Next Article in Journal
Cell Therapy with Human Reprogrammed CD8+ T-Cells Has Antimetastatic Effects on Lewis Lung Carcinoma in C57BL/6 Mice
Next Article in Special Issue
Mapping miRNA Research in Schizophrenia: A Scientometric Review
Previous Article in Journal
miR-193b-3p Promotes Proliferation of Goat Skeletal Muscle Satellite Cells through Activating IGF2BP1
Previous Article in Special Issue
Multiple Pre-Treatment miRNAs Levels in Untreated Major Depressive Disorder Patients Predict Early Response to Antidepressants and Interact with Key Pathways
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Key Characteristics and Development of Psychoceuticals: A Review

by
Genaro Herrera Cano
1,
Jordan Dean
1,
Samuel Padilla Abreu
1,
Amanda Hernández Rodríguez
1,
Cyrena Abbasi
1,
Madison Hinson
2 and
Brandon Lucke-Wold
3,*
1
University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA
2
Wake Forest University School of Medicine, 475 Vine St, Winston-Salem, NC 27101, USA
3
Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(24), 15777; https://doi.org/10.3390/ijms232415777
Submission received: 28 October 2022 / Revised: 5 December 2022 / Accepted: 6 December 2022 / Published: 12 December 2022
(This article belongs to the Special Issue Molecular Pharmacology and Genomics in Psychiatry)

Abstract

:
Psychoceuticals have brought benefits to the pharmacotherapeutic management of central nervous system (CNS) illnesses since the 19th century. However, these drugs have potential side effects or lack high response rates. This review covers twenty drugs’ biochemical mechanisms, benefits, risks, and clinical trial reports. For this study, medications from seven psychoceutical organizations were reviewed and evaluated. Nineteen drugs were chosen from the organizations, and one was selected from the literature. The databases used for the search were Pubmed, Google Scholar, and NIH clinical trials. In addition, information from the organizations’ websites and other sources, such as news reports, were also used. From the list of drugs, the most common targets were serotonergic, opioid, and N-methyl-D-aspartate (NMDA) receptors. These drugs have shown promise in psychiatric illnesses such as substance abuse, post-traumatic stress disorder (PTSD), anxiety, depression, and neurological conditions, such as Parkinson’s disease, traumatic brain injury, and neuroinflammation. Some of these drugs, however, are still early in development, so their therapeutic significance cannot be determined. These twenty drugs have promising benefits, but their clinical usage and efficacy must still be explored.

1. Introduction

The neurological and psychiatric pharmacology fields have grown immensely since the 19th century and have brought considerable benefits to managing and treating various illnesses. For example, the introduction of morphine as well as other medications that induced sedation for the rapid control of agitation and aggression in psychiatric hospitals during the second half of the 19th century [1,2] sparked research into pharmacotherapeutic interventions for neurologic and mental health illnesses. By the late 1950s, the discovery of six different central nervous system (CNS) neurotransmitters (acetylcholine, dopamine, GABA, norepinephrine, serotonin, and substance P) allowed the development of pharmacotherapeutic treatments for neurological conditions, such as Parkinson’s disease, and psychiatric conditions, such as depression and schizophrenia [1,2]. In contrast, there appears to be less interest in the research and development of psychoceutical drugs within the 21st century.
The current state of psychoceutical innovation has remained motionless for a long time. Many of the drugs within this class used today were discovered more than 70 years ago [2,3]. Various factors surrounding the research and trial testing of psychoceuticals have kept the development of these drugs from moving forward. Some examples are limited pathophysiological knowledge [2,3], high rates of failure in phase III trials, poor predictive animal models [2], later returns of investment, and high costs of drug development [4,5], among others. These factors negatively impact the development of new medications in this class, yet the most important one is the shift of charge from clinical researchers to the pharmaceutical industry. Clinical researchers are restricted to supporting the pharmaceutical industry’s interests [1,6,7,8], which can increase the risk of bias. Such restriction of data to the pharmacotherapy community can potentially harm the patients these products treat. Therefore, it is crucial to monitor the development of novel psychoceuticals that aim to replace or improve pharmacotherapy in the global market.
Currently, the psychoceuticals available in the global market are accompanied by multiple side effects and have response rates of less than 50% [2]. To see a positive change in neurological and psychiatric pharmacotherapy, the clinical research community must support the moral development of novel drugs within those fields. The main focus of developing new modes of treatment should be to improve patient care management instead of supporting the pharmaceutical industry’s interests. Furthermore, the pathophysiology behind the illnesses these fields target must be further explored. A clear understanding of the biochemical mechanisms of neurological and psychiatric conditions is the key to the success of clinical drug production [4]. Although there are other issues to tackle in developing psychoceuticals, these two should be targeted first to improve neurological and psychiatric pharmacotherapy immensely.
Over the years, psychoceuticals have come a long way in medical research to explore their benefits, risks, and underlying biochemical mechanisms to achieve their outcomes. There is still room for testing regarding patient healthcare outcomes within neurological-related specialties. Currently, psychoceutical organizations are evaluating potential outcomes for old and newly engineered drugs. For this review, drug products from six different psychoceutical organizations were selected for evaluation. Nineteen drugs from the organizations and one more from the literature were selected to review for their biochemical pathway mechanisms, benefits and risks, and clinical trials.

2. Results and Discussion

A total of 110 records were selected in the search (Pubmed, NIH clinical trials, organization websites, and Google Scholar). Before assessing eligibility, five were excluded due to duplication of the records. Then, 20 records were excluded due to there being no published clinical trial studies or the results not being posted for initiated studies (Figure 1).

2.1. Biochemical Mechanisms of Drugs

The psychoceutical list is composed of drugs interacting with various CNS targets. Most of the drugs target serotonin receptors, followed by drugs that target NMDA and opioid receptors. Other targets include GLUT-8, NF-κB, α3β4 receptors, GABA receptors, and peptides (Table 1). It is essential to recognize that not all the biochemical pathways of these drugs are fully understood. Of the twenty drugs reviewed, nine target the 5-HT family of receptors (5-HT1A, 5-HT2A, 5-HT1B, 5-HT2B, 5-HT2C), two interact with opioid receptors (mu-opioid, k-opioid), and two interact with NMDA receptors. Two drugs target all three types of receptors. The other five drugs interact with one of the remaining targets. In general, serotonergic targeting drugs directly enhance the activity of these receptors or increase the serotonin levels in the synaptic cleft by blocking reuptake transporters. Opioid-targeting drugs increase the activity of the G-protein signaling and other pathways, such as the beta-arrestin and ERK/MAPK pathways. NMDA-targeting drugs act as antagonists.

2.1.1. Serotonin Receptor-Targeted Drugs

Most of the psychoceuticals reviewed fall within this target category. These drugs generally bind to CNS G-protein receptors within the 5-HT family, such as 5-HT1A, 5-HT2A, and 5-HT2B, and act agonistically [9,13,14,15,17,18]. Additionally, two of these, ecstasy and CYB005 (phenethylamine derivative [20]), block the reuptake of serotonin, increasing the serotonin levels within the synaptic cleft [12,21,22]. These drugs aim to increase the activity of serotonergic systems within the CNS.
Although these drugs are expected only to stimulate serotonergic systems, LSD demonstrates the opposite. LSD has a dual effect on brain systems, such as the locus coeruleus (LC), raphe nuclei (RN), and the brain cortex [14]. LSD’s agonistic activity on 5-HT1A receptors has inhibitory effects [14]. In contrast, the agonistic activity of 5-HT2 receptors has stimulatory effects [14]. No reports of the other eight drugs performing inhibitory or dual serotonergic activities have been found. The possibility of these drugs causing inhibitory or dual effects through multiple serotonin receptor interactions should not be disregarded.
The underlying mechanisms activated after drug–receptor interactions have still not been confirmed for all nine psychoceuticals. Research on Peyote activity has revealed that 5-HT2 receptors, especially 5-HT2C receptors coupled with G-proteins, activate phospholipase C [15]. This activation increases the cytosolic levels of IP3, causing endoplasmic reticulum calcium release into the cytosol of postsynaptic neurons [15]. It may be assumed that drugs interacting with 5-HT2 receptors (LSD, CYB003, CYB004, Psilocybin [14,16,17,18]) initiate cellular mechanisms similar to Peyote, but cannot be guaranteed at this time. For example, LSD has demonstrated a strong selectivity for the beta-arrestin pathway through 5-HT2B receptor activity [13]. At the same time, there is unbiased selectivity between the beta-arrestin and G-protein signaling pathways through interactions with 5-HT1B receptors [13]. Although these drugs target similar receptors, their underlying biochemical pathways may differ and should be investigated in future studies.
Furthermore, CNS enzymes can denature some serotonergic systems targeting psychoceuticals and impede excess system activity, but it is not always the case. The inactivation of 5-MeO-DMT by monoamine oxidase A produces an active metabolite, bufotenine, that binds with a higher affinity to 5-HT2A receptors than the parent drug [9,10]. In contrast, the endogenous hallucinogen N, N-dimethyltryptamine (DMT) can be rendered inactive by monoamine oxidase (MAO) [11]. The CNS’ capacity for drug neutralization must be considered to modulate serotonergic system activity.
Serotonergic transport blockers give another option to affect CNS serotonergic system activity indirectly. According to preclinical data, CYB005, a phenethylamine derivative, aims to inhibit serotonin transporters within the synaptic cleft [20,21,22]. On the other hand, MDMA-derivative ecstasy blocks the reuptake of serotonin and norepinephrine and enhances their release into the synaptic cleft [12]. Furthermore, chronic use of ecstasy has been shown to cause a decrease in the expression of serotonin transporters, resulting in less serotonin reuptake and recycling [12]. However, the current focus of serotonergic drug research and development is directly stimulating serotonergic system activity.

