• Psychedelic Medicine

Mental Health | +70 articles

Panorama_of_Kings_Parade_in_Cambridge%2C_UK%2C_at_St._Mary%27s.jpg


Psychedelics show promise in treating mental illness

by Tonia Moxley | Virginia Tech | 19 Nox 2021

A growing body of evidence suggests psychedelics including psilocybin and LSD show promise in providing lasting relief from symptoms for those suffering some mental health disorders. Researchers found DOI, a similar drug to LSD, reduced negative behavioral responses following fear triggers in mouse models of anxiety.

One in five U.S. adults will experience a mental illness in their lifetime, according to the National Alliance of Mental Health. But standard treatments can be slow to work and cause side effects.

To find better solutions, a Virginia Tech researcher has joined a renaissance of research on a long-banned class of drugs that could combat several forms of mental illness and, in mice, have achieved long-lasting results from just one dose.

Using a process his lab developed in 2015, Chang Lu, the Fred W. Bull Professor of Chemical Engineering in the College of Engineering, is helping his Virginia Commonwealth University collaborators study the epigenomic effects of psychedelics.

Their findings give insight into how psychedelic substances like psilocybin, mescaline, LSD, and similar drugs may relieve symptoms of addiction, anxiety, depression, and post-traumatic stress disorder. The drugs appear to work faster and last longer than current medications—all with fewer side effects.

The project hinged on Lu’s genomic analysis. His process allows researchers to use very small samples of tissue, down to hundreds to thousands of cells, and draw meaningful conclusions from them. Older processes require much larger sample sizes, so Lu’s approach enables the studies using just a small quantity of material from a specific region of a mouse brain.

And looking at the effects of psychedelics on brain tissues is especially important.

"Researchers can do human clinical trials with the substances, taking blood and urine samples and observing behaviors," Lu said. “But the thing is, the behavioral data will tell you the result, but it doesn’t tell you why it works in a certain way,” he said.

But looking at molecular changes in animal models, such as the brains of mice, allows scientists to peer into what Lu calls the black box of neuroscience to understand the biological processes at work. While the brains of mice are very different from human brains, Lu said there are enough similarities to make valid comparisons between the two.

VCU pharmacologist Javier González-Maeso has made a career of studying psychedelics, which had been banned after recreational use of the drugs was popularized in the 1960s. But in recent years, regulators have begun allowing research on the drugs to proceed.

In work by other researchers, primarily on psilocybin, a substance found in more than 200 species of fungi, González-Maeso said psychedelics have shown promise in alleviating major depression and anxiety disorders. “They induce profound effects in perception,” he said. “But I was interested in how these drugs actually induce behavioral effects in mice.”

To explore the genomic basis of those effects, he teamed up with Lu.
In the joint Virginia Tech—VCU study, González-Maeso’s team used 2,5-dimethoxy-4-iodoamphetamine, or DOI, a drug similar to LSD, administering it to mice that had been trained to fear certain triggers. Lu’s lab then analyzed brain samples for changes in the epigenome and the gene expression. They discovered that the epigenomic variations were generally more long-lasting than the changes in gene expression, thus more likely to link with the long-term effects of a psychedelic.

After one dose of DOI, the mice that had reacted to fear triggers no longer responded to them with anxious behaviors. Their brains also showed effects, even after the substance was no longer detectable in the tissues, Lu said. The findings were published in the October issue of Cell Reports.

It’s a hopeful development for those who suffer from mental illness and the people who love them. In fact, it wasn’t just the science that drew Lu to the project.

For him, it’s also personal.

“My older brother has had schizophrenia for the last 30 years, basically. So I’ve always been intrigued by mental health,” Lu said. “And then once I found that our approach can be applied to look at processes like that—that’s why I decided to do research in the field of brain neuroscience.”

González-Maeso said research on psychedelics is still in its early stages, and there’s much work to be done before treatments derived from them could be widely available.

https://neurosciencenews.com/psychedelics-doi-lsd-anxiety-19682/
 
Last edited:
IKBARS4DJ5FV7EIYZ5P5WONZDI.jpg

Marc Morgan has used LSD to help with his PTSD.

Can psychedelics treat depression and anxiety?*

by Bethany Ao | Philadelphia Enquirer

In recent months, the use of psychedelics for untreatable depression, anxiety, and PTSD has become a hot topic in mental health.

When Marc Morgan tried LSD for the first time as a teenager, he noticed that he could emotionally disconnect from traumatic memories of the sexual abuse he had suffered as a child.

“It allowed me to dissociate from the sharp pain that a lot of these memories can bring up, which caused me to shy away from processing them,” said Morgan, who has post-traumatic stress disorder (PTSD) as a result of the abuse. “I was able to confront the topic in a more analytical way that felt healthier.”

Years later, Morgan learned about microdosing ⁠— taking tiny portions of psychedelic substances, as little as a 20th of a recreational dose, to get positive effects, such as more focus and emotional balance, without negatives such as hallucinations and disorientation. He realized that taking small doses allowed him to process his emotions without the stronger, visual effects and looping thoughts.

“It’s basically like taking a cup of coffee,” said Morgan, now 30. He said that he takes a full dose once or twice a year, and microdoses about four times a year. “You’re just able to be a little more free and a little more honest with your emotions without breaking down. There’s more of a mental clarity.”

Morgan, who has lived in Philadelphia for a decade, is part of a group of people who use both full doses and microdoses of psychedelic substances to process trauma, and better deal with depression. For some, it’s because conventional antidepressants haven’t worked for them. Others choose psychedelics because it’s a more affordable option than therapy or medication.

However, medical experts don’t recommend self-medicating.

“For people interested in a treatment who can’t get into a trial, this is not the only thing out there,” said Matthew Johnson, an associate professor of psychiatry and behavioral sciences at Johns Hopkins University who has studied psychedelics for more than 15 years. “It’s one promising thing, and it’s important for people to stay in treatment."

The brain on psychedelics

In recent months, the use of psychedelics for treatment-resistant depression, anxiety, and PTSD — meaning patients do not respond to medication or therapy — has become a hot topic in the mental-health field. The Food and Drug Administration approved a nasal spray for treatment-resistant depression (TRD) last March, which uses a derivative of the hallucinogenic ketamine called esketamine. It is the first new antidepressant in decades. (It’s currently offered at a handful of clinics in the Philadelphia area.)

In November, the FDA gave psilocybin, a hallucinogenic compound found in magic mushrooms, its second “breakthrough therapy” designation in just more than a year. The designation fast-tracks the development and review of drugs.

In a 2017 study of psilocybin and depression, researchers at Imperial College London gave psilocybin therapy to 20 patients with treatment-resistant depression, who reported benefits as long as five weeks after treatment. The study found that psilocybin decreased activity in the amygdala, which processes such emotions as fear and anxiety.

“We know that when someone is on a therapeutic dose of a psychedelic, there’s a dramatic increase in communication across brain areas,” Johnson said. “My theory is that what we’re seeing with psychedelic therapy is more like what we normally associate with talk therapy. Psychedelic therapy prompts a therapeutic process, and people learn something by transcending their sense of self and getting out of their own way.”

Much less is known about the science behind microdosing psychedelic substances, but a 2019 study by researchers at the University of California-Davis found that the practice can provide relief for symptoms of depression and anxiety in rats.

In addition, a 2019 surveyof more than 1,000 people from across the world who microdosed on LSD found that repeated microdoses were followed by “improvements in negative moods, especially depression,” as well as increased positive moods and energy levels.

“There really hasn’t been substantiation of the claimed benefits of so-called microdoses,” Johnson said. “That’s not to say the claimed benefits aren’t possible.”

"The benefits people feel from microdosing might be from a combination of a strong placebo effect and some possible benefit from tinkering with the serotonin system,”
Johnson said. But he’s open to the potential benefits of microdosing, particularly for depression.

As a result of their research on how psychedelics can be used to help smokers and cancer patients, Johnson and others at Johns Hopkins have suggested that psilocybin’s FDA classification should be changed from Schedule I, which means that it has no known medical benefit, to Schedule IV, similar to prescription drugs.

Victor Pablo Acero, a 24-year-old bioengineering doctoral candidate and executive director of the Penn Society for Psychedelic Science, said that accepting both full doses and microdoses as mental-health treatments is an important step in destigmatizing the substances.

“Having a mystical experience is correlated with actually getting more clinical effects,” Acero said. “Microdosing doesn’t recapitulate the mystical experience or ego death. Also, psychedelic use ... does not magically heal you; you have to put in work and effort to integrate your experience.”

Acero said that he became interested in psychedelics once he began reading research papers about how the substances can be used in clinical settings. But he said that research in this area is still lacking.

“Funding is nonexistent,” Acero said. “Scientists are having to argue that they need funding to study the medical purposes of a substance that’s classified as having no medical purposes.”

Most people suffering from treatment-resistant depression, anxiety, or PTSD won’t be able to access the new treatments for at least a few years, as research groups run clinical trials. The trials have a limited number of spaces ⁠— the Usona Institute, a nonprofit medical research group in Wisconsin, recruited just 80 participants for a study that’s part of a Phase 2 clinical trial for psilocybin. Similarly, LSD is being evaluated in a Phase 2 clinical trial as a treatment for depression in Switzerland. MDMA, better known as ecstasy or molly, is currently in a Phase 3 large-scale clinical trial for PTSD. Upon FDA approval, the SoundMind Center, based in West Philly, will open a clinic in Cedar Park offering MDMA-assisted psychotherapy to populations with higher rates of PTSD.

While patients wait, self-medication without the aid of a therapist or medical professional is much more common, although it is not recommended. Johnson said that there are risks to psychedelic therapy for people with existing psychotic disorders, such as schizophrenia.

“Even if this stuff is approved, it’s not going to work for everyone,” Johnson said.

Changing perceptions

Aysha Ali, who struggles with depression and anxiety, began using full doses and microdoses of magic mushrooms about two years ago. Ali, 21, said antidepressants caused such side effects as appetite loss, decreased libido, and nausea. She also found that mushrooms are more accessible and cheaper for her.

“It’s definitely easier to go to bed at night,” said Ali, who lives in Wilmington and works in King of Prussia. “I’m a little more focused, and I feel like I can smile and giggle in the moment. I can feel myself going through my day a little bit better, and the days after feel so much better.”

She said that it refreshes her brain and allows her to wake up with a positive attitude.

“It helps me see that I’m not going to be stuck where I am,” Ali said. "It’s not a cure for everything, but the scientific information we’re getting now is helping people get out of this mind-set of ‘This is bad.’ ”

Mike Allebach, a photographer who lives in Montgomery County, first learned about magic mushrooms for mental health through a New York Times article last year.

“It didn’t really quite match up with my understanding of mushrooms," said Allebach, 37.

Allebach, who was struggling with depression when he read the article, said that he later tried magic mushrooms in California, where psilocybin mushrooms are decriminalized in Oakland and Santa Cruz. The trip brought him out of his depression. Since coming home, Allebach uses breath work and meditation to maintain his mental health.

“As a society, we’re in a place where the new rich is feeling calm and having peace,” he said. "And if antidepressants don’t work for someone, this could be great for someone who wants to be happy again and work on themselves.”

*From the article here :
 
Last edited:
tingri-everest-base-camp-trek.jpg



Novel 5-HT2A Psychedelic Drug Discovery Program for treating Mental Disorders*

Psilocybin Alpha | 30 Nov 2021

— Hundreds of de novo patentable psychedelic-based compounds created through discovery program —

— Company plans to announce lead molecule in first quarter of 2022 —

Bright Minds Biosciences, a biotechnology company focused on developing novel drugs for the targeted treatment of neuropsychiatric disorders, epilepsy, and pain, today provided a scientific update on the advancement of its novel 5-HT2A psychedelic program for the treatment of mental diseases.

“Our 5-HT2A psychedelic drug discovery program is showing clear signs of success,” stated Dr. Alan Kozikowski, Ph.D., Chief Scientific Officer and Co-founder of Bright Minds. “To date, we have synthesized hundreds of de novo, novel compounds in our discovery program and are currently optimizing a select handful of the most encouraging molecules to take into late preclinical development. We are currently on track to announce our lead molecule in this program in the first quarter of 2022.”

To select compounds with the best pharmacological profile to advance in clinical trials, Bright Minds uses intelligent drug design, advanced molecular modeling, together with scientifically rigorous evaluation in preclinical models, including 5-HT2 receptor pharmacology, rodent head twitch assays, and ADME/PK studies. This data-driven decision process de-risks Bright Minds’ proprietary compounds and maximizes the chance of success in clinical trials.

“Relative to the first-generation psychedelic drug class such as psilocybin,” Dr. Kozikowski continued, “we are looking to engineer novel compounds that possess single-digit nanomolar potency at the 2A receptor, with comparable or significant selectivity over the 2C receptor, and de minimis activity at the 2B receptor, if any. Effectively, we aim to bring a more potent and safer investigational drug forward, with a significantly shorter ‘trip time,’ and that is exactly what we have found with these compounds. Importantly, all these molecules are New Chemical Entities, or NCEs, that will enjoy 20+ years of patent protection.”

Ian McDonald, Bright Minds’ Chief Executive Officer and Co-founder, stated, “Bright Minds has, what we believe to be, the largest and most advanced psychedelic drug discovery program in history. The resultant portfolio of compounds we have generated from these research efforts has a range of distinct profiles and broad applicability across a host of known diseases, including depression and PTSD. Through our scientific approach to psychedelic medications, we are committed to developing treatments for devastating illnesses and potentially bettering the lives of hundreds of millions of patients. The need has never been more urgent, and we see a significant commercial opportunity for Bright Minds.”

Bright Minds’ novel psychedelic compounds comprise a variety of different chemical scaffolds, and not just those comparable to psilocybin. This differentiated strategy enables a tailored approach to generating diverse pharmacological profiles and empowers Bright Minds to create customized solutions to a variety of medical diseases with high unmet need.

About Bright Minds
Bright Minds is focused on developing novel transformative treatments for neuropsychiatric disorders, epilepsy, and pain. Bright Minds has a portfolio of next-generation serotonin agonists designed to target neurocircuit abnormalities that are responsible for difficult to treat disorders such as resistant epilepsy, treatment resistant depression, PTSD, and pain. The Company leverages its world-class scientific and drug development expertise to bring forward the next generation of safe and efficacious drugs. Bright Minds’ drugs have been designed to potentially retain the powerful therapeutic aspects of psychedelic and other serotonergic compounds, while minimizing the side effects, thereby creating superior drugs to first-generation compounds, such as psilocybin.

*From the article here :
 
Last edited:
120421_psychedelics_feat-1440x700.jpg



Can psychedelics meet their potential for treating mental health disorders?*

by Laura Sanders | Science News | 3 Dec 2021

Kanu Caplash was lying on a futon in a medical center in Connecticut, wearing an eye mask and listening to music. But his mind was far away, tunneling down through layer upon layer of his experiences. As part of a study of MDMA, a psychedelic drug also known as molly or ecstasy, Caplash was on an inner journey to try to ease his symptoms of post-traumatic stress disorder.

On this particular trip, Caplash, now 22, returned to the locked bathroom door of his childhood home. As a kid, he used to lock himself in to escape the yelling adults outside. But now, he was both outside the locked door, knocking, and inside, as his younger, frightened self.

He started talking to his younger self. “I open the door, and my big version picks up my younger version of myself, and literally carries me out,” he says. “I carried myself out of there and drove away.”

That self-rescue brought Caplash peace. “I got out of there. I’m alive. It’s all right. I’m OK.” For years, Caplash had experienced flashbacks, nightmares and insomnia from childhood trauma. He thought constantly about killing himself, he says. His experiences while on MDMA changed his perspective. “I still have the memory, but that anger and pain is not there anymore.”

Caplash’s transcendent experiences, spurred by three therapy sessions on MDMA, happened in 2018 as part of a research project on PTSD. Along with a handful of other studies, that research suggests that when coupled with psychotherapy, mind-altering drugs bring some people immediate, powerful and durable relief.

120421_psychedelics_inline1.jpg

In 2018, Kanu Caplash participated in a clinical trial of MDMA as a treatment for PTSD. The experience brought him relief, he says. Today he is a college student who works as a mental health advocate.

Those studies, and the intense media coverage they received, have helped launch psychedelic medicine into the public conversation in the United States, England and elsewhere. Academic groups devoted to studying psychedelics have sprung up at Johns Hopkins, Yale, New York University Langone Health, the University of California, San Francisco and other research institutions. Private investors have ponied up millions of dollars for research on psychedelic drugs. The state of Oregon has started the process of legalizing therapeutic psilocybin, the key chemical in hallucinogenic mushrooms; lawmakers in other states and cities are considering the same move.

New ways to help people with PTSD, depression, anxiety and other mental health disorders are desperately needed. An estimated 30 percent of people with depression, for instance, don’t get relief from current treatments. Psychedelics, some researchers and clinicians believe, may help.

“The promise is incredible,” says Monnica Williams, a psychologist at the University of Ottawa, who ran the clinical trial Caplash participated in at UConn Health in Farmington. “Psychedelics have the potential to really completely revolutionize mental health and change everything.”

But a cloud of questions hovers over the research. It’s not known how the therapy works or who it might work for. Even if these new treatments perform well in clinical trials, the drugs, and the mind-bending experiences they bring, won’t appeal to everyone. What’s more, the drugs may not be available, or they may cost too much.

Psychedelic drugs, including MDMA, psilocybin and the hallucinogen LSD, which is also being studied as a treatment for depression and other mental health disorders, are illegal under federal law, classified as Schedule 1 substances by the U.S. government — with high potential for abuse and no currently accepted medical use. Many people may be reluctant to take an illegal drug that lowers their defenses and makes them vulnerable, no matter how great the promise of healing.

Social and legal hurdles, barriers to access and scientific questions make it unlikely that psychedelics will replace current mental health treatments, many experts agree. More likely is that with enough research, psychedelic substances will become another tool for doctors and therapists.

Caplash remembers what MDMA did for him right after his sessions. “I wasn’t as angry as I was before. My muscles were a lot less tense. I could literally see clearer,” Caplash says. “As I went through the study, I was also becoming a different person.”

The benefits are still with him. Caplash no longer thinks of suicide. A biology major at the University of Connecticut, he has big dreams and advocates for more accessible mental health care for others. “I feel like I’m at peace, to an extent,” he says. “I know who I am and what I want to do.”

A winding path

Psychedelic drugs are not new. Scientists at the pharmaceutical company Merck made MDMA in 1912. Swiss chemist Albert Hofmann synthesized LSD in 1938, and Aldous Huxley popularized his experience on mescaline, a psychedelic compound made by certain cacti, in his 1954 book The Doors of Perception. When people talk about the psychedelic renaissance, they often begin with Hofmann and Huxley, says Sutton King, who advocates to include Indigenous voices in discussions about psychedelics and is an Afro-Indigenous member of the Menominee and Oneida Nations of Wisconsin.

But the story of psychedelics starts long before then. Indigenous communities around the world have used psilocybin and other consciousness-changing compounds for healing for thousands of years.

