• Psychedelic Medicine

PARKINSON'S | +50 articles

tingri-everest-base-camp-trek.jpg



This brain protein may be the key to preventing the loss of dopamine neurons in Parkinson’s*

by Gerard O'Keeffe and Aideen Sullivan | The Conversation | 27 Dec 2020

Parkinson’s disease, a brain disorder that affects over 10 million people worldwide, is caused by the gradual loss of dopamine neurons. The loss of these neurons leads to involuntary tremors, stiffness and balance problems. While there are drugs to treat these symptoms, no drugs exist to slow the progression of the disease. However, we found a brain protein that may be able to prevent the loss of dopamine neurons. This discovery could be important for developing treatments.

For many years, scientists have been investigating the use of neurotrophic factors to slow the progression of Parkinson’s disease. These proteins are normally found in the brain and play an important role in protecting and nurturing different types of neurons, including dopamine neurons, which are critical for controlling movement.

In 1993, one neurotrophic factor, called glial cell line-derived neurotrophic factor (GDNF), was found to protect dopamine neurons in laboratory tests. Following extensive laboratory studies in which GDNF displayed much benefit, clinical trials were started in the early 2000s.

In these trials, GDNF was administered directly into the brains of Parkinson’s patients. Promising results were reported from the early trials, in which small numbers of patients all received GDNF treatment. Researchers became excited about the potential of using neurotrophic factors to treat Parkinson’s disease.

But to prove that a treatment is effective, it must be tested in clinical trials in which patients are randomly allocated to receive the experimental drug or a placebo. A GDNF clinical trial was established, but unfortunately, it showed that treating the brain with GDNF did not significantly improve movement symptoms in patients with Parkinson’s when compared with patients who received the placebo.

Despite attempts to improve the delivery of GDNF to the brain, a 2019 placebo-controlled clinical trial of GDNF still produced disappointing results. This was a huge blow to the Parkinson’s community and has led to researchers questioning the potential benefit of neurotrophic factors.

But our research has found promise in another neurotrophic factor, called GDF5. This neurotrophic factor is related to GDNF, but it exerts its effects on dopamine neurons by working in a different way. GDF5 plays an important role in the normal development and functioning of dopamine neurons. Our laboratory studies have shown that GDF5 has protective effects on these neurons, which are as potent as the effects of GDNF.

Our most recent study, published in the journal Brain, found that GDF5 had beneficial effects in a rat model of Parkinson’s, in which GDNF was previously shown to be ineffective. This particular rat model allowed us to more closely mimic human Parkinson’s disease than those rat models that had been used in the earlier studies on GDNF – and which had lead to the clinical trials being approved.

For our study, we administered an excess of alpha-synuclein (a protein that is thought to be involved in Parkinson’s) in the brain to replicate Parkinson’s disease. We then delivered the gene to produce human GDF5 protein to the brain. Six months later, we counted the numbers of dopamine neurons in the brain. We found that about 40-50% of dopamine neurons had died in the untreated group, but this was not seen in the group treated with GDF5. We also found that GDF5 increased the amount of dopamine in the brain. Our next step is to study what stage of the disease it’s best to deliver GDF5 to the brain to slow the disease’s progression.

One reason that researchers have put forward to explain the failure of the GDNF clinical trials is that a protein called RET may be destroyed in the brain when a person develops Parkinson’s. RET is needed for GDNF to act on dopamine neurons. But GDF5 acts through a different pathway – so does not need RET. Our study also found that the cell components needed for GDF5 to act on dopamine neurons are not destroyed by Parkinson’s disease.

The most important findings that we have made are that GDF5 has protective effects on dopamine neurons in the best known laboratory model of Parkinson’s and that the cell components needed for GDF5 to work are not destroyed by Parkinson’s disease. These are very promising results and mean that the search for a new therapy for Parkinson’s focusing on neurotrophic factors should continue.

 
Last edited:
Iboga-Eye.jpg



PharmaTher in clinical trial of Ketamine for Parkinson's-related Dyskinesia*

by Aisha Abdullah PhD | Parkinsons News Today | 23 Apr 2021

PharmaTher is seeking regulatory approval to launch a clinical trial of the painkiller ketamine for levodopa-induced dyskinesia — the uncontrolled, involuntary movements that affect as many as 80% of Parkinson’s patients following treatment.

The company, which focuses on the research and development of psychedelic pharmaceuticals, has submitted an investigational new drug (IND) application to the U.S. Food and Drug Administration (FDA) to initiate a study of low-dose ketamine, an anesthetic available by prescription in the U.S.

The proposed Phase 2 study would evaluate the safety, efficacy, and pharmacokinetics — or how a drug moves into, through, and out of the body — of ketamine in low doses for levodopa-induced dyskinesia (LID).

Dyskinesia is associated with the long-term use of levodopa, a Parkinson’s treatment that increases dopamine levels in the brain. Data shows that as many as 80% of Parkinson’s patients on levodopa will develop dyskinesia after 10–12 years of treatment.

If the results of the proposed trial are positive, PharmaTher plans to request a meeting with the FDA to obtain an agreement for a Phase 3 clinical study under the IND’s expedited regulatory pathway.

“The submission of our IND application with the FDA is an important milestone as it provides us with a solid foundation to advance our ambition in commercializing ketamine and unlocking its therapeutic potential through novel uses, formulations and delivery methods,” Fabio Chianelli, CEO of PharmaTher, said in a press release.

Chianelli said PharmaTher also is investigating ketamine as a potential treatment for other neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS), as well as for mental illness and chronic pain.

Ketamine is approved by the FDA as an anesthetic and pain-relieving agent. In preclinical research conducted by scientists at the University of Arizona, low-dose ketamine was found to alleviate abnormal movements in a mouse model of LID.

PharmaTher announced in October that it had entered an exclusive licensing agreement with the University of Arizona to develop ketamine as a treatment for LID in Parkinson’s patients.

Late last year, the company filed a request to meet with the FDA to receive guidance on advancing the clinical development of ketamine at sub-anesthetic doses to treat LID.

The planned Phase 2 trial is slated to enroll up to 36 participants at up to eight study sites in the U.S., according to PharmaTher. The participants will be randomly assigned to receive either low-dose ketamine or the sedative midazolam for eight weeks.

The primary goal of the study will be a change in the Unified Dyskinesia Rating Scale total score from the study’s start (baseline) to week eight, or about two months.

The study’s secondary goals include a change in the Unified Dyskinesia Rating Scale total objective, motor, and dyskinesia scores, and total daily off times after eight weeks of treatment. Off times are periods when Parkinson’s symptoms return despite medication use.

*From the article here :
 
Last edited:
parkinsons-inflammation-neurosicces.jpeg

The study bolsters the idea that inflammation is linked to Parkinson’s disease,
and points to differences in how men and women respond to the disease.


Inflammation seen in earliest stages of Parkinson’s*

Study finds evidence of inflammation in the blood of patients during the early stages of Parkinson’s disease. The findings support the theory that inflammation is a driver of the neurodegenerative disorder. The effect was most noticeable in women with Parkinson’s.

University of Alabama | Neuroscience News | 29 Apr 2021

New research shows evidence of inflammation in the blood of Parkinson’s disease patients during the earliest stages of the disease, lending support to theories that inflammation is a major driver of PD.

The findings, from researchers at the Alabama Udall Center at the University of Alabama at Birmingham, were published April 13 in npj Parkinson’s Disease, part of the Nature Partner Journal series in partnership with the Parkinson’s Foundation.

UAB is one of six National Institutes of Health-funded Morris K. Udall Centers of Excellence in Parkinson’s Disease Research.

“There is a growing body of evidence suggesting that inflammation plays a major role in the development and progression of Parkinson’s disease,” said David Standaert, M.D., Ph.D., chair of the UAB Department of Neurology in the School of Medicine and senior author of the study. “This is one of the first studies to pinpoint inflammation in the blood in patients with early PD, supporting the idea that systemic immune system activation is present early in PD.”

Standaert also says the study revealed interesting differences in the inflammation signature in women compared to men. While PD is much more common in men, there is increasing evidence that there are also other differences between the sexes in the symptoms and course of the disease. The findings bolster the idea that different approaches to treatment are needed based on the sex of the patient.

The study enrolled 34 patients, 21 males and 13 females. Eighteen had early Parkinson’s disease and 16 were healthy age-matched controls. Those with PD were within two years of symptom onset and had not begun taking anti-Parkinsonian medications.

“The majority of studies into the role of inflammation in PD have been conducted in patients with long-standing disease and wide variations in disease severity. Most had also received a number of different treatments for the disease,” said first author Samantha Carlisle, Ph.D., with the UAB Center for Clinical and Translational Science.

We looked at newly diagnosed patients to improve our understanding of whether inflammation is present early on, or develops as the disease progresses.”

The study examined blood monocytes, cells that are derived from bone marrow, which have been linked to the development of PD. They appear to have a central role in immune signaling through engulfing, processing and presenting foreign antigens for recognition by the adaptive arm of the immune system, thus turning on the immune system in the presence of PD.

The research team used molecular approaches to examine the state of activation of peripheral blood monocytes in human PD. They found evidence of inflammation in blood monocytes, with an unexpectedly strong effect in women.

“We found that there was a striking effect of sex on monocyte gene expression, with increased inflammatory activation of monocytes in females with PD,” said Hongwei Qin, Ph.D., associate professor in the UAB Department of Cell, Integrated and Developmental Biology.

“In males, the activation patterns were more diverse. This indicates the importance of systemic monocyte activation in PD, and the importance of studies which examine how men and women respond in different ways to the disease.”

The Parkinson’s Foundation estimates there are nearly one million people living with Parkinson’s disease in the United States and more than 10 million worldwide. Men are 1.5 times more likely to have PD than women.

“In addition to the greater frequency of PD in men, there are other differences between men and women, in frequency of tremor and imbalance, response to medications and cognitive features,” Standaert said.

“Identification of an inflammatory signature in the blood of PD patients opens the door to novel approaches to both biomarkers and treatment as well as an increased understanding that therapies for PD need to be tailored to the patient.”

Standaert says the study of inflammation in Parkinson’s disease is a major focus of the Alabama Udall Center.

*From the article here :
 
iu


FDA approval of Ketamine for the treatment of Parkinson’s Disease*

Psilocybin Alpha | 17 May 2021

PharmaTher, a psychedelics biotech company, has announced the FDA has approved the company’s application to proceed with a Phase 2 clinical trial to evaluate the safety, efficacy and pharmacokinetics of ketamine in the treatment of levodopa-induced dyskinesia in patients with Parkinson’s disease. PharmaTher expects to begin enrolling patients in the Phase 2 clinical trial in Q3-2021. Assuming the Phase 2 clinical trial is positive, the Company will request a meeting with the FDA to discuss its plan and obtain an agreement to move to a Phase 3 clinical study next year.

Fabio Chianelli, Chief Executive Officer of PharmaTher, said, “The FDA’s acceptance of our IND application for ketamine to treat Parkinson’s disease is a significant milestone for us. The FDA IND is our first of many we will aim to obtain, and we are one of the few psychedelics-focused biotech companies that have an IND approved by the FDA for a recognized psychedelic drug. The IND paves the way for us to expeditiously evaluate ketamine and other psychedelics via the FDA regulatory pathway in various mental illness, neurological and pain disorders. We are committed to building a rich product pipeline of novel uses, formulations and delivery methods of psychedelics, and with our FDA IND in place, we now have the foundation in making PharmaTher a global leader in psychedelic-based therapeutics.”

Parkinson’s disease is a debilitating disorder that affects over 1 million people in the U.S. and more than 7 million people worldwide. There is currently no cure for Parkinson’s disease, although some drug combinations are used to treat the disease symptoms.

About the Phase 2 clinical trial

The clinical trial is titled “A Multi-Center, Phase II, Randomized, Double-Blind, Prospective, Active Placebo-Controlled Trial of Sub-Anesthetic Ketamine to Treat Levodopa-Induced Dyskinesia in Subjects with Parkinson’s Disease.” It is anticipated that up to eight clinical sites in the U.S. will randomize a total of up to 36 subjects to the investigational product (ketamine). The primary end-point of the study is the change in the Unified Dyskinesia Rating Scale total score from Baseline to Week 8. Secondary endpoints of the study include the change in Total Objective Scores of the UDysRS, total daily OFF times as assessed by subject-completed 24-hour diaries and change in the UPDRS total and sum scores of motor and dyskinesia from Baseline to Week 8. Because LID can markedly affect a Parkinson patient’s everyday activities, a reduction in LID could improve the patient’s quality of life.