2.1.2. NMDA Receptor-Targeted Drugs

The psychoceuticals under this category, ketamine and arketamine (an enantiomer of ketamine), induce antidepressant effects by inhibiting NMDA receptors [24,26]. Ketamine’s non-competitive antagonistic activity on these receptors results in glutamate surges stimulating secondary AMPA receptor (AMPAr) activity as well [26,27]. This secondary activation is necessary to achieve ketamine’s effects [25]. While the underlying molecular mechanism of arketamine is still unclear, previous studies have shown this medication to have more potent and longer-lasting antidepressant effects than ketamine and other enantiomer forms [24,25].
The role of AMPArs after exposure to arketamine should be explored. If the parental structure requires AMPAr activation potentiating pathways, such as mTORC1 and BDNF, for antidepressant effects to occur [27], then it calls into question what mechanisms arketamine induces for similar yet more potent effects to arise. Two clinical trials will be conducted using perampanel, an AMPAr antagonist, in patients with treatment-resistant major depressive disorder, hoping to confirm the role of AMPAr in arketamine’s effects [25].

2.1.3. Opioid Receptor Targeted Drugs

The drugs targeting opioid receptors, deu-mitragynine (a major alkaloid component of kratom [28]) and salvinorin A (a non-alkaloidal component of Salvia divinorum [30]), regulate pathways initiated by G-proteins, while also stimulating other mechanisms, such as the beta-arrestin and ERK/MAPK pathways. Kratom, a mu-opioid receptor (MOR) agonist with low potency, has been demonstrated to emit ‘atypical’ opioid effects (no respiratory depression, emesis, or shortness of breath in animal models) through G-protein agonistic activity and limited recruitment of beta-arrestin after opioid receptor-dependent activation [28,29]. Studies with rodent models alternatively suggest mitragynine and its derivatives do not directly activate opioid receptors [29], which brings into question how the drug initiates the activity of MORs to deliver analgesic effects.
In contrast, salvinorin A stimulates analgesic effects by acting as a selective agonist of k-opioid receptors (KOPr) [30]. Through this receptor, adenylate cyclase is inhibited, beta-arrestin is mediated, and the ERK/MAPK pathways are activated [31]. Salvinorin A also has decreased dopaminergic activation in the dorsal and ventral striata [31], suggesting a reduced risk for addiction compared to other opioids, such as morphine. These drugs can potentially help treat individuals suffering from opioid addiction by decreasing the sense of reward from opioid consumption or reducing the risk of lethal side effects.

2.1.4. Multi-Receptor Targeted Drugs

The two psychoceuticals, ibogaine and noribogaine, are perplexing, as both can interact with opioid, serotonergic, and glutamatergic systems [32,33,34,35]. These systems include kappa-opioid, mu-opioid, NMDA, and serotonin reuptake receptors [32,33,34,35]. Ibogaine, however, can interact with nicotinic and sigma-2 receptors [32,33], while noribogaine cannot [34]. The ability to interact with multiple receptor systems can be a clinical concern, as unintended effects may occur, such as bradycardia and long QT syndrome [46].
The anti-addictive effects of ibogaine and noribogaine appear to be governed by the activity of the k-opioid receptor [34]. Rat model studies demonstrated that ibogaine’s k-opioid agonistic activity decreased cocaine and morphine self-administration [32]. In addition, noribogaine’s administration to human opioid-dependent subjects exhibited a decrease in withdrawal symptoms [47], most likely due to k-opioid agonistic activity [35]. The exact underlying mechanism that achieves this for both drugs remains unclear [32,34,35,47]. Avidor-Reiss’ study presented k-opioid agonist’s effects on adenylyl cyclase (AC) activity [48]. Acute exposure to kappa opioid agonists can potentiate the inhibition of AC, while chronic exposure can lead to the sensitization of AC activity [48]. The drug’s exact biochemical mechanisms should be researched extensively before further conclusions can be made. Additionally, these two drugs do not solely rely on opioid agonism [34], although the roles of other receptors in the anti-addictive effects are not entirely understood [32]. Exploring their role is key to understanding the potential effects of ibogaine and noribogaine.

2.1.5. Other Receptor Targeted Drugs

Five of the twenty psychoceuticals evaluated in this review have mechanisms that do not involve serotonergic, opioid, or glutamatergic systems. The mechanisms for three of these drugs, SLS-007 (peptidic drug), zolunicant HCl (MM-110), and deuterated etifoxine, require further investigation. SLS-007 targets alpha-synuclein’s NAcore (non-amyloid component core) and inhibits protein aggregation in Parkinson’s patients, showing therapeutic potential in mouse models [40,41]. The mechanism by which it inhibits protein aggregation has not currently been reported. It is suspected that SLS-007 acts as an intrabody binding to alpha-synuclein monomers to prevent their oligomerization and, consequently, stop the formation of neurotoxic fibrils [40,49]. Zolunicant HCl is known to be a potent inhibitor of nicotinic alpha-3-beta-4 receptors [44,50] and manages the regulation of dopamine levels during substance withdrawal [51]. The underlying mechanism of this drug has yet to be reported. Deuterated etifoxine aims to boost depressant effects through the positive allosteric stimulation of the GABA receptor’s β 2 and β 3 subunits [45]. In addition, increased mitochondrial TSPO promotes neurosteroid synthesis [45], although the significance of inducing these effects is still under study. More research on the mechanisms of these three drugs should be performed to form conclusions about their biochemical behavior.
Compared to the research on these three drugs, there is more knowledge on the biochemical pathways of the other two psychoceuticals: Trehalose (SLS-005) and N-acetylcysteine (NAC).
Trehalose enhances autophagy and lysosomal activation through an mTOR-independent pathway and nuclear translocation of transcription factor EB [37,38,39]. The light chain 3–II (LC3-II) increase in the cytosol indicates this enhancement [36,37,38,39]. Promoting the fusion of autophagosomes to lysosomes, LC3-II leads to the degradation of autophagosome contents [36,37,38,39]. However, some observations suggest that trehalose can inhibit autophagic flux as well [38]. Therefore, the dual behavior of trehalose should be investigated further.
Lastly, NAC has been hypothesized to exert cytoprotective effects in the CNS [43]. Often used to increase intracellular glutathione for paracetamol overdose [42], the drug’s anti-inflammatory properties may allow for the modulation of inflammatory mechanisms in other areas of the body. By suppressing nuclear factor kappa B (NF-κB) activity, the compound reduces levels of cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukins (IL-6 and IL-1B) [42]. Using the drug’s anti-inflammatory properties can potentially reduce brain damage from exerted neuroinflammation.

