavioral Neuropsychiatry (1969) that the results considered as a whole do point to a utility of administering LSD to autistic children. “The significance of seemingly contradictory results”, say Mogar and Aldrich, “has often been obscured by the persistent search for static, ‘drug-specific’ reactions to LSD”. This is an interesting point; despite that the results don’t indicate significance in an experimental sense, there may still be a therapeutic utility. Mogar and Aldrich report that the greatest therapeutic benefit was related to “(a) the degree of active therapist involvement with the patient; (b) an opportunity to experience meaningful objects and interpersonal activities; and (c) congenial settings that were reasonably free of artificiality, experimental or medical restrictions, and mechanically administered procedures” (1969). In practice clinical therapy is usually far removed from theory. It could be that testing LSD, itself being a highly unpredictable drug, in combination with the therapy dynamic is too hard to substantiate. Mogar and Aldrich conclude that “the administration of LSD is inextricably embedded in a larger psychosocial process which should be optimized in accordance with particular treatment goals”.

Considering the recent growth of interest into this area of research, these older and rather obscure studies deserve to be excavated from the psychedelic research literature. Researchers at LA BioMed (Los Angeles Biomedical Research Institute) are now constructing a study which is said to test the already established anecdotal therapeutic relationship between MDMA (3,4-methylenedioxy-N-methylamphetamine) and autism in adults. The study is the latest in an expanding program of research into the therapeutic use of MDMA funded by the nonprofit Multidisciplinary Association for Psychedelic Studies (MAPS). “This new study will give us a chance”, says Charles Grob head researcher at LA BioMed (2014), “to determine the actual effects of differing dosages of medication that we know for certain is pure MDMA on adults on the autism spectrum. If the results of this research warrant further investigation, data from this study will be used to design additional clinical trials”. Now that the limitations for research into the psychedelic experience and its therapeutic effects are being removed and LSD is once again an object of study, these previously published results could serve for the production of new hypotheses.


 
[1] See (Abramson, 1960; Bender, et al., 1962; Bender, et al., 1963; Fisher & Castile, 1963; Freedman, et al., 1962; Rolo, et al., 1965; Simmons, et al., 1966).
[2] A common psychedelic dosage of LSD ranges from 100 to 200 mcg, a strong dose being 200 to 600 mcg.
 
References
Abramson, H.A. (Ed.) (1960). The Use of LSD in Psychotherapy. New York: Josiah Macy Foundation.
Bender, L., Faretra, G., & Cobrinik, L. (1963). LSD and UM-L treatment of hospitalized disturbed children. Recent Advances in Biological Psychiatry, 5, 84-92.
Bender, L., Goldschmidt, L., & Sankar, S.D.V. (1962). Treatment of autistic schizophrenic children with LSD-25 and UML-491. Recent Advances in Biological Psychiatry, 4, 170-177.
Cholden, L., Kurland, A., & Savage, C. (1955). Clinical reactions and tolerance to LSD in chronic schizophrenia. Journal of Nervous and Mental Disease, 122, 211-216.
Cohen, S., & Eisner, B. G. (1959). Use of lysergic acid diethylamide in a psychotherapeutic setting. AMA Archives of Neurology & Psychiatry, 81(5), 615-619.
Freedman, A.M., Ebin, E.V., & Wilson, E.A. (1962). Autistic schizophrenic children: An experiment in the use of d-lysergic acid diethylamide (LSD-25). Archives of General Psychiatry, 6, 203-213.
Gettys, T. (2014). MDMA Helps Reduce Social Anxiety for Autistic Adults, and Researchers Want to Find Out How. MAPS. Retrieved at: http://www.maps.org/media/view/mdma_helps_reduce_social_anxiety_for_autistic_adults_and_researchers_w/
Grof, S. (1980). LSD Psychotherapy. California: Hunter House Publishers.
Mogar, E. R., & Aldrich, W. R. (1969). The Use of Psychedelic Agents with Autistic Schizophrenic Children. Behavioral Neuropsychiatry, 1(8), 44-50.
Rolo, A., Krinsky. L.W., Abramson, H.A., & Goldfarb, L. (1965). Preliminary method for study of LSD with children. International Journal of Neuropsychiatry, 1, 552-555.
Sewell, R. A., Ranganathan, M., & D’Souza, D. C. (2009). Cannabinoids and psychosis. International Review of Psychiatry, 21(2), 152-162.
Simmons, J.Q., Leiken, SoJ., Lovaas, Q.I., Schaffer, B., & Perloff, B. (1966). Modification of autistic behavior with LSD-25. The American Journal of Psychiatry, 122, 1201-1211.

30 April - Q&A with Rick Strassman

X