The Company has assembled a prolific scientific and clinical team experienced in Parkinson’s disease, including Dr. Scott Sherman and Dr. Torsten Falk from the University of Arizona, Dr. Alberto Espay from the University of Cincinnati and Dr. Robert Hauser from the University of South Florida.

Ketamine’s potential in treating Parkinson’s Disease

Ketamine is an FDA-approved NMDA receptor-modulating drug that is widely used as an anesthetic agent either alone or in combination with other anesthetic agents. The possible therapeutic effect of low-dose ketamine on LID was noted in a retrospective analysis of PD patients who received ketamine for pain relief. During this analysis, it was observed that the patients experienced an improvement in LID lasting several weeks beyond treatment. These results were corroborated in a test of low-dose ketamine in a rodent LID model, and this possible effect has also been examined in a controlled study. Ketamine may also have additional benefits in the treatment of pain and depression, which are frequent comorbidities of Parkinson’s disease.

From the article here :
 
Last edited:
parkinsons-disease-gel-neurosicences-public.jpg



New hydrogel offers hope to defeat Parkinson’s*

Australian National University | Neuroscience News | 3·Aug 2021

Summary: A new hydrogel that acts as a gateway to transfer stem cells into the brain and facilitate repair to damaged tissue may effectively treat neurological conditions like Parkinson’s disease and stroke.

Researchers from The Australian National University (ANU), in collaboration with The Florey Institute of Neuroscience and Mental Health, have developed a new type of hydrogel that could radically transform how we treat Parkinson’s disease.

The gel also offers hope for patients who have suffered from other neurological conditions such as strokes.

The new material is made from natural amino acids – the building blocks of proteins – and acts as a gateway to facilitate the safe transfer of stem cells into the brain and restore damaged tissue by releasing a growth-enabling protein called GDNF.

By putting the stem cells into a gel, they are exposed to less stress when injected into the brain and are more gently and successfully integrated.

“When we shake or apply energy to the hydrogel, the substance turns into a liquid which allows us to inject it into the brain through a very small capillary using a needle,” Professor David Nisbet, from the ANU John Curtin School of Medical Research (JCSMR), said.

“Once inside the brain, the gel returns to its solid form and provides support for the stem cells to replace lost dopamine neurons.”

Professor Clare Parish, Head of the Stem Cell and Neural Development Laboratory at The Florey Institute, said: “Through use of the hydrogel technique we demonstrated increased survival of the grafted dopamine neurons and restored movement in an animal model of Parkinson’s disease.”

Although dopamine-related drugs are a readily used treatment for people living with Parkinson’s disease, many have undesirable side effects that are exacerbated with time.

“The stem cell transplant delivered in this hydrogel on the other hand avoids many of these side effects and could provide a one-off intervention that can sustain dopamine levels for decades to come,” Professor Parish said.

The gel also offers hope for patients who have suffered from other neurological conditions such as strokes. Image is in the public domain

Professor Nisbet said the hydrogel has the potential to also treat patients who have suffered a stroke and could even be used to treat damaged knees or shoulders, following successful animal trials.

“When we introduced the gel technology with the stem cells we saw huge improvement in the animals’ coordinated paw movement and overall motor function recovery,” he said.

The hydrogel technology is cost-effective and easy to manufacture on a mass scale, and it’s hoped the treatment could soon be made available in hospitals, but it must first undergo clinical trials.

“We must do our due diligence and ensure we check all the right boxes regarding safety, efficacy and regulatory approval before we can take this technology into the clinic, but we hope it can be available for use in the not-too-distant future,” Professor Parish said.

About this Parkinson’s disease research news :

Source: Australian National University
Contact: George Booth – Australian National University

*From the article here :
 
Last edited:
ketamine_SophieHolmes_contrib_photo.jpeg


Ketamine as a potential treatment for depression in patients with Parkinson’s, study*

by Selin Nalbantoglu | Yale News | 4 Mar 2022

Yale researchers are currently conducting the first clinical trial that tests the use of ketamine as an antidepressant for patients with Parkinson’s disease.

Sophie Holmes, an assistant professor of psychiatry and neurology at the Yale School of Medicine, is spearheading this research. According to Gerard Sanacora, the director of the Yale Depression Research Program and a professor of psychiatry at the medical school, research on the use of ketamine as an effective antidepressant in rodents has been widely available for over 20 years. However, no researchers have been able to translate the findings in mice into clinical research for human use.

“I’ve been using neuroimaging to research depression for the past 10 years, but since coming to Yale have been particularly interested in ketamine — given its ability to rapidly reduce symptoms of depression,” Holmes wrote in an email to the News. “Specifically, I wanted to understand how it works in the brain — what are the mechanisms that underlie its therapeutic effects? Can we capture these changes using brain imaging?”

Sanacora explained that, at the clinical level, depression often occurs alongside Parkinson’s disease. At the same time, standard antidepressants such as Prozac or Zoloft are often less effective in patients suffering from Parkinson’s. In addition, neurodegenerative processes, such as those induced by Parkinson’s disease, may be halted or reversed by a drug like ketamine that induces neuroplasticity, the brain’s ability to form new synapses.

Previous studies have shown that rodents in chronic stress models, meaning models designed to emulate the conditions of depression, undergo visible neurodegeneration. In these cases, the administration of ketamine led to increased synaptic density in these rodents.

“Animal work consistently shows that chronically stressing out rats results in a loss of synaptic connections and depressive-like behavior,” Holmes wrote. “In other words, synaptic loss, especially in brain regions associated with emotional regulation, is thought to underlie depression. We think ketamine works by restoring these lost synapses and forming new pathways that can bypass rigid pathways associated with negative thought patterns and depression.”

However, Sanacora added, there is no way to ask the rodents how they are feeling, so researchers have struggled to quantify the efficacy of ketamine treatments.

Holmes added that work in animal models has proven that ketamine enhances synapses and has “neuroprotective” effects. Since ketamine works by increasing synaptic density and Parkinson’s disease is characterized by a loss of synapses, the decision to study ketamine as a potential solution for the neurodegenerative actions of the disease was logical.

The current trial is split into two experimental methods. According to Sanacora, the first section is a randomized placebo control clinical trial. Since the Federal Drug Administration has already approved the use of S-ketamine in treating depression, researchers can actually administer ketamine to patients at Yale New Haven Hospital.

“Patients with Parkinson’s and symptoms of depression will have a course of ketamine or placebo treatment,” Sanacora said. “It will be completely randomized, whether a patient gets active ketamine or a placebo. The primary measure there is how well their depressive symptoms respond.”

Holmes added that 50 patients will either receive six “infusions” of ketamine or a placebo over the course of the next three weeks.

Then, the second experimental method is to select a subset of participants for brain imaging. The researchers will use MRIs or PET scans to measure the effects of the ketamine infusions on the patients’ brains.

“A lot of research suggests that ketamine works by increasing the number of synaptic connections in the brain, making the brain more ‘malleable’ and allowing new pathways to form, which can encourage new, more helpful ways of thinking,” Holmes wrote. “Using PET imaging, we will explore whether changes in synaptic density after ketamine treatment underlie any therapeutic effects.”

According to the U.S. National Library of Medicine, the ketamine infusion will be replaced by a saline infusion for the patients receiving a placebo. In addition, the clinical trial is currently in phase 2, meaning that researchers are “gathering preliminary data on whether a drug works in people who have a certain condition/disease” — in this case, Parkinson’s disease.

This current trial is addressing a major unmet need in medical care for patients with Parkinson’s disease, according to Sanacora. He emphasized that there is a strong need for effective antidepressant treatments for patients suffering from this disease, yet there currently is no alternative to the standard treatments.

“Depression takes a major toll on patients with Parkinson’s disease.” Sanacora said. “The ability to develop something that can mitigate that feeling is so important, especially as the population ages.”

For Holmes, this trial also has personal significance. Her father was diagnosed with Parkinson’s disease around five years ago. By watching his experience and reading to learn more about the disease, she realized how “pervasive” and “debilitating” depression can become for patients suffering from Parkinson’s disease.

The study began on Nov. 23, 2021 and is planned to end in August 2024.

 
Last edited:
Ibogaine Treatment for Parkinson's



A heartfelt, emotional, and true testimonial about overcoming Parkinson's using Ibogaine Treatment. Ibogaine Therapy. Ibogaine has been used therapeutically for several indications, particularly detoxification from opiates. It has been shown to decrease the self-administration of stimulants, opiates, and alcohol, as well as to significantly reduce the withdrawal symptoms from opiates after a single administration. Ibogaine is a naturally occurring psychoactive substance found in plants in the Apocynaceae family such as Tabernanthe iboga, Voacanga Africana and Tabernaemontana undulata. It is a psychedelic with dissociative properties.​
 
tingri-everest-base-camp-trek.jpg



Psychedelics and Parkinson’s: Improving patient quality of life*

by Emily Jarvie | Truffle Report | 24 Jan 2022

In recent years, research has shown that psychedelic drugs offer new and exciting potential treatments for various physical and mental health conditions. One promising area is the use of psychedelic-assisted therapy to treat Parkinson’s Disease: a progressive neurodegenerative disorder that occurs when the nerve cells in the brain that control movement stop working or die. Truffle Report has put together a brief review of prospective treatments and clinical trials involving psychedelics and Parkinson’s Disease.

More than 10 million people worldwide live with Parkinson’s. While both men and women can be diagnosed with the disease, this condition affects 50 percent more men than women.

Common symptoms of the disease include tremors, muscle stiffness or muscle rigidity, slowness of movement, dry mouth, and difficulty walking or talking. It can also lead to Parkinson’s dementia, cognitive impairment, sleep problems, and mental health issues such as depression and anxiety.

Sadly, there is no cure for the condition. The range of disease symptoms a person with Parkinson’s can experience makes standardizing medical treatment protocols challenging. Current treatments include medications and drug therapy, and supportive therapies such as physical therapy or speech therapy, surgery, and lifestyle modifications.

However, several psychedelic drugs have shown therapeutic effects when it comes to improving the daily life of Parkinson’s patients, including DMT, ketamine, psilocybin, and LSD. This research offers promise for much more than psychedelics and Parkinson’s disease, and could also lead to breakthroughs in other neurodegenerative diseases and movement disorders through psychedelic-assisted therapy.

Could DMT be a possible treatment for Parkinson’s?

Off the back of new research, there is speculation that DMT could be used to treat neurodegenerative disorders and neurological diseases, such as Parkinson’s or Alzheimer’s Disease.

A 2020 preclinical study using mice found that DMT stimulates adult neurogenesis — the growth and development of new nerve cells in the brain — and improves spatial learning and memory tasks. Researchers concluded: “DMT’s ability to modulate brain plasticity indicates its therapeutic potential for a wide range for psychiatric and neurological disorders, among which are neurodegenerative diseases.”

In addition, Vancouver based-biotechnology company Akome Biotech, a wholly-owned subsidiary of Core One Labs, believes its psychedelic drug formulation can be used to treat Parkinson’s. The company’s preliminary research shows that its DMT formula, known as AKO004, can counter or mitigate neurodegeneration.

While both of these developments are promising, we will have to wait until more research is completed to know for sure if DMT could become a viable treatment option for Parkinson’s.

Ketamine for levodopa-induced dyskinesia: Encouraging Deep Brain Stimulation

A standard and effective treatment for Parkinson’s. is the drug levodopa which helps patients replenish their brain’s supply of dopamine. However, a common potential side effect of this treatment is levodopa-induced dyskinesia (LID): involuntary movements such as rapid jerking and twisting, which can range from inconvenient to incapacitating. This condition, once established, is difficult to treat and can adversely affect a patient’s quality of life.

But there is a growing body of research that shows low doses of ketamine may have beneficial effects in reducing the effects of LID. A 2016 study found patients displayed a “prominent and prolonged” improvement in LID following ketamine infusion. The authors concluded that low-dose ketamine might act as a “chemical deep brain stimulation” and recommended further investigation into its use as a treatment for LID.

Another 2016 preclinical study using a rodent model investigated ketamine infusion for LID. While the researchers noted that an acute injection of ketamine did not reduce abnormal involuntary movements (such as those caused by LID), when ketamine was given for 10 hours, there was a dose-dependent reduction of abnormal involuntary movements. “Sub-anesthetic ketamine infusion could be a novel long-term treatment modality to reduce LID and should be further investigated,” the researchers concluded.

Building on this research, in 2021, the Food and Drug Administration (FDA) also approved PharmaTher’s Investigational New Drug application to proceed with a Phase 2 trial for ketamine as a treatment for LID. This trial is ongoing, and PharmTher says, pending positive Phase 2 results, it plans to move to a Phase 3 study in Q3, 2022.