2.2. Pre-Clinical/Clinical Benefits and Risks of Drugs

Pre-clinical and clinical studies testing the 20 psychoceuticals have shown benefits related to substance abuse, PTSD, anxiety, depression, and other diseases (Table 2). Although promising therapeutic results have been indicated for these drugs, there are reports of potential adverse effects from some of them. The nature of these side effects include psychiatric (agitation, panic attacks), neurological (ataxia, sensory dissociation), gastrointestinal (nausea/vomiting, epigastric pain), cardiovascular (tachycardia, hypertension), endocrine (increased cortisol, prolactin), and systemic (chills, headaches) signs and symptoms. Studies on Zolunicant HCl, CYB005, CYB004, and CYB003 were not found in the literature search, so this information was obtained through the websites of their respective pharmaceutical companies.
As commonly used analgesics, such as opioids, have a high risk of being addictive, psychoceuticals that are en route to being approved may eventually replace opioids in clinical practice. For instance, Kratom exhibits analgesic effects by acting as an MOR agonist [28,71]. In addition, psychedelics have also been demonstrated to help reduce cravings for patients undergoing withdrawal from opioid use. For example, MM-110 (zolunicant HCl) and noribogaine are currently being investigated for their anti-craving properties with opioid withdrawal through the inhibition of nicotinic α3β4 receptors [44] or activation of k-opioid receptors [34,55,56]. Ibogaine also has anti-craving effects for opioid withdrawal by activating the k-opioid receptor. In addition, ibogaine also alleviates the withdrawal symptoms of certain recreational drugs, such as cocaine [34,46].
Another of the most significant benefits observed with these drugs is their use in psychological disorders. Various studies have demonstrated improvement in humans after being exposed to these drugs. Some of the medications that have been shown to decrease depression and anxiety are LSD [52], peyote [58], ketamine [67], arketamine [24,63], N, N-dimethyltryptamine [11], psilocybin [64], and deuterated etifoxine [59,60]. Administration of 5-methoxy-N, N-dimethyltryptamine (5-MeO-DMT) has reduced suicidal ideations, planning, and attempts [53,54]. Ecstasy has shown an attenuated amygdala response in humans who are shown negative stimuli and an extinction of fear memories in mice [65,66]. Lastly, salvinorin A has been shown to create feelings of calm and relaxation, leading to an improved mood [30,61]. In contrast, it had endocrine effects that significantly raised cortisol and prolactin levels [30].
Besides these compounds, new ones have been engineered by combining novel psycoceutical molecules with controllable drug delivery systems through a deuterated process for possible mental health treatments. Three drugs that researchers have been working on are CYB003 (for depressive disorder), CYB004 (for anxiety disorders), and CYB005 (for neuroinflammation). CYB003 is a deuterated psilocybin analog that has received FDA IND clearance to enter clinical development [16]. CYB004 is a deuterated DMT being used to conduct phase I in-human studies [18]. Its preclinical studies demonstrated that it could potentially treat anxiety disorders [18]. Finally, CYB005 is a phenethylamine derivative currently in preclinical development [55], which has the potential to treat neuroinflammation and psychiatric conditions [20].
Other drugs reviewed have shown to be beneficial in areas outside of psychological disorders. For example, SLS-007, a peptidic inhibitor being tested in a preclinical study, can stop the propagation and seeding of α-synuclein aggregates, which could help in Alzheimer’s disease, Parkinson’s disease, and Lewy body dementia [40,70]. SLS-005, a Trehalose injection, is currently used in a phase II clinical trial for Alzheimer’s patients to test its treatment safety and efficacy [72]. In addition, it has been shown that it protects against oxidative stress, increases levels of chaperone molecules, and enhances autophagy [62]. Animal studies have also shown that it is neuroprotective and efficacious in improving functional outcomes following a traumatic brain injury (TBI) [62]. Finally, N-acetylcysteine, a well-established drug known to be used in patients with COPD, cystic fibrosis, or those with a Tylenol overdose, has been theorized to show beneficial effects in patients with schizophrenia by improving cognition [68,69].
Even though these compounds have shown promise pre-clinically and clinically, their usage can have potential side effects which are important to be aware of. These side effects can vary from mental state/consciousness alterations to physiological changes. The following are the general risks of consumption of these compounds:
  • LSD includes delusions, visual hallucinations, distortion of one’s sense of time and identity, impaired depth and time perception, artificial sense of euphoria or certainty, distorted perception of the size and shape of objects, movements, colors, sounds, touch, and the user’s body image, severe, terrifying thoughts and feelings, fear of losing control, fear of death, and panic attacks [52].
  • Reports of 5-MeO-DMT show agitation and tachycardia with periodic reports of hyperthermia, seizures, coma, increased serum creatinine, and life-threatening experiences, such as cardiac arrest and possible death [53].
  • Ibogaine has side effects such as nausea, headache, and visual changes; the most common side effects are reported in [46].
  • Noribogaine’s most common adverse events are headache and epistaxis [56].
  • Peyote is classified as possessing the highest potential for use disorder and misuse [73].
  • Salvinorin A has adverse outcomes, including fear, panic, paranoia, agitated delirium, sadness, irritability, augmented perspiration, and chills [30]. In addition, consumers can feel a lack of insight, making them susceptible to harm, which can occur during the performance of complex tasks, such as driving [30]. Gastrointestinal malaises (e.g., nausea and vomiting) have also been reported [30].
  • Arketamine’s side effects include a slight rise in blood pressure, with a mean increase of 16.7 mmHg for systolic BP and 11.9 mmHg for diastolic [63]. Other adverse effects are ataxia, sensory dissociation, hyperactivity, and conditioned-place preference [24].
  • Ecstasy’s reported side effects are heat strokes, relapsing of pre-existing schizophrenia [65], and depression [65,74]. This drug also has cardiovascular side effects, such as cardiac contractile dysfunction and vasoconstriction [75].
  • Oral N-acetylcysteine (NAC) may cause nausea, vomiting, diarrhea, flatus, and gastroesophageal reflux [69]. In addition, IV NAC can cause rate-related anaphylactoid reactions [69].
  • Ketamine’s side effects include nausea/vomiting and epigastric pain. A rapid ketamine IV injection can cause transient apnea, cystitis, and contracted bladder [67].
  • N, N-dimethyltryptamine’s side effects include increased levels of corticotropin, cortisol, prolactin, and growth hormone when administered to human volunteers [11].
  • Kratom has shown mental health effects, primarily withdrawal symptoms [71].

2.3. Clinical Trials

The chosen psychoceuticals to review are in different stages of development (pre-clinical trial, phase I clinical trial, phase II clinical trial). Therefore, the information available from each one can vary depending on how far in development they are. Of the 20 drugs, Kratom, SLS-007, N, N-dimethyltryptamine, Zolunicant HCl, CYB005, CYB004, CYB003, deuterated etifoxine, and SLS-005 do not currently have completed clinical trials.
Five of the eleven drugs have been tested for therapeutic effects in psychiatric-related illnesses. LSD has been shown to have anxiolytic, antidepressant, and anti-addictive properties [76]. However, LSD’s clinical research during the 1960s and 1970s was halted due to the rise in its recreational use [76]. A recent phase-II clinical trial reported some improvement in anxiety for individuals with advanced-stage life-threatening disease in the full-dose LSD group [77]. Similarly, there have been self-reports of 5-MeO-DMT improving anxiety and depression along with no changes in memory, attention, and cognitive function [78,79]. In treatment-resistant depression, arketamine and psilocybin significantly decreased depressive symptoms [63,80]. Adverse effects of headache, nausea, and dizziness after psilocybin treatment were reported [80]. Besides anxiety and depression, PTSD treatment has also been under evaluation. Ecstasy has been reported beneficial to the treatment of PTSD in a randomized phase-III clinical trial with significant improvements in CAPS-5 scores in the ecstasy group compared to the control group, along with no significant adverse effects in the ecstasy group [66]. Other research on this drug is heavily confounded, as many ecstasy users are polydrug users, meaning they take other illicit drugs [81,82,83]. Studies that have attempted to identify simple dose–response effects have been futile [81,82,83]. Even though these substances have portrayed positive outcomes in their utilization to treat certain psychiatric illnesses, more observations are needed to confirm their safe therapeutic use.
The use of psychoceuticals in substance abuse disorders has also undergone clinical testing. There are mixed findings about ibogaine and noribogaine regarding their therapeutic usage in managing opioid craving and withdrawal symptoms [84,85,86]. These two drugs have shown a reduction in craving and withdrawal symptoms [87,88], yet other studies have found that they do not have significant effects on opioid withdrawal [89]. Clinical research on ibogaine is challenging in the United States due to its classification as a Schedule I substance [90]. While salvinorin A data for substance abuse treatment have not been found, the dose-related effects in healthy hallucinogen-using adults suggest this drug may be used for substance abuse conditions. A double-blind, placebo-controlled study demonstrated that the drug’s peak effects occurred two minutes after administration and rapidly dissipated [91]. The subjects were followed up with one month after the trial, and no persisting effects were observed [91]. Given that this drug produces both subjective and cognitive effects and some classic hallucinogen effects [91], its usage may be important in therapeutic settings.
NAC is reported to have positive clinical outcomes between the remaining three drugs, while peyote and ketamine’s therapeutic use must be further observed. First, NAC has shown potential in treating vascular and non-vascular neurological disorders, such as traumatic brain injury and cerebral ischemia [43]. Second, peyote has not elicited significant psychological or cognitive deficits in Navajo Native Americans [92]. These findings may not be generalizable to illicit hallucinogen users [92]. Third, the results and conclusions of a trial in China administering esketamine, a ketamine enantiomer, in patients undergoing major surgery, which evaluated outcomes such as remission rate three days post-operation and depression-related scores, were not reported in [93].
Overall, there are mixed findings on the therapeutic effects of the 11 drugs in the studies found within the search. Some of the drugs have shown positive treatment outcomes in animal- or human-model studies. In contrast, others have not demonstrated any significant clinical effects or have reported adverse effects. Therefore, more trials on these drugs’ safety and efficacy should be performed.