“The traditional Indigenous Nations … have had these connections to these medicines,” says King, who is cofounder and president of the Urban Indigenous Collective, a nonprofit advocacy group in New York City.

Belinda Eriacho, a wisdom carrier of Dine’ (Navajo) and A'shiwi (Zuni) descent, believes that psychedelic drugs, called sacred plant medicines by some Indigenous groups, are catalysts to help align mental, physical, spiritual and emotional health. “We were the knowledge keepers,” she says. “A lot of our understanding about these medicines is through practical experiences. They are not something you can read in a book.”

But around the middle of the 20th century, medical researchers, dissatisfied with existing mental health treatments, began trying to quantify these drugs’ effects on mental states. A flurry of research yielded promising hints, but many of those early attempts didn’t yield solid data. Some experiments were poorly designed; worse, some were deeply unethical, forcing high doses of psychedelics on people who were incarcerated or experiencing psychosis. Many of those study subjects were people of color.

In the 1960s, social and political sentiments began turning against these drugs — and the counterculture they represented — in both non-Indigenous and Indigenous communities. The U.S. government criminalized the use of psychedelics to keep them out of the public’s hands. The new restrictions kept the drugs out of researchers’ hands, too.
Changing minds

Psilocybin can change brain activity, but it’s not clear if these shifts relate to mental health. In a study in healthy people, red and orange show where psilocybin boosted brain connectivity compared with a placebo. Those regions are important for handling information coming in from the senses. Blue shows where connections were weaker in people who took psilocybin.​

The brain on psilocybin

right and left side brain scans show areas where psilocybin boosted connectivity

K.H. Preller et al/Biological Psychiatry 2020

That social shift stigmatized the drugs and whatever promise they held, says neuroscientist Rachel Yehuda, who has studied PTSD for decades at the Icahn School of Medicine at Mount Sinai in New York City. Current drug treatments for PTSD, such as antidepressants or sleep medications, don’t work well for some people, she says. These medicines may help with symptoms, but don’t get at the root of the problem. Psychedelics might do more, she’s come to realize.

Two years ago, when Yehuda began studying psychedelic drugs, she faced a lot of skepticism. But those dismissals have disappeared. “The general attitude in academic medicine right now is, ‘Gosh, let’s try it. Let’s see. Maybe it will be good. Wouldn’t that be nice?’ ”

In some ways, psychedelics can outperform approved psychiatric drugs such as Prozac and other selective serotonin reuptake inhibitors, or SSRIs. And so far, the data suggest psychedelics work quickly, appear to be safe and have lasting effects, says Atheir Abbas, a psychiatrist and neuroscientist at Oregon Health & Science University in Portland. “That is hard to come by. I think that is extremely exciting.”

In the last five years, a handful of high-quality, albeit small, studies have suggested tremendous benefits from the psychedelic psilocybin for depression, anxiety and PTSD. The studies differ in their details, but many follow a similar arc. Generally, the studies begin with talk therapy sessions, followed by several therapy sessions in which participants are under the influence of a psychedelic drug. More psychotherapy comes afterward. At certain points in the process, researchers measure the participants’ symptoms.

Psilocybin-assisted therapy quickly reduced signs of depression among 24 participants with moderate or severe depression, scientists reported in 2020 in JAMA Psychiatry. Four weeks after two psilocybin sessions, 71 percent of the participants had maintained a drop of at least 50 percent in their scores on a depression rating scale called the GRID-HAMD.​

Depression drop

People who received two psilocybin sessions with psychotherapy had fewer symptoms of depression one week after their second dose, at week 5, and at week 8 than people who had not yet received the treatment. Error bars show the variation among individual responses.​

Effects of immediate versus delayed psilocybin therapy on depression

line graph showing that people who received immediate psilocybin treatment had lower depression scores than those who had not yet been treated

C. Chang
Source: A.K. Davis et al/JAMA Psychiatry 2020

Earlier work showed psilocybin could help with depression and anxiety in patients facing life-threatening cancer; the benefits were still there about four years after the psilocybin treatment, researchers reported in the Journal of Psychopharmacology in 2020.

MDMA therapy seemed to have similar effects in an international study of 90 people with PTSD. Two-thirds of those who got MDMA no longer qualified for a PTSD diagnosis at the end of the trial, compared with about one-third of participants who received placebos. Those results appeared May 10 in Nature Medicine.

Collectively, the studies offer strong hints that psilocybin, MDMA and other psychedelic drugs can help in these research settings, says Scott Thompson, a neuroscientist at the University of Maryland School of Medicine in Baltimore. “All of them are giving the same sort of signal,” he says.

"We’re in a moment when Western research is converging with the long history of Indigenous knowledge," King says. "There is a place for both approaches, “two truths,” as King puts it, when it comes to helping people with psychedelic drugs."

King adds that the excitement coming from laboratories is accompanied by fear for some Indigenous people: fear of their ceremonies being appropriated, fear of worsening access to their medicines and fear of these substances being misunderstood. “It’s an exciting time, but it’s also a scary time for Indigenous peoples,” she says.

The healing components

One big question is how the various psychedelic drugs work. “If you look at all the knobs people are turning, it’s really not known what’s critical and what’s not,” Abbas says. The talk therapy that often goes with the drugs, the psychedelic trip or other drug effects could all be important.

Some people suspect that the psychotherapy is the healing component, not the drug itself. And plenty of data show that therapy works well for some people. Many of the key trials so far have been testing the drug in tandem with intense psychotherapy. Caplash, for instance, had multiple therapy sessions before, during and after his MDMA experiences. Similarly intense therapy sessions happened for people in the trial of psilocybin for depression. The drugs might make a person open to exploring some of the painful events of their past during therapy.

Others suspect that the psychedelic trip itself, the hallucinatory experience, is a crucial part of the treatment. That’s a hard question to study, but not impossible, Yehuda says. “We need the science to understand what transformation looks like, and how a drug facilitates it,” she says. “We’ve been afraid of studying the biology of altered states, or even consciousness, because it feels so wonky and so unscientific and so subjective and woo woo. But I think we should be up for this challenge.”

Other scientists are focusing on the actions of the drugs themselves, by looking inside the brain. Thompson, for instance, suspects that it’s possible to get the benefits and skip the trip by restricting cells’ responses to serotonin, a chemical messenger that’s thought to be involved in the hallucinations. His recent study in mice hints that this just might be possible.

In the mice, Thompson and colleagues blocked a sensor thought to spark psilocybin-related hallucinations. When this serotonin-detecting receptor, called 5-HT2A, was blocked, the mice appeared to stop tripping (their heads no longer twitched). Yet, psilocybin still had antidepressive effects on the mice, restoring a lost preference for sugar water. The result raises the idea that the antidepressant effects associated with psychedelics can come without the hallucinations, the researchers reported in the April 27 Proceedings of the National Academy of Sciences.

"If a similar thing could happen in people, you could retain the benefits but block the trip,” Thompson says. “The trip is the thing that makes you have to spend all day in the hospital with a facilitator there, and makes it expensive, and keeps psychedelics from being widely used,” Thompson says. “Being able to block the trip would lower the barriers in many ways.”

Eriacho, who has used psychedelics for her own healing, says the holistic experience is what matters. Psychedelic medicines allow a person to go within, to the pockets of the mind where traumas lurk, and begin to heal. “That is what it’s done for me,” she says. But the ceremony, the ritual and the wider context were all keys to her healing.

120421_psychedelics_inline4.jpg

In a research setting, psychedelic drugs are paired with psychotherapy that
involves cycles of introspection and supportive talking.


Who will benefit?

The trials completed so far have been small and included mostly white volunteers. Williams and colleagues tallied participants in psychedelic studies from 2008 to 2017. Of the 282 people who took part (and for whom race or ethnicity data were available), just 50 were not white, the researchers wrote in 2018 in BMC Psychiatry.

People of color have been underrepresented in these studies for many reasons, says Williams, who designed her study at UConn Health to specifically look at the effects of MDMA-assisted therapy in people of color. Caplash, who is Indian-American, was the first and only person to go through the process at UConn. Soon after his participation, the trial was stopped at that site. Other enrolled volunteers there were unenrolled, says Williams, now at the University of Ottawa.

“This is a very marginalized and vulnerable group,” she says. “There were fears that if anything really bad happened [during the trial], it would reflect very badly on the university.” UConn Health spokesperson Lauren Woods says that is false. The clinical trial was stopped for a “variety of reasons,” she wrote in an e-mail to Science News, declining to provide specifics.

Without strong efforts to create inclusive studies that actively recruit a diverse group of people into the trials, train therapists of color and consider racial trauma, psychedelic drugs will not be accessible to everyone who might benefit from them, Williams says. “This needs to be an important part of what we do,” she says. “We can’t keep doing it the same way it was done in the past.”

Yehuda aims to design treatments for “the populations who need it the most,” she says. That includes combat veterans with PTSD. She and her colleagues are beginning to enroll 60 veterans in a clinical trial at the James J. Peters Veterans Affairs Medical Center in the Bronx, N.Y., where Yehuda directs the Mental Health Patient Care Center. “We are hoping to enroll a lot of ethnically diverse and racially diverse people, because we already serve them,” Yehuda says. “We’ve done a lot of the trust work already.”

As part of her studies, Yehuda plans to get at the question of who the drugs might work for, and why, in part by scrutinizing biological differences between people who get relief from the psychedelics and people who don’t. “We’re going to ask these somewhat tougher questions,” she says. “There is going to be a science about who MDMA is particularly good for.”

Meanwhile, the people making laws and policies are impatient for the science to yield answers. In 2020, Oregon voters approved Measure 109, which provides a sanctioned way to offer people psilocybin-assisted therapy. The state’s Psilocybin Advisory Board, which includes doctors, scientists and even a mushroom biologist, has two years to figure out how the state will regulate that effort.

Abbas is on that advisory board. Speaking personally and not on behalf of that board, he says designing a system for psilocybin-based therapy is immensely complex. “It’s not just who, but it’s how you identify those folks, it’s how you regulate the providers, how you regulate the psilocybin.”

Though studies so far have mainly focused on the performance of psychedelic drugs from a scientific perspective, other considerations are important, too. "Perspectives from Indigenous peoples will be sought out," Abbas says.

“There needs to be this meeting of the minds, and the openness that the scientific and Western way of thinking is not always the right way,” Eriacho says. Clinical trials, for instance, often rely on narrowly defined clinical surveys to capture symptoms. “Those are Western concepts,” she says, and from her perspective, those metrics miss a lot. “You don’t have a very comprehensive way of looking at what an individual may be experiencing,” she says.

What Oregon’s psilocybin program will ultimately look like is still anyone’s guess. The same is true for use of other psychedelics. There is no single story to be found amid all of these diverse and intersecting perspectives — Indigenous traditions, Western medicalization, social movements, eager private investors.

In the place of one story, we have many, including that of Kanu Caplash, who feels healed by an experience enabled by a psychedelic drug. His transformation was deeply personal. Yet it gives us a glimpse of something possible and powerful.

*From the article (including citations) here :
 
Last edited:
Panorama_of_Kings_Parade_in_Cambridge%2C_UK%2C_at_St._Mary%27s.jpg

Cambridge

Psychedelics and other Psychoplastogens for Treating Mental Illness

Maxemiliano V. Vargas(1), Retsina Meyer(2), Arabo A. Avanes(3), Mark Rus(2), David E. Olson(2,4,5,6)

1Neuroscience Graduate Program, University of California, Davis, Davis, CA, United States
2Delix Therapeutics, Inc., Concord, MA, United States
3Biochemistry, Molecular, Cellular, and Developmental Biology Graduate Program, University of California, Davis, Davis, CA, United States
4Department of Chemistry, University of California, Davis, Davis, CA, United States
5Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Sacramento, Sacramento, CA, United States
6Center for Neuroscience, University of California, Davis, Davis, CA, United States

Psychedelics have inspired new hope for treating brain disorders, as they seem to be unlike any treatments currently available. Not only do they produce sustained therapeutic effects following a single administration, they also appear to have broad therapeutic potential, demonstrating efficacy for treating depression, post-traumatic stress disorder (PTSD), anxiety disorders, substance abuse disorder, and alcohol use disorder, among others. Psychedelics belong to a more general class of compounds known as psychoplastogens, which robustly promote structural and functional neural plasticity in key circuits relevant to brain health. Here we discuss the importance of structural plasticity in the treatment of neuropsychiatric diseases, as well as the evidence demonstrating that psychedelics are among the most effective chemical modulators of neural plasticity studied to date. Furthermore, we provide a theoretical framework with the potential to explain why psychedelic compounds produce long-lasting therapeutic effects across a wide range of brain disorders. Despite their promise as broadly efficacious neurotherapeutics, there are several issues associated with psychedelic-based medicines that drastically limit their clinical scalability. We discuss these challenges and how they might be overcome through the development of non-hallucinogenic psychoplastogens. The clinical use of psychedelics and other psychoplastogenic compounds marks a paradigm shift in neuropsychiatry toward therapeutic approaches relying on the selective modulation of neural circuits with small molecule drugs. Psychoplastogen research brings us one step closer to actually curing mental illness by rectifying the underlying pathophysiology of disorders like depression, moving beyond simply treating disease symptoms. However, determining how to most effectively deploy psychoplastogenic medicines at scale will be an important consideration as the field moves forward.​

Introduction​

Theories regarding the etiology of depression and related neuropsychiatric diseases have evolved considerably in recent years. One of the oldest and most widely known theories posits that chemical imbalances in the brain are largely responsible for the development of neuropsychiatric diseases. Support for this theory originated with the observation in the 1950's that administration of the natural product reserpine induces depression. Reserpine depletes monoamine levels through inhibition of vesicular monoamine transporters, leading to subsequent degradation by monoamine oxidase (MAO). Support for the chemical imbalance hypothesis (also known as the monoamine hypothesis) was further bolstered by findings that MAO inhibitors, tricyclics, and selective-serotonin reuptake inhibitors (SSRIs)—compounds that elevate synaptic levels of monoamines—all seemed to alleviate depressive symptoms. However, several pieces of evidence have emerged suggesting that the chemical imbalance hypothesis of depression is a drastic oversimplification.

Reduction of monoamines through acute tryptophan or phenylalanine/tyrosine depletion does not induce depression in healthy subjects (5), which questions the causal role of “chemical imbalances” in depression. However, the primary issue with the monoamine hypothesis of depression lies in the temporal discrepancy between the acute effects of traditional antidepressants and their delayed therapeutic responses. Tricyclics, MAO inhibitors, and SSRIs all increase synaptic levels of monoamines in the brain within minutes, while it takes weeks before the antidepressant effects become apparent (Figure 1). In a patient population at heightened risk for suicide, the need for rapid-acting antidepressants is self-evident.

FIGURE 1
fpsyt-12-727117-g001.jpg

Figure 1. The antidepressant effects of psychoplastogens (e.g., ketamine and psilocybin) are more rapid and sustained than those of traditional antidepressants (e.g., fluoxetine). Data adapted from three clinical trials evaluating the effects of fluoxetine, ketamine, and psilocybin (8) for treating depression. Dashed lines represent the efficacy of either 1 or 8 weeks of fluoxetine treatment.

While traditional antidepressants acutely increase synaptic levels of monoamines, their chronic administration leads to changes in structural neuroplasticity that are contemporaneous with their clinical therapeutic effects. In fact, evidence suggests that chronic administration of traditional antidepressants can re-wire the brain, with chronic fluoxetine promoting ocular dominance plasticity in the visual cortex of adult rats. Such induced plasticity (iPlasticity) has been hypothesized to play a major role in the actions of essentially all antidepressant treatments including slow-acting traditional antidepressants, transcranial magnetic stimulation, electroconvulsive therapy, exercise, and acute sleep deprivation. Moreover, people (particularly males) with the brain-derived neurotrophic factor (BDNF) Val66Met single nucleotide polymorphism—a condition that reduces activity-dependent BDNF release—are more likely to experience chronic depression. These results lend substantial support to the neuroplasticity hypothesis of depression (sometimes referred to as the neurotrophin hypothesis). This hypothesis provides a strong conceptual framework for understanding mental illnesses as disorders of neural circuits induced by a combination of genetic and environmental factors (Figure 2). The corollary being that compounds capable of rectifying these circuit pathologies can potentially serve as a powerful, disease-modifying therapeutics.

FIGURE 2
fpsyt-12-727117-g002.jpg

Figure 2. Genetic and environmental factors lead to the cortical atrophy observed in depression, which includes retraction of dendritic branches and loss of dendritic spines. These structural changes are reversed by psychoplastogens.

Psychedelics—molecules with “mind-manifesting” properties—include pharmacologically diverse compounds such as dissociatives (e.g., ketamine), classic hallucinogens (e.g., LSD, psilocybin, DMT), and entactogens (e.g., MDMA). Several psychedelics have emerged as some of the most promising treatments for re-wiring pathological neural circuitry. Given their unusually robust abilities to produce rapid and long-lasting changes in neuronal structure and function following a single administration, these compounds have been classified as psychoplastogens—a term we coined to describe this new class of therapeutic compounds. Unlike traditional antidepressants, psychoplastogens produce both fast-acting and sustained beneficial behavioral effects after a single administration (Figure 1). Here, we present evidence that directly targeting cortical circuits with psychoplastogens has the potential to produce enduring therapeutic responses in depression and co-morbid diseases. First-generation psychoplastogens are all hallucinogenic—they cause people to perceive things that are not real—which has important implications for how these medicines must be administered and how many patients could ultimately benefit from these treatment approaches. In this regard, non-hallucinogenic psychoplastogens offer significant advantages, with the potential to reach much larger patient populations and even replace traditional antidepressants as first-line treatments.​

Harnessing Structural Plasticity to Treat Mental Illness​

Depression and related neuropsychiatric diseases are often viewed as stress-related disorders given the fact that they can be precipitated or exacerbated by chronic stress. In animals, chronic stress results in the prolonged release of glucocorticoids and leads to hypertrophy of the amygdala and nucleus accumbens, atrophy of the hippocampus and prefrontal cortex (PFC), and functional impairment of the PFC. Given the importance of the PFC in cognition and mediating top-down control over subcortical brain regions, these changes in neural circuitry are believed to underlie the deficits in learning/memory, mood, motivation, and reward seeking that are characteristic of depression and related disorders.

Postmortem studies have demonstrated that patients with depression and related mental illnesses have lower BDNF and/or TRKB mRNA levels, reduced cortical neuron size, lower synaptic protein levels, decreased mTOR signaling, and fewer dendritic spines/synapses in the PFC. Clinical imaging studies have confirmed the results of these studies, demonstrating robust structural and functional deficits in the PFC across a range of disorders including depression, bipolar disorder, anxiety, obsessive compulsive disorder (OCD), schizophrenia, PTSD, alcohol abuse disorder, and substance abuse disorder. More recently, the advent of [11C]UCB-J and [18F]UCB-J has opened up new opportunities for using positron emission tomography (PET) imaging to measure the density of the synaptic protein SV2A in vivo. Using these new tools, depression severity was found to inversely correlate with SV2A density, and this neuronal atrophy was associated with aberrant network function as measured by magnetic resonance imaging (MRI) functional connectivity. Similar results were observed in patients with schizophrenia. Thus, substantial evidence points to the PFC as the convergence point underlying the pathophysiology of many neuropsychiatric diseases.