Ketamine Therapy for depression in Parkinson’s patients

Ketamine has also shown the potential to treat common comorbidities associated with Parkinson’s, including depression (which affects about 60 percent of all people diagnosed with the disease) and chronic pain. “Sub-anesthetic ketamine could provide a multifaceted benefit to individuals with Parkinson’s Disease in the future,” the researchers behind a 2020 study on ketamine and LID noted. A 2021 study on depressive-like behavior in animal models of Parkinson’s also found ketamine had therapeutic benefits for non-motor symptoms of the disease, including an ability to reverse memory impairment and depressive-like behavior due to its antidepressant effects.

Yale University’s Department of Psychiatry is currently recruiting patients for the world’s first clinical study evaluating ketamine for the treatment of depression in Parkinson’s. The Phase 2 trial will compare the efficacy of ketamine to placebo in about 50 individuals and will also use cutting-edge brain imaging to study the effects of ketamine on synaptic connections and functional networks in the brain. It is expected the trial will be completed in August 2024.

Announcing the trial in November 2021, co-principal investigator Dr. Sophie E. Holmes said: “The findings of this research could change the way that Parkinson’s is treated, leading to the discovery of fast-acting and effective treatments that in turn improve the quality of life for the many individuals with Parkinson’s Disease that suffer from depression.”

This research has been funded by a $2 million donation from the Michael J. Fox Foundation for Parkinson’s Research. This foundation, founded and named after American actor Michael J. Fox (who was diagnosed with early-onset Parkinson’s in 1991), is a significant funding source for Parkinson’s research worldwide.

Psilocybin and LSD for wellbeing, depression, and anxiety in people with Parkinson’s

Two other psychedelic drugs being researched for their potential to improve the lives of patients with Parkinson’s are psilocybin and LSD. Psilocybin is especially promising, given its recently proven ability to spur the growth of neural connections and new brain cells.

One Phase 2B study based at Maastricht University in the Netherlands, sponsored by Silo Pharma, is set to evaluate the effect of repeated low doses of both psilocybin and LSD on wellbeing, emotional and cognitive attention, and biological markers of neuroplasticity. Although this study was announced back in December 2020, it is yet to commence.

Psilocybin’s potential for helping Parkinson’s patients is also being explored in a University of California, San Francisco Phase 2 trial. This trial will evaluate the effectiveness of oral psilocybin therapy for depression and anxiety for people with Parkinson’s. It is expected this study will be completed by December 2022.

*From the article here :
 
hallucinations-drug-abuse_f_600x250.jpg



LSD and Parkinson's

Drug Times | 14 Dec 2020

In a very real sense the circle is closing with respect to Albert Hofmann's hope, expressed at the time of his discovery of LSD. Because of its psychotomimetic action (ability to mimic certain mental illnesses), the drug might prove useful in their treatment. In fact, LSD has been employed to that end over the years by some psychiatrists, often with beneficial results.

Recently, scientists at the School of Medicine of the University of California at Los Angeles have made some significant discoveries about the interaction of LSD with dopamine, one of the neurotransmitter agents in the brain, that may lead not only to a better understanding and eventual treatment of schizophrenia, the mental disorder to which the LSD "high" is a kind of temporary analogue, but even of such physically, rather than mentally, crippling disorders as Parkinson's disease. The investigators, Drs. Sidney Roberts and Kern von Hungen and Diane F. Hill, determined that adenyl cyclase, an enzyme in nervous tissue that is stimulated by naturally occurring neurotransmitter agents, is also stimulated by the action of LSD on receptors for one of these neurotransmitters, dopamine. In addition, LSD blocked the stimulatory actions of dopamine and other neurotransmitters such as serotonin and norepeninephrine. These are themselves structurally closely related to powerful plant growth hormones; dopamine, moreover, has also been identified with the giant saguaro cactus of Arizona and northern Mexico.

Schizophrenia is thought to be a disease of dopamine hyperactivity; victims of Parkinson's disease, on the other hand, suffer from dopamine insufficiency, which is partially offset nowadays by the administration of a new drug, L-dopa, often in combination with Tofranil or some other amphetamine. The adenyl cyclase experiments enabled the UCLA team to show that dopamine receptors are present in the higher regions of the brain, which are concerned with the more complex experiences and thus are more likely to be the seat of alternate states of consciousness, or "hallucinations." Their work, report the UCLA investigators, makes it appear that the psychotic mimicking effects of LSD, first noted by Hofmann more than thirty years ago, may also be related to hyperactivity of brain dopamine systems. These insights have obvious implications for work on new drugs for schizophrenia on the one hand and Parkinson's disease on the other; recognition of their biochemical kinship was, of course, still far off in the distant future when Hofmann correctly predicted the ultimate benefits of LSD for brain research. Nor did he suspect at the time that "primitive" psychotherapy had been making effective use of a natural compound very like LSD for hundreds, perhaps thousands, of years.

"Lysergic acid," Hofmann (1967) has explained, "is the foundation stone of the ergot alkaloids, the active principle of the fungus product ergot. Botanically speaking ergot is the sclerotia of the filamentous fungus Claviceps purpurea which grows on grasses, especially rye. The ears of rye that have been attacked by the fungus develop into long, dark pegs to form ergot. The chemical and pharmacological investigation of the ergot alkaloids has been a main field of research of the natural products division of the Sandoz laboratories since the discovery of ergotamine by A. Stoll in 1918. A variety of useful pharmaceuticals have resulted from these investigations, which have been conducted over a number of decades. They find wide application in obstetrics, in internal medicine, in neurology and psychiatry."

 
ibogaine-and-parkinsons.jpg



Does Ibogaine work for Parkinson’s Disease?

by Shea Prueger & Dr. Benjamin Malcolm, PharmD, MPH, BCPP | Psychable

Ibogaine has been used for years in the treatment of Substance Use Disorders, which are often associated with drugs that directly or indirectly increase dopamine. It appears ibogaine can provide a ‘resetting’ effect to mesocortical dopamine circuits affected by addiction. However, ibogaine also modulates dopamine neurocircuits in the striatum that are lost in persons with Parkinson’s disease. The discovery of ibogaine being able to promote increases in neurotrophic factors such as GDNF (glial cell line-derived neurotrophic factor), has sparked interest in its potential as a treatment in Parkinson’s disease. Though ibogaine has not yet been scientifically tested to treat Parkinson’s, various personal stories have come out about the potential benefits of ibogaine treatment.***​

Background of Parkinson’s Disease

Parkinson’s disease is a chronic and progressive neurodegenerative disorder that affects motor control and is caused by the loss of dopamine neurons located in the substantia nigra, as well as the deterioration of the central and peripheral nervous system. The main symptoms are related to motor control and include tremors, difficulty with speech, slow movement, rigidity, and postural instability.

Parkinson’s affects about 1% of the population over 60 years old, with men being affected more commonly than women. Parkinson’s diagnosis increases with age and only 4% of all cases are in people under the age of 50. Parkinson’s disease is the second most common neurodegenerative disorder after Alzheimer’s. Parkinson’s symptoms cause physical discomfort and motor problems first and with a gradual increase to emotional and cognitive deficits. Depression is extremely common in persons with Parkinson’s and the illness can also lead to dementia.

There is no cure for Parkinson’s, but there are treatments to help manage symptoms like levodopa/carbidopa or dopamine agonists (like ropinirole), which work by stimulating dopamine receptors in the brain. These drugs can have side effects, including hallucinations and dyskinesia, and are not always successful at alleviating symptoms or preventing disease progression. Parkinson’s patients have few palliative care options and poor quality of life is often expected for those in the late stages of their terminal diagnosis.​

Parkinson’s Disease and Neurotrophic Factors

A few lines of research have investigated the potential of increasing neurotrophic factors like GDNF in the treatment of Parkinson’s disease. GDNF and other neurotrophic factors stimulate the growth of dopaminergic fibers in the brain. There is evidence that GDNF improves the survival of dopaminergic neurons, which in turn could improve symptoms of Parkinson’s.

Unfortunately, research with neurotrophic factors and Parkinson’s is limited. The delivery of trophic factors such as GDNF directly into the brain has some issues. They cannot cross the blood-brain barrier so cannot be administered by mouth. Administration by injection is invasive and carries risks of missing the target tissues.

Therefore, it may be a preferable strategy to use medications, such as ibogaine, that can release GDNF via oral administration.
Ibogaine and Neurotrophic Factors

Ibogaine’s mechanism of action is not fully understood. However, ibogaine and its metabolite noribogaine increase levels of neurotrophic factors such as GDNF (glial cell line-derived neurotrophic factor), which helps neurons survive by protecting them from damage and death. Evidence suggests that ibogaine modulates GDNF in areas of the brain affected by Parkinson’s disease, namely nigrostriatal dopamine circuits. Ibogaine may have neuroprotective qualities that could promote the survival or even regeneration of motor and dopaminergic neurons lost in Parkinson’s. Ibogaine is also theorized to modulate and regulate other neurotrophic factors such as BDNF and NGF. The discovery that ibogaine can collectively increase and modulate several neurotrophic factors is an exciting subject for future studies on its relationship to neurodegenerative diseases.
Ibogaine and Parkinson’s

Anecdotally, there are stories weaved through the ibogaine treatment world that ibogaine has reported the improvement of symptoms in those with Parkinson’s disease as well as other neurologic illnesses such as traumatic brain injuries, peripheral neuropathy, and fibromyalgia. Theoretically, ibogaine in a low, repeated structured dosing may stimulate and increase GDNF, eliminating the side effects of medications or the difficulty of replicating brain infusions, as other studies have done. A low dose regimen may also decrease some of the risks, and unpleasant side-effects that come with ibogaine treatment, although this also needs further study to confirm safety and benefits.

Some studies suggest that ibogaine signals between neurotrophins, dopamine, and sigma receptors. There are also theories that a misbalance of signaling between neurotrophins and dopamine sites might be related to Parkinson’s or other neurodegenerative diseases. Ibogaine should be further studied for its ability to help address these neurological issues.
Conclusion

Ibogaine has mechanisms of action along with anecdotal reports that suggest potential benefits in Parkinson’s disease. However, more research needs to be done before it can be recommended as an option for treating symptoms of Parkinson’s Disease.

***Disclaimer:

Currently, there is no scientific research on ibogaine and Parkinson’s. This article is meant to be theoretical and as an advocate for further research to occur. Further research will be hindered if people are reckless and die or get hurt as a result of experimentation.

There may be providers who say they can treat Parkinson’s, please refer to our guide to finding a safe ibogaine center to thoroughly vet providers as there are stories in the ibogaine community of reckless behavior with Parkinson’s and other neurodegenerative diseases that have resulted in adverse medical events.

Please, do not attempt to order ibogaine online and attempt self-treatment for anyone with Parkinson’s. Ibogaine can be fatal and fatalities are documented even in low doses. With that being said, this author is an advocate for scientific research on ibogaine treatment and Parkinson’s.

 
Is ibogaine a promising new treatment for Parkinson's?

by Jonathan Dickinson | Global Ibogaine Therapy Alliance

A research team at Columbia University soon hopes to show whether or not ibogaine, an extract of iboga, a psychoactive West-African plant medicine, may have unique benefits in the treatment of Parkinson's disease. The majority of existing research on ibogaine has explored its increasing use over the past several decades as an experimental treatment for substance use disorders, and particularly for opiate detoxification. But in addition to ibogaine's anti-addictive properties, which have been explored in early clinical trials and observational research, there's reason to believe that ibogaine may be beneficial in Parkinson's treatment. Columbia's two-year animal study represents the first time that researchers will probe for a direct link. Parkinson's disease, which is recognized as the second most common neurodegenerative disorder after Alzheimer's, is caused by the loss of a specific type of brain cells called dopaminergic neurons from a small and concentrated area in the mid-brain. This results in gradually increasing symptoms that include motor effects like tremors, muscle stiffness, difficulties with speech, severe lack of coordination and balance, as well as non-motor symptoms such as dementia, depression and others.

According to the Parkinson's Disease Foundation the condition affects an estimated 7 to 10 million people worldwide. And although treatments are available that offer some symptomatic relief, there is currently no known method of reversing the effects of the disease.

The exact mechanisms of action of ibogaine are still obscure, but the theory for ibogaine's benefit for Parkinson's rests on the fact that amongst its other effects, it has been shown to increase the production of a protein called glial cell-derived neurotrophic factor, or GDNF. This and other neurotrophic factors have been shown to stimulate the production of dopaminergic fibers throughout the brain, and some existing research shows that GDNF in particular improves the recovery of dopaminergic neurons and leads to an improvement of Parkinson's symptoms.

Previous methods used to increase the expression of GDNF were limited gene therapy and direct brain infusion, but according the research abstract, the research team, led by Dr. Serge Przedborski, President of the World Parkinson Coalition, is exploring whether ibogaine could provide "a safer and more convenient means to enhance GDNF production in the brain."