2.4. Limitations

This review does not represent an overall image of current research and development of psychoceuticals. It involves 20 drugs from six pharmaceutical organizations. Additionally, some of the drugs listed are still early in trial testing. Therefore, there are not much data on the therapeutic effects of these products, leading to the necessity of filling gaps in information using the organizations’ websites, which can lead to bias in their performance.

2.5. Future Directions

Creating a picture of the current psychoceutical availability in the global pharmaceutical market is a considerable next step. Evaluating the drugs’ clinical benefits and potential risks can illustrate if their positive outcomes outweigh their negative effects and, if they do, by how much. Additionally, it is essential to determine which psychoceutical drugs currently in the market may be replaced by drugs that are in clinical development. Finally, for those new prospective medications, it would be beneficial to compare trial results (effective concentration dose, clinical outcomes, adverse effects) between them and those already in the global market.

3. Methods

PRISMA statement registration was not obtained for this study but will be obtained for related studies in the future. Seven websites of psychoceutical organizations (Psycheceutical, GH Research, Atai Life Sciences, Cybin, Compass, MindMed, and Seelos Therapeutics) were utilized to evaluate the drugs they were researching or developing for clinical treatment of neurological and psychiatric illnesses. This review aimed to assess a list of 20 psychoceuticals. From those websites, a list of 19 drugs was composed to evaluate a combination of old and new engineered medications. One drug was obtained from the literature to refrain from restricting this study’s evaluation of newly engineered drugs. For psychoceutical assessment, the biochemical mechanisms, benefits and risks, and clinical trials were searched.
Out of the seven websites, only six presented psychoceuticals, while the organization that did not, Psycheceutical, only presented novel drug-delivery methods, which do not fall in line with the aim of this review. From GH Research and Compass, one drug was selected from each, 5-MeO-DMT (GH Research) and psilocybin (Compass). From three organizations, two to three drugs were selected: LSD and Zolunicant HCl (MindMed); Deuterated Psilocybin analog, Phenethylamine derivative, and Deuterated DMT (Cybin); SLS-002, Trehalose, and SLS-007 (Seelos Therapeutics). As a percentage of the drugs selected were psychedelics, such as LSD and psilocybin, an additional drug within that category was searched in PubMed, inputting the word psychedelic, and Peyote was chosen (Figure 2).
For biochemical mechanisms, the pathways or actions of these molecules were searched for in PubMed. In this search, phrases used included “drug name” and one of the following: biochemistry, mechanism, biochemical mechanism, and biochemical pathway. For the medications not found in the search or missing details, other sources on Google Scholar were used along with information provided by the organizations’ websites.
As if for the biochemical mechanisms, the benefits and risks of these twenty medications were searched for in PubMed. In the search, phrases included the “drug name” and one of the following: benefits, clinical benefits, risks, and potential risks. For the medications not found, information from sources found in Google Scholar search and the organization websites were used.
Only Pubmed, NIH clinical trial databases, and news reports for the clinical trial and trial review search were strictly used. Information from the organizations’ websites was excluded to avoid bias.

4. Conclusions

The 20 psychoceuticals presented in this review have shown pre-clinical or clinical promise. Many of these drugs, however, are far from entering the market. Although the organizations publish an overview of their results on their websites, finished clinical trial studies in the past literature and from the NIH clinical trial database for numerous drugs in the list were not found. The targets of these drugs have been identified, but some underlying biochemical mechanisms still need to be confirmed or researched thoroughly before conclusions may be made. Additionally, more than half of the drugs on the list have side effects that are important to consider for future patient treatment use. Still, they may improve the treatments of multiple illnesses and conditions, ranging from neurologic, such as TBI, to psychiatric, such as substance abuse disorder. The biochemical mechanisms, benefits, and risks were not all found in the literature search. Information from the organizations’ websites had to be considered, as many of these are newly engineered.
The application of these drugs within neuro-related specialties, including neurology and neurosurgery, requires further exploration and research. It is crucial to recognize that only a small subset of psychoceuticals was reviewed. It is worth reviewing the applications of other drugs within neuro-related and psychiatric fields.

Author Contributions

Record Search and Review: G.H.C., J.D., S.P.A., A.H.R., C.A. and M.H. Writing—Original Draft Preparation: G.H.C. Writing—Review and Editing: G.H.C., J.D., S.P.A., A.H.R. and M.H. Supervision: B.L.-W. All authors have read and agreed with the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this manuscript.

Conflicts of Interest

The authors report no conflict of interest.