In imaging studies of traditional antidepressant treatment response, increased cortical thickness and cerebral blood flow in the PFC correlate with efficacy. Next-generation, psychoplastogenic antidepressants also modulate PFC function with both ketamine and psychedelics increasing PFC activation as measured by 18F-FDG PET imaging. In fact, antidepressant outcomes following ketamine treatment correlate well with PFC activation. There is some evidence in humans that psychoplastogens can impact brain structure as well. Chronic use of the psychedelic tisane ayahuasca is associated with thickening of the anterior cingulate cortex, and ketamine treatment has been shown to rescue atrophy of the inferior frontal gyrus observed in MDD and PTSD patients.

Structural plasticity studies in preclinical animal models support the findings in humans suggesting that cortical neuron structure/function plays a key role in depression and related neuropsychiatric disorders. Cortical neuron atrophy and dysfunction is observed in rodents following chronic corticosterone administration, chronic unpredictable mild stress, chronic restraint stress, and chronic social defeat stress. These structural changes are accompanied by depressive phenotypes related to motivation, anxiety, and anhedonia. Moreover, antidepressants appear to rectify these structural changes by promoting structural plasticity in the PFC.

Nature uses BDNF to induce structural plasticity in many neuronal populations, and direct administration of BDNF into the rodent brain has been shown to alleviate several depressive phenotypes, curb addiction, and enhance fear extinction. Conversely, disruption of BDNF signaling in the brain can block the behavioral effects of antidepressants. BDNF heterozygous mice are resistant to the effects of traditional antidepressants and the psychoplastogen ketamine does not produce antidepressant-like effects in BDNF inducible knockout animals. The BDNF Val66Met single-nucleotide polymorphism causes major structural atrophy and functional deficits in the PFC and blocks the synaptogenic effects of ketamine. Both rodents and humans with BDNF Val66Met polymorphisms exhibit impaired fear extinction learning and reduced mPFC activity during extinction, phenotypes that are common in stress-related disorders.

All antidepressant treatments impact BDNF/TrkB signaling in some way. Chronic, but not acute, administration of several traditional antidepressants has been shown to significantly increase mRNA levels of BDNF and/or TrkB as well as increase levels of cAMP response element binding protein (CREB), a transcription factor that regulates the expression of several proteins important for plasticity including BDNF. Other studies have found that antidepressants from a variety of chemical families (e.g., SSRIs, SNRIs, tricyclics, etc.) all increase activation of TrkB and its downstream effector CREB. Moreover, the effects of the traditional antidepressant fluoxetine on synaptic plasticity and fear extinction have been shown to be dependent on BDNF. Recent evidence suggests that several antidepressants, including traditional antidepressants, may directly interact with the TrkB receptor to facilitate TrkB signaling. Taken together, the importance of BDNF/TrkB signaling in the therapeutic effects of traditional antidepressants is clear, even if these agents must be administered chronically to achieve robust regulation of this pathway.

Psychoplastogens are also known to impact BDNF/TrkB signaling, but in contrast to traditional antidepressants, they do so rapidly after a single administration. Ketamine increases BDNF protein translation and its antidepressant effects are absent when administered to inducible BDNF knockout mice or homozygous mice harboring the BDNF Val66Met mutation. Ketamine and psychedelics modulate cortical neuron function by increasing dendritic spine and synapse density in the PFC; however, ketamine's effects on structural plasticity appear to last for approximately a week while psilocybin's effects seem to be more durable lasting for at least a month. Though the primary molecular targets of ketamine and serotonergic psychedelics are distinct, their downstream pharmacology overlaps, requiring AMPA receptor, TrkB, and mTOR activation to elicit changes in neuronal structure and function. Moreover, their effects seem to be Cmax driven, as very short stimulation periods (15 min−1 h) are sufficient to induce sustained changes in cortical neuron structure. Psilocybin has also been shown to increase the density of SV2A in vivo as measured by PET imaging. Importantly, Liston et al. recently used a photoactivatable Rac1 to demonstrate that ketamine-induced spine growth in the PFC was causally related to long-lasting antidepressant effects of the drug in rodents. In humans, the subjective effects of ketamine wane after a few hours, but the antidepressant response continues to increase over several days. This time course is consistent with what we know about how ketamine and other psychoplastogens alter neuronal structure over time. An exceedingly short stimulation period (<1 h) is sufficient for psychoplastogens to activate cortical neuron growth mechanisms that can last for several days.

Several volatile anesthetics, such as isoflurane, nitrous oxide, propofol, and xenon, may produce rapid antidepressant effects. Xenon is not a small molecule, and by definition, is not a psychoplastogen. Other volatile anesthetics might be considered psychoplastogens if additional studies in larger patient populations confirm that they produce sustained therapeutic effects after a single administration. The data for isoflurane are encouraging as isoflurane produces rapid antidepressant effects after a single administration in both preclinical and in a subset of patients suffering from treatment-resistant depression. Moreover, these effects appear to be mediated via TrkB signaling and subsequent increases in dendritic spine density in the PFC and hippocampus. Thus, preliminary evidence suggests that isoflurane may be considered a psychoplastogen.​

Possible Explanations for the Broad Therapeutic Potential of Psychoplastogens​

While the beneficial effects of psychoplastogens can last for months following a single administration, and these medicines have demonstrated efficacy across a range of neuropsychiatric disorders including depression, PTSD, and addiction, they are not panaceas. Their broad therapeutic utility likely arises from their ability to impact the structure/function of layer V pyramidal neurons in the PFC. As the PFC is a key hub impacted in most neuropsychiatric disorders, it is not surprising that psychoplastogens have proven useful for a variety of indications. Indeed, with the advent of opto- and chemogenetics, systems neurobiology has developed a much deeper understanding of what circuits control behavior. The PFC is known to exert top-down control over a variety of subcortical regions, and recent research has identified a number of circuits originating in the PFC that control behaviors relevant to the treatment of depression, anxiety, and addiction (Figure 3).

FIGURE 3
fpsyt-12-727117-g003.jpg

Figure 3. Opto- and chemogenetic experiments have revealed a number of circuits originating in the PFC that are relevant to the treatment of neuropsychiatric disorders. Arrows indicated excitatory projections.

In mice subjected to social defeat stress, optogenetic stimulation of the ventral medial PFC increased social interaction and reduced anhedonia as measured via the sucrose preference test. Deisseroth et al. later found that optogenetic stimulation of medial PFC neurons projecting to the dorsal raphe nucleus (DRN) decreased immobility in the forced swim test. The forced swim test is a preclinical assay for antidepressant potential with high predictive validity, and activation of this PFC → DRN circuit likely mediates the robust effects of psychoplastogens on forced swim test behavior. While the National Institutes of Mental Health (NIMH) has stressed that the FST is not a model of depression, it is a powerful behavioral readout for activation of the PFC → DRN circuit. The DRN is a serotonergic nucleus that has been implicated in major depressive disorder, and recent evidence suggests that acute activation of serotoninergic neurons in the DRN increase active coping to inescapable stress. Dzirasa et al. demonstrated that optical stimulation of layer V pyramidal neurons in the PFC expressing channelrhodopsin-2 was sufficient to produce an antidepressant-like response in the forced swim test and suppress anxiety-like behavior in the elevated plus maze for over 10 days after the last optical stimulation session. This type of long-lasting anxiolytic effect is reminiscent of sustained effects observed following a single administration of a psychoplastogen. Finally, Duman et al. found that optogenetic stimulation of the infralimbic cortex produces rapid and sustained antidepressant-like effects comparable to ketamine in the forced swim, novelty-suppressed feeding, and sucrose preference tests. Moreover, microinfusion of ketamine into the infralimbic cortex produces comparable antidepressant-effects as systemic administration of ketamine and inactivation of the infralimbic cortex with muscimol was sufficient to block the antidepressant-like effects of systemic ketamine.

In addition to relieving symptoms associated with depression, psychoplastogens have also demonstrated efficacy for treating PTSD—a disorder that can involve dysfunction of the amygdala (AMY). The medial PFC and amygdala are well-connected, and bi-directional communication between these structures is likely involved in modulating responses to emotional stimuli. In fact, optogenetic stimulation of ventral medial PFC projections to the basomedial amygdala decreases fear responses and facilitates fear extinction learning. Chemogenetic inhibition of PFC neurons projecting to the amygdala is sufficient to impair fear extinction learning. As PTSD has often been described as a disorder of impaired fear extinction, the therapeutic effects of psychoplastogens might result from their ability to strengthen PFC → AMY circuits mediating top-down control of fear responses.

Like fear extinction, drug-cue extinction is believed to involve neurons in the PFC. This is perhaps unsurprising given the large body of neuroimaging data suggesting that PFC hypofunction is a hallmark of addiction. Chemogenetic activation of ventral medial PFC neurons projecting to the nucleus accumbens (NAc) shell was able to reduce cue-induced reinstatement of drug-seeking behavior. Moreover, chronic cocaine self-administration has been shown to decrease the intrinsic excitability of pyramidal neurons in the PFC. Optogenetic stimulation of these neurons prevented compulsive drug-seeking while silencing these neurons promoted drug-seeking behavior despite being paired with aversive foot shocks. Similarly, pharmacological activation and inactivation of neurons in the infralimbic cortex suppressed and enhanced reinstatement of drug-seeking behavior, respectively. Optogenetic experiments have also revealed that ventral medial PFC projections to the NAc shell are involved in suppressing ethanol self-administration in the presence of aversive stimuli. A PFC → NAc circuit appears to be involved in compulsive food-seeking behavior as well, given that chemogenetic inhibition of this circuit led to compulsive food seeking even in the presence of aversive foot shocks. While PFC → NAc circuits have been well-established in controlling drug-seeking behavior, more recently, Tye et al. demonstrated that a PFC projection to the dorsal periaqueductal gray (dPAG) may also play an important role in addiction. Specifically, they showed that optogenetic activation of a PFC → dPAG circuit prevented compulsive alcohol consumption.

Psychoplastogens produce robust, fast-acting, and long-lasting effects on structural plasticity in the PFC. This may explain why they have demonstrated efficacy in many preclinical rodent behavioral tests involving PFC circuitry including the forced swim test and fear extinction learning. However, achieving circuit-level selectivity is a key challenge in the design of optimized psychoplastogens with minimal to no side effects. The issue with non-selective activation of BDNF/TrkB signaling is apparent from chronic stress studies demonstrating that enhanced BDNF/TrkB signaling in the amygdala leads to maladaptive plasticity resulting in overactivation of this brain region and exacerbated anxiety and fear responses. Furthermore, compounds that promote plasticity in the mesolimbic pathway could have pro-depressive and/or addictive properties.

Psychoplastogens that target the 5-HT2A receptor have advantages over NMDA receptor antagonists like ketamine, as 5-HT2A receptors exhibit a relatively selective expression profile. With the exception of the claustrum, the highest density of 5-HT2A receptors is in layer V pyramidal neurons of the PFC, which are precisely the neurons that are most impacted in stress-related neuropsychiatric diseases. In rodents, this expression pattern has been confirmed using immunohistochemistry, light and electron microscope immunocytochemistry, in situ hybridization receptor autoradiography, and transgenic mice expressing EGFP under control of the 5-HT2A receptor promoter. A similar pattern of 5-HT2A receptor expression has been shown in human post-mortem tissue using both autoradiography and in situ hybridization. Additionally, PET imaging has revealed a high density of 5-HT2 receptors in the frontal and temporal cortices of the human brain. The high genetic localization of 5-HT2A receptors to excitatory neurons in layer V of the PFC is perhaps why animals do not typically self-administer classic serotonergic psychedelics and most psychedelics are not considered to be addictive.​

Can the Intoxicating Effects of Psychedelics Be Removed to Create More Scalable Therapeutics?​

At high doses, psychedelics reliably induce both hallucinations and mystical-type experiences. Currently, it is unclear if the mystical-type experiences they induce are necessary for their therapeutic effects in humans. Moreover, it is unclear if the intoxicating effects of psychedelics can be decoupled from their therapeutic properties. This critical question has profound implications for healthcare, as hallucinogenic treatments will inevitably be more limited in scope given safety and cost considerations. Many patients describe psychedelic-induced “peak” or “mystical” experiences as being among the most meaningful events of their lives, and the intensity of these events correlates with therapeutic responses. While these events could provide patients with valuable insight relevant to their disease symptoms, it is important to remember that correlation does not imply causation, and mystical-type experiences could simply be an epiphenomenon associated with 5-HT2A receptor activation. Activation of 5-HT2A receptors also promotes structural and functional neuroplasticity, which could be the primary driver of the sustained behavioral effects following a single administration of ketamine or serotonergic psychedelics.

Given that 5-HT2A receptor activation is associated with both psychoplastogenic effects and mystical-type experiences, it is challenging to determine exactly how much each contributes to the therapeutic properties of psychedelics. However, a number of key pieces of evidence suggest that intoxicating subjective effects are not necessary to achieve some level of therapeutic efficacy. In patients treated with ketamine, floating sensations did not correlate with PFC activation as measured by 18F-PET, demonstrating that it is possible to activate cortical circuits to produce antidepressant responses without inducing dissociative effects. In fact, several studies have demonstrated that intraoperative ketamine administration can improve postoperative mood even though the patients were unconscious at the time of administration. This strongly suggests that the dissociative experience itself is not necessary for ketamine to produce antidepressant effects.

Additionally, there is preclinical evidence suggesting that R-ketamine is a potent psychoplastogen that induces longer-lasting antidepressant-like effects than the S-enantiomer (i.e., Spravato®) despite having a lower affinity for the NMDA receptor and producing fewer dissociative effects. Similarly, a metabolite of R-ketamine lacking dissociative properties has also demonstrated robust antidepressant-like effects in rodents. Future clinical trials using these non-hallucinogenic agents will prove informative when assessing the role of subjective effects in ketamine treatment response.

While R-ketamine is still in the early stages of clinical development, MDMA has demonstrated robust results in a recently disclosed Phase III trial. As an atypical psychedelic of the entactogen family, MDMA produces robust effects on cortical neuron growth, and facilitates fear extinction learning through a 5-HT2-dependent mechanism without inducing psilocybin- or ketamine-like perceptual effects or dissociation. In fact, only about 20% of recreational MDMA users report experiencing any visual hallucinations, and these are relatively mild compared to those induced by psilocybin and LSD. While MDMA does induce a “blissful state,” likely due to its effects on monoamine efflux, it does not produce mystical-type experiences as measured using a variety of scales related to altered states of consciousness. This is in stark contrast to drugs like ketamine and psilocybin.

As an entactogen, MDMA is pharmacologically distinct from classic serotonergic psychedelics like psilocybin and LSD. However, there is clinical evidence suggesting that non-hallucinogenic analogs of classic psychedelics can also produce therapeutic effects. Lisuride, a non-hallucinogenic structural analog of LSD, has been shown to have an antidepressant properties in the clinic. Moreover, lisuride has demonstrated some efficacy in preclinical models as well. Lisuride has a polypharmacology profile that includes activation of D2 and 5-HT1A receptors in addition to 5-HT2 receptors, and thus, it is currently unclear what receptor(s) mediate its antidepressant effects.

In addition to the clinical evidence supporting the efficacy of non-hallucinogenic psychoplastogens, mounting preclinical data suggest that the beneficial effects of psychoplastogens can be dissociated from their hallucinogenic effects. In 2019, we demonstrated that low doses of DMT produce beneficial effects in rodent behavioral paradigms relevant to treating depression and PTSD comparable to high doses. The low dose was predicted to be subhallucinogenic in humans based on allometric scaling while the high dose was predicted to be hallucinogenic. However, the data on human psychedelic microdosing are equivocal, and therapeutic benefit of low doses of psychedelics remains to be demonstrated in a well-controlled clinical study. Moreover, administration of low doses of hallucinogenic compounds is not an ideal therapeutic strategy, as these compounds still possess the potential for abuse. Fortunately, through careful chemical design, we were able to engineer several non-hallucinogenic analogs of psychedelics with beneficial properties. These compounds do not induce a head-twitch response—a behavioral proxy for hallucinations in mice that correlates exceptionally well with hallucinogenic potency in humans.

Our two most advanced compounds—AAZ and TBG—have demonstrated robust plasticity-promoting properties and produce sustained (>1 week) antidepressant-like effects following a single administration in both environmental (chronic unpredictable stress) and genetic (VMAT2 heterozygous mice) models of depression as measured via behavioral tests relevant to motivation, anhedonia, anxiety, and cognitive flexibility. TBG has also been shown to have anti-addictive properties in models of alcohol and opioid use disorders. Moreover, a single dose of TBG was able to completely rescue circuit-level dysfunction induced by chronic stress, which included deficits in dendritic spine density, calcium dynamics, and interneuron function. The psychoplastogenic effects of TBG and AAZ may involve activation of 5-HT2 receptors, though detailed mechanistic studies have not yet been reported.

One of the most important questions to address is the durability of psychoplastogen effects. In both humans and rodents, the antidepressant responses of ketamine appear to last for about 1 week. This correlates well with the effects of the drug on dendritic spine density. In contrast, the mood-elevating properties of psilocybin seem to last significantly longer, as do its effects on neuronal structure. Currently, it is unclear exactly how long the effects of non-hallucinogenic psychoplastogens will last following a single administration, and head-to-head comparisons with ketamine and psilocybin are warranted to help establish optimal dosing frequency.​

Patient Populations Best Suited for Hallucinogenic and Non-hallucinogenic Approaches​

Given that ~20% of American adults suffer from a mental illness in a given year, the economic burden of these disorders is estimated to be in the hundreds of billions of dollars annually. Moreover, the current standard of care treatments suffer from slow onset (e.g., 7 week average response time with citalopram), low efficacy (only 27% of depressed patients achieved remission after 12 weeks of citalopram treatment as measured by HAM-D), and often intolerable side-effects, it is clear that we need to expand psychiatry's arsenal to include a variety of new approaches. Both hallucinogenic and non-hallucinogenic psychoplastogens have important roles to play in the fight against mental illness, but it is critical to identify which patient populations are best served by these types of treatment. First, many patients may not want to participate in psychedelic-assisted therapy given that hallucinogens can induce acute anxiety and may lead to challenging experiences (40). For those who are open to psychedelic-assisted therapy, significant barriers to treatment still exist.

Currently, there are over 300 clinical trials registered on clinicaltrials.gov to study the effects of psychedelics in humans with the majority of recent studies focusing on the effects of psilocybin and MDMA. However, only a fraction of patients who volunteer for psychedelic-assisted therapy are permitted to participate. Exclusion criteria for these trials typically include cardiovascular and mental health risks that could potentially be exacerbated by psychedelics. For example, psychedelic-assisted therapy is generally contraindicated for people with a family history of psychotic disorders or complex psychiatric comorbidities to avoid the possibility of triggering a first episode of psychosis or precipitating suicidal behaviors.