Ibogaine's clinical development

Tabernanthe iboga, ibogaine's primary plant source, has been used for millennia in Gabon and other parts of West Africa as a medicine and initiatory sacrament. In 1962, a 19-year-old named Howard Lotsof was the first Westerner to discover that ibogaine, iboga's primary active ingredient, could mitigate withdrawals from opiates, and provide unique psychological benefits including relief from cravings for many who take it.

Lotsof's later advocacy work during the late eighties and early nineties resulted in the U.S. National Institutes of Health (NIH) funding early clinical trials. While this proved a promising and uncharacteristic support for public research, the NIH eventually withdrew funding because of the complexity of the trials and litigation between the investigators.

Despite the lack of funding for clinical development, the research that has been done has continued to be supportive. Some researchers believe that ibogaine's release of GDNF may play a role in its more lasting psychological benefits by potentiating a state of neuroplasticity that some refer to as a kind of "neurological second childhood." To some extent, the rapid growth of new neurons may help to repair and rebuild neurological pathways that have been damaged by addiction.

Years later, several research teams are slowly preparing Phase 1 and 2 trials on human patients to continue to explore ibogaine's use in addiction treatment. Re-initiating those trials has remained difficult for many reasons, partly because there is little profit motive for pharmaceutical drug developers, and to some extent due to reports of fatalities that have occurred in close conjunction with ibogaine administration.

The risks involved in ibogaine addiction treatment stem largely from the fact that it is unregulated and there are huge variations in the level of care provided at ibogaine treatment centers. However, many therapy providers and researchers agree that its use can be made safe with proper screening and in-treatment monitoring.

One of those doctors, Dr. Jeffrey Kamlet, a Miami medical doctor and board certified addiction specialist has been researching ibogaine for over 20 years, and is a strong advocate for medical supervision. According to Dr. Kamlet, "Ibogaine is the most important discovery in the field of opiate dependency in the history of addiction medicines, and I'm confident that it will one day be a main stay treatment for many addictions."

Even with this in mind, some advocates believe that ibogaine's application as a treatment for Parkinson's may prove more straightforward for research. In the case of addiction therapy, ibogaine is most often administered in a single large dose, but there is anecdotal evidence that the repeated use of very low sub-perceptual doses, which are generally agreed to be well tolerated in patients, could be sufficient to reverse Parkinson's symptoms over time.

One promising case

Patient D* was 69 years old in 2012 when he was diagnosed with Atypical Parkinson's, a Parkinson's-like syndrome that does not include the characteristic palsy. By last year Patient D's symptoms had advanced to the point where his facial muscles felt frozen. He had difficulty finding his balance, talking or using his hands. As a writer and artist, he noted that emotionally it was the first time in his life he had lost his desire to do anything creative.

In December 2014, Patient D was treated with CKBR-12, an experimental natural health product and "ibogaine-derivative" at a medical center in Rosarito, Mexico. He took a small dosage twice a day for 30 days, and after the first two weeks began to notice that he could use his fingers to pick up objects again. After a month he had seen a gradual improvement in all of his symptoms to the point where he could carry on normal conversation, and coordinate previously impossible tasks such as buttoning his shirt.

In a post-treatment interview that was published on YouTube, Patient D says, "It's difficult to explain what Parkinson's is, but you lose your edge. And I've got my edge back in a few areas. They may be small things to some, but they are big things for me. I'm anxious to see what happens next."

When we spoke, to Patient D, he said that he had continued to have improvements with further treatment. His case has been reviewed by several doctors, including Dr. Susanne Cappendijk of Florida State University Medical Center, who relayed some of the results at a conference at the New York Academy of Sciences on April 27th.

Ibogaine as a prescription medicine

Much like ibogaine's use in addiction treatment, the current use of ibogaine for Parkinson's and other neurodegenerative disorders is illegal in the U.S. due to its Schedule 1 status. In most countries, including Canada and Mexico, this is not the case. Yet, even in places where ibogaine can be administered, treatments are considered experimental until they undergo the full battery of demanding clinical trials.

This widening berth of ibogaine research may help to build a more encouraging case for its approval as a prescription medicine. But even if both applications hold efficacy, as long as one is granted FDA approval, the other could speculatively be administered "off-label."

The clinical trial process is bulky and challenging for many medications, including useful new antibiotics, because they cost millions and are slow to develop. If medications that are advantageous from a public health perspective don't come packaged with a lucrative business plan there few big pharmaceutical companies willing to invest. One of the main reasons that ibogaine hasn't been funded by the existing addiction treatment industry is that it is only administered once, or a few times, rather than as an ongoing regimen.

In 2009, New Zealand became the first country in the world to offer ibogaine as a prescription on an experimental basis, a trend that may eventually follow elsewhere. Now, in Vermont, addiction rates have become so severe that the governor dedicated his entire State of the State speech to the issue. This prompted members of the Vermont State Legislature to table a bill that would allow the operation of a non-profit ibogaine detox center in the state.

Even in the midst of these isolated examples, and without broad regulatory reform, FDA approval is needed to make ibogaine treatments more widely available. Researchers who are studying ibogaine for addiction treatment have to look past a $35 billion U.S. addiction treatment industry that believes it possesses effective modalities, even despite a general increase in rates of addiction. Parkinson's researchers, on the other hand, may have the benefit of a number of large foundations that are established to fund research in yet another field where few other drugs can claim similar promise.

https://www.ibogainealliance.org/ne...omising-new-treatment-for-parkinsons-disease/
 
Last edited:
Ketamine_Study_Suggests_Psychedelic_Could_Treat_Parkinsons_Disease_Psychedelic_Spotlight-1200x800.jpg



Ketamine could treat Parkinson’s, study*

by James Hallifax | Psychedelic Spotlight | 23 March 2022

PharmaTher had the goal of evaluating “the safety, tolerability and efficacy of low-dose ketamine infusion for the treatment of LID in patients with Parkinson’s Disease.” On all three counts, it appears to have exceeded expectations.
It seems that ketamine is having a moment. Originally discovered in 1962, the dissociative psychedelic has been used mainly as an anesthetic (pain killer) in the medical establishment for decades.

In recent years, however, its medical uses have ballooned, as evidence emerges that its potential benefits range from treating depression, to addiction, to ALS. Across North America, ketamine clinics have been sprouting like dandelions in spring, legally treating mental health conditions such as depression.

Now, evidence is growing that the psychedelic can also treat a debilitating aspect of Parkinson’s Disease.

Today, PharmaTher announced topline results of their Phase 2 study using ketamine to treat Levodopa-Induced Dyskinesia (“LID”) in patients with Parkinson’s Disease.

Before getting to the results, let’s take a moment to understand LID. Basically, when a patient shows symptoms of Parkinson’s — a brain disorder that can cause uncontrollable shaking, and difficulty with walking, balance, and coordination — the most common treatment is a drug called Levodopa. And while this drug is effective in treating the symptoms, over time the side effects of Levodopa treatment start to mirror that of Parkinson’s itself. After five years of treatment, roughly half of patients will experience abnormal and uncontrollable bodily movements. Within 10 years, it will affect nearly everyone. This is LID, and it is currently basically untreatable.

Enter ketamine.

In the current study, PharmaTher had the goal of evaluating “the safety, tolerability and efficacy of low-dose ketamine infusion for the treatment of LID in patients with Parkinson’s Disease.” On all three counts, it appears to have exceeded expectations.

Starting with efficacy, PharmaTher says the study “demonstrated that 100% of patients treated with ketamine demonstrated a reduction in dyskinesias (LID).” Now, while this is certainly impressive, it must be noted that PharmaTher’s early topline data did not specify by how much, on average, patients saw the dyskinesias symptoms decrease; only that they did. Full data is expected to be released in June.

On safety and tolerability, “the study demonstrated that ketamine was well tolerated with no serious adverse events reported. All adverse events were mild or moderate and reflected the expected side-effects of ketamine administration.”

So, despite not having the full data available, we can say that the results are very positive indeed. I am looking forward to writing about the full data in June.

Though the public doesn’t yet have the full data, PharmaTher is confident that it is positive enough to greenlight a Phase 3 trial this year. This trial will use PharmaTher’s proprietary ketamine intravenous product, KETARX™. As a reminder, a drug must pass through two Phase 3 trials before being approved to treat a condition. Though, as ketamine itself is already legal, doctors can prescribe it “off-label,” meaning that doctors, lacking other options, could likely already try this therapy.

Parkinson’s is a tragically large problem. More than 1 million people in the United States, and more than 7 million people world-wide, suffer from the disease. And while the drug Levodopa does certainly help them, the long-term consequences of taking it leave much to be desired. Hopefully, future studies continue to show ketamine’s effectiveness, and these 7 million plus individuals can get the help they deserve.

From a business perspective, the treatment market for LID is estimated to be more than $3 billion annually in the US alone. If PharmaTher can tap into this market, the company, currently with a tiny market cap of $15 million CAD, could see big days ahead. However, it must be noted that many companies produce and deliver ketamine, so even if PharmaTher proves its effectiveness in treating LID, it would face stiff competition.

Stepping back, this trial illustrates a BIG TREND in psychedelic medicines that Psychedelic Spotlight will be watching closely over the coming years: the transition of psychedelic medicines from treating solely mental health conditions, to treating physical health conditions. While we certainly do have a mental health crisis, battling traditional health issues is also important. Recent studies have shown that psychedelics may be useful in treating conditions as far ranging as eating disorders, to chronic pain, to strokes, and Alzheimers.

At the end of the day, one of our goals as a species should be to lessen the pain and despair found on our planet. With each passing month, as more clinical data is released, it appears that psychedelic medicines such as ketamine, psilocybin, MDMA and LSD can help us achieve this lofty goal. We must put aside the stigmas of the past, and begin to view these molecules as medicines, rather than dangerous drugs that must be illegal.

*From the article here :
 
Last edited:
bioNewsTX_ShakingThingsUp%E2%80%93MaryBethSkylis_logo_210919_02_v1-e1572631496869.jpg



Could CBD help to ease Parkinson’s symptoms?

by Mary Beth Skylis | Parkinson's News Today | 19 Feb 2020

As a Colorado resident, I often wonder about the healing power of plants. We live in a time when the pharmaceutical industry is booming. Pills exist to tame nearly any symptom, but they often can have unwanted side effects. The side effects of plants, however, may be less harsh, or even nonexistent. Cannabidiol (CBD) is a great example of this phenomenon.

What is CBD?

CBD is a compound derived from the cannabis plant and is commonly sold in oils and foods. Depending on the product, CBD could potentially treat pain, anxiety, depression, insomnia, and inflammation, among other issues. Additionally, research suggests that CBD potentially could be useful for other conditions, including improving well-being and quality of life in Parkinson’s disease (PD).

Unlike tetrahydrocannabinol (THC), CBD generally has relaxing effects. Users do not feel “stoned” or intoxicated.

Why is CBD controversial?

The use of CBD is legally gray, as marijuana is illegal at the federal level. However, the 2018 U.S. Farm Bill legalized the use of CBD produced via the cultivation of hemp with THC levels below 0.3 percent.

You also can use a medical marijuana card to obtain CBD in some states. Nevertheless, a few states currently forbid the use of CBD. Check to see if CBD is legal in your state here.

Because CBD is unregulated at the federal level, it can be difficult to determine the amount of THC in certain products. Purchasing CBD products from reputable brands that conduct third-party testing is currently the safest option.

What might CBD do for those with Parkinson's?

CBD has shown potential in early studies for reducing dyskinetic activity in people with PD and treating motor symptoms in various neurodegenerative conditions.

According to a 2018 review study published by the journal Frontiers in Pharmacology, “Cannabidiol is a non-psychotomimetic compound from Cannabis sativa that presents antipsychotic, anxiolytic, anti-inflammatory, and neuroprotective effects.” Data also suggest that CBD could potentially play a protective role in the treatment of certain movement disorders. Results are promising, but further studies are needed to clarify the efficacy of CBD.

Our experience

My dad kept hearing about the potential benefits of CBD. He doesn’t like the sensation of getting high, so he investigated products that would yield similar benefits without the possibility of intoxication. Eventually, he purchased two tinctures that he consumed orally for several weeks. He doesn’t believe the tinctures had a substantial impact on his everyday life, but I’m not ready to let him stop hoping.

Of course, it is important to consult your physician before trying CBD or any other treatment.

58c9d2dfc1618467103160c303f09ebf.jpeg


Mary Beth Skylis is a freelance writer specializing in personal narratives. Her work focuses on the outdoors and the transformative powers of nature. When her dad was diagnosed with Parkinson's in 2013, her search to understand the disease materialized through language. She now writes to help others understand the disease and hopes to teach how to be a compassionate caregiver.

 
Last edited:
tingri-everest-base-camp-trek.jpg



Psychedelics as prospective therapeutics for neurodegenerative disorders*

Urszula Kozlowska, Charles Nichols, Kalina Wiatr, Maciej Figiel | https://doi.org/10.1111/jnc.15509 | 13 September 2021

The studies of psychedelics, especially psychedelic tryptamines like psilocybin, are rapidly gaining interest in neuroscience research. Much of this interest stems from recent clinical studies demonstrating that they have a unique ability to improve the debilitating symptoms of major depressive disorder (MDD) long-term after only a single treatment. Indeed, the Food and Drug Administration (FDA) has recently designated two Phase III clinical trials studying the ability of psilocybin to treat forms of MDD with "Breakthrough Therapy" status. If successful, the use of psychedelics to treat psychiatric diseases like depression would be revolutionary. As more evidence appears in the scientific literature to support their use in psychiatry to treat MDD on and substance use disorders (SUD), recent studies with rodents revealed that their therapeutic effects might extend beyond treating MDD and SUD. For example, psychedelics may have efficacy in the treatment and prevention of brain injury and neurodegenerative diseases such as Alzheimer's Disease. Preclinical work has highlighted psychedelics’ ability to induce neuroplasticity and synaptogenesis, and neural progenitor cell proliferation. Psychedelics may also act as immunomodulators by reducing levels of proinflammatory biomarkers, including IL-1β, IL-6, and tumor necrosis factor-α (TNF-α). Their exact molecular mechanisms, and induction of cellular interactions, especially between neural and glial cells, leading to therapeutic efficacy, remain to be determined. In this review, we discuss recent findings and information on how psychedelics may act therapeutically on cells within the central nervous system (CNS) during brain injuries and neurodegenerative diseases.
------------------------------------------------------------------------------​

INTRODUCTION

We are in the midst of a renaissance of research into a class of drugs named psychedelics. This class of drugs was made illegal to use or possess worldwide in the late 1960s, but is now making a comeback as a possible clinical therapy for treating psychiatric conditions such as treatment-resistant depression (TRD), post-traumatic stress disorder (PTSD), and other neuropsychiatric diseases. There is no doubt that psychedelics influence essential functions of the Central Nervous System (CNS). Therefore, they are increasingly recognized and being studied as therapeutic agents for psychiatric disorders. In modern pharmacology, the term "psychedelic" refers to a class of CNS active drugs that primarily produce their effects through serotonin 5-HT2A receptor activation. Classic psychedelics are the natural products: psilocybin, DMT, 5-MeO-DMT, mescaline, and the semi-synthetic ergot derivative lysergic acid diethylamide (LSD). Non-classic psychedelics are newer derivatives of these classic compounds and also include DOx and 2C compounds such as (R)-DOI and 2C-B. CNS active drugs that can produce similar perceptual alterations such as ketamine, MDMA, and THC are not pharmacologically considered psychedelics because their effects are not mediated primarily through the 5-HT2A receptor. However, recent phase-III-clinical trial data on MDMA-assisted psychotherapy demonstrated therapeutic benefits in patients with severe PTSD.

Research using psychedelics was essentially banned worldwide in the late 1960s and early 1970s, and this class of drug labeled dangerous with no medical value. Fortunately, research in this field has gained interest in recent years, and clinical trials in several areas show promise for these drugs as potential new therapeutics. For instance, so-called "magic mushrooms" are a well-known natural source of the classic psychedelic tryptamine psilocybin. Although known and used for millennia, psilocybin itself was not isolated until 1957 by Albert Hoffman from Psylocibe mexicana, who first synthesized it in 1958. Psilocybin itself is a prodrug, rapidly converted to the active form, psilocin, in the body. Another classic psychedelic compound, DMT, is found in significant concentrations in several plants such as Mimosa tenuiflora, Psychotria viridis, and Diplopterys cabrerana, among others. It is also produced in the mammalian body but at low levels. DMT was first synthesized in 1931 and isolated in 1942 from M. tenuiflora by Oswaldo Gonçalves de Lima. Its psychoactive properties, however, were not confirmed until 1956. The β-carboline and monoamine oxidase inhibitors (MAOi) harmine, tetrahydroharmine, and harmaline in Banisteropsis caapi are often used to facilitate the oral activity of DMT in the Amazonian brew ayahuasca, which has also recently been studied for therapeutic benefits.

This review will discuss the current state of the art of how psychedelics influence neural tissue homeostasis and activity. We hypothesize that psychedelics can also be used as therapeutics in the treatment of neurodegenerative diseases and brain injuries. We will mainly focus on neuroimmunology and how data from recent research in the context of neuroinflammation support the hypothesis that psychedelics may have a beneficial outcome in restoring the balance of neural tissue function. In this context, we will also discuss psychedelic-induced neuroplasticity, neurogenesis, and gliogenesis. We propose that psychedelic research in studies of neurodegeneration may be beneficial for future development in this field. We hope that this review will provide information useful to support future psychedelic research in the area of regenerative medicine and the treatment of neurodegenerative disorders and brain injuries.

PSYCHEDELIC INDUCED NEUROPLASTICITY AND NEUROGENESIS

The term "neural plasticity" describes changes in functional neural connectivity. The mechanisms are mostly associated with neural cells, but the process reaches beyond the plasticity of neural synapses. The adaptive changes in the fMRI-measurable macro-scale come from changes in local micro-scales within multicellular interactions, involving neurons, astrocytes, microglia, and oligodendrocytes. These cellular interactions are characterized by complicated homeostatic processes employing both paracrine and direct cell-to-cell communication. Neural plasticity is still poorly understood, but some mechanisms have already been described. Psychedelics may induce a so-called elevated brain entropy state, resulting in an increased ability to learn and "unlearn" certain information. Such action may be therapeutic, and is likely associated with increased neural plasticity mechanisms at the cellular level. Acute changes in the density and complexity of synaptic architecture induced by psychedelics and 5-HT2A receptor activation have been shown by several investigators in both in vitro and in vivo models. These changes involve multiple mechanisms. For example, increases in spine density and morphology can involve direct signaling downstream of 5-HT2A receptor stimulation by psychedelics through serotonylation and activation of Rac1 and kalirin-7, or indirect modulation of synaptic architecture by elevated glutamate levels acting through BDNF/TrkB and mTOR signaling. A feature of psychedelic therapy is the long-lasting effect after only a single treatment. The reason for this is unclear, but likely involves changes in gene expression and/or epigenetic factors underlying the maintenance of neural processes normalized by treatment. There are several known genes involved in synaptic plasticity whose expression is changed in response to psychedelics. Neurogenesis may also be a factor; the administration of DMT induces neural progenitor cells proliferation and adult hippocampal neurogenesis in vivo via activation of Sigma-1 receptors in C57BL/6 mouse. Taken together, neurotrophic signaling and neuroplasticity promoting pathways activated by psychedelics are hypothesized to be key to the mechanism(s) of action for therapeutic effect(s).

A potential key mechanistic component not taken into account for nearly all proposed models is the involvement of microglia for therapeutic effect, as most if not all attention has been focused on neurons in the mechanism of action of psilocybin and other psychedelics. Microglia are tissue-specific, self-renewable CNS macrophage-like cells that are different from other cell types since they appear in the brain and spinal cord during fetal development in the process of primitive hematopoiesis. During their life-long residency inside the CNS environment, microglia assume specific immune cell characteristics and functions. Microglia are very mobile, continually scanning the environment, ready to respond to injury and infections, and take an active part in synaptic rearrangement and neural tissue regeneration. They modulate the deletion of unnecessary connections and the formation of new ones. It is tempting to speculate that psychedelics may stimulate neural plasticity through microglia regulation, especially since many receptors targeted by certain psychedelics like psilocybin and LSD are also present on microglia, including 5-HT2A, 5-HT2B, and 5-HT7 receptors, and the Sigma-1 receptor. Interestingly, in vitro application of DMT and 5-MeO-DMT to monocyte-derived dendritic cells (moDCs) reduce mRNA and protein expression of IL-1β, IL-6, TNF-α, IL-8, and increase expression of regulatory and tolerogenic IL-10.

Another interesting phenomenon is the reciprocity in the dynamics of neuron–microglia interactions. For instance, activation of NMDA receptors on a single neuron's dendrites can stimulate the growth of microglial extensions. Further research may help better understand how neuronal–microglia interactions affect learning and memory, neurodegeneration, and possibly the progression of certain mental illnesses. Microglia may be regulating synaptic pruning or growth by signals from neurons themselves. These regulatory signals may rely on the electrochemical transmission or the complement system, which is also involved in the process of synaptic pruning. During this process, unnecessary synapses are tagged with specific complement proteins that are detected and phagocytized by microglia. Errors in this process during childhood may lead to the development of autism, schizophrenia, or mental retardation, and in adult also result in degenerative diseases. The cellular phenotype and activity of microglia, the involvement of the complement system, and the neuronal signals relevant to synaptic plasticity upon psychedelics stimulation are probably critical aspects of the psychedelic therapeutic mechanism.

Our unpublished data suggest that psilocin increases the protein expression of triggering receptor expressed on myeloid cells 2 (TREM2) on microglia while reducing p65, TLR4, and CD80 proinflammatory markers. TREM2 is involved in the regulation of several microglial functions, including phagocytosis and synaptic refinement. Microglia deficient in TREM2 expression results in synaptic pruning defects, increased excitatory neurotransmission, and reduced long-range functional connectivity. The down-regulation of TREM2 was also observed in brain samples of patients suffering refractory epilepsy. According to our pilot data, psychedelics may prevent neuronal damage in microglia-neuron co-culture, however, it is presently unknown if the protective mechanisms of psychedelics are mediated by microglial TREM2.

PATHOLOGICAL MECHANISMS IN NEURODEGENERATION—A POTENTIAL TARGET FOR PSYCHEDELICS

Because the brain is very fragile and hardly an accessible organ, only limited therapeutic approaches can be proposed for the treatment of brain-specific neurodegeneration. These can be pharmacological, stem cell, or gene therapy approaches. Unfortunately, results to date with these approaches have not been very successful. Interestingly, one recently proposed solution is the application of traditional psychiatric drugs because they have been shown to prevent neural loss and stimulate neurogenesis, Neuroprotection and induction of neurogenesis may be a fruitful avenue to treat MDD as the histopathology of depressed individuals sometimes show signs of subtle neurodegeneration. For example, post-mortem brain studies have revealed neural loss and atrophy in the prefrontal cortex (PFC) and the hippocampus. Neural protective mechanisms (e.g., neuroprotection, neurogenesis, neuroplasticity) have been shown to be induced by psychedelics, which are effective in the treatment of MDD. Here, we discuss pathologies that occur in neurodegenerative disorders that may potentially be targeted by psychedelics for therapeutic effect. We speculate that their application at early disease stages may result in the delay of pathological symptoms.

Oxidative cell damage is often a reported in brain-specific neurodegeneration. This damage usually occurs because of an imbalance between free radicals, reactive oxygen species (ROS), and reactive nitrogen species (RNS), and the presence of antioxidants and antioxidative proteins, such as superoxide dismutases (SOD), hioredoxin peroxidases (TRXPs), glutathione peroxidases (GPXs). In a typical situation, if reactive species are held in balance, they play an essential role in regulating essential cellular processes including phagocytosis, apoptosis, and cellular signaling. However, when cells are unable to neutralize excesses of reactive molecules, these molecules may induce damage to mitochondria, cellular and nucleolar membrane, and DNA, and over time result in organ and/or tissue degeneration. Elevated oxidative stress and disruption in redox balance are observed in many psychiatric conditions such as MDD, schizophernia, bipolar disorder, anxiety disorder. Importantly, disruption of redox homeostasis occurs in the pathology of ALS, PD, AD, and DNA repeat expansion disorders such as HD and SCAs.

In the psychedelic brew ayahuasca, two components, harmine and harmaline, are monoamine oxidase inhibitors with antioxidant properties and have the capability to induce gliogenesis and neural progenitor cell migration. Another anti-oxidative effect of psychedelics such as psilocybin and/or DMT may come as a result of 5-HT1A receptor activation. The 5-HT1A receptor agonist 8-OH-DPAT induces expression of the anti-oxidative factor metallothionein-1/-2 (MT-1/-2) and Nfr2. In retina pigment epithelial cell line (ARPE-19), 8-OH-DPAT reduces damage caused by paraquat, an oxidative herbal agent, through elevation of MT1, heme oxygenase-1 (HO1), NAD(P)H: quinone acceptor oxidoreductase 1 (NqO1), superoxide dismutase 1 and 2 (SOD1, SOD2), and catalase (Cat) mRNA expression. 8-OH-DPAT also reduces oxidative stress damage in retinal pigment epithelium/choroid in Sod2 knockout mice.