References

  1. Ban, T.A. Academic psychiatry and the pharmaceutical industry. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2006, 30, 429–441. [Google Scholar] [CrossRef] [PubMed]
  2. Zimmer, L. Fundamental and clinical neuropharmacology, a terra incognita with constantly expanding frontiers. Fundam. Clin. Pharmacol. 2021, 35, 501–505. [Google Scholar] [CrossRef] [PubMed]
  3. Becker, R.E.; Seeman, M.V.; Greig, N.H.; Lahiri, D.K. What can triumphs and tribulations from drug research in Alzheimer’s disease tell us about the development of psychotropic drugs in general? Lancet Psychiatry 2015, 2, 756–764. [Google Scholar] [CrossRef] [Green Version]
  4. Spedding, M. New directions for drug discovery. Dialogues Clin. Neurosci. 2006, 8, 295–301. [Google Scholar] [CrossRef] [PubMed]
  5. Huskamp, H.A. Managing psychotropic drug costs: Will formularies work? Health Aff. 2003, 22, 84–96. [Google Scholar] [CrossRef] [Green Version]
  6. Cosgrove, L.; Vannoy, S.; Mintzes, B.; Shaughnessy, A.F. Under the Influence: The Interplay among Industry, Publishing, and Drug Regulation. Account. Res. 2016, 23, 257–279. [Google Scholar] [CrossRef]
  7. Carrier, F.; Banayan, D.; Boley, R.; Karnik, N. Ethical challenges in developing drugs for psychiatric disorders. Prog. Neurobiol. 2017, 152, 58–69. [Google Scholar] [CrossRef] [PubMed]
  8. Hirshbein, L. Looking back to the future of psychopharmacology. J. Nerv. Ment. Dis. 2012, 200, 1109–1112. [Google Scholar] [CrossRef]
  9. Shen, H.W.; Jiang, X.L.; CWinter, J.; Yu, A.M. Psychedelic 5-Methoxy-N,N-Dimethyltryptamine: Metabolism, Pharmacokinetics, Drug Interactions, and Pharmacological Actions. Curr. Drug Metab. 2010, 11, 659–666. [Google Scholar] [CrossRef] [Green Version]
  10. Yu, A.M. Indolealkylamines: Biotransformations and Potential Drug–Drug Interactions. AAPS J. 2008, 10, 242–253. [Google Scholar] [CrossRef]
  11. Carbonaro, T.M.; Gatch, M.B. Neuropharmacology of N,N-dimethyltryptamine. Brain Res. Bull. 2016, 126, 74–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. National Institute on Drug Abuse. What Are MDMA’s Effects on the Brain? National Institute on Drug Abuse: Rockville, MD, USA, 2017. Available online: https://nida.nih.gov/publications/research-reports/mdma-ecstasy-abuse/what-are-mdmas-effects-on-brain (accessed on 16 September 2022).
  13. Andén, N.E.; Corrodi, H.; Fuxe, K.; Hökfelt, T. Evidence for a central 5-hydroxytryptamine receptor stimulation by lysergic acid diethylamide. Br. J. Pharmacol. 1968, 34, 1–7. [Google Scholar] [CrossRef] [PubMed]
  14. Passie, T.; Halpern, J.H.; Stichtenoth, D.O.; Emrich, H.M.; Hintzen, A. The pharmacology of lysergic acid diethylamide: A review. CNS Neurosci. Ther. 2008, 14, 295–314. [Google Scholar] [CrossRef] [PubMed]
  15. Dinis-Oliveira, R.J.; Pereira, C.L.; da Silva, D.D. Pharmacokinetic and Pharmacodynamic Aspects of Peyote and Mescaline: Clinical and Forensic Repercussions. Curr. Mol. Pharmacol. 2019, 12, 184–194. [Google Scholar] [CrossRef] [PubMed]
  16. Cybin. CYB003. Cybin. Available online: https://cybin.com/cyb003/ (accessed on 20 September 2022).
  17. Schmidt, C.J.; Sorensen, S.M.; Kenne, J.H.; Carr, A.A.; Palfreyman, M.G. The role of 5-HT2A receptors in antipsychotic activity. Life Sci. 1995, 56, 2209–2222. [Google Scholar] [CrossRef] [PubMed]
  18. Cybin. CYB004. Cybin. Available online: https://cybin.com/cyb004/ (accessed on 20 September 2022).
  19. Barker, S.A. N, N-Dimethyltryptamine (DMT), an Endogenous Hallucinogen: Past, Present, and Future Research to Determine Its Role and Function. Front. Neurosci. 2018, 12, 536. [Google Scholar] [CrossRef] [Green Version]
  20. Cybin. Development Pipeline. Cybin. Available online: https://cybin.com/development-pipeline/#:~:text=CYB005%3A%20Discovery%2DPhase%20Phenethylamine%20Derivative (accessed on 20 September 2022).
  21. Phenethylamine. RxList. Published 17 September 2019. Available online: https://www.rxlist.com/phenethylamine/supplements.htm (accessed on 24 September 2022).
  22. Irsfeld, M.; Spadafore, M.; Prüß, B.M. β-phenylethylamine, a small molecule with a large impact. Webmedcentral 2013, 4, 4409. [Google Scholar]
  23. About Psilocybin Therapy: Compass Pathways. Available online: https://compasspathways.com/our-work/about-psilocybin-therapy/ (accessed on 27 September 2022).
  24. Zanos, P.; Highland, J.N.; Liu, X.; Troppoli, T.A.; Georgiou, P.; Lovett, J.; Gould, T.D. (R)-Ketamine exerts antidepressant actions partly via conversion to (2R,6R)-hydroxynorketamine, while causing adverse effects at sub-anaesthetic doses. Br. J. Pharmacol. 2019, 176, 2573–2592. [Google Scholar] [CrossRef]
  25. Wei, Y.; Chang, L.; Hashimoto, K. Molecular Mechanisms Underlying the Antidepressant Actions of Arketamine: Beyond the NMDA Receptor. Mol. Psychiatry 2021, 27, 559–573. [Google Scholar] [CrossRef]
  26. Matveychuk, D.; Thomas, R.K.; Swainson, J.; Khullar, A.; MacKay, M.A.; Baker, G.B.; Dursun, S.M. Ketamine as an antidepressant: Overview of its mechanisms of action and potential predictive biomarkers. Adv. Psychopharmacol. 2020, 10, 2045125320916657. [Google Scholar] [CrossRef]
  27. Derakhshanian, S.; Zhou, M.; Rath, A.; Barlow, R.; Bertrand, S.; DeGraw, C.; Kaye, A.D. Role of Ketamine in the Treatment of Psychiatric Disorders. Health Psychol. Res. 2021, 9, 25091. [Google Scholar] [CrossRef] [PubMed]
  28. Atai Life Sciences. Deu-Mitragynine. Atai Life Sciences. Available online: https://www.atai.life/programs/deu-mitragynine/ (accessed on 28 September 2022).
  29. Shukla, A.K. Dwivedi-Agnihotri, H. Structure and function of β-arrestins, their emerging role in breast cancer, and potential opportunities for therapeutic manipulation. Adv. Cancer Res. 2020, 145, 139–156. [Google Scholar] [CrossRef] [PubMed]
  30. Brito-da-Costa, A.M.; Dias-da-Silva, D.; Gomes, N.G.M.; Dinis-Oliveira, R.J.; Madureira-Carvalho, Á. Pharmacokinetics and Pharmacodynamics of Salvinorin A and Salvia divinorum: Clinical and Forensic Aspects. Pharmaceuticals 2021, 14, 116. [Google Scholar] [CrossRef] [PubMed]
  31. Roach, J.J.; Shenvi, R.A. A review of salvinorin analogs and their kappa-opioid receptor activity. Bioorg. Med. Chem. Lett. 2018, 28, 1436–1445. [Google Scholar] [CrossRef]
  32. Glick, S.D.; Maisonneuve, I.S. Mechanisms of antiaddictive actions of ibogaine. Ann. N. Y. Acad. Sci. 1998, 844, 214–226. [Google Scholar] [CrossRef]
  33. Healthtown. Pharmacodynamics, Pharmacokinetics and Chemistry of Ibogaine in Ibogaine Treatment: Health Town; (Canada) 2021. Available online: https://healthtown.ca/pharmacodynamics-pharmacokinetics-and-chemistry-of-ibogaine-in-ibogaine-treatment/#:~:text=The%20mechanism%20of%20Ibogaine%20includes%20action%20like%20opioid (accessed on 28 September 2022).
  34. Atai Life Sciences. Ibogaine & Noribogaine. Atai Life Sciences. Available online: https://atai.life/programs/ibogaine/ (accessed on 28 September 2022).
  35. Maillet, E.L.; Milon, N.; Heghinian, M.D.; Fishback, J.; Schürer, S.C.; Garamszegi, N.; Mash, D.C. Noribogaine is a G-protein biased κ-opioid receptor agonist. Neuropharmacology 2015, 99, 675–688. [Google Scholar] [CrossRef] [Green Version]
  36. Seelos Therapeutics. SLS-005 (Trehalose). Available online: https://seelostherapeutics.com/sls-005-trehalose/ (accessed on 30 September 2022).
  37. Rusmini, P.; Cortese, K.; Crippa, V.; Cristofani, R.; Cicardi, M.E.; Ferrari, V.; Poletti, A. Trehalose induces autophagy via lysosomal-mediated TFEB activation in models of motoneuron degeneration. Autophagy 2019, 15, 631–651. [Google Scholar] [CrossRef]
  38. Lee, H.J.; Yoon, Y.S.; Lee, S.J. Mechanism of neuroprotection by trehalose: Controversy surrounding autophagy induction. Cell Death Dis. 2018, 9, 712. [Google Scholar] [CrossRef] [Green Version]