In two recent psilocybin clinical trials, ~95% of all volunteers were eliminated on the basis of exclusion criteria. For comparison, ~25% of participants were excluded from two recent major depressive disorder (MDD) trials of the non-hallucinogenic compound vortioxetine (Figure 4). If these strict exclusion criteria are deemed necessary by the FDA and payers, they will drastically limit the number of patients who could potentially benefit from this treatment paradigm, especially when you consider the high genetic heritability and co-morbidity of neuropsychiatric disorders (Figure 5). Between 33 and 66% of patients suffering from MDD have a psychiatric comorbidity that could potentially exclude them from psychedelic-assisted therapy

FIGURE 4
fpsyt-12-727117-g004.jpg

Figure 4. Percentage of patients excluded from recent large MDD trials of the hallucinogenic drug psilocybin and the non-hallucinogenic drug vortioxetine. A comprehensive analysis of all psychedelic trials is beyond the scope of this review.

FIGURE 5
fpsyt-12-727117-g005.jpg

Figure 5. Percentage of MDD with various comorbidities. Data for bipolar disorder, psychosis, substance use disorder, personality disorders, and suicidality were obtained from references, respectively.

Identifying factors that can predict how patients will respond to psychedelic-assisted therapy will be important for maximizing efficacy and reducing adverse events. For example, patients who exhibit apprehension, preoccupation, or baseline attachment avoidance appear to be more likely to have challenging experiences following administration of a psychedelic drug. Using these metrics to pre-screen patients should improve the safety and efficacy of psychedelic-assisted therapy; however, stratification of patients in this manner will inevitably reduce the addressable patient population, again, emphasizing the need for a non-hallucinogenic first-line treatment against depression and related disorders.

Presumably, treatment with non-hallucinogenic psychoplastogens would not be limited by comorbidities or other factors that might exclude someone from participating in psychedelic-assisted therapy. For these reasons, and others described below, it seems reasonable for non-hallucinogenic psychoplastogens to potentically be used as first-line treatments, assuming that they demonstrate greater efficacy in humans than the standard of care (i.e., traditional antidepressants). Psychedelic-assisted therapy could be reserved for patients who have not responded to any other medicine. Indeed, some patients may benefit from the mystical-type experiences occasioned by psychedelics as many people rate these experiences as among the most meaningful in their lives. Such a positive experience could have a variety of effects on patients including, but not limited to, improving the relationship between therapist and patient, helping patients to gain insight about their condition, or producing a powerful placebo effect. It is challenging to design truly double-blind placebo-controlled clinical trials with psychedelics given their profound subjective effects. Despite efforts to employ “active placebos” (e.g., niacin or a low-dose of a psychedelic), many patients and clinicians can still correctly distinguish between a high dose of a psychedelic drug and an active placebo. Thus, the exact role of placebo effects in the overall efficacy of psychedelic-assisted therapy has not been firmly established. Even so, an enhanced placebo effect can potentially be leveraged to treat severely depressed patients who have not been helped by other means.​

Healthcare System Issues With the Psychedelic-Assisted Psychotherapy Model​

In addition to comorbidities and genetic predispositions, the basic mechanics of healthcare systems are likely to be major factors limiting the number of patients who will ultimately be able to receive hallucinogen-based therapeutics. Given their profound effects on perception, these drugs necessitate administration in a clinical setting where the patient can be observed by medical professionals. In 2019, Johnson & Johnson's SpravatoⓇ (esketamine nasal spray) became the first hallucinogenic psychoplastogen treatment approved by the FDA for refractory depression, and in 2020 the indication was expanded to include adult major depressive disorder. SpravatoⓇ has to be administered in a clinic under supervision due to the known risks of serious adverse outcomes resulting from disassociation and sedation as well as the potential for abuse. After intake and medical screening, the patient is enrolled in the SpravatoⓇ REMS (Risk Evaluation and Mitigation Strategy) program. SpravatoⓇ is self-administered intranasally in the presence of a healthcare professional, and the patient is monitored for the next 2 h. Patients receive two treatments a week for the 1st month, and once a week or once every 2 weeks after that. For racemic ketamine administered intravenously, clinics follow the NIMH trial protocol of six infusions administered over a two-to-3-week period in an outpatient clinic or medical facility. Boosters are given every 3–5 weeks after that. Intravenous infusion takes about 40 min, and guidance is provided not to drive, operate any dangerous machinery, or make any important decisions until the day after a ketamine treatment. The requirement for administration in a clinical setting drastically increases the cost of both racemic ketamine and SpravatoⓇ.

Though psychedelics are expected to be administered less frequently than ketamine given their robust effects, they too must be administered under the care of a healthcare professional. Current research-based psychedelic-assisted therapy has three phases: preparation, treatment, and integration. Given that psychedelics have the potential to cause dangerous behaviors due to potential negative psychological reactions such as anxiety, fear, panic, or psychosis, a team of two professional therapists is required to be in attendance to supervise, but minimally interact with the patients throughout the course of the drug's action. Moreover, it is recommended that multiple healthcare workers be involved in all three stages to ensure that professional boundaries are maintained. The preparation session establishes the alliance between the therapist and the participant. Treatment sessions, which typically last 6–8 h for psilocybin-assisted therapy allow the participants to have a peak experience within a set and setting thought to be most conducive to optimizing the therapeutic effect. The integration session is meant to help the participant process, rationalize, and gain insight from the hallucinogenic experience. Although psychedelic therapies have demonstrated outstanding benefits in several clinical trials, the cost-effectiveness and overall accessibility of such therapies raises major concerns.

The costs associated with treating mental illness with hallucinogenic psychoplastogens is extremely high compared to the standard of care (Figure 6). For example, the initial month of ketamine therapy costs from $4,720 to $6,785, and subsequent monthly therapy can range from $2,360 to $3,540. Additional costs associated with patients taking time off work to receive treatment and to travel to appropriately staffed/equipped clinics must also be considered.

FIGURE 6
fpsyt-12-727117-g006.jpg

Figure 6. Estimated quarterly (3 months) costs (L = low, M = medium, H = high) for daily administration of non-hallucinogenic take-home traditional antidepressants (blue) and intermittent dosing of clinically administered, dissociative psychoplastogen (ketamine) treatments (red). Note: generic ketamine costs include drug, administration (IV), and required monitoring, while Prozac®, Zoloft®, Celexa®, and Spravato® costs are for the drug only. Retail costs for SSRIs reflect the low and high branded price published on goodrx.com with the medium price being calculated as an average of the two. To estimate the total cost, we used medium doses for each drug (Prozac® 20 mg, Zoloft® 50 mg, and Celexa® 20 mg,) and assumed daily dosing for 90 days. Ketamine infusion cost was sourced from ketamineclinicsdirectory.com/ketamine-infusion-cost/. This site estimates the low and high cost of a complete set of 4–6 generic ketamine infusions to be $1,600–$4,800, respectively. The medium cost was calculated as an average of the two ($3,200). Our estimate for the 3-month cost of generic ketamine assumes that no additional doses are needed beyond the initial 4–6 doses. Retail Spravato® costs were estimated based on the price of a 56 mg dose ($900) published on goodrx.com3 and dosing was based on the package insert. The label indicates Spravato® should be dosed twice a week during the induction phase (weeks 1–4; $900 × 2 × 4 = $7,200), once a week during the maintenance phase (weeks 5–8; $900 × 1 × 4 = $3,600), and once a week thereafter (weeks 9–12; $900 × 1 × 4 = $3,600). The estimated low cost of Spravato® includes the induction phase only. The estimated medium cost of Spravato® includes the induction phase and the maintenance phase. The estimated high cost of Spravato® includes the induction phase, maintenance phase, and 4 weeks of additional treatment. Note: the cost of administration and monitoring for Spravato are not publicly available, and thus not included, but these costs are anticipated to be significant.

Many people assume that psilocybin-assisted therapy will be cheaper than ketamine treatment as the antidepressant effects following a single administration of psilocybin appear to be more enduring than those of ketamine. However, the overall cost of psilocybin treatment is estimated to be close to, if not higher than, the cost of SpravatoⓇ treatment due to the increased session participation of therapists. The low-throughput nature and associated high costs of the psychedelic-assisted therapy model have been acknowledged by the community and have resulted in new studies being launched by non-profit and corporate sponsors to streamline the process and reduce costs through group and virtual therapy sessions. However, it is currently unclear if such approaches will be as safe and efficacious as the current model.

The cost associated with rolling out the psychedelic-assisted therapy model poses a great hurdle to its effective implementation in the current mental health ecosystem. In 2019, the Institute for Clinical and Economic Review (ICER) issued a recommendation that deemed SpravatoⓇ to deliver a “low value for money” according to their value-assessment framework. Furthermore, the UK price watchdog agency, the National Institute for Health and Care Excellence (NICE), refused to endorse SpravatoⓇ therapy for inclusion as a reimbursable drug on the UK's National Health System (NHS). Although NICE acknowledged the drug's efficacy for relieving the symptoms of depression, the agency also commented that the “introduction of esketamine into clinical practice in the NHS will be complex because the structure and delivery of services would need to be changed. Estimates of the costs of providing the clinical service for esketamine were highly uncertain.” As these price watchdog agencies become progressively more influential in the decision-making process of payers, their recommendations will likely lead to payers raising their medical criteria for coverage, potentially jeopardizing patient access to psychedelic-assisted therapy for financial reasons. We are already seeing racemic ketamine being administered off-label for a variety of neuropsychiatric disorders without reimbursement from insurance companies. This leads to an inequity issue, where only wealthy individuals who can afford the out-of-pocket costs have access to this type of treatment.

In addition to the financial burdens to the patients, understaffing of qualified psychotherapy practitioners is likely to be one of the biggest issues for nationwide implementation of psychedelic-assisted therapy. The FDA requires all U.S. therapists to have at least a master's degree, and current best practices require a minimum of two therapists to be present during psychedelic sessions. Moreover, all the therapists who participate in the psilocybin and MDMA clinical development programs are required to hold a professional license and demonstrate clinical experience in psychotherapy or mental health counseling. Although institutes such as the California Institute of Integral Study (CIIS), Multidisciplinary Association for Psychedelic Studies (MAPS), and corporate programs from companies like COMPASS Pathways offer short-term training programs for psychedelic-assisted counseling, there is a huge demand and supply gap for competent therapists considering that the estimated prevalence of treatment-resistant depression (TRD) in the U.S. is around 2.8 million people. Moreover, given that set and setting are well-known to influence the subjective effects of psychedelics, clinical centers with appropriate facilities will need to be established.​

Democratizing Access to Psychoplastogenic Medicines​

The limitations associated with hallucinogenic medicines could prevent many patients from benefiting from the growing body of psychedelic-inspired research related to pathological circuit remodeling. By eliminating the need to treat patients in the clinic, non-hallucinogenic psychoplastogens—which would presumably be available from retail pharmacies much like traditional antidepressants—would reduce the complexity of treatment administration and have the potential to greatly expand access of patients to psychoplastogenic medicines. These molecules produce the same types of long-lasting structural and functional changes in the brains of preclinical animals that follow administration of ketamine or serotonergic psychedelics. Assuming that they are efficacious in humans, non-hallucinogenic psychoplastogens would not bear the same administrative costs and limitations as first-generation hallucinogenic psychoplastogens. When comparing the known or predicted yearly healthcare costs associated with ketamine, psilocybin, SSRIs, and non-hallucinogenic psychoplastogen treatments, it is clear that the non-hallucinogenic approaches will be more cost-effective (Figure 6).

Non-hallucinogenic psychoplastogens could potentially meet the demands of patients who fail to respond to monoaminergic agents, or even replace them as more effective first-line treatments with fewer side-effects. In preclinical animal models, like in humans, traditional monoaminergic antidepressants require chronic administration to achieve robust efficacy. Such chronic dosing paradigms inevitably lead to a host of undesirable side-effects that include weight gain, sexual dysfunction, and gastrointestinal problems, with many patients refusing to take traditional antidepressants due to their side-effects. In contrast, non-hallucinogenic psychoplastogens, like their hallucinogenic counterparts, produce sustained therapeutic behavioral responses in preclinical animal models after a single administration. Thus, the need for chronic dosing in humans will likely be obviated resulting in fewer undesired side-effects. However, a major challenge for the field will be to determine exactly what frequency of dosing will be most effective. Fortunately, new imaging tools have the potential to identify biomarkers of psychoplastogen efficacy. These include relatively new positron emission tomography (PET) tracers that can non-invasively measure the effects of psychoplastogens on synaptic vesicle density in vivo.

It is quite possible that insight gained from mystical-type experiences coupled with changes in neurocircuitry is responsible for the large effect size and durability of psychedelic-assisted therapy. However, if even a fraction of the efficacy or durability could be achieved using compounds that do not induce mystical-type experiences or hallucinations, a much larger patient population could benefit. While mystical-type experiences will undoubtedly be beneficial for some patients, they may not be necessary for all patients. Thus, if non-hallucinogenic psychoplastogens can demonstrate efficacy in the clinic as robust as their effects in preclinical models, their advantages over both traditional monoaminergic antidepressants and hallucinogenic psychoplastogens should position them as first-line treatment options (Figure 7).

FIGURE 7
fpsyt-12-727117-g007.jpg

Figure 7. Comparison of traditional antidepressant treatments with hallucinogenic and non-hallucinogenic psychoplastogen treatments. *Currently, no clinical trials have been conducted with non-hallucinogenic psychoplastogens, and thus, their fast-acting and long-lasting effects refer to preclinical testing only. **The scalability and cost-effectiveness of non-hallucinogenic psychoplastogens are based on the assumption that they will produce clinical efficacy greater than the standard of care (i.e., traditional antidepressants).

Conclusion​

The ability to selectively modulate neural circuits using small molecule psychoplastogens opens up new horizons in neuropsychiatry focused on healing pathological neural circuitry rather than masking disease symptoms. This type of circuit-based approach represents a fundamental shift in how we might treat a number of neuropsychiatric diseases and has important implications for the future of CNS drug discovery. Given the history of neuropsychiatry and the intractable nature of brain disorders, we need to take advantage of every available tool in our therapeutic arsenal including both hallucinogenic and non-hallucinogenic psychoplastogens. Ketamine and psilocybin have demonstrated that it is possible to produce long-lasting beneficial changes in neural circuitry using small molecule drugs, and they have forged a path for future, optimized psychoplastogens to take their place. If we ever hope to heal the nearly 20% of the population suffering from a mental illness, we must find innovative ways to reduce healthcare costs and broaden patient access to psychoplastogenic medicines. Non-hallucinogenic psychoplastogens have the potential to be truly scalable solutions to many of the problems facing neuropsychiatry.

 
Last edited:
another-psychedelic-and-its-potential-psychiatric-promise-350064-1280x720.webp


DMT and its Potential Psychiatric Promise

by Samoon Ahmad | Technology Networks | 31 Mar 2022

Psychedelics’ potential is rivaled only by their power. They are extremely potent substances that have been used in various cultures dating back thousands of years, largely for ritualistic and spiritual purposes. In addition, they have traditionally been called upon to offer those who wish to gain insight into realms believed to be inaccessible to the conscious mind.

This is more or less the same reason why interest in psychedelics has skyrocketed among clinicians, researchers, and the general public. For clinicians, these drugs are being touted as potential groundbreaking treatment options for a host of psychiatric and inflammatory conditions as they can potentially treat symptoms that are extremely difficult to manage with conventional medicines. For researchers, studying psychedelics can help them understand how these substances can eliminate the interference of ego structures and conscious experiences in their subjects to unveil the most fundamental dynamics of the mind, thereby offering new insights into our understanding of neuroscience, consciousness, and how the two intersect. For the general public, they offer relief from symptoms of hobbling psychiatric conditions, fuel for philosophical musings, and the opportunity to take part in deeply meaningful experiences. As has been frequently reported in research dating back to at least 2006, psychedelic sessions are frequently ranked among the top five most meaningful experiences in individuals’ lives even more than a year later.

Psychiatrists are very excited about the potential of psychedelics, particularly with respect to drugs like psilocybin, LSD, and MDMA. Despite the excitement about adding these substances to psychiatrists’ toolkits, one of the most potent psychedelic compounds available, DMT has received very little attention from clinicians until recently.

While there is no question that DMT holds enormous potential, as clinicians we must first understand how to harness it.

What is DMT?

DMT is a classic psychedelic that can induce profound changes in sensory perception (hallucinations), feelings of euphoria, and radical alterations in typical thought patterns. On the neurochemical level, DMT behaves similar to other psychedelics (e.g., LSD, psilocybin, mescalin) by not only binding to serotonin 2A (5-HT2A) receptors in the central nervous system, but also interacting with other serotonergic (5-HT1A, 5-HT2C, and 5-HT7) and glutaminergic receptors, sigma-1 receptors, and trace-amine associated receptors. DMT also interacts with acetylcholine and dopamine signaling.2 Interestingly, DMT exists naturally in the brains of mammals, including humans, though the reason is unclear. Some have speculated that it may play a role in altering consciousness to allow for dream states, spiritual or mystical experiences, or even passage from life onto death. Currently, there is no evidence to support these claims, and it is not even clear if DMT exists in concentrations high enough to produce measurable effect.

DMT is the main ingredient in medicines that have traditionally been prepared and consumed as teas by indigenous peoples from throughout South America well before any European contact. The most well-known example is ayahuasca. In conjunction with DMT (from Psychotria viridis), ingredients in these teas include naturally occurring monoamine oxidase inhibitors, which delay onset and extend the duration of DMT’s effects for several hours. If one takes synthetic DMT, which is often inhaled using a vape pen, the effects may take a few minutes to manifest and may peak within five to ten minutes before beginning to wear off. The entire experience may only last 30 minutes or so.

What are the effects of DMT?

One of the most promising areas of research for psychedelics in recent years has been its potential role in mitigating anxiety and depression among terminal cancer patients suffering with existential crisis. Unfortunately, there are few pharmacotherapies or interventions in the field of psychiatry that can be used to help patients as they struggle to come to terms with their own mortality. “The ego, faced with the prospect of its own extinction, turns inward and becomes hypervigilant, withdrawing its investment in the world and other people,” Michael Pollan wrote in his paean to psychedelics, How to Change Your Mind. Psychedelics offer the opportunity to suspend the ego temporarily, which can be experienced initially as terrifying, but it is followed by a gradual sense of surrender, acceptance, love, and a greater sense of unity with the universe as the ego dissolves and one experiences simulated death and rebirth. Psychedelics are not treating end-of-life anxiety and depression pharmacologically; rather, they are treating these conditions phenomenologically (i.e., through the subjective experience it induces).

Though DMT acts upon the same serotonin receptors as other classical psychedelics, the DMT experience is unique. With smoked DMT, the ego is not just dissolved in a transcendent experience; it is obliterated.

According to DMT users, the visible world is often replaced by extremely vivid kaleidoscopic patterns of color. Furthermore, users report being transported to what they describe as another plane of reality with the presence of godlike creatures, and many of those who have taken DMT have reportedly interacted with these creatures. An online survey conducted by researchers at Johns Hopkins School of Medicine involving 2,561 individuals (median age 32 years; 77% male) who reportedly had encounters with these ostensibly autonomous beings found that half of participants who considered themselves atheists before the experience no longer did afterwards, and more than half of total participants believed that these beings are conscious, intelligent, benevolent and continued to exist after the encounter in a real but different reality.

Characterized by Davis as an “ontological shock,” over half of the participants claimed that the DMT experience was one of the most spiritually significant and meaningful moments in their lives. Many respondents also reported profound changes in outlook and positive changes in life satisfaction and subjective well-being.
Potential Directions for DMT Research

Clinical trials of DMT are only now beginning, so it will likely be years before we even start to glean its full potential. However, years of research has shown us that the beneficial psychological effects of psychedelics appear to be tied to their long-term subjective effects and how they change patient perspective. The important question to ask is that if DMT imparts its effect through ego annihilation, wouldn’t it make sense that it could be even more effective than other psychedelics?

Theoretically, yes.

Like other psychedelics, it may prove capable of easing existential distress among terminally ill patients and promoting subjective well-being. It may even help individuals with substance use disorders overcome their dependence. A pharmaceutical company based in the United Kingdom, in conjunction with Imperial College London’s Centre for Psychedelic Research, initiated a phase I trial earlier this year to see if DMT could be used to treat major depressive disorder.

However, there are potential roadblocks to DMT’s use in a clinical setting, too—at least in its smoked or vaped form. As Stephen Ross, one of my colleagues at New York University who has studied psychedelics extensively, observed during a conversation we had about frontiers of psychedelic research, psychedelics are only part of the equation in a larger model of medication-assisted psychotherapy. Patients should not simply be given these extremely potent drugs, and then left on their own to process the experience. They need guidance from people who have the tools and training to place it into a larger narrative.

To properly provide care, staff members need to be trained to be effective guides to patients who are experiencing the acute effects of the drugs and to help them contextualize and absorb the experience. Furthermore, patients need to be psychologically prepared for the experience and will likely require several sessions before and after the experience to effectively process it. Given the enormous difference between the acute effects of DMT and LSD or psilocybin, it stands to reason that protocols and models that provide the best therapeutic experience are not even remotely interchangeable.

To fully harness the therapeutic power of DMT, as well as psychedelics in general, research will need to focus not only on the potential conditions it can help treat or the neurophysiological effects of the drug, but how the drug is administered and how patients are guided through the experience. We will need to embrace the role of navigator, rather than simply impartial facilitator of awareness, growth, and psychological development of our patients.

About the author:Samoon Ahmad, M.D., is Professor of Psychiatry at NYU Grossman School of Medicine.

*From the article here :
 
Last edited:
giphy.gif



Psychedelic drugs could treat depression, other mental illnesses

by David E. Olson | The Conversation | 20 June 2018

Strictly speaking, a psychedelic is a “mind-manifesting” drug – a definition that’s open to interpretation. They tend to produce perceptual distortions or hallucinations by activating 5-HT2A receptors. Our research group has found that compounds typically regarded as psychedelics, like LSD and DMT, as well as those that are sometimes called psychedelics, like MDMA, and those that are not usually called psychedelics, like ketamine, are all capable of profoundly impacting neuronal structure.

Psychedelics vs. Psychoplastogens

Our group has coined the term “psychoplastogen” to refer to such compounds, and we believe that these molecules may hold the key to treating a wide variety of brain diseases.

Our studies on neurons grown in dishes, as well as experiments performed using fruit flies and rodents, have demonstrated that several psychoplastogens, including psychedelics and ketamine, encourage neurons to grow more branches and spines. It seems that all of these compounds work by activating mTOR – a key protein involved in cell growth.

The biochemical machinery that regulates mTOR activity is intricate. As we tease apart how psychedelics and other psychoplastogens turn on mTOR signaling, we might be able to engineer compounds that only produce the therapeutic effects on neuronal growth while bypassing pathways that lead to undesired hallucinations.

The field has known for some time now that psychedelics can produce lasting positive effects on brain function, and it’s possible that these long-lasting changes result from the psychoplastogenic effects of these drugs. If true, this would suggest that psychoplastogens might be used to repair circuits that are damaged in mood and anxiety disorders.
Panacea or poison?

Many diseases, such as depression and anxiety disorders, are characterized by atrophy of dendritic branches and spines. Therefore, compounds capable of rapidly promoting dendritic growth, like psychedelics, have broad therapeutic potential. The number of papers demonstrating that psychedelics can produce therapeutic effects continues to grow every year.

Panacea or poison?

However, we should temper our enthusiasm because we do not yet know all of the risks associated with using these drugs. For example, it’s possible that promoting neuronal growth during development could have negative consequences by interfering with the normal processes by which neural circuits are refined. We just don’t know, yet.

Similarly, it is unclear what effects psychoplastogens will have on the aging brain. It’s important to keep in mind that excessive mTOR activation is also associated with a number of diseases including autism spectrum disorder (ASD) and Alzheimer’s disease.

We need to understand how these powerful compounds affect the brain, in both positive and negative ways, if we hope to fully comprehend the fundamental laws governing how the nervous system works and how to fix it when it doesn’t.

 
psilocybin-1000.jpg



Microdosing found to improve mental health*

University of British Columbia | Neuroscience News | 23 Nov 2021

An international study led by UBC Okanagan researchers suggests repeated use of small doses of psychedelics such as psilocybin or LSD can be a valuable tool for those struggling with anxiety and depression.

The study, recently published in Scientific Reports, demonstrated fewer symptoms of anxiety and depression, and greater feelings of wellbeing among individuals who reported consuming psychedelics in small quantities, or microdosing, compared to those who did not.

Microdosing involves regular self-administration of psychedelic substances in amounts small enough to not impair normal cognitive functioning.

Considering this is the largest psychedelic microdosing study published to date, the results are encouraging, says UBCO doctoral student and lead author Joseph Rootman.

“In total, we followed more than 8,500 people from 75 countries using an anonymous self-reporting system—about half were following a microdosing regimen and half were not,” Rootman explains. “In comparing microdosers and non-microdosers, there was a clear association between microdosing and fewer symptoms of depression, anxiety and stress—which is important given the high prevalence of these conditions and the substantial suffering they cause.”

The study is also the first to systematically examine the practice of stacking, or combining microdoses of psychedelics with other substances like niacin, lions mane mushrooms and cacao, which some believe work in conjunction to maximize benefit.

Rootman works with Dr. Zach Walsh, a psychology professor in UBCO’s Irving K. Barber Faculty of Arts and Social Sciences. Dr. Walsh says it’s an exciting time for research in this area.

“These findings highlight adults who are microdosing to treat their mental health conditions and enhance their wellbeing—rather than simply to get high,” says Dr. Walsh. “We have an epidemic of mental health problems, with existing treatments that don’t work for everyone. We need to follow the lead of patients who are taking these initiatives to improve their wellbeing and reduce suffering.”

Microdosing involves regular self-administration of psychedelic substances in amounts small enough to not impair normal cognitive functioning. Image is in the public domain

Study co-author Kalin Harvey is the chief technology officer of Quantified Citizen, a mobile health research platform. He says this study highlights the potential of citizen science.

“The use of citizen science allows us to examine the effects of behaviors that are difficult to study in the lab due to regulatory challenges and stigma associated with the now discredited ‘war on drugs.'”

According to the Canadian Mental Health Association, one in five Canadians personally experience a mental health problem or illness each year. This is one of the many reasons Dr. Walsh says conducting innovative psychological research is imperative.

“These cross-sectional findings are promising and highlight the need for further investigation to better determine the impacts of factors like dosage and stacking,” explains Dr. Walsh.

“While the data is growing to support the use of psychedelics like psilocybin in large doses to treat depression and addiction—our data also helps to expand our understanding of how psychedelics may also help in smaller doses.”

Original Research: Open access.
Adults who microdose psychedelics report health related motivations and lower levels of anxiety and depression compared to non-microdosers” by Joseph M. Rootman et al. Scientific Reports

*From the article here :
 
GettyImages-501927692.jpg



Are psychedelics the future of mental health care?*

by Maya Singer VOGUE | 19 Feb 2021

At seven o’clock on a recent evening, I dim the bedroom lights, call out a reminder to my boyfriend to rouse me in an hour with a gentle tap, and close the door. “Have a great trip,” I hear him say from the living room as the two ketamine tablets I’ve pressed into the pockets of my cheeks dissolve, leaving a bitter residue. Minutes later, I’m flying over water that reflects a sourceless golden light. Am I the light? The thought triggers a sensation of being stretched like taffy in all directions. It’s not my body being stretched – I don’t have a body anymore – but the immaterial me moving in tune with the ambient music in my headphones. I stretch and spread until at last I’ve dissolved – pixelated – at which point a small voice in my head calls out, “Do you really think this will help you quit smoking?”

The last time I was on ketamine, I was hooked up to an IV following surgery. This time, the drug – in general medical use as an anaesthetic since 1970 – arrived on my doorstep courtesy of Mindbloom, a new telemedicine company specialising in ketamine-based psychedelic therapy. This was no shady dark-web deal. Prescribed by a psychiatric nurse practitioner following an extensive intake evaluation, and compounded by a licensed pharmacy, the ketamine came bundled with an eye mask, a hardbound journal, and a blood-pressure cuff that I was instructed to use before and after dosing, to test my vitals. The tablets themselves were housed in a mirrored pouchette with the tagline ACHIEVE YOUR BREAKTHROUGH spelled out in sleek, sans serif font. I was tempted to post a shot to Instagram, but I had a Zoom call with my psychedelic-integration coach in half an hour, and I wanted to meditate first.

Welcome to the brave new world of psychedelic wellness. After decades underground, hallucinogens such as ketamine, LSD, psilocybin, and MDMA are getting a fresh look from the medical establishment, thanks to myriad studies suggesting silver bullet-like efficacy in the treatment of anxiety, depression, and addiction, among other ailments. MDMA, renowned for its bliss-inducing effect – hence the street name “ecstasy” – is on course to be approved for the treatment of post-traumatic stress disorder (PTSD) in the US within the next year or two. Synthetic forms of psilocybin, the active compound in magic mushrooms, were given “breakthrough” designation by America’s Food & Drug Administration (FDA) in 2018, allowing for the fast-tracking of drug trials. Meanwhile, this past November, Oregon became the first state to legalise psilocybin for medical use, an advance not lost on the investors flocking to start-ups like MindMed and Compass Pathways, both of which are developing psilocybin treatments in anticipation of a cannabis-style psychedelics boom. A mental health revolution is at hand – and it’s long overdue, according to experts such as Frederick Streeter Barrett, PhD, assistant professor of psychiatry and behavioural sciences at Johns Hopkins School of Medicine and a faculty member at the university’s recently opened Centre for Psychedelic & Consciousness Research.

“The current model for treating problems like anxiety and depression just isn’t very good,” Barrett says. “Patients take pills every day, for years, and these medications not only have nasty side effects, they often don’t even work. But with psychedelics-assisted therapy, there’s the potential to truly alter someone’s life with just one or two sessions, because you’re getting at suffering at the source.”

I’m not suffering, exactly, but for lack of more technical language, I’ve kind of been freaking out. Straining to maintain a productive work schedule under lockdown, I fell back into the habit of smoking as I write – and soon thereafter, the habit of trying to quit. The addiction struck me as fundamentally psychological. If I was so hooked on nicotine, why did I reach for my American Spirits only when I was stuck at my desk, staring down a deadline? But reach for them I did, and the harder I worked not to – with the aid of gum, apps, hypnosis, you-name-it – the more fixated I became on the fear that I simply could not write without cigarettes. I was starting to feel truly hopeless when I stumbled across a news item about studies showing that with the aid of psilocybin, long-time smokers were quitting cold turkey and sticking with it at rates that put all other remedies to shame; two-thirds of participants in one recent study were confirmed cigarette-free after one year.

mirror.jpg


Intrigued, I did a little more digging and discovered that ketamine – a dissociative hallucinogen that is already legal for supervised medical use, including in the treatment of depression – seemed to draw out the mind in a way similar to psilocybin by putting the brain in a “neuroplastic” state, explains Julie Holland, MD, a New York-based psychiatrist and the author of the 2020 book Good Chemistry. “They have different chemical properties, but both ketamine and psilocybin have an ego-dissolving effect, where you’re breaking the mental loop that’s symptomatic of conditions like depression and anxiety and addiction, and allowing the brain to form new connections.”

Maybe a little ego-dissolution was the answer, I mused as I stamped out another butt in the ashtray next to my laptop and Googled “ketamine therapy – New York.”

“The truth is, we don’t really know how this stuff works,” Michael Pollan, author of the bestselling psychedelics primer How to Change Your Mind, tells me. “A leading theory is that psychedelics quiet the brain’s ‘default-mode network’, and that opens up new pathways for thought.” As Pollan goes on to explain, the default-mode network is where “the ego has its address” – it’s the part of our brains where we construct the narrative of who we are and, thus, the place we get stuck in destructive thought patterns about ourselves. “That could be ‘I’m a worthless person who doesn’t deserve love,’ or it could mean telling yourself that you can’t get through the day without smoking,” Pollan continues. “Either way, the idea is that, by muffling those thoughts, psychedelics help you out of the rut.”

Pollan’s précis on the science of psychedelics is reassuringly down-to-earth. For years, I’d been put off by the drugs’ woo-woo connotations, and to judge by the refined, minimalist aesthetics of new ketamine-therapy chains such as Field Trip Health, which has serene locations in New York City, Toronto, Atlanta, Chicago, and Los Angeles, I’m not the only person with zero interest in a tie-dye mental makeover. It’s all a far cry from Timothy Leary and The Electric Kool-Aid Acid Test. But Leary – who famously conducted psychedelics experiments at Harvard in the early 1960s, before he ran afoul of the law and, in turn, helped prompt the criminalisation of psilocybin and LSD – does continue to exert an influence. His “set and setting” theory is a cornerstone of all contemporary psychedelics-aided therapy. “Set basically refers to mindset, going into your journey, and setting is your environment,” explains Ronan Levy, who cofounded Field Trip in 2019 after establishing – then selling – Canada’s largest network of cannabis clinics. “They matter as much as the drug you’re taking,” he continues. “You need to be in a place – mentally and physically – where you feel inspired and at ease.”

Because I’d chosen to work with Mindbloom, thanks to their Covid-friendly process, the setting for my four, hour-long treatments, was my bedroom. To be perfectly clear, I wasn’t microdosing. Nor was I popping a pill just to see what colours spilled out of my head. Prior to receiving my Mindbloom package, I spent over an hour on Zoom with a board-certified psychiatric nurse practitioner who quizzed me on everything from my family medical history to my typical responses to stress. (According to Mindbloom founder and CEO, Dylan Beynon, about 35 per cent of potential patients are screened out at this point, for reasons such as past experience of psychosis or, at the other end of the spectrum, not meeting the threshold for a diagnosis of anxiety and/or depression.) “Set” was established in conversation with Laura Teodori, my psychedelic-integration-support coach, who – after obtaining confirmation from my boyfriend that he’d check on me every 20 minutes – helped me formulate an “intention” for the trip immediately after our call. My goal, we ascertained, was to recall moments in my life when I could create without smoking. With that in mind, I tucked the tablets inside my mouth, pressed play on the Mindbloom-curated soundtrack that would be piping through my headphones, lowered my eye mask, and waited for my default-mode network to go off-line.

Hallucinogens come in many forms, from the low-dose ketamine I was taking to wallop-packing plant medicines, like ayahuasca and ibogaine and peyote, that have been used in sacred rituals for hundreds, perhaps thousands of years (and are illegal in the US). But a feature common to all is the sense of coming into contact with the cosmic. “It’s like there’s no boundary between you and others, or you and the universe,” notes Johns Hopkins’s Barrett, saying that "virtually all subjects in psilocybin studies have reported such a feeling of oneness. Some people call this an experience of God, or nirvana.” I went into my first ketamine journey matter-of-factly, with a problem to solve, and even so, that first trip commenced with a vision of the world rewinding, a kind of reverse big bang that exposed the heretofore invisible filaments connecting everyone and everything. The vision moved me – tears puddled behind my eye mask – and then it yielded to more personal impressions, such as a recurring image of myself, age six or seven, playing with my dollhouse.

“What do you think was important about the dollhouse?” Teodori asked me in our post-trip call. I was still pretty woozy as we Zoomed – the effect wore off by the next day – but suddenly, it was like a light bulb went on in my head. “I think…I think I was remembering what it felt like to create without smoking,” I told her. “When there wasn’t any pressure, and I could just play.”

This download is part of integration, another cornerstone of modern psychedelic medicine. “The goal is to take advantage of the neuroplastic state, which lasts for about a week after dosing,” explains Beynon. “You want the changes in your brain to stick, so the question becomes, ‘How do you turn these new thoughts into new behaviours?’” For me, this entailed finding ways to get back in touch with that dollhouse sense of play.

psychology-2706902_1280-1-1200x630.jpg


Easier said than done. My ketamine experiences were clarifying and often even profound, but they didn’t change certain nerve-racking facts of life, such as that I write for a living and thus have deadlines to meet if I wish to pay my bills. Or that it’s hard – like, really hard – to stay motivated in the midst of a global pandemic, when each day brings fresh spurs to panic and depression. “There’s a huge mental health crisis happening parallel to, and in response to, this pandemic,” notes Benjamin Brody, MD, assistant professor of clinical psychiatry at Weill Cornell Medicine in New York, and chief of the Division of Inpatient Psychiatry at the university hospital, where ketamine infusions are typically administered. “People who are grieving, people who have lost jobs, people who are feeling disconnected, whose lives have been upended .…” With demand for care rising “across the board”, as Brody notes,"it’s no surprise that psychiatrists such as Amanda Itzkoff, MD, are seeing a huge uptick in inquiries about ketamine therapy. But it may or may not be the right tool for every job, Itzkoff points out."

“The thing is, if you got laid off and you don’t know how you’re going to pay rent, ketamine won’t change that,”
says Itzkoff, an early adopter who has been providing ketamine infusions at her Manhattan practice since 2014. “It doesn’t remove the external pressures. But when you’ve got someone with severe depression, who has kind of given up, then there’s real promise in this treatment.” Itzkoff cites the example of a former patient, a high-powered attorney and mother of two, who was on disability and “almost catatonic” when they began working together. “She had to be retrieved from this state,” recalls Itzkoff. “By breaking the negative thought loop – even temporarily – you show someone it’s possible to feel another way. And that,” she adds, “can be channeled toward getting people back on their feet.”

Chad Kuske didn’t just get back on his feet following his first psilocybin treatment a year and a half ago; he experienced what he calls an immediate and profound “sense of meaning and a desire to live”. A former Navy SEAL, Kuske, 40, had tried psychoanalysis and various pharmaceuticals before being medically retired from the service in 2017. Reentering civilian life, he found himself using drugs and alcohol as a way of coping with the anxiety, depression, and alienation that he now comprehends as the symptoms of PTSD. “Nothing else had worked. And I knew that sooner or later, if I kept doing things the same way, my life would be over – either literally or metaphorically, like I’d wind up in jail,” Kuske explains. “The mushrooms helped me see my situation clearly: I was in hell, but it was a hell of my own creation, and I could make the choice whether to stay there and suffer or leave and start the work of changing.”

One of the key insights Kuske has taken away from his trips – and from his integration process, which is ongoing – is that he’s not alone in struggling to meet the challenges of daily life. Likewise, Itzkoff suggests that the feeling of interconnectedness induced by psychedelic therapy – and near-psychedelics, such as ketamine and MDMA – may help alleviate the isolation brought on by Covid. It may also play a role in helping the people hardest hit by the pandemic recuperate: Nautilus Sanctuary, a nonprofit psychedelics-research and training centre in New York, is already planning a study exploring the use of MDMA to treat frontline workers with severe PTSD – one of the many inquiries to expand on the drug’s groundbreaking FDA trials sponsored by MAPS, which entered phase three in 2017. Other studies sponsored by the organisation have focused on veterans in Israel and the US, and the Department of Veterans Affairs has exhibited a willingness to approve such studies, so long as they are safe, beneficial, and scientifically sound.

This kind of conservative approach is merited, notes Weill Cornell’s Brody. “I’m very concerned about this atmosphere, that the floodgates are opening. I work with ketamine, a drug that’s been in use for decades, and even there, we don’t know all that much about its long-term effects,” says Brody, who provides ketamine-infusion therapy only to patients in whom he’s observed severe, treatment-resistant depression – and who was positively aghast when I relayed rumours that self-dosing with inhalers of esketamine, a synthetic form of the drug given FDA approval for supervised use in 2019, was all the rage in LA. “Ketamine is a serious drug!” he reiterates. “This isn’t a spa service. It’s not like getting Botox. And what worries me about all these clinics popping up is that people are going to start thinking about it that way.”

Brody is hardly alone in fearing the commercialisation of psychedelics – a trend that, if canny investors like Peter Thiel, a backer of Compass Pathways, are correct, is on pace to increase rapidly. “It’s a unique space because so much of the technology has been developed by Indigenous healers,” notes Pip Deely, cofounder of the venture-capital firm Delphi, which is eyeing investments in psychedelics start-ups and supporting a new psilocybin-legalisation campaign in Hawaii. “We see a lot of dread that if this all goes the way of cannabis, the people who have been doing this work the longest will be cut out of the conversation, and those Indigenous roots will be erased.” Unprompted, I hear a version of this concern from one Berkshires-area healer who, for legal reasons, prefers to remain unnamed; she tells me that, although she supports expanding access to psychedelics, she worries about the experience becoming pro forma and “clinical.”

img.jpg


Though they come at their misgivings from opposite angles, both traditional healers and Brody are wary of psychedelics’ getting marketed as a quick fix – and in all honesty, I’m the target demographic for that pitch. When I sat down at my computer to fill out Mindbloom’s candidate questionnaire, what I wanted was to detangle a few mental wires. By the time I’d completed my final ketamine treatment, I’d come to realise that those wires were crossed very deep down. My writing-while-smoking problem was really a problem with the little voice in my head telling me that I’m not good enough, I haven’t achieved enough, I’m falling behind. As I wrote in my integration journal after my second session, “Every little deal is a big deal.” I added a frowny face to underline the point.

I can’t blame Mindbloom for my failure of mindset. All my conversations with Teodori were oriented around getting me to probe the heart of my fears, and she was diligent in supporting me as I attempted to integrate the lessons of my journeys into daily life, checking in with me every few days via text and reminding me that she was always available to talk. Alas, I didn’t take her up on that offer as often as I should have – I had a ton of writing to do! – and in the end, I felt changed but not transformed. Which could be a me thing, or it could be a drug thing. Barrett of Johns Hopkins pointed me to studies from the university’s Centre for Psychedelic & Consciousness Research indicating that, where smoking cessation is concerned, the more “mystical” the trip, the more effective the treatment. “There’s a big difference between a low dose of ketamine and taking what we call a ‘breakthrough’ dose of psilocybin,” he notes. “That’s where you’re really going to break down your sense of self.”

Is that what I want? Is that what we all want, in some subconscious way? “There’s a spiritual hunger these medicines satisfy,” Pollan points out, and I can attest that once you’ve visited the astral plane, you want to go back. Most hallucinogens are not physically addictive, but the psychedelic experience is itself addicting. I spoke to numerous people for this story who described their encounters with psychedelics as “life-changing” in ways large and small; one woman even credited peyote with restoring movement to her paralysed arm. But Ann Watson’s account is the most relatable. A former VP and fashion director at Henri Bendel in New York City, and now a cochair of The Vaquera Group, a global marketing firm, Watson, 52, is also a self-described “explorer” who, like me, kept a pretty tight lid on her deepest, darkest feelings – until she began working with psychedelics 12 years ago. “My childhood was chaotic; there was a lot of abandonment, but I didn’t associate with the word trauma, because I thought it was reserved for people who have experienced things like rape or war. But I was seeking something,” explains Watson, who tried a variety of treatments to relieve an “ever-present vibration of anxiety,” including counselling and prescribed antidepressants, before experimenting with a long list of psychedelics. Eventually she arrived at a treatment plan with a doctor in Los Angeles she sees four times a year for guided psilocybin trips; she also microdoses psilocybin on a more regular basis, mixing magic mushrooms with Lion’s Mane. “It’s an ongoing process,” she tells me. “The thing is, once you start looking inward, you realise there’s always more to see.”

Perhaps that is the main takeaway from my own journey: that I’m just at the start of it. I have work to do on myself. But in the meantime, I also have work to do, period – as in, I’m on deadline for this piece. And I regret to inform you that as I write these words, I am indeed smoking.

*From the article here :
 
shulgins-2.jpg



"On the precipice of a mental health treatment revolution"*

by Lucas Richert, Ph.D. | Psychology Today

In early 2014, Scientific American called for further inquiries into certain psychedelic drugs. It also criticized American drug regulators for limiting access to LSD (Lysergic acid-diethylamide), ecstasy (MDMA), and psilocybin.

Then, a few months afterward, Science described how scientists were rediscovering psychedelics – both as objects of basic research and as therapeutic applications. The basic thrust was that “more and more researchers are turning back to psychedelics” to treat depression, obsessive-compulsive disorder, various addictions, and other categories of mental illness.

One psychiatrist, Ben Sessa, has christened this moment a psychedelic renaissance. It’s not the first time commentators have used the term. And he isn’t alone in applying that label to the present. Such sites as Vice, The New Yorker, and Psychology Today have all used the phrase. It has become increasingly common.

This past July, Matt Lamkin (again in Scientific American) suggested that psychedelic medicines were definitely coming down the pike but the law itself wasn’t ready. “The prospect of psychedelic drugs gaining approval as treatments,” he argued, “will force a reckoning for our existing system of drug control.”

An even more enthusiastic writer suggested that “we are on the precipice of a mental health treatment revolution, and the ball’s already rolling.”

The ball is certainly rolling but it’s worthwhile reckoning with the history and interpretation of such psychedelics. Biographies of drugs, just like people, can evolve. It has simply taken time.

Such organizations as MAPS and the Heffter Research Institute, had formed partnerships with laboratories based in the U.S. as well as Switzerland, meaning psychoactive drugs had stronger institutional champions.

The subsequent advance of the psychedelic research agenda can also be partially explained by a change in personnel at the FDA to a newer generation of regulators, according to David Nichols, an emeritus professor of pharmacology at Purdue University and a co-founder of the Heffter Research Institute.

Psychedelic medicine aged – and got more serious. “The psychedelic revival” as Andrew Brown, writing in The Spectator named it, grew apart from the “wackier end of the pro-LSD lobby.”

A full-blown comeback?

For all the progress that LSD has made in the past decade, though, there have been limited scientific gains and, as yet, no marketable prescription drugs. Many questions remain.

Still, researchers are now examining LSD as a treatment for alcoholism, post-traumatic stress disorder, cluster headaches, and as a supplement to palliative care. Neuroscience, as you’d expect, is more involved than in previous studies.

The literature concerning LSD and other psychedelics has undoubtedly grown over the past decade. In very recent years, numerous scholars including Patrick Barber, Alexander Dawson, Erika Dyck, Mike Jay, Matt Oram, and others have explored psychedelic medicines. That includes me, too. Meanwhile, Michael Pollan has produced a bestselling account of psychedelic science.

The histories are evolving. Mike Jay’s new book, Mescaline, forces readers to think about the cultural and biomedical elements of the cactus-derived drug. And he wonders why historians and others have given mescaline so little attention. Erika Dyck asks why psychedelics stories are so short on women and people of color. Dawson’s analysis of peyote weaves ethnicity and Mexican identity into the evolving story.

Dr. Hofmann’s “problem child,” then, has reemerged and is being reevaluated in a new political, economic, and social context. Could we consider LSD a turbulent teen? Perhaps in mid-life crisis? Perhaps. To be sure, LSD has certainly matured, according to Nicolas Langlitz in his book Neuropsychedelia: The Revival of Hallucinogen Research since the Decade of the Brain. It has grown “from wonder and shame to inquiry.”

This is crucial. Yet, we should also be aware that the psychedelic renaissance is broad, loaded with nuance, and about so much more than LSD. There are going to be many more shocking twists.

*From the article here :
 
Last edited:
photo-1535037532858-83180dabddb0



Why it's so hard to find mental health counseling right now

by Lenny Bernstein | Washington Post | 6 Mar 2022

Angelle Haney Gullett lost her father in September and knew she would need grief counseling. She contacted 25 therapists in the Los Angeles area, where she lives, between early October and Christmas, neatly tracking her efforts on a spreadsheet.

None would accept a new client. In most cases, their waiting lists were closed as well, even though Gullett was willing to pay hundreds of dollars in cash for each session. She spent February’s Super Bowl in tears, watching the Cincinnati Bengals, the team her father rooted for.

“I’m in a big city. I’m in L.A. We have a lot of therapists,” she said. “So it’s just kind of wild to me that that many people are at capacity.”

It has been difficult to find mental health counseling in much of the United States for years, long before the coronavirus pandemic began. But now, after two years of unrelenting stress, turmoil and grief, many people seeking help are confronting a system at or beyond capacity, its inadequacy for this moment plainly exposed.

It is even more difficult to find specialized care for children or those with lower income. Assistance of any kind is in short supply in rural areas, where all health-care choices are more limited than they are for residents of cities and suburbia. Those hoping to find a Black or Latino therapist face even more limited options.

While all of those circumstances have long been true, the pandemic has significantly worsened conditions, according to mental health practitioners, officials at professional associations, people seeking care and a wide variety of data.

“It’s the worst it’s ever been,” said Kelly Roberts, director of Graduate Programs in Human Sciences at Oklahoma Christian University in Edmond. “I’ve never seen it like this.”

The pandemic has caused nearly two years of collective trauma. Many people are near a breaking point.

At Boston Medical Center, the safety net hospital for the city, staff recently began contacting parents of children who joined the 170-person wait list in April 2021 — a 10-month wait for a chance to receive services, said Christine M. Crawford, a child psychiatrist at the center who is also the associate medical director for the National Alliance on Mental Illness.

At a 20-practitioner branch of the nonprofit agency OhioGuidestone in Columbus, the wait list now holds 150 to 200 names, nearly double the norm.

The federal government’s mental health and substance abuse referral line fielded 833,598 calls in 2020, 27 percent more than in 2019, before the pandemic began. In 2021, the number rose again, to 1.02 million.

When the American Psychological Association surveyed its members last fall, it found a surge in demand and new referrals, particularly for anxiety, depressive and trauma-related disorders. Yet 65 percent of the more than 1,100 psychologists who responded said they had no capacity for new patients and 68 percent said their wait lists were longer than they were in 2020.

In December, U.S. Surgeon General Vivek H. Murthy issued an advisory on “the urgent need to address the nation’s youth mental health crisis.” And on Feb. 1, a Senate committee held a hearing to address the nation’s growing mental health and substance abuse problem.

“Mental health problems often lag a stressor,” said Mitch Prinstein, chief science officer of the American Psychological Association, which represents 133,000 psychologists, researchers, educators and others. “This is not a surprise.”

Prinstein and 35 colleagues predicted the current situation with remarkable clarity in a “call to action” in the American Psychologist journal in August 2020.

“The toxic psychosocial stressors that the pandemic has created (e.g., physical risks, daily disruptions, uncertainty, social isolation, financial loss, etc.) are well known to affect mental health (and thereby also physical health) adversely, and collectively encompass many characteristics that have been identified as having the greatest negative effects,” they wrote.

It is nearly impossible to generalize about the state of the entire mental health profession in a society as vast and varied as the United States. Hundreds of thousands of specialists work in a wide variety of settings, including schools, hospitals, private companies, nonprofit agencies, free-standing clinics, outpatient settings and private offices.

Traditional one-on-one talk therapy is offered by practitioners with significantly different levels of training, including clinical psychologists, who have doctoral degrees; psychiatrists, who hold medical degrees and can prescribe medication; clinical social workers, marriage and family therapists, licensed professional counselors and others.

The frustration some people encounter when trying to book time with one of them points to the serious consequences of the two-year-old pandemic and a mental health system that simply isn’t built to handle current demands.

In Avon, Ind., 24-year-old Rowan Welch, a team leader for a bank, said he has contacted every provider within 50 miles on his insurance plan’s network without success. “I am still looking, after looking in a 50-mile radius,” Welch said. “There’s not much more I can do.”

In New York, Flora Stamatiades, a gig worker who relies on insurance she purchased through the state’s exchange, said she has been looking for a therapist on and off for more than a year. She is using a list provided by the Actors Fund, which is available to her through her job as a covid safety manager for a show now in rehearsal. She is on a list for a possible opening this spring.

“It never, ever occurred to me that I would not be able to find a therapist when I needed one,” said Stamatiades, 55, who left her job with the Actors’ Equity Association in 2018 after 24 years. “I might have to wait a few months. That occurred to me. That would have been okay."

“This is not actually a new problem,”
she added. “It’s just desperate now.”

And Toni Powell, an associate director of clinical quality for UnitedHealthCare in Austin, said she has contacted about 50 providers, online and by telephone, without finding one who can help with the behavioral problems her 5-year-old granddaughter is experiencing. Powell is raising the child, who she said suffers significant impairment from exposure to alcohol in the womb, cystic fibrosis, trauma and other conditions.

The child needs to be seen in person, an obstacle to finding therapy from providers who largely moved online when the pandemic began, she said. Powell recently began meeting with one specialist online, in the hope that the practitioner will see her granddaughter in person in the next few months.

“There are not enough [providers] or their specialty does not include what you need for your kid,” she said. “And then of course the pandemic.”

Gullett, a 47-year-old screenwriter, also wanted to speak with a counselor in person, which substantially narrowed her chances of finding someone. Now she is competing with an untold number of people who see the chance to emerge from the pandemic and its impact on their psyches.

“We’re all living through a mass trauma event, and that includes therapists, so their capacity to help is understandably impacted,” she said. “And so many more people need access to care.”

Unable to find a counselor, she has turned to self-care: yoga, journaling, books on grief and the online apps that have sprung up as stopgaps. Los Angeles County offered residents free subscriptions to one. She believes other people may find help in support groups, even ones offered online. But for her, the alternatives have been insufficient.

“I just really need some guide rails to go through this process,” she said, “and that has not been available.”

In interviews, therapists and experts cited the obvious causes of the backlog: the psychological and emotional toll of long-term disruption during the pandemic, prolonged uncertainty about the future, fear of infection, financial stress from lost jobs, grief over the deaths of loved ones, loss of social interaction and, especially for children, the unnatural circumstance of isolation at home.

But they also noted other factors. People who might never have sought therapy are now more willing to consider it in the wake of the pandemic, led by a younger generation that sees less stigma in acknowledging mental illness, said Saul Levin, chief executive of the American Psychiatric Association, which represents more than 37,000 psychiatrists.

“Covid in some ways exposed mental illness to the general public that the majority always ignored,” Levin said. “People have been accessing mental health care a lot more now because of covid.”

Yet insurance coverage and training slots for new psychiatrists lag far behind the demand, Levin said. In his State of the Union address last week, President Biden proposed a large increase in mental health providers. In a fact sheet, the White House noted that more than one third of Americans live in designated mental health professional shortage areas.

“We must dramatically expand the supply, diversity, and cultural competency of our mental health and substance use disorder workforce — from psychiatrists to psychologists, peers to paraprofessionals — and increase both opportunity and incentive for them to practice in areas of highest need,” the White House said.

"Online therapy, a boon to patients and clinicians, especially in rural areas, has dramatically reduced no-shows and dropouts by offering the convenience of counseling at the touch of a button. This has cut into many counselors’ availability to accept new clients," experts said.

Some patients also need more time to reach their goals now, said Roberts, the Oklahoma therapist, which also delays the opening of new slots.

“It’s like the boxcars are all crashed and piling up behind, and everyone is trying to figure out how to get them back on track,” she said.

D. Giovanni Scott, a private practitioner in The District, noted that people gave up insurance and access to employee assistance programs when they lost or left jobs during the pandemic. To accommodate demand, she said, she offers a few people the option of biweekly therapy sessions when it is appropriate, and she tries to retain some insurance-only clients despite the low reimbursement rate. She keeps her waiting list short, unwilling to offer unreasonable hope to people seeking an opening.

Scott, who is Black, and others said racial injustice and 2020′s protests against police violence have also sent more people to their doors.

Crawford, the Boston child psychiatrist, said the return to school has allowed teachers and others to assess the needs of children who have been seen only by their parents in recent months. The result has been a large surge in referrals to a mental health workforce that has not expanded.

“Other adults and teachers are able to lay eyes on these kids,” she said, and they are “bringing up concerns about behavioral issues and emotional issues.”

According to the American Academy of Child and Adolescent Psychiatry, there are just 8,300 practicing child psychiatrists in the United States for the more than 15 million young people who need them. Pediatricians are doing initial assessments, Crawford said, with only the most severe cases reaching her. Children considered a danger to themselves or others are routinely held in hospital emergency rooms for a week or two waiting for beds in appropriate settings, she said.

Some providers hope the crisis will be used to overhaul a mental health system that was created after World War II and has always been crisis-oriented, by infusing mental health awareness and techniques into schools, the workplace and other parts of society. "Simply doing more of the same won’t work," Prinstein said. "Biden’s plan contains some elements of revamping the system."
And there is no doubt that the United States needs more practitioners, experts said.

“The nation is in the middle of a mental health epidemic, and it is up to behavioral health agencies to find solutions that will attract and retain employees who can provide access to quality mental health care,” Brant Russell, president and CEO of OhioGuidestone, the nonprofit agency, said in a statement.

But it won’t be soon enough for Gullett and others searching for help today. “Most people have given up. Nobody is looking right now that I’m aware of,” she said. “There seems to be no expectation that it’s going to change.”

Lizzy Raben and Andrew Van Dam contributed to this report.


*From the article here :
 
Last edited:
1*4aSCBwPIh0RM0-w19nmVgQ.jpeg


AI maps psychedelic experiences to regions of the brain, opening new route to psychiatric treatments*

The Conversation | Neuroscience News | 20 Mar 2022

For the past several decades, psychedelics have been widely stigmatized as dangerous illegal drugs. But a recent surge of academic research into their use to treat psychiatric conditions is spurring a recent shift in public opinion.

Psychedelics are psychotropic drugs: substances that affect your mental state. Other types of psychotropics include antidepressants and anti-anxiety medications. Psychedelics, however, are unique in their ability to temporarily induce intense hallucinations, emotions and disruptions of self-awareness.

Researchers looking into the therapeutic potential of these effects have found that psychedelics can dramatically reduce symptoms of depression and anxiety, PTSD, substance abuse and other psychiatric conditions. The intense experiences, or “trips,” that psychedelics induce are thought to create a temporary window of cognitive flexibility that allows patients to gain access to elusive parts of their psyches and forge better coping skills and thought patterns.

Precisely how psychedelics create these effects, however, is still unclear. So as researchers in psychiatry and machine learning, we were interested in figuring out how these drugs affect the brain. With artificial intelligence, we were able to map people’s subjective experiences while using psychedelics to specific regions of the brain, down to the molecular level.

ai-psychedelics-neuroscinecss-public.jpg


Mapping ‘trips’ in the brain

Every psychedelic functions differently in the body, and each of the subjective experiences these drugs create have different therapeutic effects. Mystical type experiences, or feelings of unity and oneness with the world, for example, are associated with decreases in depression and anxiety. Knowing how each psychedelic creates these specific effects in the body can help clinicians optimize their therapeutic use.

To better understand how these subjective effects manifest in the brain, we analyzed over 6,000 written testimonials of hallucinogenic experiences from Erowid Center, an organization that collects and provides information about psychoactive substances.

We transformed these testimonials into what’s called a bag-of-words model, which breaks down a given text into individual words and counts how many times each word appears. We then paired the most commonly used words linked to each psychedelic with receptors in the brain that are known to bind to each drug.

After using an algorithm to extract the most common subjective experiences associated with these word-receptor pairs, we mapped these experiences onto different brain regions by matching them to the types of receptors present in each area.

We found both new links and patterns that confirm what’s known in the research literature. For example, changes in sensory perception were associated with a serotonin receptor in the visual cortex of the brain, which binds to a molecule that helps regulate mood and memory. Feelings of transcendence were connected to dopamine and opioid receptors in the salience network, a collection of brain regions involved in managing sensory and emotional input. Auditory hallucinations were linked to a number of receptors spread throughout the auditory cortex.

Our findings also align with the leading hypothesis that psychedelics temporarily reduce top-down executive function, or cognitive processes involved in inhibition, attention and memory, among others, while amplifying brain regions involved in sensory experience.

Why it matters

The U.S. is going through a profound mental health crisis that has been exacerbated by the COVID-19 pandemic. Yet there have been no truly new psychiatric drug treatments since Prozac and other selective serotonin reuptake inhibitors, the most common type of antidepressants, of the 1980s.

Our study shows that it’s possible to map the diverse and wildly subjective psychedelic experiences to specific regions in the brain. These insights may lead to new ways to combine existing or yet to be discovered compounds to produce desired treatment effects for a range of psychiatric conditions.
Author: Galen Ballentine and Sam Friedman
Source: The Conversation

*From the article here :
 
Last edited:
healthcare-research-mushrooms.jpg


Can psychedelics provide a breakthrough in mental health care?*

by Ben Hargreaves | PharmaPhorum | 2 Mar 2022

There is a growing pipeline of psychedelic therapies and an expanding body of science to suggest that they may be able to provide an alternative to existing mental health treatments. Ben Hargreaves discovers why medical research in the area has shifted from being highly controversial to being backed by major pharma companies and venture capital.

The image of psychedelic drugs within medical research is changing. Although the development of LSD began with Albert Hofmann of Sandoz Laboratories and saw extensive research in the 1950s, the drug became closely associated with the counterculture movement of the 60s and Timothy Leary’s call to “turn on, tune in, drop out.” Following this period, the study of psychedelic treatments continued but at the fringes of medical research.

But today this is rapidly changing as a greater number of companies and research institutes explore potential uses for mental health disorders. One reason behind the resurgence in interest is a small but growing body of clinical research, with various psychedelic treatments showing promising outcomes in treating a range of medical conditions, such as depression and addiction.

The term ‘psychedelic treatments’ incorporates a broad spectrum of different compounds, from LSD, psilocybin and DMT, to MDMA, ibogaine and ketamine. The latter compound has already been approved for treatment-resistance depression as the product Spravato (esketamine), after being developed by Janssen. This success has seen other traditional pharma companies, such as Otsuka Pharmaceutical, take a greater interest in the space and spurred the investment required to progress the field further.

A psychedelic renaissance

One of the reasons that interest has developed again in psychedelic therapies is the crisis being faced in mental health disorders globally. According to the World Health Organization, each year 25% of individuals in Europe will suffer from depression or anxiety, with neuropsychiatric disorders accounting for 26% of the burden of disease in the European Union. It is a similar story in the US, with the National Institute of Health estimating that nearly one in five adults lives with a mental illness.

A spokesperson for atai Life Sciences, a biotech developing a range of compounds to treat mental health conditions, told pharmaphorum that the challenge of treating mental health issues had brought about a ‘psychedelic renaissance’ in the scientific community. They noted that this represented “a significant shift from half a century ago when legislative rulings limited research”, with this new era for the drugs “marked by an increase in research, education and growing understanding of the potential of these compounds.”

Similarly, Dr. Dan Karlin, chief medical officer at MindMed, another biotech working in the psychedelic research space, outlined to pharmaphorum: “in engaging with the scientific and medical communities, whether it’s to build out our scientific advisory board or to hire senior drug developers, I’ve been pleasantly surprised at the prevailing open-mindedness towards the work we’re pursuing.”

The reason research interest is high in the area is because psychedelic therapies are achieving strong results in the area of mental health. Compass Pathways, a biotech that is progressing a psilocybin-based therapy, COMP-360, through clinical trials, recently published results showing that patients taking a higher dose version of its treatment demonstrated a -6.6 difference on the MADRS depression scale against a 1mg comparator dose at three weeks. A recent John Hopkins Medicine study showed that treatment with psilocybin relieved major depressive disorder for at least a year for certain patients.

A growing pipeline

With promising pre-clinical and clinical stage results, there are more and more therapeutic candidates being developed in this space. A recent publication showcased that the number of active clinical trials running that are researching various psychedelic therapies has increased rapidly from 2003 to 2021. The number of active clinical trials studying psilocybin alone has increased more than three-fold from 2017.

However, Dr. Guy Goodwin, chief medical officer at Compass Pathways, told pharmaphorum that some caution is required when analysing smaller scale studies that attract media attention. He explained that although there have been a number of ‘high profile’ publications about the use of psychedelics, “they are not the kind of studies that would gain regulatory approval.”

He continued to say, “In other words, they are small and have few safeguards against bias. We need larger, controlled studies to generate rigorous evidence.” By contrast, Compass Pathways’ recently completed phase 2b trial was the largest study into the therapeutic benefits of psilocybin yet undertaken.

Trials of this scale have been made possible by the increased investment flowing into the area. In 2020, an investment fund was created for the first time in the UK to support the development of psychedelic treatments. The founders of this VC fund had previously backed atai Life Sciences, which itself is a part owner of Compass Pathways. All three biotechs previously mentioned, atai Life Sciences, MindMed and Compass Pathways, launched IPOs on the Nasdaq recently – with the two former companies listing in 2021, and the latter in 2020.

Paving the way to patients

There is more research and more money than ever going into psychedelic therapies, but questions remain about the pathway they will take to reach patients. In the US, many of the psychedelic treatments being studied are Schedule 1 drugs by the Drug Enforcement Administration (DEA), which defines the drugs listed as having no accepted medical use and a high potential for abuse.

Dr. Karlin explained why this could create barriers to be overcome by the companies working in the space: “When the FDA approves a new drug it has to be rescheduled by the DEA. But, as MAPS notes, 23 states plus the District of Columbia do not automatically reschedule just because the DEA does, creating potential delays and confusion in the rollout of treatments.”

However, when asked about the potential barriers in this area, Dr. Goodwin stressed the major challenges to getting the treatments to patients will be to generate the evidence and the data required for approval. From that point, the companies involved will have to “demonstrate the value to patients and health systems”, so that the treatments will be reimbursed and accessible to patients. With GW Pharma previously achieving approval for a cannabis-based treatment, despite the DEA noting cannabis as a Schedule 1 drug, there is precedent for taking forward a therapeutic candidate with a similar classification. The challenge, as noted by Dr. Goodwin, will be to prove that psychedelic therapies can be considered safe and effective. With more treatments entering later stages of clinical trials, the answer to these questions could be arriving sooner rather than later – though more than 80 years after Albert Hofmann first synthesised LSD and began wondering about its potential for psychotherapeutic use.

*From the article (including references) here :
 
Last edited:

Researching Psychedelics for Psychiatric Disorders​




Frederick S. Barrett, PhD, is affiliated with the Center for Psychedelic and Consciousness Research at Johns Hopkins University.

Dr Barrett spoke with Nick Andrews at TEDMED about the research that has been conducted by his center on the impact of psychedelics, or hallucinogens, on psychiatric disorders.

Take-home points:

  • Barrett transitioned into neuroscience research through his interest in the effect of music on human emotions and the brain.​
  • Up until 1970, psychedelics were widely used in clinical research, with over 1000 academic papers published about their use. For example, psychedelics were used as a model for schizophrenia and helped identify the role of serotonin in psychosis. They were also studied to treat addiction and as a treatment for existential anxiety in cancer. In 1971, psychedelics were declared illegal under the United Nations Convention on Psychotropic Substances.​
  • Dr Roland Griffiths and a group at Johns Hopkins have led the way in re-establishing clinic research using psychedelics.​
  • In the next 10 years, Barrett would like to have a clear understanding of the effect size of psychedelics on mood and substance use disorders.​
  • Psychedelic agents have a novel therapeutic quality: Studies support that a few or even one exposure to a psychedelic compound, which has a short-term biological effect, leads to a long-lasting therapeutic effect such as remission of mood disorder or change in personality characteristics. The clinical outcomes are mediated by the intensity of the psychedelic experience.​
Summary:
  • The Center for Psychedelic and Consciousness Research is working to discern which medical indications have the most promise of being treated with psychedelics. Their goal is a balanced and rational approach to psychedelic research, and subsequent treatment considering the societal and political contexts around these drugs.​
  • Barrett trained in music education and psychology, and has been a musician all his life. He moved into neuroscience during graduate school and used music as a tool to study emotions and the brain.​
  • Music, meditation, and psychedelics have a similar flow component that inspires converging research questions, as well as a desire to analyze the brain and understand this experience that is central to consciousness.​
  • Music is fundamental to the human experience, and it's exciting to describe the neural circuitry of how music affects the brain and emotions.​
  • Music is useful in therapy because it can regulate emotions. There has long been an overlap of the use of psychedelics and music in therapy. A prime example of this is Guided Imagery and Music (GIM), a specialized form of therapy that arose from work done by Helen Bonny, who developed a protocol for using music to regulate emotions during psychedelic experiences.​
  • It will be interesting to see whether (and how) psychedelics are efficacious in treating an array of substance use disorders. If effective, they would be a single-use treatment for addiction to substances that interact with diverse neural circuits.​

 
UW-to-trial-psyilocybin-for-frontline-healthcare-workers_Banner.jpg


Frontline healthcare workers to receive psilocybin therapy*

A small, Seattle-based study will look to see if the psychedelic can alleviate the pandemic’s mental health impact.

by B. David Zarley | Freethink | 20 Dec 2021

The University of Washington School of Medicine has begun to enroll frontline healthcare workers in a small study testing the effects of psychedelic-assisted therapy for depression and anxiety.

The pandemic has taken a toll, physically and psychologically, on workers in an already stressful profession.

“We’re looking for doctors, nurses and advance-practice providers who have clinically significant symptoms of depression and anxiety,” lead investigator Anthony Back said. “We hope to help them address their feelings of grief, inadequacy and moral distress as a result of caring for COVID patients.”

Subjects will be broken into placebo and psilocybin groups, with all getting therapy sessions with specially trained therapists, as well, in the first study of psilocybin-assisted therapy for clinicians.

“We really need to explore what the things are we can do to change up the mental well-being of our populace,” Back told Seattle’s KING-TV.

“I think we’re totally at a tipping point.”

hero-visit2.jpg


A second pandemic: Frontline healthcare workers had high rates of mental health issues before the pandemic, Jessi Gold, an assistant professor of psychiatry at Washington University in St. Louis, told CNN.

“Health care workers are not people who had good mental health before Covid,” Gold said. “It’s not like Covid came and all of a sudden we’re having problems. We had longstanding problems.”

But the pandemic has exacerbated the psychiatric struggles of a career already rife with burnout and anxiety.

A national poll by the Kaiser Family Foundation (KFF) and the Washington Post found that a majority of frontline healthcare workers say that the stress and anxiety brought on by the pandemic’s relentless march has negatively impacted their mental health; 30% have received, or feel they need, mental health treatment.

The survey also found medical staff were suffering from lack of sleep, frequent head and stomach aches, and are consuming more alcohol. A survey by Mental Health America found emotional exhaustion to be their most common feeling.

John Hick, a doctor at Minnesota’s Hennepin Healthcare, told reporters that the experience is like “drinking from a fire hose.” And to top it all, clinicians are encountering more hostility from patients and family members.

“Now people are yelling at you and telling you that you don’t know anything,” Gold told CNN. “So it’s like a fire hose in a setting that hates you.”

“What I hear my colleagues say is, ‘I don’t think I can keep doing this,’”
UW’s Back said.
Previous studies have shown potential for the use of psilocybin treatment for depression, taken in controlled conditions and and with therapy.

Psilocybin vs. SARS-2: Previous studies have shown potential for the use of psilocybin treatment for depression, taken in controlled conditions and in addition to therapy.

The UW trial, first announced early this year, will be a small one, enrolling only 30 volunteers, split into placebo and psilocybin groups. A research-backed questionnaire will confirm their depression symptoms, and be administered again at the end of the study to assess the drug’s impact.

All of the participants will have two 90-minute sessions with therapists to gain a rapport and learn about their upcoming experience, preparation work researchers have found to be important in psychedelically-assisted therapies.

On their third visit, volunteers will either receive synthesized psilocybin or a placebo — placebos being one of the trickier aspects of studying psychedelics, since patients often can tell after the fact what they received.

After their trip (or not), both groups will have three more sessions to discuss their experiences and how to apply them — what psychedelic therapists refer to as “integration.”

There is a feeling in the psychedelic world that these new treatment methods are not just exciting but needed. And there may be few people more in need of mental health help right now than healthcare workers.

*From the article here :
 
MHAM-psychedelics-therapy.png



Psychedelic-assisted therapy is transforming the way we think about mental health care

How mushrooms, LSD, MDMA, and ketamine could help treat mental illnesses.

by Stav Dimitropoulos | SELF | 24 May 2022

Since the 1970s and the barrage of scientific studies that have explored the therapeutic effects of psychedelics, the mind-bending substances have exhibited immense promise in transforming how we think about mental health care, most notably when it comes to understanding how people can heal from depression, anxiety, addiction, and post-traumatic stress disorder (PTSD).

These substances, broadly thought of as psychedelic drugs, include LSD, psilocybin, and MDMA. All three of them are Schedule I drugs in the U.S., which is the federal government’s most restrictive classification for drugs, indicating that they have a high potential for abuse and no accepted medical uses. Not only does this scheduling make these drugs illegal at federal level, but it has also historically restricted the research that experts can do with them.

But now the global scientific community is going through something of a “psychedelic renaissance.” And research continues to show that, when used in controlled therapy settings, these substances have the potential to profoundly alter the future of mental health care.

What does psychedelic therapy actually look like?

“We have had success in demonstrating that psychedelics tackle depression as well as the mental distress associated with life-threatening illnesses, addictions, and more,” Alan Davis, PhD, director of the Center for Psychedelic Drug Research and Education at Ohio State University, tells SELF.

In a 2022 study published by Dr. Davis and his colleagues in the Journal of Psychopharmacology, 24 people with moderate to severe depression received two doses of psilocybin, spaced two weeks apart, in combination with supportive talk therapy, either immediately after a screening or after an eight-week wait. The researchers followed up with the participants intermittently over a year following their second dose—and the results were exciting: 58% of the people in the study achieved remission from their depression, while 75% of the people in the study responded to the treatment with dramatically-reduced depression symptoms. “This is pretty remarkable considering that the best treatments we have right now require daily medication for the rest of your life or extensive psychotherapy, and even then they’re not as effective,” Dr. Davis says.

This research follows a similarly-designed November 2020 study from some members of the same scientific team. The findings were equally promising: After everyone received psilocybin-assisted therapy, 71 percent of the participants (17 people) were in remission from depression four weeks later.

While both of these studies are encouraging, it’s worth noting that they include relatively small sample groups (although they are generally on par with other studies in this area), and further research that includes diverse groups of people will be needed to truly understand the benefits of psilocybin-assisted therapy, as well as other psychedelic-assisted therapies, on a larger scale.

With that said, in both experiments the participants didn’t just take a capsule and watch their depression vanish into thin air. The interventions outlined a model for a psychedelic-assisted therapeutic approach, which requires a structured and supervised setting and well-trained providers.

In the 2022 study, for example, participants were required to first have at least six hours of preparatory meetings with two trained credentialed facilitators. After that they underwent two day-long sessions with psilocybin administration, two weeks apart. During the sessions, in which a participant lies on a couch with headphones on while listening to music, the facilitators are on hand to respond to their needs. After the sessions the participant has another two to three hours of follow-up meetings with the facilitators.

The patients in the 2020 study also received a total of 11 hours of supportive psychotherapy alongside the drug treatment to make sense of the experience and move forward with their lives. The entire treatment process lasted about eight weeks. This protocol is similar—but not totally identical—to that of other psychedelic-assisted therapy studies. But what they all have in common is extensive access to support before, during, and after the drug experience.

How does psychedelic therapy work?

For many people antidepressants can be an effective way to treat the symptoms of depression, various forms of anxiety, and other mental health issues, especially when used alongside talk therapies. But these medications often need to be taken for years or even a person’s entire lifetime, and they can come with a risk of potential side effects, such as sleep issues, agitation, loss of appetite, and a reduced desire for sex, among others.

Psychedelic-assisted therapy appears to be effective in the long term after a few short, intense weeks to months of treatment; some research suggests therapeutic effects can begin after a single session. These drugs can cause side effects, too, such as nausea, increased heart rate, and hallucinations (depending on the substance). But when the drugs are used in this short-term way, those side effects generally occur during or shortly after the drug experience, which is one reason why supervision and support during that time are so essential.

Like antidepressants, psychedelics have effects on specific neurotransmitters, including serotonin. But experts think psychedelics’ effectiveness is also due to their ability to reconfigure our neural architecture. “For instance, brain imaging studies show that, over time, neurons in the brain’s prefrontal cortex atrophy in people with depression. But the controlled use of psychedelics may actually change this,” Linda Strause, PhD, professor of human nutrition at the University of California San Diego and an executive in the drug development industry, tells SELF. "Psychedelics may stimulate the growth of new neurons as well as new connections between them, which suggests they may be able to help treat the symptoms of depression, PTSD, and other mental health issues," she says.

“Hallucinogenic substances also have the ability to reset our default mode network (DMN) immediately and for long periods of time,” David J. Mokler, PhD, a neuropharmacologist, professor emeritus at the University of New England, and advisor at psychedelic biotechnology company Havn Life, tells SELF. "The DMN is a network of connected parts of the brain that are active when you’re not specifically thinking about or working on anything, such as when you’re daydreaming. Altering the way the DMN functions seems to help long-term with depression, particularly the depression that doesn’t seem to respond to the classic treatments we usually give,” Dr. Mokler says.

These biological changes likely reflect the psychological—and sometimes spiritual—changes that can occur during the psychedelic experience.

“When people take psychedelics, they often have spiritual experiences and a new awareness of their emotions, relationships, and career, and it is this combination that seems to bring the most results,” Dr. Davis says. "In this way, psychedelic therapy can offer someone the chance to experience a radically different perspective on their life, one that might help them realize important aspects of their environment, relationships, or thinking patterns that could be contributing to their depression, for instance."

However, it’s crucial to understand that it’s not just the drugs that are likely to be helpful; the follow-up sessions with facilitators are also important so that patients can “integrate” their experience, as some researchers put it, and draw productive meaning from what they explored while in their psychedelic state.

Various drugs have psychoactive properties—but which ones hold the most therapeutic potential?

Ketamine

Some experts hesitate to classify ketamine as a genuine psychedelic, Dr. Davis included. Ketamine is an injectable, short-acting anesthetic that comes in a clear liquid or white powder, but it’s lumped in with psychedelic drugs due to its possible hallucinogenic effects. That’s why it’s sometimes referred to as a “dissociative anesthetic”—it can warp sensory perceptions like sight and sound and make a person feel disconnected from their surroundings or detached from feelings like pain.

Researchers theorize that ketamine influences the activity of glutamate, an abundant neurotransmitter in the central nervous system that is thought to have significant effects on various health conditions, including mood disorders like depression, but more research is needed to understand the correlation.

As a Schedule III non-narcotic substance, ketamine is the only drug under the psychedelics umbrella with legalized, medically-accepted use at the time of publication. The U.S. Food and Drug Administration (FDA) first approved esketamine, a ketamine-based nasal spray, for treatment-resistant depression in 2019. That’s the caveat: It’s only been approved for that specific mental health condition. Ketamine-assisted therapy used to treat other mental health issues—such as PTSD, which often triggers depression—would be considered “off-label” use by the FDA. Off-label means a health care provider prescribes an approved drug to treat a condition it was not specifically approved to treat, prescribes the drug in a different dosage than what has been approved, or administers the drug in a different way than it has been approved. While ketamine’s off-label use concerns some experts for various reasons, including patient safety, the practice continues to be incredibly common in the medical community as a whole—one in five prescriptions are considered to be off-label use, according to the Agency for Healthcare Research and Quality.

Currently, ketamine is the focus of at least 99 studies for the treatment of mental health conditions with a heavy focus on depression and pain-related health issues like chronic headaches. Emerging research continues to explore ketamine’s therapeutic potential for conditions like PTSD, social anxiety, and obsessive-compulsive disorder as well—all of which hold potentially exciting developments for ketamine’s future use.

MDMA

MDMA, which is classified as both a stimulant and a psychedelic, became popular in the rave and electronic dance music culture of the 1980s and 1990s. It’s commonly sourced from safrole oil, an essential oil derived from the sassafras tree, which is an aromatic tree endemic in North America, and it’s most commonly distributed in tablet form.

The drug’s signature effects are feelings of emotional connectedness, closeness, warmth, and empathy, which tend to last for three to six hours. MDMA can also cause an increased heart rate, sweating, blurred vision, restlessness, and (in rare cases) a life-threatening increase in body temperature.

Much of the research looking at the therapeutic potential of MDMA-assisted therapy comes from MAPS, a nonprofit research and educational organization founded in 1986 that is committed to furthering the understanding of psychedelics and marijuana; it is known for its MDMA-assisted therapy training program for eligible mental health professionals. In 2021, MAPS finalized the first Phase 3 trial of the use of MDMA-assisted psychotherapy for people with PTSD. The study showed that over 67% of the participants who received MDMA-assisted therapy no longer met the criteria for PTSD. The organization’s protocol received a Breakthrough Therapy designation from the FDA in 2017, meaning the treatment has shown the potential to be more effective compared to current medications used to treat severe PTSD.

While most of the clinical trials focused on MDMA are further exploring the drug’s impact on people with PTSD, experts are also investigating its potential in treating other mental health issues, such as anxiety associated with a life-threatening illness.

Psilocybin

Hallucinogenic magic mushrooms have an ancient history. Even Stone Age artists in Africa and Europe, pre-Columbian sculptors across North, Central, and South America, and ancient populations of the Sahara desert depicted what experts believe to be mushrooms in their art.

Today, psilocybin, the chemical compound derived from certain dried or fresh hallucinogenic mushrooms, has been both decriminalized and legalized for therapeutic use in Oregon only. Starting in January 2023, the state will make the drug available in qualified settings with the oversight of a trained physician and therapist, Dr. Strause says.

Researchers believe that, like other psychedelics, psilocybin mingles with a certain serotonin receptor (5-HT2A), which sets off a “mystical-like hallucinatory effect.” Psilocybin’s effects typically hit within 20 minutes and last for about six hours, and the drug can cause mood changes and visual and auditory hallucinations, as well as nausea, vomiting, sleepiness, and muscle weakness. Large doses, in particular, may elicit feelings of panic.

In recent investigational studies, researchers have found psilocybin-assisted therapy to be helpful for people with treatment-resistant depression as well as depression and anxiety in people with life-threatening diseases. And in the past few years, the FDA gave two psilocybin-assisted therapy programs Breakthrough Therapy status.

LSD

First accidentally synthesized by Swiss chemist Albert Hofmann in 1938, LSD is a potent, perception-changing synthetic chemical. It’s created from the lysergic acid found in ergot, which is a fungus that infects rye. (And yes, the CIA did investigate LSD as a potential psychological weapon during the Cold War.) An LSD experience can typically last up to 12 hours, in which a person may experience hallucinations, distortions in sensory perceptions and time, and positive or negative changes in mood. It can also cause physical side effects like higher or lower body temperatures, sweating, dry mouth, high blood pressure, or muscle tension, among others.

Researchers have been looking at LSD’s potential to help people with mental health issues since the 1950s, but those studies stalled when the federal government prohibited LSD, largely because it became inextricably linked to the 1960s counterculture. However, recent small studies looking at LSD-assisted therapy have found that it has potential benefits for people dealing with alcoholism and anxiety in people with life-threatening diseases. In January 2022, the FDA cleared a Phase 2(b) clinical trial of a generalized anxiety disorder treatment that uses a “pharmacologically optimized form of LSD.”

Will psychedelic-assisted therapy be accessible in the future?

As research continues, various forms of psychedelic-assisted therapy could eventually reach FDA approval. But like any modern medicine, accessibility will be a huge concern. Here are a few potential challenges the scientific community is grappling with—and how experts are thinking about making these innovative therapies more available to everyone.
Drug scheduling limitations

Most drugs under the psychedelic umbrella still have Schedule I status, which “imposes a ceiling on many policy recommendations,” especially when it comes to federal funding, researchers who authored a 2021 paper in the journal Nature Medicine argue. “Legislation is the biggest obstacle,” Dr. Davis says. “It won’t be until we have FDA approval and/or other legislative changes that we will be able to see advances and the destigmatization of psychedelics.”

Rescheduling drugs isn’t an easy process, but some advocacy groups are trying to work toward just that. Experts especially have their eyes on psilocybin, since it’s naturally derived from fungi, has a low toxicity risk, has a low potential for dependence, and is already decriminalized in several other countries. One global coalition—the International Therapeutic Psilocybin Rescheduling Initiative—is advocating to reschedule psilocybin under the United Nations 1971 Convention on Psychotropic Substances, an international treaty that aims to limit the abuse of certain psychoactive drugs. This rescheduling would help nix psilocybin’s international research restrictions, potentially opening the doors for its medical use. In the U.S., anyone is able to petition the Drug Enforcement Administration to reschedule a particular substance, which may prompt an FDA review of its current state in research.

Potential for gatekeeping

As clinical trials continue to unveil exciting possibilities, some stakeholders are already trying to monopolize psychedelics through patents, both for the compounds themselves and for the therapeutic practices of producing or administering them. This has ignited criticism from medical and legal experts as well as Indigenous communities, some of which have used certain psychedelics for hundreds (possibly even thousands) of years, the authors of the Nature Medicine paper explain. “Restricting patents on psychedelics may be necessary to promote their role in the meaningful advancement of mental health care,” they write. “U.S. law prohibits patents on products of nature…. Some might also think of psychedelics as tools of discovery that should be free to all and reserved exclusively to none.”

Need for professional, standardized training

New and exciting courses related to psychedelic-assisted therapy training, such as the course provided by MAPS, are on the rise—but not without a few challenges. “There are training programs that have started but, because the treatments aren’t FDA approved and because the credentialing for this profession has not been established yet, there is nobody to accredit those programs,” Dr. Davis explains.

Currently that means there are no “standards” for training, he says—which is vital for high-quality administration of any drug. For psychedelics, specialized training will help ensure a consistent therapeutic approach, safe and ethical practices, and culturally sensitive care, one 2021 study that explored a training program for psilocybin-assisted therapy concluded. “Eventually, when the FDA approves these treatments, there will then be a creation of an accreditation body,” Dr. Davis says. “Then there will be an establishment of what constitutes adequate training and expertise.”

Necessary insurance coverage

Those who stand to benefit from psychedelic-assisted therapy the most will likely be those who lack access, possibly due to socioeconomic barriers like insurance coverage. When—not if—these therapies become legal, they’re likely to be very expensive. Estimates for a future course of MDMA-assisted treatment, for example, hover around $15,000, according to a 2021 research paper published in Harm Reduction Journal. Experts theorize that establishing a clear prescription model—meaning psychedelic-assisted therapies would require a licensed prescriber like a physician—could help circumvent this issue. But that roadmap may not be the best approach for everyone as discussions around who should be eligible to administer these drugs continue.

Even with these challenges, one hurdle the scientific community continues to face is lifting the stigma surrounding these drugs. The psychedelic renaissance is really a return to decades-old research and even older traditional spiritual practices. We have a long way to go before they become widely accepted, but we’re getting closer with each clinical trial.

“I believe very strongly that education will lift the stigma and improve access, whatever that looks like, quicker than anything else will,” Dr. Strause says. “And I do believe that the word is getting out.”

 
mRfAMf6.jpg



Psilocybin as mental health therapy? Here’s what I found.

Interest in and use of the drug is growing, but much is still unknown.

by Steven Petrow | Washington Post | 5 Sep 2022

A few weeks ago, I mentioned to a friend that I was interested in learning more about psychedelics, especially how they might help me with depression and anxiety. That’s a broad category of plant medicines including psilocybin (“magic”) mushrooms, MDMA (ecstasy), DMT (Dimitri or the Businessman’s Trip), ketamine (“special K”) and some others.

I’d been hesitant to be open about my search, because I’m old enough to remember the warnings about “bad trips” that scramble your brain. Imagine my surprise when my friend told me he’d recently taken his first “trip,” which he described as life-changing.

I asked him — a real estate developer living in Northern California, married with kids — why he decided to try a psychedelic substance. “My work felt increasingly stale and meaningless,” he explained to me over a beer. “Despite a massive amount of reflection and coaching around how to break the rut, I felt as though I was still off track.”

He and the others who have used these medicines spoke on the condition of anonymity because most of these psychedelics are Schedule I substances, meaning they are illegal to manufacture, buy, possess or distribute.

When I confided my interest in psychedelics to a few other friends, several said they had tried the drugs and experienced several benefits: from easing anxiety to finding spiritual insights to combating depression and, among some with cancer, helping to reduce the fear of dying.

They are hardly outliers. According to a new YouGovAmerica study, “one in four Americans say they’ve tried at least one psychedelic drug,” amounting to some 72 million U.S. adults. (The study included the medicines mentioned earlier, plus LSD, mescaline and salvia.) Was I missing a beat by not getting onboard?

When I queried my psychiatrist about participating to help improve my mental health, he was supportive, with two caveats: Do it with a trained therapist or guide, and do your best to ensure that the substance is what it’s said to be.

These days, it’s hard not to see, hear or read about the use of psychedelics, whether it’s Michael Pollan’s best-selling book (and accompanying Netflix documentary) “How to Change Your Mind,” online advertisements for psychedelic spa “trips,” underground therapists (also referred to as “sitters” or “guides”) with websites promising consciousness-expanding journeys, and a DIY online ketamine program — with a medical professional tethered by videoconferencing — that you can do at home. (Ketamine was approved by the Food and Drug Administration in 1970 as an anesthetic/analgesic, which makes it legal to prescribe. For over 20 years, it has been prescribed off-label for depression, anxiety and other mental health issues. A derivative of ketamine, called esketamine — sold as Spravato — was approved by the FDA in 2019 specifically for depression.)

In biggest advance for depression in years, FDA approves novel treatment for hardest cases

Recent clinical trials and studies, which have garnered big headlines, have shown efficacy in treating a variety of conditions, such as depression, addiction, obsessive-compulsive disorder and post-traumatic stress disorder. And an increasing number of studies are underway.

Intrigued but cautious, I wanted to know: How should I approach this in a smart and safe way? I started by interviewing Rick Doblin, founder and executive director of the Multidisciplinary Association for Psychedelic Studies (MAPS). He reminded me that, other than ketamine, none of these generally illegal psychedelics are approved by the FDA, so he would talk only about “minimizing risks.”

“I don’t want people to think that this is like going on a carnival ride,” he said. “There’s always a risk.”

Matthew Johnson, a psychiatry professor at the Johns Hopkins Center for Psychedelic and Consciousness Research who has conducted numerous studies on psychedelics, also spoke to the issue of safety. To that point, Hopkins’s clinical trials screen out those with schizophrenia, bipolar disorder or severe heart disease.

I mentioned that, like millions of Americans, I’m taking an antidepressant (an SSRI or selective serotonin reuptake inhibitor), which he explained would probably mute the effect of psilocybin or MDMA. To partake of psychedelics, he told me I’d want to taper off the SSRI first, which is best done with medical supervision (and which I’ve had trouble with in the past).

I wasn’t suicidal, until suddenly, terrifyingly I was

I also have heart disease, so he cautioned me to speak with my cardiologist (who texted me that he knows nothing about the use of psychedelics). In other words, these drugs are not for everyone.

Johnson reiterated that despite public testimonials about the positive therapeutic effects of psychedelic usage, “there are dangers, and it is illegal.” Was he trying to discourage me? “I don’t encourage anyone to do this on their own,” he said.

After researching this column, I’m not interested in taking this journey on my own. But assuming I’d have a guide or therapist, where would I start?

Doblin suggested that anyone with a “clinical indication” (such as depression, PTSD or anxiety), should go to ClinicalTrials.gov to find and possibly participate in nearby studies. Recently, when I checked the database for “psilocybin” studies in the United States, 67 trials came up. All are being conducted at well-known academic medical centers, which means the studies are done with pure drugs, approved by the FDA and licensed by the Drug Enforcement Administration, which means the trials have regulatory approval.

What about people without a clinical indication, but who are on a spiritual quest? Here are the considerations I gleaned:

Set and setting: Over and over I heard this phrase, which refers to finding a healthy mind-set and a relatively safe environment. For obvious reasons, Johnson called the rooftop of a tall building a bad idea, as is being around cars or sharp objects. Robert Mitchell, who has practiced psychedelic therapy and administered plant medicines for 30 years and has treated “hundreds of clients,” said “the most important thing is that you feel safe, comfortable and will not be disturbed.” Based in Los Angeles, he said he often has clients rent a cabin in the Santa Monica mountains, which serves as a “sacred space.”

Find an experienced, trusted therapist: If you’re seeking a psychedelic guide, word of mouth may be helpful. My friend, the real estate developer, said “for a first timer, I feel resolute in advising that others find a guide, ideally referred by someone you trust.” Hopkins’s Johnson urges people not to take one of these psychedelic medications alone; although there are still risks, it’s less risky when someone is there who has knowledge of the identity of the substance and the dose. (He said this can be especially critical for psilocybin mushrooms, which are known to have a great variation in their potency.) New programs are available, such as the Psychedelic-Assisted Therapies and Research Certificate Program at the California Institute of Integral Studies, intended to serve the growing need for skilled psychedelic therapists to meet demand.

Ask questions ahead of time: Many therapists include a preparation session before any journey or treatment begins. Questions to discuss in the prep session include a discussion of the therapist’s background and expertise, your intention in taking a psychedelic medicine (and which one), your personal health history, how they might handle a problem that arises (such as a medical side effect or a “bad” trip), the sourcing of the medicine, and, of course, the fee. A Colorado woman gave me this advice, “I would make sure to work with a therapist who has experience and a clear protocol for using psychedelics, including pre-journey discussions and post-journey integration appointments.”

Know what you’re ingesting: Doblin said there’s one DEA-licensed facility in the United States: Drug Detection Laboratories. It accepts anonymous samples of illegal drugs and will analyze them and post the results online. (You send it in with a specific code and pay a fee for the analysis.) Barring that, you’ll want to talk with potential guides about the source of their substances. Mitchell told me he knows where his psilocybin mushrooms are farmed and can vouch for their purity. In the end, one friend who had two psilocybin sessions said he “had to rely on the guide and the trust engendered.” That’s always going to be imperfect.

Do your homework: MAPS is an educational nonprofit group whose first phase 3 study — on the effective use of psilocybin for severe cases of PTSD — was published last year in Nature Medicine, a top peer-reviewed journal. The organization publishes information about the functions, uses and legality of psychedelics. It offers an introductory course, Psychedelic Fundamentals. Another resource is the “MAPS Code of Ethics for Psychedelic Psychotherapy,” which discusses psychological and physical risks.

So will I be taking a psychedelic journey?

I’m reading everything I can get my hands on and talking to everyone I can about their experiences. I’m also remembering the legal issues. Yes, Pollan and others are trying psychedelics and writing about their experiences — and not being arrested or having their careers derailed or apparently suffering ill effects — but that shouldn’t be taken as carte blanche for the rest of us.

I’ll check back in here in a few months, so stay tuned.

 
Top