Endoplasmatic reticulum stress (ERS)

Some psychedelics (e.g., DMT) target the Sigma-1 receptor, which is reported to protect cells from various insults. ER stress induces up-regulation of Sigma-1 expression and modulates the action of PERK, IRE1α, and ATF6 proteins in mitochondria-associated membrane (MAM). Stimulation of Sigma-1 may prevent ERS-mediated cellular apoptosis by regulation of ATF4, ATF6/ C/EBP homologous protein (CHOP), and the balance between Bax and Bcl-2 in granulosa cells. Because ERS damage is reported in MDD and several neurodegenerative disorders, targeting Sigma-1 receptors with psychedelics is proposed as a novel therapeutic strategy.

Blood–brain barrier disruption

The vasculature system in the brain is equipped with a special feature called the blood–brain barrier (BBB). The BBB is composed of a tight layer of astrocytes, is selectively permeable, and separates the intracerebral circulatory system from the peripheral blood to protect the brain against chemical and biological insults. The BBB also contains other cells types, such as microglia cells, perivascular macrophages, and pericytes. The whole structure is embedded in the basal membrane, with extracellular matrix secreted by endothelial cells and pericytes. The endothelial cells inside the blood capillaries form tight junctions (TJ), multi-protein complexes composed of occludins, claudins, and tight junction proteins ZO-1, -2, -3. Breakdown of this system is associated with brain-specific damage and neurodegeneration, and may be the cause of serious illness. Breakdown can originate from prolonged exposure to oxidative stress and/or immune cell activity. For example, microglia inflammatory cytokines acting via IL-1β on Sonic Hedgehog (SHH) can down-regulate tight-junction proteins in astrocytes, resulting in BBB leakage. Moreover, suppressing SHH in astrocytes leads to increased secretion of proinflammatory chemotactic proteins and immune cell activation. Microglia also secrete IL-1β via inflammasome-dependent mechanisms in response to proinflammatory cytokines, DAMPS, β-amyloid, or other toxic protein aggregates. In a C57BL/6 healthy male mouse model, chronic social stress causes BBB disruption via claudin-5 down-regulation, which leads to the infiltration of proinflammatory factors and depression-like behaviors. Disruption of the BBB is also observed in a genetic mouse model of schizophrenia, and may be involved in bipolar disorder pathology. BBB disruption is also observed in multiple neurodegenerative disorders including ALS, AD, and SCA3.

Conceptually, inflammation-based BBB leakage could be prevented to some degree by the presence of psychedelics. The drugs N,N-DMT and 5-MeO-DMT, applied into LPS and polyI:C – activated dendritic cells in vitro, result in down-regulation of expression of IL-1β, IL-6, IL-8, TNF-α, and up-regulation of IL-10 as measured by mRNA and protein expression through stimulation of Sigma-1 receptors. This observation has also been confirmed in vivo in a Wistar rat model of stroke, where N,N-DMT administration significantly decreased IL-1β, IL-6, and TNF-α, but increased IL-10, measured by mRNA and protein expression. Furthermore, N,N-DMT-treated rats demonstrate improved motor skills post-stroke. Although not validated yet in brain tissues, several psychedelics, including DOI, LSD, and psilocybin, have been shown to have potent anti-inflammatory effects of suppressing many of these same proinflammatory biomarkers in peripheral tissues, and they may represent effective therapies for inflammation-related neuropathologies.

Oligodendrocyte pathology

Oligodendrocytes protect and support neurons and their axons by providing myelin that improves electric signal transmission. Unfortunately, their active role in immune response and neural regeneration has long been overlooked. Microglia and oligodendrocytes actively work to regulate each other's functions. Moreover, oligodendrocytes’ pathology occurs in many neurodegenerative diseases, including Alzheimer's and Parkinson's Disease, ALS, Multiple Sclerosis, Spinal Cord Injury, but also non-degenerative psychiatric conditions including MDD, schizophrenia, and Alcohol Use Disorder. For example, in MDD, abnormalities in oligodendrocyte density are observed in the PFC and amygdala. Interestingly, oligodendrocytes are extremely vulnerable to oxidative stress and prolonged exposure to proinflammatory factors secreted by microglia. The previously discussed psychedelic-mediated reduction in cytokine secretions may play a protective role in myelin and oligodendrocyte cell survival. Certain psychedelics can target Sigma-1 receptors, which are essential in stimulating OPC differentials. Together, these findings indicate that more attention should be paid to the influence of psychedelics on oligodendrocyte biology.
PSYCHEDELICS AS IMMUNOMODULATORS

The study of psychedelics at target receptors and the activation of effector pathways have brought new, but still limited, insights into their immunomodulatory potential (see Table 1). Classic psychedelics like LSD, DMT, 5-Meo-DMT, and psilocin have the potential to interact with several 5-HT receptor subtypes, Sigma-1R, and TAAR, which are present in CNS and other tissues, including cells of innate and adaptive immunity like macrophages, monocytes, dendritic cells, and T cells. These receptors are mediators of immunological response, and serotonin is considered a critical factor in immune homeostasis. Therefore, psychedelics can regulate both adaptive and innate immune responses. A review of putative molecular mechanisms in which psychedelics may act as immunomodulators was published by Szabo, emphasizing cross-talk between pattern recognition receptors (PRR), such as Toll-Like Receptor 4 (TLR4), 5-HTRs, and Sigma-1R, and regulation of inflammatory response via NFκB/IRF signal transduction pathways. Although these modulations result in changes, Sigma-1, and TAAR seem to play a crucial role in immune response, and all three of them can be stimulated by psychedelics.
Psychedelic ligandsReceptorEffectLiterature
(R)- DOI5-HT2AIn smooth muscle cells in vitro:

Prevention of Nf-κβ nuclear translocation and inhibition of nitric-oxide synthase activity

In aortic arch and small intestine in vivo:

Down-regulation of TNF-α-mediated Cx3CL1, Icam-1, Vcam-1, MCP-1, IL-6, IL-1β

In OVA-treated asthma-model lung in vivo:

Suppression of Th2-related genes: Mcp-1, Il-13, Il-5, and Gm-csf,

Inhibition of neutrophil infiltration,

Inhibition of mucus hyperproduction
Yu et al. (2008)

Nau et al. (2013)

Nau et al. (2015
DMT,

5-MeO-DMT
Sigma-1Down-regulation of: IL-1 β, IL-6, TNF-α, IL-8

Up-regulation of: IL-10

(gene and protein expression)

Decrease of Th1 and Th17 activation after E. coli or H1N1 co-culture
Szabo et al. (2014)
5-MeO-DMT5-HT2A (?)

5-HT2c (?)
In human cerebral organoid in vitro model:

Down-regulation of Nf-κβ pathways (involved in the immune response)

Down-regulation of Nuclear Factor of Activated T cells (NFAT) (T-cell activation, stem cell differentiation)

Modulation of Gaq-Rho-ROCK pathway (cytoskeletal rearrangement, phagocytosis)
Dakic et al. (2017)
DOI, DMT, Psilocin5-HT2B(Reported with BW723C86 5-HT2B ligand)

In CD1+ moDC down-regulation of: TNF-α, IL-6, IL-8/CXCL8/ IP-10/CXCL10 after TLR2, and TLR6/7 activation,

In TLR3-activated CD1+ moDC: down-regulation of CD80, CD83, CD86 (anti-inflammatory, tolerogenic)

Prevention of Th1, Th17 lymphocyte polarization
Szabo et al. (2018)
DMTSigma-1In in vivo model of stroke:

Down-regulation of: IL-1β, IL-6, IL-8, TNF-α, NOS, APAF-1 (proinflammatory, proapoptotic)

Up-regulation of BDNF and IL-10 (tolerogenic, neurogenic)
Nardai et al. (2020)
(Hypothetically) DOI, DMT, Psilocin, LSD, mescalineTAAR1, TAAR2(Not tested yet with psychedelics)

Lymphocyte migration, increase in IL-4 secretion, Th1/Th2/Th3 phenotype modulation, mediation of IgE-secretion
Babusyte et al. (2013)

5.1 5-HT receptors

Serotonin is one of the most critical factors during fetal brain development and neurogenesis, and is responsible for the formation of axons and dendrites, and adult axonal regeneration. Serotonin receptors are present on most, if not all, types of cells in the CNS. In neurons, for example, their activation can influence cellular membrane polarization states through multiple mechanisms. Serotonin also plays significant roles aside from being a neurotransmitter. There are several receptor subtypes expressed in mammalian peripheral tissues and cells outside the CNS, including adaptive and innate immune cells. Serotonin itself has an endocrine effect on the regulation of whole-body homeostasis, such as heart rate, intestinal motility, and last but not least: the immune response.

Although 5-HTRs are primarily described as activators of proinflammatory pathways, they surprisingly have anti-inflammatory properties when activated by certain, but not all, psychedelics. The selective 5-HT2 receptor agonist (R)-DOI, reduces mRNA expression of proinflammatory adhesion molecules ICAM-1 and VCAM-1 as well as mRNA levels for proinflammatory cytokines MCP1, IL-1β, and IL-6 in various tissues like intestine and aorta, and circulating levels of IL-6 in TNF-α treated mice. Several of these findings were confirmed in a high fat-fed ApoE−/− knockout moue model of cardiovascular and metabolic disease. An increase in levels of VCAM-1, IL-6, and MCP-1 mRNA expression was observed in animals fed a high-fat and -cholesterol "Western diet" compared to control mice fed regular food, and this increase was prevented in mice fed the Western diet and treated with (R)-DOI. The precise mechanism underlying why 5-HT2A receptors, which are widely described as inflammation inducers, induce anti-inflammatory processes after activated by (R)-DOI and some other psychedelics is not known. The hypothesis proposed by Flanagan and Nichols involves the concept of functional selectivity, in which different ligands induce slightly different conformations of the receptor to recruit different sets of effector pathways. Psychedelics are hypothesized to recruit and activate anti-inflammatory effector signaling pathways, whereas serotonin itself recruits proinflammatory pathways. In rodent models of allergic asthma, nasal administration of (R)-DOI at a very low dose (EC50: ~0.005 mg/kg) completely prevents symptoms, including airways hyperresponsiveness, pulmonary inflammation, and mucus overproduction in response to allergen. Further examination of the lung tissue revealed prevention of eosinophilia and a reduction in Th2 cell recruitment. Interestingly, the behavioral potency of different psychedelics does not correlate with anti-asthma efficacy. Significantly, therapeutic drug levels in these models are orders of magnitude lower than the levels necessary to induce measurable behavioral responses. These findings suggest that subperceptual levels of some psychedelics may be an exciting alternative to currently available steroid drugs in the treatment of asthma and other inflammatory-related disease.

Given the high level of expression of 5-HT2A receptors in the brain on multiple cell types, it may be that psychedelics have similar anti-inflammatory properties against neuroinflammation. In vitro application of 5-MeO-DMT in human cerebellar organoids results in down-regulation of NF-κβ and nuclear factor of activated T cells (NFAT) pathways, as well as modulation of the Gαq-RhoA-ROCK pathway involved in cytoskeleton rearrangement and phagocytosis.

Classical psychedelics have mid affinity for and efficacy at 5-HT2B receptors. Interestingly, the activation of this receptor type with the agonist (BW723C86) regulates immune responses in CD1+ monocyte-derived dendritic cells (moDC). The application of BW723C86 resulted in down-regulation of CD80, CD83, and CD86 proinflammatory molecules on CD1+ moDC. Furthermore, stimulation of 5-HT2B down-regulates TLR2, TLR3, and TLR7/8-mediated proinflammatory cytokine protein expression. It also prevents moDC-mediated activation of T cells toward inflammatory Th1 and Th-17 phenotypes. Furthermore, certain immune stimulators such as the molecule polyI:C, which is a TLR3 agonist, up-regulate the expression 5-HT2B receptor protein. Together, these observations suggest that 5-HT2B agonism may participate in some aspects of their anti-inflammatory mechanism. However, in the allergic asthma model, (R)-DOI was not effective in reducing pulmonary inflammation in the 5-HT2A receptor knockout mouse indicating that for at least asthma and pulmonary inflammation 5-HT2A receptor activity is necessary and sufficient for therapeutic effect.

5.2 Sigma-1 receptor

The sigma-1R is a transmembrane protein located in mitochondria and the endoplasmatic reticulum (ER). It is abundantly present within the CNS in neurons, astrocytes, oligodendrocytes, and microglia, where it mediates a neuroprotective effect. Sigma-1R activity promotes neural function and survival via modulation of Ca2+ homeostasis, mitigation of oxidative stress, regulation of gliosis, neuroplasticity, and glutamate activity.