  39. Sarkar, S.; Davies, J.E.; Huang, Z.; Tunnacliffe, A.; Rubinsztein, D.C. Trehalose, a Novel mTOR-independent Autophagy Enhancer, Accelerates the Clearance of Mutant Huntingtin and α-Synuclein *. J. Biol. Chem. 2007, 282, 5641–5652. [Google Scholar] [CrossRef] [Green Version]
  40. Seelos Therapeutics, Inc. Seelos Therapeutics Receives a Notice of Allowance in the U.S. for an Additional Patent for SLS-007. Available online: https://www.prnewswire.com/news-releases/seelos-therapeutics-receives-a-notice-of-allowance-in-the-us-for-an-additional-patent-for-sls-007-301549493.html (accessed on 19 September 2022).
  41. SLS-007 (Peptidic Inhibitor). Seelos Therapeutics. Available online: https://seelostherapeutics.com/sls-007/ (accessed on 19 September 2022).
  42. Dos Tenório, M.C.S.; Graciliano, N.G.; Moura, F.A.; de Oliveira, A.C.M.; Goulart, M.O.F. N-Acetylcysteine (NAC): Impacts on Human Health. Antioxidants 2021, 10, 967. [Google Scholar] [CrossRef]
  43. Bavarsad Shahripour, R.; Harrigan, M.R.; Alexandrov, A.V. N-acetylcysteine (NAC) in neurological disorders: Mechanisms of action and therapeutic opportunities. Brain Behav. 2014, 4, 108–122. [Google Scholar] [CrossRef] [PubMed]
  44. O’Brien, D. MindMed Reports Topline Data from Phase 1 Trial of MM-110 in Development for the Treatment of Opioid Withdrawal. MindMed: New York, NY, USA, 2022; Available online: https://mindmed.co/news/press-release/mindmed-reports-topline-data-from-phase-1-trial-of-mm-110-in-development-for-the-treatment-of-opioid-withdrawal/ (accessed on 22 September 2022).
  45. Deu-Etifoxine. Atai Life Sciences. Available online: https://atai.life/programs/deu-etifoxine/#:~:text=Enter%20GABA%20Therapeutics%E2%80%99%20flagship%20compound%2C%20deuterated%20etifoxine.%20History (accessed on 24 September 2022).
  46. Corkery, J.M. Ibogaine as a treatment for substance misuse: Potential benefits and practical dangers. Prog. Brain Res. 2018, 242, 217–257. [Google Scholar] [CrossRef] [PubMed]
  47. Glue, P.; Cape, G.; Tunnicliff, D.; Lockhart, M.; Lam, F.; Hung, N.; Friedhoff, L. Ascending Single-Dose, Double-Blind, Placebo-Controlled Safety Study of Noribogaine in Opioid-Dependent Patients. Clin. Pharmacol. Drug Dev. 2016, 5, 460–468. [Google Scholar] [CrossRef] [PubMed]
  48. Avidor-Reiss, T.; Zippel, R.; Levy, R.; Saya, D.; Ezra, V.; Barg, J.; Vogel, Z. κ-Opioid receptor-transfected cell lines: Modulation of adenylyl cyclase activity following acute and chronic opioid treatments. FEBS Lett. 1995, 361, 70–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Brundin, P.; Dave, K.D.; Kordower, J.H. Therapeutic approaches to target alpha-synuclein pathology. Exp. Neurol. 2017, 298, 225–235. [Google Scholar] [CrossRef] [PubMed]
  50. Zolunicant (MM-110) | Nicotinic α3β4 Receptor Inhibitor | MedChemExpress. MedchemExpress.com. Available online: https://www.medchemexpress.com/zolunicant.html (accessed on 22 September 2022).
  51. Programs & Research. MindMed. Available online: https://mindmed.co/programs-research/#scroll_section-2 (accessed on 22 September 2022).
  52. Bodnár, K.J.; Kakuk, P. Research ethics aspects of experimentation with LSD on human subjects: A historical and ethical review. Med. Health Care Philos. 2019, 22, 327–337. [Google Scholar] [CrossRef]
  53. Lancelotta, R.L.; Davis, A.K. Use of Benefit Enhancement Strategies among 5-Methoxy-N,N-Dimethyltryptamine (5-MeO-DMT) Users: Associations with Mystical, Challenging, and Enduring Effects. J. Psychoact. Drugs 2020, 53, 237–281. [Google Scholar] [CrossRef]
  54. Davis, A.K.; Barsuglia, J.P.; Lancelotta, R.; Grant, R.M.; Renn, E. The epidemiology of 5-methoxy-N, N-dimethyltryptamine (5-MeO-DMT) use: Benefits, consequences, patterns of use, subjective effects, and reasons for consumption. J. Psychopharmacol. 2018, 32, 779–792. [Google Scholar] [CrossRef]
  55. Cybin. Psychedelics to TherapeuticsTM. 2022. Available online: https://s28.q4cdn.com/259445127/files/doc_presentation/2022/Jan/Cybin-Psychedelics-to-Therapeutics2.0.pdf (accessed on 10 October 2022).
  56. Glue, P.; Lockhart, M.; Lam, F.; Hung, N.; Hung, C.-T.; Friedhoff, L. Ascending-dose study of noribogaine in healthy volunteers: Pharmacokinetics, pharmacodynamics, safety, and tolerability. J. Clin. Pharmacol. 2014, 55, 189–194. [Google Scholar] [CrossRef]
  57. Mash, D.C. Breaking the cycle of opioid use disorder with Ibogaine. Am. J. Drug Alcohol Abus. 2017, 44, 1–3. [Google Scholar] [CrossRef]
  58. Uthaug, M.V.; Davis, A.K.; Haas, T.F.; Davis, D.; Dolan, S.B.; Lancelotta, R.; Ramaekers, J.G. The epidemiology of mescaline use: Pattern of use, motivations for consumption, and perceived consequences, benefits, and acute and enduring subjective effects. J. Psychopharmacol. 2022, 36, 309–320. [Google Scholar] [CrossRef]
  59. Golani, L.K.; Divović, B.; Sharmin, D.; Pandey, K.P.; Mian, M.Y.; Cerne, R.; Witkin, J.M. Metabolism, pharmacokinetics, and anticonvulsant activity of a deuterated analog of the α2/3-selective GABAkine KRM-II-81. Biopharm. Drug Dispos. 2022, 43, 66–75. [Google Scholar] [CrossRef]
  60. Witkin, J.M.; Lippa, A.; Smith, J.L.; Jin, X.; Ping, X.; Biggerstaff, A.; Cook, J.M. The imidazodiazepine, KRM-II-81: An example of a newly emerging generation of GABAkines for neurological and psychiatric disorders. Pharmacol. Biochem. Behav. 2022, 213, 173321. [Google Scholar] [CrossRef] [PubMed]
  61. Listos, J.; Merska, A.; Fidecka, S. Pharmacological activity of salvinorin A, the major component of Salvia divinorum. Pharmacol. Rep. 2011, 63, 1305–1309. [Google Scholar] [CrossRef] [PubMed]
  62. Portbury, S.D.; Hare, D.J.; Finkelstein, D.I.; Adlard, P.A. Trehalose improves traumatic brain injury-induced cognitive impairment. PLoS ONE 2017, 12, e0183683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Leal, G.C.; Bandeira, I.D.; Correia-Melo, F.S.; Telles, M.; Mello, R.P.; Vieira, F.; Quarantini, L.C. Intravenous arketamine for treatment-resistant depression: Open-label pilot study. Eur. Arch. Psychiatry Clin. Neurosci. 2021, 271, 577–582. [Google Scholar] [CrossRef] [PubMed]
  64. Johannesdottir, A.; Sigurdsson, E. The use of psilocybin for treatment-resistant depression. Laeknabladid 2022, 108, 403–410. [Google Scholar] [CrossRef] [PubMed]
  65. Holland, J. Ecstasy: The Complete Guide: A Comprehensive Look at the Risks and Benefits of MDMA; Inner Traditions/Bear & Co.: Rochester, VT, USA, 2001; Available online: https://books.google.com/books?hl=en&lr=&id=CUCcyklcO00C&oi=fnd&pg=PP12&dq=ecstasy+risks&ots=2Fr82FHvpe&sig=OwI7oLniItoG4ZsNbyCl4zytI9k#v=onepage&q=ecstasy%20risks&f=false (accessed on 5 October 2022).
  66. Mitchell, J.M.; Bogenschutz, M.; Lilienstein, A.; Harrison, C.; Kleiman, S.; Parker-Guilbert, K.; Doblin, R. MDMA-assisted therapy for severe PTSD: A randomized, double-blind, placebo-controlled phase 3 study. Nat. Med. 2021, 27, 1025–1033. [Google Scholar] [CrossRef]
  67. Gao, M.; Rejaei, D.; Liu, H. Ketamine use in current clinical practice. Acta Pharmacol. Sin. 2016, 37, 865–872. [Google Scholar] [CrossRef] [Green Version]
  68. Schwalfenberg, G.K. N-Acetylcysteine: A Review of Clinical Usefulness (an Old Drug with New Tricks). J. Nutr. Metab. 2021, 2021, 9949453. [Google Scholar] [CrossRef]
  69. Ershad, M.; Vearrier, D. N Acetylcysteine. Nih.gov, 19 March 2019. Available online: https://www.ncbi.nlm.nih.gov/books/NBK537183/ (accessed on 13 October 2022).
  70. Vidović, M.; Rikalovic, M.G. Alpha-Synuclein Aggregation Pathway in Parkinson’s Disease: Current Status and Novel Therapeutic Approaches. Cells 2022, 11, 1732. [Google Scholar] [CrossRef] [PubMed]