Stimulation of Sigma-1R in oligodendrocyte progenitor cells (OPC) results in oligodendrocyte differentiation (Hayashi & Su, 2004), and stimulation in astrocytes improves the BDNF secretion. This suggests that targeting Sigma-1R may be a promising therapeutic strategy for psychiatric and neurodegenerative conditions. Interestingly, Sigma-1R activity is involved in the transition between M1-like proinflammatory and M2-like proregenerative and tolerogenic microglia phenotypes. These mechanisms are not very well understood. Moreover, as microglia are cells of complicated biology and are somewhat difficult to study, the concept of M1/M2 polarization may be too simplistic to address many aspects of microglia function. It has been recently proposed that microglia displaying a proinflammatory phenotype are crucial for their role in neural tissue reorganization and regeneration.

DMT is an agonist of Sigma-1Rs, and found to be produced by specific tissues in the brain. Szabo et al. observed that the application of DMT into human iPSC-derived cortical neuron cultures in vitro resulted in better survival under hypoxia conditions, but that the protective effect vanished after Sigma-1R gene knockdown with siRNA. This suggests that DMT can protect cells from hypoxia-induced apoptosis via Sigma-1 receptor stimulation. This observation was later confirmed in an in vivo rat model of stroke, where continuous administration of DMT reduced the size and number of lesions, and decreased levels of IL-1β while up-regulating IL-10, and BDNF protein and mRNA levels. Similar results were found with another Sigma-1 agonist (PRE-084) after embolic stroke to significantly reduce the size of lesions, improve neuronal deficits, and reduce concentrations of some proinflammatory cytokines while elevating levels of some anti-inflammatory cytokines like IL-10. Interestingly, application of the Sigma-1R selective antagonist (MR309), had similar neuroprotective effects. It is tempting to speculate that elevated anti-inflammatory cytokine levels after DMT administration in these stroke models may be caused by Sigma-1R mediated changes in microglia phenotypes. For example, in a study by Moritz et al., stimulation of Sigma-1R "switched off" activated microglia and made them migrate away from the location of damaged tissue. Moreover, in the LPS-treated microglial BV2 cell line, application of Sigma-1R agonist SKF83959 results in the prevention of M1-like phenotype switching by microglia, and a decrease in TNF-α, IL-1β, and inducible NOS levels. A similar effect was reported in a model of traumatic brain injury (TBI), and Parkinson's Disease.

TAAR

Trace amine-associated receptors (TAARs) are G-protein-coupled receptors abundantly present in the CNS. In most vertebrates, they exist in nine isoforms. Only TAAR1 has been studied in-depth, however. This receptor is relatively non-selective and has an affinity for endogenous trace amines as well as the classical neurotransmitters serotonin and dopamine, and multiple psychoactive drugs, including amphetamines, ergoline derivatives, psilocin, DMT, and mescaline. TAAR1 is a modulator of neurotransmission induced by canonical dopamine, serotonin, and glutamine receptors, and its aberrations and rare variants may contribute to the etiology of schizophrenia. TAAR1 is also expressed in non-CNS tissues such as the thyroid, stomach, pancreas, and intestine, where it may regulate body functions in an endocrine manner.

Although abnormalities in TAARs expression or function may be related to the development of schizophrenia, data suggest involvement in additional neuropsychiatric conditions. For example, stimulation of TAAR1 in an experimental model of Parkinson's Disease results in L-DOPA-related dyskinesias, and TAAR1 knockout mice are reported to be more vulnerable to various substance addiction. Targeting TAAR1 has also been suggested as a possible therapeutic target for the treatment of bipolar depression, fibromyalgia syndrome, and diabetes.

TAAR are found in immune cells and can elicit immunomodulatory effects; however, our knowledge about TAARs and immune responses is limited. TAAR1 is expressed in polymorphonuclear leukocytes (PMN), T cells, and B cells, whereas TAAR2 is also abundant on NK cells and monocytes. In T cells, stimulation of TAAR1 and TAAR2 receptors induce IL-4 production and modulation of Th1, Th2, and Th3 markers, whereas silencing of these receptors reduces IgE secretion in B cells after induction with trace amines. TAAR1 and TAAR2 are also reported to be involved in PMN chemotactic migration. DMT, (R)-DOI, d-LSD, and 5-MeO-DMT are TAAR1 agonists, and it is, therefore, possible that psychedelics may regulate immune cells to respond and regulate neural tissue homeostasis via TAAR1 activation.

RESEARCH PERSPECTIVES FOR PSYCHEDELICS IN PREVENTING NEURODEGENERATION

In this review, we have highlighted the beneficial outcomes of psychedelic treatment for MDD. Our primary focus was on processes in neural tissue microenvironments which can be affected by psychedelics. These include the induction of neurogenesis and neuroplasticity and reduction of inflammation and oxidative stress. These characteristics of psychedelics may play crucial roles in restoring long-term healthy homeostasis in depressed patients. We also emphasized potential areas of therapeutic actions in brain-specific neurodegeneration in which psychedelics may be beneficial. These include oxidative stress, inflammation, BBB disruption, and loss of oligodendrocytes and myelin.

In Europe, around 7 million people suffer from dementia-related disorders, and the aging of society is expected to double this number by 2040. Around 15 mln people worldwide and 1,12 mln in Europe experience a stroke every year, with 5 mln of those being fatal incidents (650 000 in Europe), and another 5 mln of patients suffer post-stroke severe disability. Each year 10 mln new dementia cases are being diagnosed globally, and 60–70% of them are Alzheimer's Disease. In 2015 ALS was diagnosed in 222 801 people worldwide, and that number is predicted to grow by 69%, reaching 376 674 new ALS cases annually by 2040. Therefore, neurodegenerative diseases are a serious and growing burden for modern societies. Moreover, the development of effective therapeutics lags behind other fields such as cardiovascular diseases and cancer. Therefore it is a top priority to search for novel candidates for therapeutic approaches to revert these dire statistics.

Psychedelics represent such a novel approach. Certain psychedelics, with demonstrated therapeutic efficacy for psychiatric disorders in clinical trials, have been used safely for centuries by indigenous populations. The beneficial therapeutic dosages of these substances have been shown to be well-tolerated, and they present a favorable safety profile in treating a variety of disorders. Clinical trials are investigating therapeutic efficacy for anorexia nervosa, the early stage of Alzheimer's Disease, and traumatic brain injury, among other CNS disorders.

Psychedelics (e.g., DOI, DMT, LSD) promote structural plasticity via BDNF signaling and are thus proposed as potential therapeutics for MDD and related disorders. Neuropsychiatric disorders are well known to be associated with atrophy of neurons and abnormal neuronal circuits. Among neurodegenerative disorders, commonalities in pathological characteristics may be seen in polyQ disorders such as spinocerebellar ataxia type 3 (SCA3).

Similar to many other neurodegenerative disorders, the down-regulation of BDNF is observed in SCA3 cells and in dentate neurons of SCA3 patients. Moreover, several essential proteins belonging to the BDNF signaling pathway are also down-regulated in mouse models. For example, Rac1, acting downstream of BDNF and TrkB, is down-regulated in the cerebral cortex and cerebellum of young SCA3 mice. Together with another down-regulated protein, RhoA, Rac1 mediates proplasticity properties evoked by BDNF by facilitating sLTP (structural long-term potentiation) and regulating actin cytoskeleton in dendritic spines. Furthermore, MAPK1 (a.k.a. Erk) and MAP2K1 are also down-regulated in the young SCA3 mice, and Erk signaling is down-regulated in MDD. Importantly, BDNF is an immediate upstream regulator of the MAPK (mitogen-activated protein kinase) cascade. Activation of MAPK (ERK) signaling by neurotrophins is involved in long-term synaptic plasticity and the structural remodeling of the spines in the excitatory synapses.

Another protein necessary for BDNF signal transduction to the nucleus is Pea15, which is also down-regulated in SCA3 mice. Silencing of Pea15 results in inhibition of BDNF retrograde signaling. Pea15 acts as a scaffolding protein for PLD1, RSK2, and ERK1/2, and the formation of this complex is triggered by BDNF in cortical neurons. Therefore, the potential use of psychedelics could affect the levels of several proteins, which are down-regulated in the SCA3 model probably by an increase in BDNF which may promote synaptic plasticity. The role of BDNF signaling in the survival of neurons has been well documented in other neurodegenerative disorders, such as Huntington's, Parkinson's, and Alzheimer's disease.

Psychedelics have also been shown to reduce oxidative stress, which is a significant issue in neurodegenerative disorders. Several oxidative stress biomarkers are elevated in models of neurodegenerative disorders. Other anti-oxidative proteins, which play an essential role in reducing oxidative stress by breaking down ROS (Gstp1, Sod2, Fth1), are down-regulated in the SCA3 model. Moreover, Txn down-regulation in SCA3 mice might increase the vulnerability of neurons to ROS. Therefore, psychedelics’ anti-oxidative properties could be a beneficial component of a therapeutic strategy for SCA3 and other neurodegenerative disorders.

The strategy of serotonergic signaling modulation by psychedelics in SCA3 is also strongly supported by studies showing a therapeutic effect for SCA3 through 5-HT1AR activity, which is activated by several psychedelics including psilocybin and LSD. Targeting of the 5-HT1A serotonin receptor orthologue SER-4 in C. elegans ameliorates motor dysfunction and reduced mutant ATXN3 aggregation. Furthermore, treatment with partial agonists of 5-HT1A receptors has been demonstrated to reduce ataxia, pain, insomnia, and depressive symptoms in patients with SCA3 and other forms of SCA. The SSRI citalopram has beneficial therapeutic effects in animal models of SCA3 and preclinical trials. Thus, activation of serotonergic signaling in SCA3 patients with psychedelic agents is a promising therapeutic strategy.

Non-hallucinogenic psychedelics approach

The “hallucinogenic” effects of psychedelics have been proposed to be directly associated with their therapeutic potential in psychedelic-assisted psychotherapeutical approach as the subjective peak intensity has a high correlation with therapeutic efficacy. However, in patients suffering brain-specific neurodegeneration, psychotropic effects of psychedelics may be a serious limitation, especially if because of the disease physiology, the medicine would have to be administrated more often and in higher doses. Furthermore, correlation is not causation, and subjective peak experiences may merely indicate that sufficient drug has been administered to produce therapeutic efficacy at cellular and molecular targets and circuits. Although ibogaine is not classified as a psychedelic, it is a type of hallucinogen that may have therapeutic efficacy to treat substance use disorder. Recently, analogs of ibogaine have been reported by two different investigators to attenuate behaviors associated with substance abuse in rodent models, in a similar way to ibogaine. One is peer reviewed, and the other awaiting peer review. Because of the low toxicity and lack of hallucinogenic properties of these new molecules, they represent potential non-hallucinogenic derivatives of hallucinogenic parent molecules with therapeutic effect. With regard to psychedelics, their demonstrated potency in multiple animal models of disease suggests it may not be necessary to eliminate hallucinogenic behaviors from effective molecules because the dose is so low that effects on behaviors would not be seen at relevant therapeutic levels. Regardless, work by Flanagan et al. suggest that it may be possible to engineer hallucinogenic effects away from therapeutic effects to develop non-hallucinogenic 5-HT2A receptor agonists with anti-inflammatory potential. Development of a 5-HT2A receptor agonist therapeutic devoid of hallucinogenic effects would conceivably allow higher levels to be used, especially for those with weaker potencies, which may allow for greater efficacy in certain circumstances.

Prospect implications for regenerative medicine

Besides the prevention and regulation of pathology in neurodegenerative disease, the immunomodulatory properties of psychedelics may also be relevant to regenerative medicine. Neural Stem/Progenitor Cell (NSC/NPC) transplantation is a recently developed and promising therapeutic tool. However, the limitation of such a strategy is poor graft survival because of immune response. Our recent study revealed that DMT and psilocin down-regulate CD80 co-stimulatory molecule expression on the surface of microglial cells, with and without LPS stimulation. The co-stimulatory signal is crucial for recruiting adaptive immune cells; therefore, blockade of co-stimulatory molecules an attractive immunosuppressive strategy. The unique properties of psychedelics in suppressing inflammatory responses, and promoting neural survival and plasticity, could be a strong rationale for the hypothesis that psychedelics might support grafted cells and facilitate their survival for therapeutic benefits.