  71. Swogger, M.T.; Walsh, Z. Kratom use and mental health: A systematic review. Drug Alcohol. Depend. 2018, 183, 134–140. [Google Scholar] [CrossRef] [PubMed]
  72. Neuroscience Trials Australia, Seelos Therapeutics, Inc. A Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Treatment Effects and Safety of SLS-005 (Trehalose Injection, 90.5 mg/mL for Intravenous Infusion) in Participants with Alzheimer’s Disease (AD); clinicaltrials.gov; Neuroscience Trials Australia: North Melbourne, Australia, 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT05332678 (accessed on 6 October 2022).
  73. Preuss, C.V.; Kalava, A.; King, K.C. Prescription of Controlled Substances: Benefits and Risks. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  74. Boot, B.P.; McGregor, I.S.; Hall, W. MDMA (Ecstasy) neurotoxicity: Assessing and communicating the risks. Lancet 2000, 355, 1818–1821. [Google Scholar] [CrossRef] [PubMed]
  75. Fonseca, D.A.; Ribeiro, D.M.; Tapadas, M.; Cotrim, M.D. Ecstasy (3,4-methylenedioxymethamphetamine): Cardiovascular effects and mechanisms. Eur. J. Pharmacol. 2021, 903, 174156. [Google Scholar] [CrossRef]
  76. Dos Santos, R.G.; Osório, F.L.; Crippa, J.A.; Riba, J.; Zuardi, A.W.; Hallak, J.E. Antidepressive, anxiolytic, and antiaddictive effects of ayahuasca, psilocybin and lysergic acid diethylamide (LSD): A systematic review of clinical trials published in the last 25 years. Ther. Adv. Psychopharmacol. 2016, 6, 193–213. [Google Scholar] [CrossRef] [Green Version]
  77. Multidisciplinary Association for Psychedelic Studies. LSD-assisted Psychotherapy in Persons Suffering from Anxiety Associated with Advanced-Stage Life Threatening Diseases; A Phase-II, Double-Blind, Placebo-Controlled Dose-Response Pilot Study. clinicaltrials.gov; Multidisciplinary Association for Psychedelic Studies: San Jose, CA, USA, 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT00920387 (accessed on 8 October 2022).
  78. Davis, A.K.; So, S.; Lancelotta, R.; Barsuglia, J.P.; Griffiths, R.R. 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT) used in a naturalistic group setting is associated with unintended improvements in depression and anxiety. Am. J. Drug Alcohol. Abuse 2019, 45, 161–169. [Google Scholar] [CrossRef]
  79. Reckweg, J.; Mason, N.L.; van Leeuwen, C.; Toennes, S.W.; Terwey, T.H.; Ramaekers, J.G. A Phase 1, Dose-Ranging Study to Assess Safety and Psychoactive Effects of a Vaporized 5-Methoxy-N, N-Dimethyltryptamine Formulation (GH001) in Healthy Volunteers. Front. Pharmacol. 2021, 12, 760671. [Google Scholar] [CrossRef]
  80. Globenewswire. COMPASS Pathways Presents Largest Ever Study of Psilocybin Therapy at American Psychiatric. Market Wire News. 23 May 2022. Available online: https://marketwirenews.com/news-releases/compass-pathways-presents-largest-ever-study-of-psil-7816213443619056.html (accessed on 12 October 2022).
  81. Check, E. Psychedelic drugs: The ups and downs of ecstasy. Nature 2004, 429, 126–128. [Google Scholar] [CrossRef]
  82. Cole, J.C.; Sumnall, H.R. Altered states: The clinical effects of Ecstasy. Pharmacol. Ther. 2003, 98, 35–58. [Google Scholar] [CrossRef]
  83. De La Torre, R.; Farre, M.; Ortuno, J.; Mas, M.; Brenneisen, R.; Roset, P.N.; Cami, J. Non-linear pharmacokinetics of MDMA (‘ecstasy’) in humans. Br. J. Clin. Pharmacol. 2000, 49, 104–109. [Google Scholar] [CrossRef]
  84. Mash, D.C.; Duque, L.; Page, B.; Allen-Ferdinand, K. Ibogaine Detoxification Transitions Opioid and Cocaine Abusers Between Dependence and Abstinence: Clinical Observations and Treatment Outcomes. Front. Pharmacol. 2018, 9, 529. [Google Scholar] [CrossRef] [PubMed]
  85. Brown, T.K.; Alper, K. Treatment of opioid use disorder with ibogaine: Detoxification and drug use outcomes. Am. J. Drug Alcohol. Abus. 2018, 44, 24–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Cappendijk, S.L.; Dzoljic, M.R. Inhibitory effects of ibogaine on cocaine self-administration in rats. Eur. J. Pharmacol. 1993, 241, 261–265. [Google Scholar] [CrossRef]
  87. Schenberg, E.E.; de Castro Comis, M.A.; Alexandre, J.F.M.; Chaves, B.D.R.; Tófoli, L.F.; da Silveira, D.X. Treating drug dependence with the aid of ibogaine: A qualitative study. J. Psychedelic Stud. 2017, 1, 10–19. [Google Scholar] [CrossRef]
  88. Köck, P.; Froelich, K.; Walter, M.; Lang, U.; Dürsteler, K.M. A systematic literature review of clinical trials and therapeutic applications of ibogaine. J. Subst. Abuse Treat. 2022, 138, 108717. [Google Scholar] [CrossRef] [PubMed]
  89. dos Santos, R.G.; Bouso, J.C.; Hallak, J.E.C. The antiaddictive effects of ibogaine: A systematic literature review of human studies. J. Psychedelic Stud. 2017, 1, 20–28. [Google Scholar] [CrossRef]
  90. Brown, T.K. Ibogaine in the treatment of substance dependence. Curr. Drug Abuse Rev. 2013, 6, 3–16. [Google Scholar] [CrossRef] [PubMed]
  91. MacLean, K.A.; Johnson, M.W.; Reissig, C.J.; Prisinzano, T.E.; Griffiths, R.R. Dose-related effects of salvinorin A in humans: Dissociative, hallucinogenic, and memory effects. Psychopharmacology 2013, 226, 381–392. [Google Scholar] [CrossRef]
  92. Halpern, J.H.; Sherwood, A.R.; Hudson, J.I.; Yurgelun-Todd, D.; Pope, H.G., Jr. Psychological and cognitive effects of long-term peyote use among Native Americans. Biol. Psychiatry 2005, 58, 624–631. [Google Scholar] [CrossRef]
  93. Zhou, Y.; Ma, B.; Sun, W.; Wang, J.; Fu, Y.; Wang, A.; Han, R. Effect of esketamine on perioperative depressive symptoms in major surgery patients (PASSION II): Study protocol for a randomized controlled trial. BMJ Open 2022, 12, e056713. [Google Scholar] [CrossRef]
Figure 1. Flow Search Diagram.
Figure 1. Flow Search Diagram.
Ijms 23 15777 g001
Figure 2. Psychoceuticals selected and their sources.
Figure 2. Psychoceuticals selected and their sources.
Ijms 23 15777 g002
Table 1. Biochemical structures, mechanisms, and receptor functions of twenty psychoceutical drugs.
Table 1. Biochemical structures, mechanisms, and receptor functions of twenty psychoceutical drugs.
Drug NameChemical StructureTarget TypeMethod of ActionCitations
5-Methoxy-N,N-dimenthyltryptamine
(5-MeO-DMT)
Ijms 23 15777 i001Serotonin Receptor5-MeO-DMT acts as a 5-HT analog in the serotonin system with a high affinity for the 5-HT1A receptor.Shen et al. [9]
Yu [10]
N,N-dimethyltryptamine (DMT)Ijms 23 15777 i002DMT activates pharmacological receptors, such as serotonin receptors, within the synaptic cleft after its secretion as a neurotransmitter. Although the exact molecular pathway of DMT remains unclear, the effects of DMT as a modulator for serotonergic systems are currently being tested.Carbonaro, Gatch [11]
MDMA-Derivative (Ecstasy)Ijms 23 15777 i003MDMA and derivatives increase serotonin and norepinephrine release while blocking their reuptake within the synaptic cleft.National Institute on Drug Abuse [12]
Lysergic Acid Diethylamide
(LSD)
Ijms 23 15777 i004LSD functions to increase serotonin concentration and acts as a 5-HT1A and 5-HT2 receptor agonist. LSD has inhibitory effects acting upon 5-HT1A receptors and stimulating effects acting upon 5-HT2 receptors.Andén et al. [13]
Passie et al. [14]
PeyoteIjms 23 15777 i005
* Mescaline, the active component of Peyote
Mescaline, the active component of Peyote, functions as a serotonin receptor agonist, especially 5-HT2 receptors, with a higher affinity for 5-HT2C receptors than for 5-HT2A receptors and 5-HT2B receptors. Peyote increases cytosolic levels of IP3 through Gq coupling of G-proteins to activate phospholipase C, stimulating calcium channel opening. Dinis-Oliveira et al. [15]
Deuterated Psilocybin Analog
(CYB003)