Therapeutic perspectives for microdosing

One dosing method of psychedelics is the use of so called “microdoses”—very low concentrations of various psychedelics that do not reach the threshold of perceivable behavioral effects. This is usually 10% of active recreational doses (e.g., 10–15 µg of LSD, or 0.1–0.3 g of dry “magic mushrooms”) taken up to three times per week. This regimen is popular in underground settings without medical guidance. Microdosing is believed to improve the creative thinking, cognitive function, and overall psychological well-being, and is described mostly in the context of self-application by healthy enthusiasts. There have been few rigorously controlled studies of microdosing, and the therapeutic effects of psychedelic microdoses for the treatment of psychiatric disorders are questionable. According to a self-blinding study involving 191 healthy volunteers, in which the mood changes were measured using various questionaries, the authors concluded that anecdotal psychological improvements are more likely associated with the placebo effect rather than drug effect. Furthermore, Family et al. reported that repeated administration of LSD (5–20 µg) in healthy individuals in a blinded placebo-controlled clinical trial produced no significant changes in several cognitive outcome measures. However, a recent study using fMRI showed that 13 µg of LSD changes connectivity inside the limbic circuits 90 min after drug administration compared to the placebo control, which was associated with positive mood changes as measured with a Positive and Negative Affect Schedule (PANAS). Each of these reports has been in healthy individuals, and there have been no rigorous and controlled studies to date on microdosing in patients with diagnosed depressive disorder. The application of psychedelic microdosing in the context of the treatment of brain-specific neurodegenerative disorders has not been yet directly investigated, however, researches speculate that it may influence the hippocampal neurogenesis. Importantly, a Phase I feasibility and safety study on repeated low-dose LSD administration has been conducted in an elderly healthy population in preparations for later phase clinical trials to treat Alzheimer's Disease.

CONCLUSION

Psychedelics stimulate neuro- and gliogenesis, reduce inflammation, and ameliorate oxidative stress. Therefore, they are promising candidates for future therapeutics for psychiatric, neurodegenerative, and movement disorders. Importantly, psychedelics hold the promise of being disease-modifying therapeutics, and not simply just providing symptomatic relief. Current clinical trials have demonstrated both safety and efficacy for their therapeutic use in controlled clinical settings, and psilocybin, has even been designated with “Breakthrough Therapy” status by the FDA in the United States for two different Phase III clinical trials. Often only just one or two therapeutic administrations produce profound and persistent effects. Preclinical research has shown promise in several disease models for both psychiatric and non-psychiatric diseases. Therefore, the use of psychedelics as therapeutics is very promising and should be further developed, paying special attention in the future to prospect applications in neurodegenerative diseases.

*From the article (including references) here :
 
Last edited:
c8853884-43fe-474d-b32e-061d9fcfe61c-GettyImages-593319838.jpg


Researchers focus on use of psilocybin to treat Parkinson’s and MS*

by Clara Furlong | Silo Pharmaceuticals | USA Today | 17 Feb 2021

In 2006, Johns Hopkins researchers published a groundbreaking study on the effects of a single dose of psilocybin, a naturally occurring psychedelic compound found in more than 200 species of mushrooms. The team had to set up “unusually rigorous scientific conditions and measures” amid highly restrictive regulatory policies, which were partially due to unfavorable media coverage from the recreational use of “shrooms” in the 1960s and ‘70s that had resulted in misperceptions of risk.

The landmark study provided a scientific framework to better understand how hallucinogenic drugs target the brain’s serotonin receptors, and the researchers announced future plans to investigate how psilocybin might be used to treat conditions like cancer-related anxiety and depression, and other mental health conditions. Since then, more than 60 peer-reviewed articles in scientific journals further validated the safety and enduring positive effects of a unique class of pharmacological compounds known as psychedelics.

The US Food and Drug Administration (FDA) has since granted breakthrough therapy designation for psilocybin to determine its viability for treating major depressive disorder, opening enrollment to patients for clinical studies in 2019.

5c1ea3a0-855d-4f64-abc0-2e274f015b5f-GettyImages-1211251035_1.jpg


“Psilocybin therapy studies have had a promising start, but research is still in its infancy,” said Dr. Josh Wooley, an Associate Professor in the Department of Psychiatry and Behavioral Sciences at the University of California, San Francisco (UCSF). His Bonding and Attunement in Neuropsychiatric Disorders (BAND) Laboratory is studying why people with mental illnesses have trouble with social connection.

Dr. Wooley recently joined the scientific advisory board of Silo Pharmaceuticals, a development stage biopharmaceutical company that focuses on merging traditional therapeutics with psychedelic research.

“Our mission here at Silo Pharmaceuticals is to identify assets to license and fund the research which we believe will be transformative to the well-being of patients and the health care industry,” said Eric Weisblum, its Chairman and Chief Executive Officer.

18628214-f2a8-499b-95fd-5e03469cf60e-GettyImages-1266746971.jpg


Silo Pharmaceuticals is addressing an enormous need to develop and test novel treatments for major depressive disorder, bipolar depression, chronic pain and mood symptoms associated with Parkinson's disease. Researchers believe Psilocybin therapy under supervised care may be a more effective treatment and offer patients an avenue to mainstream back into society.

“Our approach is unique in that we have already filed four provisional patents for the delivery of anti-inflammatory therapeutics couples with psilocybin,” added Weisblum. He is actively guiding his company through the different stages of research, licensing, partnerships and eventually clinical trials.

Silo Pharmaceuticals currently has the exclusive rights to license patents owned by the University of Maryland Baltimore for the treatment of neuroinflammatory disease and can utilize the university’s research for potential multiple sclerosis treatments by delivering psilocybin to the central nervous system.

2acd4537-c045-4ac7-9610-1b5e8191168b-GettyImages-1182436133.jpg


Silo Pharmaceuticals has also entered into an investigator-sponsored study agreement with Maastricht University of the Netherlands for the use of psilocybin and LSD to treat Parkinson’s disease.

For Weisblum, working in this industry has been particularly personal.

“I’ve watched one of my best friends’ father-in-law suffer through the diagnosis, getting support and education from the community, and living life to the fullest before eventually losing the battle," he said. “A close friend of mine who is a doctor lost both of his parents to Parkinson’s. With so many lives impacted by this disease, I was moved to do my part and to shepherd the financial resources of my company to improve quality of life.”

“Every member of our advisory board and management team is fully committed to this vision,”
said Weisblum. “The positive reception that we’ve been getting from both the scientific community and the investment community validates our mission of taking psilocybin therapy studies out of the lab and turning them into real-world applications and treatments.”

To learn more about Silo Pharmaceuticals, please visit the website at silopharma.com
*From the article here :
 
wol-banner-ibogaine-for-parkinson.jpg


Ibogaine for Parkinson’s Disease: Exploring the Science*

by Martha Allitt and Lynn Marie Morski, MD, Esq. | Way of Leaf | 5 Jun 2022

Ibogaine is a psychedelic compound found in the root bark of the Western African shrub Iboga. It has a strong hallucinogenic effect when consumed and has been used in African spiritual traditions throughout history.

Western medicine has recently found interest in ibogaine as a therapeutic tool for drug addiction. However, scientific evidence has suggested ibogaine could also help patients with Parkinson’s disease (PD).

PD affects one in every hundred of the over-sixties population. With current treatments causing a wide range of unpleasant side effects, could ibogaine provide a solution?

Clinical uses of Ibogaine

People have used ibogaine for healing purposes for several thousand years in West-Central Africa. The bark from the Iboga tree is central to the spiritual ceremonies of the Bwiti tradition of Gabon, where people use ibogaine as a tool for personal development, physical healing, and communication with spirits and ancestors.

The use of ibogaine in Western medicine was pioneered by the American researcher Howard Lotsof in 1962. Lotsof discovered ibogaine could prevent withdrawals from opiates and reduce drug craving. Consequently, this led to a series of research studies beginning from the late 80s highlighting ibogaine as an effective treatment for substance misuse disorders.

Ibogaine is illegal in the US, UK, and much of Europe. However, in certain countries, patients can legally access ibogaine-assisted therapy at special ibogaine treatment clinics. Ibogaine therapy is mainly used to treat addiction disorders but is also available for, and has proved helpful in treating, psychological problems such as depression, anxiety, and OCD.

Could Ibogaine be a promising new treatment for Parkinson’s?

Parkinson’s disease (PD) is a nervous system disease characterized by motor impairment, although patients often experience physical and cognitive (mental) symptoms. PD is more common in men than women, and symptoms generally appear later in life (after around 60).

Motor symptoms of PD include:​
  • Tremor: shaking of the limbs (most often beginning in the hands and arms and gradually moving across the body)​
  • Bradykinesia: slowness of movement​
  • Rigidity: stiffness of the muscles, which can cause muscle cramps​
  • Problems with balance​
  • Anosmia: loss of sense of smell​
  • Erectile dysfunction​
Cognitive symptoms of PD include:Depression​
  • Anxiety​
  • Mild cognitive impairment: problems with memory and problem solving​
  • Dementia: severe memory loss, hallucinations, and deluded thinking​

What causes Parkinson’s?

Researchers have shown that in the brains of patients with PD are Lewy bodies, clumps found inside cells made up of a protein called alpha-synuclein. The presence of Lewy bodies stops nerve cells in the brain from receiving nutrients and oxygen, causing them to die. In turn, this causes cells that communicate with the affected cells to die.

In PD, Lewy bodies commonly affect a brain region called the substantia nigra. Nerve cells in this region communicate with the chemical messenger dopamine, creating signals to other parts of the brain involved in creating and controlling movement. Hence when dopamine-producing cells of the substantia nigra are lost in PD, the pathways involved in movement become impaired, and patients experience motor deficits.

Cells that produce the chemical messenger norepinephrine are also affected in PD. Loss of norepinephrine-producing cells may account for some of the PD cognitive symptoms.

It is unknown what causes alpha-synuclein to aggregate and form Lewy bodies, although some risk factors have been linked. These include:​

How could Ibogaine help?

Evidence has shown that ibogaine causes cells to increase the production of neurotrophins. Neurotrophins are proteins that help nerve cells grow and survive. By increasing neurotrophins, ibogaine could help replace lost cells in PD.

We’ll cover the specific research in this area below.

Studies

One experiment showed that ibogaine increased the production of two neurotrophins. These are the glial-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) in the neural circuits affected by PD.

Research has shown that GDNF administration can recover dopamine-producing cells lost in PD. These pieces of evidence have led researchers to use GDNF in clinical trials of Parkinson’s disease. In one study using human patients, direct brain injection of GDNF in PD patients improved the patient’s motor symptoms and daily living and increased their brain levels of dopamine.
Increasing neurotrophins by administering ibogaine could help reverse some of the cell death and symptoms associated with PD.

Evidence has suggested problems with BDNF may be linked to PD. A study in mice showed that removing the BDNF receptor caused increased alpha-synuclein aggregation and increased the death of dopamine-producing nerve cells.

The experimental evidence for GDNF and BDNF in PD suggest increasing these neurotrophins by administering ibogaine could help reverse some of the cell death and symptoms associated with PD.

Patient Case Report

One patient was diagnosed with PD aged 69 years old. His symptoms included difficulty with balance, talking, and using his hands. In 2014 the patient was treated with an ibogaine-derivative, CKBR-1, which he took in small doses twice a day as part of a clinical trial.

After a month of taking CKBR-1, he saw an improvement in all his symptoms and was able to do things he hadn’t previously, such as using his fingers to pick up objects, have full conversations, and button his shirt.*

There have been no clinical studies to date investigating whether ibogaine can improve Parkinson’s disease. However, early-stage research is currently underway at Columbia University.

Safety

Ibogaine can cause cardiac arrhythmias (irregular heartbeat), which in some cases can be fatal. As such, patients with a personal and family history of heart-related conditions are strongly advised against taking ibogaine.

Fatalities linking ibogaine administration combined with other drugs of abuse have also been reported. As such, mixing ibogaine with other drugs should be avoided.

Anecdotal evidence suggests that only small doses of ibogaine may be necessary to treat PD. The dosage is the polar opposite of ibogaine therapy for addiction, where patients take large doses. By taking smaller amounts, patients may be less likely to face risks related to ibogaine.

Researchers are currently attempting to manufacture versions of ibogaine that work in a similar mechanism but don’t have the same toxic or hallucinogenic effects.

Summary

PD is a debilitating disorder that severely affects a patient’s quality of life. As such, scientists are desperately trying to find effective treatments. Researchers are beginning to investigate whether ibogaine could be effective in addressing PD.

The use of ibogaine for PD treatment is still largely theoretical, with no current published study that supports the claim. However, there is strong evidence regarding ibogaine’s effect on neurotrophins. Now, scientists are taking steps to see whether or not ibogaine can reverse the damage and symptoms associated with PD.

*From the article here :
 
Top