*Once converted to psilocin, CYB003 functions as a partial agonist of 5-HT2A receptors, a subtype of 5-HT2 receptors that act as serotonin and G protein-coupled (GCP) receptors.Cybin [16]
Schmidt et al. [17]
Deuterated DMT
(CYB004)
*CYB004 acts as an agonist of 5-HT2A serotonin receptors. Uptake of DMT occurs through serotonin uptake transporters (SERT) and is sequestered into synaptic vesicles via monoamine transporters. Cybin [18]
Barker [19]
Carbonaro, Gatch [11]
Phenethylamine derivative
(CYB005)
*CYB005 inhibits serotonin and dopamine transporters within the synaptic cleft. Cybin [20]
RxList [21]
Irsfeld et al. [22]
Psilocybin
(COMP360)
Ijms 23 15777 i006COMP360 acts as an agonist of 5-HT2A receptors, a subtype of 5-HT2 receptors that act on serotonin and G protein-coupled (GCP) receptors.Compass [23]
Schmidt et al. [17]
(R)-Ketamine (Arketamine)Ijms 23 15777 i007NMDA ReceptorArketamine acts as an NMDA receptor inhibitor with long lasting effects. Although the metabolism of Arketamine to 2R,6Rhydroxynorketamine (HNK) is known to have antidepressant effects, the exact molecular mechanisms of arketamine remain unclear. Zanos et al. [24]
Wei et al. [25]
SLS-002 (Ketamine)*SLS-002 functions as an NMDAr antagonist that binds to GABAergic interneurons, resulting in the disinhibition of glutamatergic neurons. A glutamate surge follows, stimulating AMPA receptors to potentiate BDNF and mTORC1 signaling pathways. Matveychuk et al. [26]
Derakhshanian et al. [27]
Deu-mitragynine (Kratom)*Opioid ReceptorDeu-mitragynine acts as a mu-opioid (MOR) agonist with the ability to interact with G-protein coupled receptors, resulting in its analgesic effects. Deu-mitragynine has been shown to exhibit opioid-receptor-dependent analgesic effects and G-protein-based agonists of MOR.Atai Life Sciences [28]
Shukla et al. [29]
Salvinorin AIjms 23 15777 i008Salvinorin A acts as an agonist of k-opioid receptors (KOPr) and is coupled to Gi/o proteins. Salvinorin A inhibits adenylyl cyclase and activates beta-arrestin mediated pathways and ERK/MAPK pathways.Brito-da-Costa et al. [30]
Roach, Shenvi [31]
12-methoxyibogamine
(Ibogaine)
Ijms 23 15777 i009Serotonin Receptor
NMDA Receptor
Opioid Receptor
Ibogaine functions as a kappa opioid receptor agonist, NMDA receptor antagonist, serotonin uptake receptor antagonist, and nicotinic receptor antagonist. Ibogaine’s underlying mechanism remains unclear.Glick et al. [32]
Healthtown [33]
Atai Life Sciences [34]
12-hydroxyibogamine
(Noribogaine)
Ijms 23 15777 i010Noribogaine is the main metabolite of Ibogaine and has a higher affinity for opioid receptors than its parent compound. Noribogaine acts as an NMDA receptor antagonist, k-opioid receptor agonist, serotonin reuptake inhibitor, and µ-opioid receptor antagonist. The exact mechanism behind its effects on k-opioid receptors remains unclear. Maillet et al. [35]
Atai Life Sciences [34]
Trehalose
(SLS-005)
Ijms 23 15777 i011GLUT8 ReceptorTrehalose stabilizes proteins, enhances autophagy, and enhances lysosomal pathways through mTOR-independent pathways. By increasing cytosolic levels of light chain 3-II, the inner and outer membranes of autophagosomes prepare for lysosome fusion. SLS-005 must act upon the mammalian trehalose transporter (GLUT8) for trehalose-induced autophagy in hepatocytes, but is unknown in neuronal cells. Seelos Therapeutics [36]
Rusmini et al. [37]
Lee et al. [38]
Sarkar et al. [39]
SLS-007*PeptideSLS-007 inhibits protein aggregation by targeting alpha-synuclein’s non-amyloid component cores (NAcore). Inc ST [40]
Seelos Therapeutics [41]
N-acetylcysteine (NAC)Ijms 23 15777 i012NF-κB NAC increases intracellular glutathione (GSH) concentration, modulates glutamatergic, neurotrophic, and inflammatory pathways, and suppresses NF-κB activity.Tenório et al. [42]
Bavarsad [43]
Zolunicant HCL
(MM-110)
*α3β4 receptors
MM-110 functions as an inhibitor for nicotinic α3β4 receptors and regulates dopamine levels during withdrawal.
O’Brien [44]
Deuterated Etifoxine
(Etifoxine-d3)
Ijms 23 15777 i013GABA receptors Deuterated etifoxine targets GABA channel subunits and projects GABAergic transmission via GABA receptors. Deuterated etifoxine can increase mitochondrial translocator protein (TSPO) to stimulate neurosteroid synthesis.Atai Life Sciences [45]
Biochemical structures, mechanisms, and receptor functions of twenty psychoceutical drugs. The table includes the chemical structures of known drugs, their receptor target type, and their biochemical method of action. Unknown chemical structures are marked with an asterisk (*).
Table 2. Highlights of the psychoceutical benefits.
Table 2. Highlights of the psychoceutical benefits.
DrugBenefitCitationsDrugBenefitCitations
LSDTreats chronic alcoholism and
anxiety
Bodnár, Kakuk [52]MM-110 (zolunicant HCl)Potential clinical utility to safely mitigate symptoms of opioid withdrawalO’Brien [44]
5-methoxy-N, N-dimethyltryptamine (5-MeO-DMT)Fewer suicidal thoughts, plans, attempts, and psychological distressLancelota, Davis [53]
Davis et al. [54]
CYB005 (Phenethylamine derivative)Treats neuroinflammation and psychiatric conditions Cybin [20]
Cybin [55]
IbogaineHumans: reduces cravings for heroin and withdrawal symptoms
Rats: reduces opioid withdrawal, as well as cocaine and heroin self-administration
Corkery [46]CYB004 (Deuterated DMT)Anxiety disorders, including generalized anxiety disorder, GAD, and social anxiety disorder, SAD.Cybin [18]
NoribogaineBlocks opioid cravings
Mice: fewer stress effects and less acute toxicity
Glue et al. [56]
Mash [57]
CYB003 (Deuterated Psilocybin Analog)Treats major depressive disorder (MDD) and alcohol use disorder (AUD).Cybin [16]
Peyote (mescaline)Improves depression, anxiety, and PTSDUthaug et al. [58]Deuterated etifoxine Treatment of anxietyGolani et al. [59]
Witkin et al. [60]
Salvinorin AFeelings of calm and relaxationListos et al. [61]
Brito et al. [30]
SLS-005 (Trehalose) Improving functional outcomes following TBIPortbury et al. [62]
ArketamineAntidepressantZanos et al. [24]
Leal et al. [63]
Psilocybin (COMP360)Psilocybin significantly reduces depressive symptomsJohannesdottir, Sigurdsson [64]
EcstasyMice: extinction of fear memories
Humans: attenuated amygdala response when shown negative stimuli
Holland [65]
Mitchell et al. [66]
KetamineAnti-depressant, pain management, anti-inflammatoryGao et al. [67]
N-acetylcysteineImproves cognition in schizophrenicsSchwalfenberg [68]
Muhammed, Vearrier [69]
SLS 007Treatment for PDInc ST [40]
Vidović, Rikalovic [70]
N,N-dimethyltryptamineAnti-anxiety/anti-psychotic via actions at the trace amino acid receptor Carbonaro, Gatch [11]KratomSubstitute for opioids among people who are addicted. Kratom also enhances mood and relieves anxietySwogger, Walsh [71]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cano, G.H.; Dean, J.; Abreu, S.P.; Rodríguez, A.H.; Abbasi, C.; Hinson, M.; Lucke-Wold, B. Key Characteristics and Development of Psychoceuticals: A Review. Int. J. Mol. Sci. 2022, 23, 15777. https://doi.org/10.3390/ijms232415777

AMA Style

Cano GH, Dean J, Abreu SP, Rodríguez AH, Abbasi C, Hinson M, Lucke-Wold B. Key Characteristics and Development of Psychoceuticals: A Review. International Journal of Molecular Sciences. 2022; 23(24):15777. https://doi.org/10.3390/ijms232415777

Chicago/Turabian Style

Cano, Genaro Herrera, Jordan Dean, Samuel Padilla Abreu, Amanda Hernández Rodríguez, Cyrena Abbasi, Madison Hinson, and Brandon Lucke-Wold. 2022. "Key Characteristics and Development of Psychoceuticals: A Review" International Journal of Molecular Sciences 23, no. 24: 15777. https://doi.org/10.3390/ijms232415777

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop