Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ISOTOPICALLY ENRICHED ANALOGS OF 5,6-METHYLENEDIOXY-2-AMINOINDANE (MDAI)
Document Type and Number:
WIPO Patent Application WO/2023/081895
Kind Code:
A1
Abstract:
Disclosed herein are isotopically enriched compounds and methods of using both isotopically and non-isotopically enriched compounds of the following formula (I), in the treatment of neurologic and brain disorders/conditions.

Inventors:
DUNCTON MATTHEW (US)
CLARK SAMUEL (US)
Application Number:
PCT/US2022/079411
Publication Date:
May 11, 2023
Filing Date:
November 07, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
TERRAN BIOSCIENCES INC (US)
DUNCTON MATTHEW (US)
CLARK SAMUEL (US)
International Classes:
A61P25/00; A61K31/36; C07D317/48; C07D317/50; C07D317/70
Other References:
ZIDKOVÁ MONIKA ET AL: "Study on the metabolism of 5,6-methylenedioxy-2-aminoindane (MDAI) in rats: identification of urinary metabolites", XENOBIOTICA, vol. 47, no. 6, 3 June 2017 (2017-06-03), UK, pages 505 - 514, XP055867643, ISSN: 0049-8254, Retrieved from the Internet DOI: 10.1080/00498254.2016.1199919
FOSTER A B: "Deuterium isotope effects in the metabolism of drugs and xenobiotics: implications for drug design", ADVANCES IN DRUG RESEARCH, ACADEMIC PRESS, LONDON, GB, vol. 14, 1 January 1985 (1985-01-01), pages 1 - 40, XP009086953, ISSN: 0065-2490
GRAHAM S TIMMINS: "Deuterated drugs: where are we now?", EXPERT OPINION ON THERAPEUTIC PATENTS, vol. 24, no. 10, 29 July 2014 (2014-07-29), GB, pages 1 - 9, XP055132523, ISSN: 1354-3776, DOI: 10.1517/13543776.2014.943184
LIEBERMAN, PHARMACEUTICAL DOSAGE FORMS, vol. I, II, 1992
LLOYD, THE ART, SCIENCE AND TECHNOLOGY OF PHARMACEUTICAL COMPOUNDING, 1999
PICKAR, DOSAGE CALCULATIONS, 1999
REMINGTON: "The Science and Practice of Pharmacy", 2003, LIPPINCOTT, WILLIAMS & WILKINS
PROGRESS IN NEURO-PSYCHOPHARMACOLOGY & BIOLOGICAL PSYCHIATRY, vol. 69, 2016, pages 49 - 59
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY
ROHATAGI, J. CLIN. PHARMACOL., vol. 35, 1995, pages 1187 - 1193
TJWA, ANN. ALLERGY ASTHMA IMMUNOL., vol. 75, 1995, pages 107 - 111
MINTO, J. PHARMACOL. EXP. THER., vol. 281, 1997, pages 93 - 102
RAO, J. BIOMATER SCI. POLYM. ED., vol. 7, 1995, pages 623 - 645
GAO, PHARM. RES., vol. 12, 1995, pages 857 - 863
EYLES, J. PHARM. PHARMACOL., vol. 49, 1997, pages 669 - 674
AL-MUHAMMED, J. MICROENCAPSUL., vol. 13, 1996, pages 293 - 306
CHONN, CURR. OPIN. BIOTECHNOL., vol. 6, 1995, pages 698 - 708
OSTRO, AM. J. HOSP. PHARM., vol. 46, 1989, pages 1576 - 1587
SHEPHERD, JKGREWEL, SSFLETCHER, ABILL, DJDOURISH, CT: "Behavioural and pharmacological validation of the elevated ''zero-maze'' as an animal model of anxiety", PSYCHOPHARMACOL, vol. 116, 1994, pages 56 - 64
"Diagnostic and statistical manual of mental disorders", 2013, AMERICAN PSYCHIATRIC ASSOCIATION
GOULD E.TANAPAT: "Stress and hippocampal neurogenesis", BIOL. PSYCHIATRY, vol. 46, 1999, pages 1472 - 1479
Attorney, Agent or Firm:
OWENS, Eric A. et al. (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. An isotopically enriched compound of the formula or a pharmaceutically acceptable salt thereof.

2. The compound of claim 1, wherein the compound is enriched in an isotope from 14C, tritium and deuterium.

3. The compound of claim 1, wherein the compound has the formula wherein at least one of Y1, Y2, Y3, Y4, Y5, Y6, Y7, Y8, Y9, Y10, and Y11 is enriched in deuterium, or a pharmaceutically acceptable salt thereof.

4. The compound of claim 1, wherein at least one of Y1, Y2, Y3, Y4, Y5, Y6, Y7, Y8, Y9, Y10, and Y11 is enriched in tritium.

5. The compound of claim 1, selected from the group

6. The compound of claim 1, having a structure selected from

7. A compound having the structure of any one of the compounds shown in Table 1 or a pharmaceutically acceptable salt thereof.

8. The compound of claim 6, wherein the compound is racemic.

9. The isotopically enriched compound of any one of claims 1-7, wherein the isotopically enriched compound is in the form of a pharmaceutically acceptable salt.

10. The compound of any one of the preceding claims, wherein the compound is optically active.

11. The isotopically enriched compound of any one of claims 1 - 10 wherein the isotopically enriched compound is in the form of a pharmaceutically acceptable salt.

12. The isotopically enriched compound of any one of claims 1 - 8 wherein the isotopically enriched compound is a free base.

13. A pharmaceutical composition comprising a compound of any one of claims 1 - 10.

14. A method for increasing neuronal plasticity, comprising contacting a neuron with an effective amount of a compound according to any one of claims 1 - 12.

15. The method of claim 14, wherein contacting comprises administering the compound to a subject.

16. A method for treating a neurological disorder, psychiatric disorder, or both, comprising contacting a subject having the neurological disorder, psychiatric disorder or both with an effective amount of a compound according to any one of claims 1 - 12.

17. The method of claim 16, wherein the neurological disorder is a neurodegenerative disorder.

67

18. The method of claim 17, wherein the neurological disorder, psychiatric disorder, or both, comprises depression, addiction, anxiety, or a post-traumatic stress disorder.

19. The method of claim 16, wherein the neurological disorder, psychiatric disorder, or both, comprises treatment resistant depression, suicidal ideation, major depressive disorder, bipolar disorder, schizophrenia, or substance use disorder.

20. The method of claim 16, wherein the neurological disorder, psychiatric disorder, or both, comprises stroke, traumatic brain injury, or a combination thereof.

21. The method of claim 16, further comprising administering to the subject an effective amount of an empathogenic agent.

22. The method of claim 21, wherein the empathogenic agent is MDMA.

23. The method of claim 14, further comprising administering a 5-HT2A antagonist to the subject.

24. The method of claim 21, wherein the 5-HT2A antagonist is selected from MDL- 11,939, eplivanserin (SR-46,349), ketanserin, ritanserin, altanserin, acepromazine, mianserin, mirtazapine, quetiapine, SB204741, SB206553, SB242084, LY272015, SB243213, blonanserin, SB200646, RS 102221, nefazodone, MDL- 100,907, pimavanserin, nelotanserin and lorcaserin.

68

Description:
ISOTOPICALLY ENRICHED ANALOGS OF 5,6-METHYLENEDIOXY-2- AMINOINDANE (MDAI)

CROSS REFERENCE TO RELATED APPLICATIONS

This application claims priority to and the benefit of U.S. Provisional Patent Application Nos. 63/276,541, filed November 5, 2021, and 63/319,252, filed on March 11, 2022. The contents of each are incorporated by reference in their entirety for all purposes.

BACKGROUND

Major depressive disorder and related neuropsychiatric diseases are among the leading causes of disability worldwide. Despite recent advances, there remains a need for new therapeutics to support treatment of debilitating neuropsychiatric diseases.

Recently, psychedelic compounds have received renewed interest for the treatment of depression and other disorders. For example, the Food and Drug Administration (FDA) recently approved the dissociative anesthetic ketamine for treatment-resistant depression, making it the first mechanistically distinct medicine to be introduced to psychiatry in nearly thirty years. Ketamine is a member of a class of compounds known as psychoplastogens. Psychoplastogens promote neuronal growth through a mechanism involving the activation of AMPA receptors, the tropomyosin receptor kinase B (TrkB), and the mammalian target of rapamycin (mTOR). As pyramidal neurons in the PFC exhibit top-down control over areas of the brain controlling motivation, fear, and reward, these effects support clinical development of psychoplastogenic compounds for their antidepressant, anxiolytic, and anti-addictive effects properties.

5,6-Methylenedioxy-2-aminoindane (MDAI):

5,6-methylenedioxy-2-aminoindane (MDAI)

6,7-dihydro-5/7-indeno[5,6-d][1 ,3]dioxol-6-amine has been reported as a synthetic analog of the psychedelic phenethylamine class of compounds. More particularly MDAI has been proposed as substitute for the empathogenic phenethylamine, MDMA. Unfortunately, problematic complications have been reported associated with MDAI administration. The present application addresses this currently unmet need. SUMMARY

Disclosed herein are isotopically enriched compounds of the formula

< J JL NH 2 Such compounds are, in certain embodiments, is enriched in an isotope, such as 14 C, tritium or deuterium. Certain examples of the disclosed compounds are represented by the formula . • , wherein at leas t t one o f f

Y 1 , Y 2 , Y 3 , Y 4 , Y 5 , Y 6 , Y 7 , Y 8 , Y 9 , Y 10 , and Y 11 is enriched in deuterium.

The foregoing and other objects, features, and advantages of the invention will become more apparent from the following detailed description. Also disclosed are methods for using the presently disclosed MDAI analogs for treating brain relates to the provision of isotopically enriched compounds with improved characteristics.

DETAILED DESCRIPTION

General

Disclosed herein are MDAI analogs, in particular, isotopically labeled MDAI analogs, or isotopologues of MDAI. The presently disclosed isotopologues are useful for the treatment of a variety of brain disorders and other conditions. Without limitation to any particular theory, it is believed that the present compounds increase neuronal plasticity, and increase at least one of translation, transcription, or secretion of neurotrophic factors. Moreover, by virtue of their isotopic enrichment, the presently disclosed compounds have improved pharmacokinetic and pharmacodynamic properties as compared to previously disclosed molecules. In certain embodiments the isotopic labels of the present compounds allow monitoring of its pharmacodynamic and ADME behavior following in vivo administration. In some embodiments, the isotopically enriched compounds described herein provide better therapeutic potential for neurological diseases than known compounds. BRIEF DESCRIPTION OF FIGURES

FIG. 1 illustrates the percentage of time spent in the open arms after MDAI compared to vehicle and chlordiazepoxide control on the elevated zero maze.

FIG. 2 illustrates the total open arm entries after MDAI compared to vehicle and chlordiazepoxide control on the elevated zero maze.

FIG. 3 illustrates the total line crossings after MDAI compared to vehicle and chlordiazepoxide control on the elevated zero maze.

FIG. 4 illustrates the frequency of HDIPS after MDAI compared to vehicle and chlordiazepoxide control on the elevated zero maze.

Terms and Abbreviations:

The term "isotopic enrichment factor" as used herein means the ratio between the isotopic abundance and the natural abundance of a specified isotope. It will be recognized that some variation of natural isotopic abundance occurs in a synthesized compound depending upon the origin of chemical materials used in the synthesis. Thus, a preparation of any compound will inherently contain small amounts of isotopologues, including deuterated isotopologues. The concentration of naturally abundant stable hydrogen isotopes, notwithstanding this variation, is small and immaterial as compared to the degree of stable isotopic substitution of compounds of this disclosure. In a compound of this disclosure, when a particular position is designated as having a particular isotope, such as deuterium, it is understood that the abundance of deuterium at that position is substantially greater than the natural abundance of deuterium, which is about 0.015% (on a mol/mol basis). A position designated as a particular isotope will have a minimum isotopic enrichment factor of at least 3000 (45% incorporation of the indicated isotope). Thus, isotopically enriched compounds disclosed herein having deuterium will have a minimum isotopic enrichment factor of at least 3000 (45% deuterium incorporation) at each atom designated as deuterium in the compound. Such compounds may be referred to herein as “deuterated” compounds.

In other embodiments, disclosed compounds have an isotopic enrichment factor for each designated atom of at least 3500 (52.5%). For example, for such disclosed compounds that are deuterium isotopologues, the compounds have an isotopic enrichment factor for each designated hydrogen atom of at least 3500 (52.5% deuterium incorporation at each designated atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation). As above, such compounds also are referred to as “deuterated” compounds.

In the compounds of this disclosure any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom. Unless otherwise stated, when a position is designated specifically as "H", the position is understood to have hydrogen at about its natural abundance isotopic composition.

The term "isotopologue" refers to a species that has the same chemical structure and formula as another compound, with the exception of the isotopic composition at one or more positions, e.g., H vs. D. Thus, isotopologues differ in their isotopic composition.

“Salt” refers to acid or base salts of the compounds used in the methods of the present invention, in particular pharmaceutically acceptable salts. Illustrative examples of pharmaceutically acceptable salts are mineral acid (hydrochloric acid, hydrobromic acid, phosphoric acid, and the like) salts, organic acid (fumaric acid, acetic acid, propionic acid, glutamic acid, citric acid, tartaric acid and the like) salts, quaternary ammonium (methyl iodide, ethyl iodide, and the like) salts. It is understood that the pharmaceutically acceptable salts are non-toxic. Additional suitable pharmaceutically acceptable salts are known to those of skill in the art. See, e.g., Remington: The Science and Practice of Pharmacy, volume I and volume II. (22 nd Ed., University of the Sciences, Philadelphia)., which is incorporated herein by reference.

The neutral forms of the compounds may be regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner. The parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the present invention.

“Pharmaceutically acceptable salt” refers to a compound in salt form, wherein the salt form is suitable for administration to a subject. Representative pharmaceutically acceptable salts include salts of acetic, ascorbic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, edisylic, fumaric, gentisic, gluconic, glucoronic, glutamic, hippuric, hydrobromic, hydrochloric, isethionic, lactic, lactobionic, maleic, malic, mandelic, methanesulfonic, mucic, naphthalenesulfonic, naphthalene- 1,5-disulfonic, naphthal ene-2, 6- disulfonic, nicotinic, nitric, orotic, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p- toluenesulfonic and xinafoic acid, and the like

“Pharmaceutically acceptable excipient” refers to a substance that aids the administration of an active agent to and absorption by a subject. Pharmaceutical excipients useful in the present invention include, but are not limited to, binders, fillers, disintegrants, lubricants, coatings, sweeteners, flavors and colors. One of skill in the art will recognize that other pharmaceutical excipients are useful in the present invention.

“Composition” refers to a product comprising the specified ingredients in the specified amounts, as well as any product, which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. By “pharmaceutically acceptable” it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation.

“Isomers” refers to compounds with same chemical formula but different connectivity between the atoms in the molecule, leading to distinct chemical structures. Isomers include structural isomers and stereoisomers. Examples of structural isomers include, but are not limited to tautomers and regioisomers. Examples of stereoisomers include but are not limited to diastereomers and enantiomers.

“Administering” refers to any suitable mode of administration, including, oral administration, administration as a suppository, topical contact, parenteral, intravenous, intraperitoneal, intramuscular, intralesional, intranasal or subcutaneous administration, intrathecal administration, or the implantation of a slow-release device e.g., a mini-osmotic pump, to the subject.

“Subject” refers to an animal, such as a mammal, including, but not limited to, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice and the like. In certain embodiments, the subject is a human subject.

“Therapeutically effective amount” or “therapeutically sufficient amount” or “effective or sufficient amount” refers to a dose that produces therapeutic effects for which it is administered. The exact dose will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, e.g. , Lieberman, Pharmaceutical Dosage Forms (vols. 1-3, 1992); Lloyd, The Art, Science and Technology of Pharmaceutical Compounding (1999); Pickar, Dosage Calculations (1999); and Remington: The Science and Practice of Pharmacy, 20th Edition, 2003, Gennaro, Ed., Lippincott, Williams & Wilkins). In sensitized cells, the therapeutically effective dose can often be lower than the conventional therapeutically effective dose for non- sensitized cells.

“Neuronal plasticity” refers to the ability of the brain to change its structure and/or function continuously throughout a subject’s life. Examples of the changes to the brain include, but are not limited to, the ability to adapt or respond to internal and/or external stimuli, such as due to an injury, and the ability to produce new neurites, dendritic spines, and synapses.

“Brain disorder” refers to a neurological disorder which affects the brain’s structure and function. Brain disorders can include, but are not limited to, Alzheimer’s, Parkinson’s disease, psychological disorder, depression, treatment resistant depression, addiction, anxiety, post- traumatic stress disorder, suicidal ideation, major depressive disorder, bipolar disorder, schizophrenia, stroke, traumatic brain injury, and substance use disorder.

“Combination therapy” refers to a method of treating a disease or disorder, wherein two or more different pharmaceutical agents are administered in overlapping regimens so that the subject is simultaneously exposed to both agents. For example, the compounds of the invention can be used in combination with other pharmaceutically active compounds. The compounds of the invention can be administered simultaneously (as a single preparation or separate preparation) or sequentially to the other drug therapy. In general, a combination therapy envisions administration of two or more drugs during a single cycle or course of therapy.

“Neurotrophic factors” refers to a family of soluble peptides or proteins which support the survival, growth, and differentiation of developing and mature neurons.

“Modulate” or “modulating” or “modulation” refers to an increase or decrease in the amount, quality, or effect of a particular activity, function or molecule. By way of illustration and not limitation, agonists, partial agonists, antagonists, and allosteric modulators (e.g., a positive allosteric modulator) of a G protein-coupled receptor (e.g., 5HT2A) are modulators of the receptor.

“Agonism” refers to the activation of a receptor or enzyme by a modulator, or agonist, to produce a biological response.

“Agonist” refers to a modulator that binds to a receptor or enzyme and activates the receptor to produce a biological response. By way of example only, “5HT2A agonist” can be used to refer to a compound that exhibits an ECso with respect to 5HT2A activity of no more than about 100 mM. In some embodiments, the term “agonist” includes full agonists or partial agonists. “Full agonist” refers to a modulator that binds to and activates a receptor with the maximum response that an agonist can elicit at the receptor. “Partial agonist” refers to a modulator that binds to and activates a given receptor, but has partial efficacy, that is, less than the maximal response, at the receptor relative to a full agonist.

“Positive allosteric modulator” refers to a modulator that binds to a site distinct from the orthosteric binding site and enhances or amplifies the effect of an agonist.

“Antagonism” refers to the inactivation of a receptor or enzyme by a modulator, or antagonist. Antagonism of a receptor, for example, is when a molecule binds to the receptor and does not allow activity to occur.

“Antagonist” or “neutral antagonist” refers to a modulator that binds to a receptor or enzyme and blocks a biological response. An antagonist has no activity in the absence of an agonist or inverse agonist but can block the activity of either, causing no change in the biological response.

Compounds:

Disclosed herein are isotopically labeled compounds of the formula , and methods for the use of such compounds in treating brain, neurological and psychiatric disorders.

The present inventors noted that MDAI does not have the drug-like pharmacokinetic and pharmacodynamic properties to support their wider use in the clinical treatment of brain disorders. In particular, serotonin syndrome has been reported associated with MDAI intoxication. See, Progress in Neuro-Psychopharmacology & Biological Psychiatry 69. 2016, 49-59. The present inventors observed that the toxicity of MDAI might be ameliorated by controlling or modifying its pharmacokinetic and pharmacodynamic properties, such as its absorption, distribution, metabolism and/or excretion (ADME) properties. In particular, the present inventors observed that the metabolic properties of MDAI could be improved upon by isotopic enrichment, in particular, deuterium or tritium enrichment. In this approach, the CYP- mediated metabolism of the molecule or to reduce the formation of undesirable metabolites is slowed, by replacing one or more protium ( 1 H) atoms with deuterium atoms. Deuterium is a safe, stable, non-radioactive isotope of hydrogen. Compared to protium, deuterium forms stronger bonds with carbon. In select cases, the increased bond strength imparted by deuterium can positively affect the pharmacokinetic properties of a drug, creating the potential for improved drug efficacy, safety, and/or tolerability. At the same time, because the size and shape of deuterium are essentially identical to those of protium, replacement of protium by deuterium would not be expected to affect the biochemical potency and selectivity of the drug as compared to the original chemical entity that contains only hydrogen. Tritium, 3 H, forms still stronger bonds with carbon than deuterium. Thus, replacement of protium with tritium also can affect the pharmacokinetic properties of a molecule. Moreover, tritium is a beta emitter, meaning that enriching a molecule with tritium allows determination of pharmacokinetic and pharmacodynamic properties of the molecule to better understand its activity and ADME properties.

Accordingly, in certain embodiments, the present invention provides an isotopically enriched compound of the formula

< J JL NH 2 Such compounds are, in certain embodiments, is enriched in an isotope, such as 14 C, tritium or deuterium. Certain examples of the disclosed compounds are represented by the formula . • , wherein at leas t t one o f f

Y 1 , Y 2 , Y 3 , Y 4 , Y 5 , Y 6 , Y 7 , Y 8 , Y 9 , Y 10 , and Y 11 is enriched in deuterium, tritium, or both. More particular embodiments of the disclosed isotopically enriched analogs have the formulas illustrated below:

10 Certain disclosed compounds have one or more chiral center. Examples of such chiral compounds include those of the structures The racemic mixtures, optically active mixtures and isolated diastereomers and mixtures thereof all are specifically contemplated herein.

In some embodiments, the present disclosure provides any one of the compounds in Table 1:

TABLE 1 Cooley Docket No.: STBI-034/02WO The compounds of the present invention, including those described in Table 1, can also be in salt forms, such as acid or base salts of the compounds of the present invention. Illustrative examples of pharmaceutically acceptable acid salts can be found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, which is incorporated herein by reference.

The present invention includes all tautomers and stereoisomers of compounds of the formulas disclosed herein, including those described in Table 1, either in admixture or in pure or substantially pure form. The compounds of the present invention can have asymmetric centers at the carbon atoms, and therefore the compounds of the present invention can exist in diastereomeric or enantiomeric forms or mixtures thereof. All conformational isomers (e.g., cis and trans isomers) and all optical isomers (e.g., enantiomers and diastereomers), racemic, diastereomeric and other mixtures of such isomers.

In addition, all physical forms of the disclosed compounds are intended herein, including the compounds of illustrated above and in Table 1, in the form of solvates, such as hydrates. Moreover, non-crystalline and crystalline forms of the isotopically enriched compounds disclosed herein, including amorphous forms, isomorphs and polymorphs are within the scope of the present invention.

Exemplary compounds according to the present invention are chiral. Such compounds can be prepared as is known to those of skill in the art can be prepared as single enantiomers, or enantiomerically enriched mixtures, or racemic mixtures as contemplated herein; such compounds having more than one stereocenter can also be prepared as diastereomeric, enantiomeric or racemic mixtures as contemplated herein. Furthermore, diastereomer and enantiomer products can be separated by chromatography, fractional crystallization or other methods known to those of skill in the art.

Pharmaceutical Compositions and Formulations

In some embodiments, the present invention provides a pharmaceutical composition comprising a compound of the present invention, such as a composition comprising a compound illustrated above or in Table 1, and a pharmaceutically acceptable excipient. Such compositions are suitable for administration to a subject, such as a human subject. The presently disclosed pharmaceutical compositions, including those described in Table 1, can be prepared in a wide variety of oral, parenteral and topical dosage forms. Oral preparations include tablets, pills, powder, capsules, liquids, lozenges, cachets, gels, syrups, slurries, suspensions, etc., suitable for ingestion by the patient. The compositions of the present invention can also be administered by injection, that is, intravenously, intramuscularly, intracutaneously, subcutaneously, intraduodenally, or intraperitoneally. Also, the compositions described herein can be administered by inhalation, for example, intranasally. Additionally, the compositions of the present invention can be administered transdermally. The compositions of this invention can also be administered by intraocular, intravaginal, and intrarectal routes including suppositories, insufflation, powders and aerosol formulations (for examples of steroid inhalants, see Rohatagi, J. Clin. Pharmacol. 35: 1187-1193, 1995; Tjwa, Ann. Allergy Asthma Immunol. 75: 107-111, 1995). Accordingly, the present invention also provides pharmaceutical compositions including a pharmaceutically acceptable carrier or excipient and the compounds of the present invention.

For preparing pharmaceutical compositions from the compounds disclosed herein, including those described in Table 1, pharmaceutically acceptable carriers can be either solid or liquid. Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible granules. A solid carrier can be one or more substances, which may also act as diluents, flavoring agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material. Details on techniques for formulation and administration are well described in the scientific and patent literature, see, e.g., the latest edition of Remington's Pharmaceutical Sciences, Mack Publishing Co, Easton PA ("Remington's").

In powders, the carrier is a finely divided solid, which is in a mixture with the finely divided active component. In tablets, the active component is mixed with the carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired. The powders and tablets preferably contain from 5% to 70% or 10% to 70% of the compounds of the present invention.

Suitable solid excipients include, but are not limited to, magnesium carbonate; magnesium stearate; talc; pectin; dextrin; starch; tragacanth; a low melting wax; cocoa butter; carbohydrates; sugars including, but not limited to, lactose, sucrose, mannitol, or sorbitol, starch from com, wheat, rice, potato, or other plants; cellulose such as methyl cellulose, hydroxypropylmethylcellulose, or sodium carboxymethylcellulose; and gums including arabic and tragacanth; as well as proteins including, but not limited to, gelatin and collagen.

If desired, disintegrating or solubilizing agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, alginic acid, or a salt thereof, such as sodium alginate.

For preparing suppositories, a low melting wax, such as a mixture of fatty acid glycerides or cocoa butter, is first melted and the compounds of the present invention are dispersed homogeneously therein, as by stirring. The molten homogeneous mixture is then poured into convenient sized molds, allowed to cool, and thereby to solidify.

Liquid form preparations include solutions, suspensions, and emulsions, for example, water or water/propylene glycol solutions. For parenteral injection, liquid preparations can be formulated in solution in aqueous polyethylene glycol solution.

Aqueous solutions suitable for oral use can be prepared by dissolving the compounds of the present invention in water and adding suitable colorants, flavors, stabilizers, and thickening agents as desired. Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethylene oxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol (e.g., polyoxyethylene sorbitol mono-oleate), or a condensation product of ethylene oxide with a partial ester derived from fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan mono-oleate). The aqueous suspension can also contain one or more preservatives such as ethyl or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose, aspartame or saccharin. Formulations can be adjusted for osmolarity.

Also included are solid form preparations, which are intended to be converted, shortly before use, to liquid form preparations for oral administration. Such liquid forms include solutions, suspensions, and emulsions. These preparations may contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents, and the like.

Oil suspensions can be formulated by suspending the compound of the present invention in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin; or a mixture of these. The oil suspensions can contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents can be added to provide a palatable oral preparation, such as glycerol, sorbitol or sucrose. These formulations can be preserved by the addition of an antioxidant such as ascorbic acid. As an example of an injectable oil vehicle, see Minto, J. Pharmacol. Exp. Ther. 281 :93-102, 1997. The pharmaceutical formulations of the invention can also be in the form of oil-in-water emulsions. The oily phase can be a vegetable oil or a mineral oil, described above, or a mixture of these. Suitable emulsifying agents include naturally occurring gums, such as gum acacia and gum tragacanth, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan mono-oleate, and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan mono-oleate. The emulsion can also contain sweetening agents and flavoring agents, as in the formulation of syrups and elixirs. Such formulations can also contain a demulcent, a preservative, or a coloring agent.

The compositions of the present invention can also be delivered as microspheres for slow release in the body. For example, microspheres can be formulated for administration via intradermal injection of drug- containing microspheres, which slowly release subcutaneously (see Rao, J. Biomater Sci. Polym. Ed. 7:623-645, 1995; as biodegradable and injectable gel formulations (see, e.g., Gao Pharm. Res. 12:857-863, 1995); or, as microspheres for oral administration (see, e.g., Eyles, J. Pharm. Pharmacol. 49:669-674, 1997). Both transdermal and intradermal routes afford constant delivery for weeks or months.

In some embodiments, the pharmaceutical compositions of the present invention can be formulated for parenteral administration, such as intravenous (IV) administration or administration into a body cavity or lumen of an organ. The formulations for administration will commonly comprise a solution of the compositions of the present invention dissolved in a pharmaceutically acceptable carrier. Among the acceptable vehicles and solvents that can be employed are water and Ringer's solution, an isotonic sodium chloride. In addition, sterile fixed oils can conventionally be employed as a solvent or suspending medium. For this purpose, any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid can likewise be used in the preparation of injectables. These solutions are sterile and generally free of undesirable matter. These formulations may be sterilized by conventional, well known sterilization techniques. The formulations may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, toxicity adjusting agents, e.g., sodium acetate, sodium chloride, potassium chloride, calcium chloride, sodium lactate and the like. The concentration of the compositions of the present invention in these formulations can vary widely, and will be selected primarily based on fluid volumes, viscosities, body weight, and the like, in accordance with the particular mode of administration selected and the patient's needs. For IV administration, the formulation can be a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. This suspension can be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation can also be a sterile injectable solution or suspension in a nontoxic parenterally-acceptable diluent or solvent, such as a solution of 1,3 -butanediol.

In some embodiments, the formulations of the compositions of the present invention can be delivered by the use of liposomes which fuse with the cellular membrane or are endocytosed, for example, by employing ligands attached to the liposome, or attached directly to the oligonucleotide, that bind to surface membrane protein receptors of the cell resulting in endocytosis. By using liposomes, particularly where the liposome surface carries ligands specific for target cells, or are otherwise preferentially directed to a specific organ, one can focus the delivery of the compositions of the present invention into the target cells in vivo. (See, e.g., Al-Muhammed, J. Microencapsul. 13:293-306, 1996; Chonn, Curr. Opin.

Biotechnol. 6:698-708, 1995; Ostro, Am. J. Hosp. Pharm. 46: 1576-1587, 1989).

Administration:

The compositions of the present invention, including those described in Table 1, can be administered by any suitable means, including oral, parenteral and topical methods. Transdermal administration methods, by a topical route, can be formulated as applicator sticks, solutions, suspensions, emulsions, gels, creams, ointments, pastes, jellies, paints, powders, and aerosols. The pharmaceutical preparation is preferably in unit dosage form. In such form the preparation is subdivided into unit doses containing appropriate quantities of the compounds of the present invention. The unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or ampoules. Also, the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.

The compound of the present invention can be present in any suitable amount, and can depend on various factors including, but not limited to, weight and age of the subject, state of the disease, and the like as is known to those of ordinary skill in the art. The compounds disclosed herein can be administered at any suitable frequency, interval and duration.

The compounds of the present invention, including those described in Table 1, can be coadministered with a second active agent. In some embodiments, co-administration can be accomplished by co-formulation, such as by preparing a single pharmaceutical composition including both the compound of the present invention and a second active agent. In other embodiments, the compound of the present invention and the second active agent can be formulated separately.

Methods of Treatment

The compounds of the present invention, such as a compound illustrated above or in Table 1, can be used for increasing neuronal plasticity. The compounds of the present invention can also be used to treat any brain disease. The compounds of the present invention can also be used for increasing at least one of translation, transcription or secretion of neurotrophic factors.

In some embodiments, a compound of the present invention, including those described in Table 1, is used to treat neurological diseases. In some embodiments, the compounds have, for example, anti- addictive properties, antidepressant properties, anxiolytic properties, or a combination thereof. In some embodiments, the neurological disease is a neuropsychiatric disease. In some embodiments, the neuropsychiatric disease is a mood or anxiety disorder. In some embodiments, the neurological disease is a migraine, headaches (e.g., cluster headache), post-traumatic stress disorder (PTSD), anxiety, depression, neurodegenerative disorder, Alzheimer’s disease, Parkinson’s disease, psychological disorder, treatment resistant depression, suicidal ideation, major depressive disorder, bipolar disorder, schizophrenia, stroke, traumatic brain injury, and addiction (e.g., substance use disorder). In some embodiments, the neurological disease is a migraine or cluster headache. In some embodiments, the neurological disease is a neurodegenerative disorder, Alzheimer’s disease, or Parkinson’s disease. In some embodiments, the neurological disease is a psychological disorder, treatment resistant depression, suicidal ideation, major depressive disorder, bipolar disorder, schizophrenia, post- traumatic stress disorder (PTSD), addiction (e.g., substance use disorder), depression, or anxiety. In some embodiments, the neuropsychiatric disease is a psychological disorder, treatment resistant depression, suicidal ideation, major depressive disorder, bipolar disorder, schizophrenia, post-traumatic stress disorder (PTSD), addiction (e.g., substance use disorder), depression, or anxiety. In some embodiments, the neuropsychiatric disease or neurological disease is post- traumatic stress disorder (PTSD), addiction (e.g., substance use disorder), schizophrenia, depression, or anxiety. In some embodiments, the neuropsychiatric disease or neurological disease is addiction (e.g., substance use disorder). In some embodiments, the neuropsychiatric disease or neurological disease is depression. In some embodiments, the neuropsychiatric disease or neurological disease is anxiety. In some embodiments, the neuropsychiatric disease or neurological disease is post-traumatic stress disorder (PTSD). In some embodiments, the neurological disease is stroke or traumatic brain injury. In some embodiments, the neuropsychiatric disease or neurological disease is schizophrenia.

In some embodiments, a compound of the present invention, including those described in Table 1, is used for increasing neuronal plasticity. In some embodiments, the compounds described herein, including those described in Table 1, are used for treating a brain disorder. In some embodiments, the compounds described herein, including those described in Table 1, are used for increasing at least one of translation, transcription, or secretion of neurotrophic factors.

In some embodiments, the present invention provides a method of treating a disease, including administering to a subject in need thereof, a therapeutically effective amount of a compound of the present invention, including those described in Table 1. In some embodiments, the disease is a musculoskeletal pain disorder including fibromyalgia, muscle pain, joint stiffness, osteoarthritis, rheumatoid arthritis, muscle cramps. In some embodiments, the present invention provides a method of treating a disease of women’s reproductive health including premenstrual dysphoric disorder (PMDD), premenstrual syndrome (PMS), post-partum depression, and menopause. In some embodiments, a single enantiomer of MDAI, e.g., a single enantiomer of isotopically enriched MDAI, e.g., deuterated MDAI, is administered to a subject in order to treat a disorder, e.g., an anxiety disorder or depressive disorder. Anxiety disorders that may be treated with a single enantiomer of MDAI, e.g., deuterated MDAI, include but are not limited to post traumatic stress disorder, generalized anxiety disorder, panic disorder, social anxiety disorder, obsessive-compulsive disorder, separation anxiety disorder, or agoraphobia Depressive disorders that may be treated with a single enantiomer include but are not limited to major depressive disorder, treatment resistant depression, persistent depressive disorder, seasonal effective disorder, premenstrual dysphoric disorder, prolonged grief disorder, bipolar depression, or psychotic depression.

In some embodiments, the compounds of the present invention, including those described in Table 1, have activity as 5-HT2A modulators. In some embodiments, the compounds of the present invention elicit a biological response by activating the 5-HT2A receptor (e.g., allosteric modulation or modulation of a biological target that activates the 5-HT2A receptor). 5-HT2A agonism has been correlated with the promotion of neural plasticity (Ly et al., 2018). 5-HT2A antagonists abrogate the neuritogenesis and spinogenesis effects of hallucinogenic compounds with 5-HT2A agonist activity, for example., DMT, LSD, and DOI. In some embodiments, the compounds of the present invention, including those described in Table 1, are 5-HT2A modulators and promote neural plasticity (e.g., cortical structural plasticity). In some embodiments, the compounds of the present invention, including those described in Table 1, are selective 5-HT2A modulators and promote neural plasticity (e.g., cortical structural plasticity). In some embodiments, promotion of neural plasticity includes, for example, increased dendritic spine growth, increased synthesis of synaptic proteins, strengthened synaptic responses, increased dendritic arbor complexity, increased dendritic branch content, increased spinogenesis, increased neuritogenesis, or any combination thereof. In some embodiments, increased neural plasticity includes, for example, increased cortical structural plasticity in the anterior parts of the brain.

In some embodiments, the 5-HT2A modulators (e.g., 5-HT2A agonists) are non- hallucinogenic. In some embodiments, non-hallucinogenic 5-HT2A modulators (e.g., 5-HT2A agonists) are used to treat neurological diseases, which modulators do not elicit dissociative sideeffects. In some embodiments, the hallucinogenic potential of the compounds described herein, including those described in Table 1, is assessed in vitro. In some embodiments, the hallucinogenic potential assessed in vitro of the compounds described herein, including those described in Table 1, is compared to the hallucinogenic potential assessed in vitro of hallucinogenic homologs. In some embodiments, the compounds described herein, including those described in Table 1, elicit less hallucinogenic potential in vitro than the hallucinogenic homologs.

In some embodiments, serotonin receptor modulators, such as modulators of serotonin receptor 2A (5-HT2A modulators, e.g., 5-HT2A agonists), are used to treat a brain disorder. The presently disclosed compounds, including those described in Table 1, can function as 5-HT2A agonists alone, or in combination with a second therapeutic agent that also is a 5-HT2A modulator. In such cases the second therapeutic agent can be an agonist or an antagonist. In some instances, it may be helpful administer a 5-HT2A antagonist in combination with a compound of the present invention to mitigate undesirable effects of 5-HT2A agonism, such as potential hallucinogenic effects. Serotonin receptor modulators useful as second therapeutic agents for combination therapy as described herein are known to those of skill in the art and include, without limitation, ketanserin, volinanserin (MDL-100907), eplivanserin (SR-46349), pimavanserin (ACP-103), glemanserin (MDL-11939), ritanserin, flibanserin, nelotanserin, blonanserin, mianserin, mirtazapine, roluperiodone (CYR-101, MIN- 101), quetiapine, olanzapine, altanserin, acepromazine, nefazodone, risperidone, pruvanserin, AC-90179, AC -279, adatanserin, fananserin, HY10275, benanserin, butanserin, manserin, iferanserin, lidanserin, pelanserin, seganserin, tropanserin, lorcaserin, ICI-169369, methiothepin, methysergide, trazodone, cinitapride, cyproheptadine, brexpiprazole, cariprazine, agomelatine, setoperone, 1- (l-Naphthyl)piperazine, LY-367265, pirenperone, metergoline, deramciclane, amperozide, cinanserin, LY-86057, GSK-215083, cyamemazine, mesulergine, BF-1, LY-215840, sergolexole, spiramide, LY-53857, amesergide, LY-108742, pipamperone, LY-314228, 5-1- R91150, 5-MeO-NBpBrT, 9-Aminomethyl-9,10-dihydroanthracene, niaprazine, SB-215505, SB- 204741 , SB-206553, SB-242084, LY-272015, SB-243213, SB-200646, RS-102221, zotepine, clozapine, chlorpromazine, sertindole, iloperidone, paliperidone, asenapine, amisulpride, aripiprazole, lurasidone, ziprasidone, lumateperone, perospirone, mosapramine, AMDA (9- Aminomethyl-9,10-dihydroanthracene), methiothepin, an extended-release form of olanzapine (e.g., ZYPREXA RELPREVV), an extended-release form of quetiapine, an extended-release form of risperidone (e.g., Risperdal Consta), an extended-release form of paliperidone (e.g., Invega Sustenna and Invega Trinza), an extended-release form of fluphenazine decanoate including Prolixin Decanoate, an extended-release form of aripiprazole lauroxil including Aristada, and an extended-release form of aripiprazole including Abilify Maintena, or a pharmaceutically acceptable salt, solvate, metabolite, deuterated analog, derivative, prodrug, or combinations thereof. In some embodiments, the serotonin receptor modulator used as a second therapeutic is pimavanserin or a pharmaceutically acceptable salt, solvate, metabolite, derivative, or prodrug thereof.

In some embodiments, non-hallucinogenic 5-HT2A modulators (e.g., 5-HT2A agonists) are used to treat neurological diseases. In some embodiments, the neurological diseases comprise decreased neural plasticity, decreased cortical structural plasticity, decreased 5-HT2A receptor content, decreased dendritic arbor complexity, loss of dendritic spines, decreased dendritic branch content, decreased spinogenesis, decreased neuritogenesis, retraction of neurites, or any combination thereof.

In some embodiments, non-hallucinogenic 5-HT2A modulators (e.g., 5-HT2A agonists) are used for increasing neuronal plasticity. In some embodiments, non-hallucinogenic 5-HT2A modulators (e.g., 5-HT2A agonists) are used for treating a brain disorder. In some embodiments, non-hallucinogenic 5-HT2A modulators (e.g., 5-FIT2A agonists) are used for increasing at least one of translation, transcription, or secretion of neurotrophic factors.

Methods for Increasing Neuronal Plasticity

Neuronal plasticity refers to the ability of the brain to change structure and/or function throughout a subject’s life. New neurons can be produced and integrated into the central nervous system throughout the subject’s life. Increasing neuronal plasticity includes, but is not limited to, promoting neuronal growth, promoting neuritogenesis, promoting synaptogenesis, promoting dendritogenesis, increasing dendritic arbor complexity, increasing dendritic spine density, and increasing excitatory synapsis in the brain. In some embodiments, increasing neuronal plasticity comprises promoting neuronal growth, promoting neuritogenesis, promoting synaptogenesis, promoting dendritogenesis, increasing dendritic arbor complexity, and increasing dendritic spine density. In some embodiments, increasing neuronal plasticity by treating a subject with a compound of the present invention, including those described in Table 1, can treat neurodegenerative disorder, Alzheimer’s, Parkinson’s disease, psychological disorder, depression, addiction, anxiety, post-traumatic stress disorder, treatment resistant depression, suicidal ideation, major depressive disorder, bipolar disorder, schizophrenia, stroke, traumatic brain injury, or substance use disorder.

In some embodiments, the present invention provides methods for increasing neuronal plasticity, comprising contacting a neuronal cell with a compound of the present invention, including those described in Table 1. In some embodiments, increasing neuronal plasticity improves a brain disorder described herein.

In some embodiments, a compound of the present invention, including those described in Table 1, is used to increase neuronal plasticity. In some embodiments, the compounds used to increase neuronal plasticity have, for example, anti- addictive properties, antidepressant properties, anxiolytic properties, or a combination thereof. In some embodiments, decreased neuronal plasticity is associated with a neuropsychiatric disease. In some embodiments, the neuropsychiatric disease is a mood or anxiety disorder. In some embodiments, the neuropsychiatric disease includes, for example, migraine, cluster headache, post-traumatic stress disorder (PTSD), schizophrenia, anxiety, depression, and addiction (e.g., substance abuse disorder). In some embodiments, brain disorders include, for example, migraines, addiction (e.g., substance use disorder), depression, and anxiety.

In some embodiments, the experiment or assay to determine increased neuronal plasticity of any compound of the present invention, including those described in Table 1, is a phenotypic assay, a dendritogenesis assay, a spinogenesis assay, a synaptogenesis assay, a Sholl analysis, a concentration-response experiment, a 5-HT2A agonist assay, a 5-HT2A antagonist assay, a 5-HT2A binding assay, or a 5-HT2A blocking experiment (e.g., ketanserin blocking experiments). In some embodiments, the experiment or assay to determine the hallucinogenic potential of any compound of the present invention, including those described in Table 1, is a mouse head-twitch response (HTR) assay.

In some embodiments, the present invention provides a method for increasing neuronal plasticity, comprising contacting a neuronal cell with a compound of the present invention, including those described in Table 1. Methods of Treating a Brain Disorder

In some embodiments, the present invention provides a method of treating a disease, including administering to a subject in need thereof, a therapeutically effective amount of a compound of the present invention, including those described in Table 1. In some embodiments, the disease is a musculoskeletal pain disorder including fibromyalgia, muscle pain, joint stiffness, osteoarthritis, rheumatoid arthritis, muscle cramps. In some embodiments, the present invention provides a method of treating a disease of women’s reproductive health including premenstrual dysphoric disorder (PMDD), premenstrual syndrome (PMS), post-partum depression, and menopause. In some embodiments, the present invention provides a method of treating a brain disorder, including administering to a subject in need thereof, a therapeutically effective amount of a compound of the present invention, including those described in Table 1. In some embodiments, the present invention provides a method of treating a brain disorder with combination therapy, including administering to a subject in need thereof, a therapeutically effective amount of a compound of the present invention, including those described in Table 1, and at least one additional therapeutic agent.

In some embodiments, 5-HT2A modulators (e.g., 5-HT2A agonists) are used to treat a brain disorder. In some embodiments, the brain disorders comprise decreased neural plasticity, decreased cortical structural plasticity, decreased 5-HT2A receptor content, decreased dendritic arbor complexity, loss of dendritic spines, decreased dendritic branch content, decreased spinogenesis, decreased neuritogenesis, retraction of neurites, or any combination thereof.

In some embodiments, a compound of the present invention, including those described in Table 1, is used to treat brain disorders. In some embodiments, the compounds have, for example, anti- addictive properties, antidepressant properties, anxiolytic properties, or a combination thereof. In some embodiments, the brain disorder is a neuropsychiatric disease. In some embodiments, the neuropsychiatric disease is a mood or anxiety disorder. In some embodiments, brain disorders include, for example, migraine, cluster headache, post-traumatic stress disorder (PTSD), anxiety, depression, panic disorder, suicidality, schizophrenia, and addiction (e.g., substance abuse disorder). In some embodiments, brain disorders include, for example, migraines, addiction (e.g., substance use disorder), depression, and anxiety. In some embodiments, the present invention provides a method of treating a brain disorder, comprising administering to a subject in need thereof a therapeutically effective amount of a compound disclosed herein, including those described in Table 1.

In some embodiments, the brain disorder is a neurodegenerative disorder, Alzheimer’s, Parkinson’s disease, psychological disorder, depression, addiction, anxiety, post-traumatic stress disorder, treatment resistant depression, suicidal ideation, major depressive disorder, bipolar disorder, schizophrenia, stroke, traumatic brain injury, or substance use disorder.

In some embodiments, the brain disorder is a neurodegenerative disorder, Alzheimer’s, or Parkinson’s disease. In some embodiments, the brain disorder is a psychological disorder, depression, addiction, anxiety, or a post-traumatic stress disorder. In some embodiments, the brain disorder is depression. In some embodiments, the brain disorder is addiction. In some embodiments, the brain disorder is treatment resistant depression, suicidal ideation, major depressive disorder, bipolar disorder, schizophrenia, stroke, traumatic brain injury or substance use disorder. In some embodiments, the brain disorder is treatment resistant depression, suicidal ideation, major depressive disorder, bipolar disorder, schizophrenia, or substance use disorder. In some embodiments, the brain disorder is stroke or traumatic brain injury. In some embodiments, the brain disorder is treatment resistant depression, suicidal ideation, major depressive disorder, bipolar disorder, or substance use disorder. In some embodiments, the brain disorder is schizophrenia. In some embodiments, the brain disorder is alcohol use disorder.

In some embodiments, the method further comprises administering one or more additional therapeutic agent that is lithium, olanzapine (Zyprexa), quetiapine (Seroquel), risperidone (Risperdal), ariprazole (Abilify), ziprasidone (Geodon), clozapine (Clozaril), divalproex sodium (Depakote), lamotrigine (Lamictal), valproic acid (Depakene), carbamazepine (Equetro), topiramate (Topamax), levomilnacipran (Fetzima), duloxetine (Cymbalta, Yentreve), venlafaxine (Effexor), citalopram (Celexa), fluvoxamine (Luvox), escitalopram (Lexapro), fluoxetine (Prozac), paroxetine (Paxil), sertraline (Zoloft), clomipramine (Anafranil), amitriptyline (Elavil), desipramine (Norpramin), imipramine (Tofranil), nortriptyline (Pamelor), phenelzine (Nardil), tranylcypromine (Parnate), diazepam (Valium), alprazolam (Xanax), or clonazepam (Klonopin).

In certain embodiments of the method for treating a brain disorder disclosed herein with a compound according to the present invention, including those described in Table 1, a second therapeutic agent agent that is an empathogenic agent is administered. Examples of suitable empathogenic agents for use in combination with an MDAI analog compound disclosed herein are selected from the phenethylamines, such as 3, 4-methylenedi oxymethamphetamine (MDMA) and analogs thereof. Other suitable empathogenic agents for use in combination with the presently disclosed compounds include, without limitation, N- Allyl-3, 4-methylenedi oxy-amphetamine (MDAL) A-Butyl-3, 4-methylenedi oxyamphetamine (MDBU) A-Benzyl-3, 4-methylenedi oxyamphetamine (MDBZ) A-Cyclopropyl methyl -3, 4-methylenedi oxyamphetamine (MDCPM) N, A-Di methyl -3, 4-methylenedi oxyamphetamine (MDDM) A -Ethyl -3, 4-methylenedi oxyamphetamine (MDE; MDEA) A-(2-Hydroxyethyl)-3, 4-methylenedi oxy amphetamine (MDHOET) A-Isopropyl-3, 4-methylenedi oxyamphetamine (MDIP) A-Methyl-3,4-ethylenedioxyamphetamine (MDMC) A-Methoxy-3, 4-methylenedi oxyamphetamine (MDMEO)

N-(2 -Methoxy ethyl)-3, 4-methylenedi oxyamphetamine (MDMEOET) alpha, alpha, A-Trimethyl-3, 4-methylenedi oxyphenethylamine (MDMP;

3.4-Methylenedioxy-A-methylphentermine)

A -Hydroxy-3, 4-methylenedi oxyamphetamine (MDOH)

3.4-Methylenedi oxyphenethylamine (MDPEA) alpha, alpha-Dimethyl-3, 4-methylenedi oxyphenethylamine (MDPH; 3,4- methylenedi oxyphentermine)

A-Propargyl-3,4-methylenedioxyamphetamine (MDPL) 1 , 3 -B enzodi oxoly 1 -N -methy Ibutanamine MBDB

3.4-methylenedi oxy -N-methyl-a-ethylphenylethylamine

3.4-Methylenedioxyamphetamine MDA

Methylone (also known as "3,4-methylenedioxy-N-methylcathinone) Ethylone, also known as 3,4-methylenedioxy-N-ethylcathinone GHB or Gamma Hydroxybutyrate or sodium oxybate

A -Propyl -3, 4-methylenedi oxyamphetamine (MDPR), and the like. In some embodiments, the compounds of the present invention, including those described in Table 1, are used in combination with the standard of care therapy for a neurological disease described herein. Non- limiting examples of the standard of care therapies, may include, for example, lithium, olanzapine, quetiapine, risperidone, ariprazole, ziprasidone, clozapine, divalproex sodium, lamotrigine, valproic acid, carbamazepine, topiramate, levomilnacipran, duloxetine, venlafaxine, citalopram, fluvoxamine, escitalopram, fluoxetine, paroxetine, sertraline, clomipramine, amitriptyline, desipramine, imipramine, nortriptyline, phenelzine, tranylcypromine, diazepam, alprazolam, clonazepam, or any combination thereof. Nonlimiting examples of standard of care therapy for depression are sertraline, fluoxetine, escitalopram, venlafaxine, or aripiprazole. Non-limiting examples of standard of care therapy for depression are citralopram, escitalopram, fluoxetine, paroxetine, diazepam, or sertraline. Additional examples of standard of care therapeutics are known to those of ordinary skill in the art.

Methods of increasing at least one of translation, transcription, or secretion of neurotrophic factors

Neurotrophic factors refers to a family of soluble peptides or proteins which support the survival, growth, and differentiation of developing and mature neurons. Increasing at least one of translation, transcription, or secretion of neurotrophic factors can be useful for, but not limited to, increasing neuronal plasticity, promoting neuronal growth, promoting neuritogenesis, promoting synaptogenesis, promoting dendritogenesis, increasing dendritic arbor complexity, increasing dendritic spine density, and increasing excitatory synapsis in the brain. In some embodiments, increasing at least one of translation, transcription, or secretion of neurotrophic factors can increasing neuronal plasticity. In some embodiments, increasing at least one of translation, transcription, or secretion of neurotrophic factors can promoting neuronal growth, promoting neuritogenesis, promoting synaptogenesis, promoting dendritogenesis, increasing dendritic arbor complexity, and/or increasing dendritic spine density.

In some embodiments, 5-HT2A modulators (e.g., 5-HT2A agonists) are used to increase at least one of translation, transcription, or secretion of neurotrophic factors. In some embodiments, a compound of the present invention, including those described in Table 1, is used to increase at least one of translation, transcription, or secretion of neurotrophic factors. In some embodiments, increasing at least one of translation, transcription or secretion of neurotrophic factors treats a migraine, headaches (e.g., cluster headache), post-traumatic stress disorder (PTSD), anxiety, depression, neurodegenerative disorder, Alzheimer’s disease, Parkinson’s disease, psychological disorder, treatment resistant depression, suicidal ideation, major depressive disorder, bipolar disorder, schizophrenia, stroke, traumatic brain injury, and addiction (e.g., substance use disorder).

In some embodiments, the experiment or assay used to determine increase translation of neurotrophic factors includes ELISA, western blot, immunofluorescence assays, proteomic experiments, and mass spectrometry. In some embodiments, the experiment or assay used to determine increase transcription of neurotrophic factors includes gene expression assays, PCR, and microarrays. In some embodiments, the experiment or assay used to determine increase secretion of neurotrophic factors includes ELISA, western blot, immunofluorescence assays, proteomic experiments, and mass spectrometry.

In some embodiments, the present invention provides a method for increasing at least one of translation, transcription or secretion of neurotrophic factors, comprising contacting a neuronal cell with a compound disclosed herein, such as a compound of the disclosed formulas or Table 1.

EXAMPLES

Exemplary compounds disclosed herein are prepared from isotopically labeled building blocks in place of non-labeled building blocks in syntheses known to those of skill in the art. General Conditions

Mass spectra were run on LC-MS systems using electrospray ionization. These were run using a Waters Acquity Classic UPLC with PDA and SQ mass detection or a Waters Acquity H-Class UPLC with PDA and QDA mass detection. [M+H] + refers to mono-isotopic molecular weights. NMR spectra were run on either a Bruker Ultrashield 400 MHz or 500MHz NMR spectrometer. Spectra were recorded at 298 K, unless otherwise stated, and were referenced using the solvent peak.

The following examples are intended to illustrate the invention and are not to be construed as being limitations thereon. Temperatures are given in degrees centigrade. If not mentioned otherwise, all evaporations are performed in vacuo, preferably between about 15 mm Hg and 100 mm Hg (= 20-133 mbar). The structure of final products, intermediates and starting materials is confirmed by standard analytical methods, e.g., MS and NMR. Abbreviations used are those conventional in the art. If not defined, the terms have their generally accepted meanings. Abbreviation

AcOH acetic acid app apparent br broad

CDCh chloroform-t/

D2O deuterium oxide d doublet d deuterium dd doublet of doublets

DBU l,8-diazabicyclo[5.4.0]undec-7-ene

DCM dichloromethane

DIPEA diisopropylethylamine

DMA dimethylacetamide

DMF N, A-di methyl form am ide

DMSO dimethyl sulfoxide

Et2O diethyl ether

EtOAc ethyl acetate

HC1 hydrochloric acid h hextet; sextet

HPLC high pressure liquid chromatography

IMS industrial methylated spirit

LC-MS liquid chromatography and mass spectrometry

MeOH methanol

MeCN acetonitrile

MS mass spectrometry m multiplet min(s) minute(s) mL milliliter(s) pL microliter(s) m/z mass to charge ratio

P pentet q quartet

N 2 nitrogen

NaHCCh sodium hydrogen carbonate

NaOH sodium hydroxide

Na 2 SO 4 sodium sulfate

NH 4 C1 ammonium chloride

NMP 7V-methyl-2-pyrrolidone

NMR nuclear magnetic resonance

Rt retention time s singlet t triplet tert tertiary

THF tetrahydrofuran

The various starting materials, intermediates, and compounds of the preferred embodiments may be isolated and purified, where appropriate, using conventional techniques such as precipitation, filtration, crystallization, evaporation, distillation, and chromatography. Salts may be prepared from compounds by known salt-forming procedures. Unless otherwise stated, all starting materials are obtained from commercial suppliers and used without further purification. More specific compounds required for the syntheses are listed below: 5,6-Methylenedioxy-l -indanone (cas 6412-87-9) purchased from Sigma Aldrich (catalogue number 657573)

If not indicated otherwise, the analytical HPLC conditions are as follows:

Instrument: LC-MS-1:

Method 2A

Column: Acquity UPLC BEH C18 2.1 x 50 mm 1.7 pm

Column Temp: 50 °C

Flow rate: 0.8 mL/min.

Eluents: A: H 2 O, 0.1% formic acid, B: MeCN Gradient: 0.0-1.8 min 2-98% B, 1.8-2.1 min 98% B, 2.1-2.5 98% A.

Method 2B

Column: Acquity UPLC BEH C18 2.1 x 50 mm 1.7 pm

Column Temp: 50 °C

Flow rate: 0.8 mL/min.

Eluents: A: H2O, 0.1% ammonia B: MeCN

Gradient: 0.0-1.8 min 2-98% B, 1.8-2.1 min 98% B, 2.1-2.5 98% A.

Instrument: LC-MS-2:

Method 2A

Column: Acquity UPLC BEH C18 2.1 x 50 mm 1.7 pm

Column Temp: 50 °C

Flow rate: 0.8 mL/min.

Eluents: A: H 2 O, B: MeCN, C: 50% H2O / 50% MeCN + 2.0% formic acid

Gradient: 0.0 - 1.7 mins 0-95% B, 5% C; 1.7-2.1 mins 95% B, 5% C

2.1-2.5 mins 95% A, 5% C.

Method 2B

Column: Acquity UPLC BEH C18 2.1 x 50 mm 1.7 pm

Column Temp: 50 °C

Flow rate: 0.8 mL/min.

Eluents: A: H2O, B: MeCN, C: 50% H2O / 50% MeCN + 2.0% ammonia (aq.)

Gradient: 0.0 - 1.7 mins 0-95% B, 5% D; 1.7-2.1 mins 95% B, 5% D

2.1-2.5 mins 95% A, 5% D.

Example 1: 4-Bromo-l,2-dideuteriooxy-benzene

A solution of 4-bromobenzene-l,2-diol (5.37 g, 28.4 mmol) in THF (15 mL) and D2O (15 mL) was stirred at rt under N2 overnight. The mixture was concentrated in vacuo. Methanol -di (15 mL) and D2O (15 mL) were added to the residue and the mixture was then stirred at rt under N2 for 3 days. The mixture was concentrated in vacuo to leave 4-bromo-l,2-dideuteriooxy -benzene (5.20 g, 96% yield) as a solid. Spectroscopic data of the title compound showed 85% deuterium incorporation. The intermediate was used in the next step without further purification. 1 H NMR (400 MHz, CDCh) 6 7.02 (d, J= 2.3 Hz, 1H), 6.93 (dd, J= 8.4, 2.3 Hz, 1H), 6.74 (d, J= 8.4 Hz, 1H), 5.32 (br. s, 0.15H), 5.17 (br. s, 0.15H).

The compounds of the following tabulated Examples (Table Exl) were prepared analogously to Example 1 from the appropriate catechol.

Table Exl

Example 2: 5-Bromo-2,2-dideuterio-l,3-benzodioxole

A solution of 4-bromo-l,2-dideuteriooxy -benzene (4.68 g, 24.5 mmol) in DCM-tfc (6.39 g, 73.5 mmol, 4.73 mL) and NMP (4.5 mL) was added dropwise over 5 min to a stirred suspension of cesium carbonate (15.97 g, 49.0 mmol) in NMP (9 mL) and D2O (1.47 g, 73.5 mmol, 1.33 mL) at rt under an atmosphere of N2. The mixture was heated to 110 °C and stirred for 2 h. The mixture was cooled to rt and then filtered. H2O (50 mL) and EtOAc (50 mL) were added to the filtrate. The separated aqueous phase was extracted with EtOAc (2 x 25 mL) and the combined organic fractions were then washed with brine (50 mL), dried over Na2SO4 and concentrated in vacuo. The residue was purified by column chromatography on silica gel, eluting with 10% EtOAc in petroleum ether, to give 5-bromo-2,2-dideuterio-l,3-benzodioxole (3.10 g, 62% yield) as an oil. Spectroscopic data of the title compound showed 99% deuterium incorporation. LC- MS (LCMS2: Method 2A): Rt 1.56 mins; MS m/z not observed; X H NMR (400 MHz, CDCh) 6 6.98 - 6.91 (m, 2H), 6.69 (d, J= 8.2 Hz, 1H). The compounds of the following tabulated Examples (Table Ex2) were prepared analogously to Example 2 from the appropriate deuterated catechol.

Table Ex2

Example 3: 3-(l,3-Benzodioxol-5-yl)prop-2-ynoic acid

Propiolic acid (1.92 g, 27.4 mmol, 1.68 mL) was added dropwise over 10 min to a stirred solution of 5-bromo-l,3-benzodioxole (5.10 g, 25.4 mmol, 3.00 mL) and DBU (8.33 g, 54.7 mmol, 8.18 mL) in de-oxygenated DMSO (50 mL) at rt under an atmosphere of N2. The mixture was stirred at rt for 5 min and then tetrakis(triphenylphosphine)palladium(0) (2.87 g, 2.49 mmol) was added in one portion. The mixture was heated to 50 °C and stirred overnight. EtOAc (125 mL), saturated aqueous NaHCOs (125 mL) and H2O(75 mL) were added to the mixture. The biphasic mixture was agitated and then filtered. The filtrate was separated and the pH of the aqueous phase was adjusted to pH ~l-2 by dropwise addition of 2M aqueous HC1 (~90 mL). The aqueous phase was extracted with DCM (3 x 150 mL) and the combined organic fractions were then dried over Na2SO4 and concentrated in vacuo. The residue was purified by column chromatography on silica gel, eluting with a gradient of 0-80% EtOAc in petroleum ether with 2% AcOH, to leave 3-(l,3-benzodioxol-5-yl)prop-2-ynoic acid (1.73 g, 37% yield) as a solid. LC-MS (LCMS2: Method 2A): Rt 1.12 mins; MS m/z 189.0 = [M-H]'; ’H NMR (400 MHz, DMSO-tA) 6 13.65 (br. s, 1H), 7.21 - 7.15 (m, 2H), 7.03 - 6.99 (m, 1H), 6.12 (s, 2H). The compounds of the following tabulated Examples (Table Ex3) were prepared analogously to Example 3 from the appropriate bromide.

Table Ex3

Example 4: 3-(l,3-Benzodioxol-5-yl)-2,2,3,3-tetradeuterio-propanoic acid

A solution of sodium carbonate (3.06 g, 28.9 mmol) in D2O (27 mL) was added to 3-(l ,3- benzodioxol-5-yl)prop-2-ynoic acid (1.83 g, 9.62 mmol) at rt under an atmosphere of N2. The mixture was stirred at rt for 30 min. Palladium on carbon (1.02 g, 0.96 mmol, 10 wt%) was added in one portion to the mixture at rt and the mixture was then heated to 50 °C. A solution of hypophosphorous acid-t/s (3.99 g, 28.9 mmol, 2.95 mL, 50 wt% in D2O) in D2O (18 mL) was added dropwise over 10 min to the mixture at 50 °C and the resulting mixture was then stirred at this temperature overnight. The pH of the mixture was adjusted to pH 1 by dropwise addition of 2M aqueous HC1 (20 mL). Chloroform (50 mL) was added and the biphasic mixture was then filtered through celite, eluting with chloroform (2 x 50 mL). The filtrate was separated and the aqueous phase was extracted with chloroform (2 x 50 mL). The combined organic fractions were dried over Na2SO4 and concentrated in vacuo to leave 3-(l,3-benzodioxol-5-yl)-2, 2,3,3- tetradeuterio-propanoic acid (1.66 g, 87% yield) as a solid. The title compound was used in the next step without further purification. LC-MS (LCMS2: Method 2A): Rt 1.18 mins; MS m/z not observed;^ NMR (400 MHz, DMSO-t/e) 6 12.08 (br. s, 1H), 6.84 - 6.76 (m, 2H), 6.67 (dd, J= 7.9, 1.7 Hz, 1H), 5.95 (s, 2H); 2 H NMR (61 MHz, DMSO-t/e) 6 2.68 (s, 2D), 2.43 (s, 2D).

The compounds of the following tabulated Examples (Table Ex4) were prepared analogously to Example 4 from the appropriate alkyne.

Table Ex4

Example 5: (£')-3-(2,2-Dideuterio-l,3-benzodioxol-5-yl)prop-2-enoic acid

Piperidine (178 mg, 2.09 mmol, 206 pL) was added dropwise over 1 min to a stirred solution of

2.2-dideuterio-l,3-benzodioxole-5-carbaldehyde (3.80 g, 25.0 mmol) and malonic acid (4.86 g, 46.7 mmol) in pyridine (10.5 mL) at rt under an atmosphere of N2. The mixture was stirred at 105 °C for 3 h. The mixture was cooled to rt and then poured into 4M aqueous HC1 (57.5 mL) at 0 °C. The mixture was stirred at 0 °C for 10 min and then the precipitated solid was collected by filtration. H2O (35 mL) was added to the solid and the mixture was then stirred at rt for 10 min before the solid was again collected by filtration. The solid was recrystallised in a 50% mixture of MeOH in H2O(160 mL), filtered and then dried under vacuum to leave (£)-3-(2,2-dideuterio-

1.3-benzodioxol-5-yl)prop-2-enoic acid (4.00 g, 82% yield) as a solid. LC-MS (LCMS2: Method 2A): Rt 1.21 mins; MS m/z 195.1 = [M+H] + ;LC-MS (LCMS2: Method 2B): Rt 0.45 mins; MS m/z 193.1 = [M-HJ’ HNMR (400 MHz, DMSO-t/e) 6 12.24 (br. s, 1H), 7.51 (d, J= 15.9 Hz, 1H), 7.37 (d, J= 1.7 Hz, 1H), 7.16 (dd, J= 8.1, 1.7 Hz, 1H), 6.95 (d, J= 8.1 Hz, 1H), 6.40 (d, J = 15.9 Hz, 1H).

Example 6: 3-(2,2-Dideuterio-l,3-benzodioxol-5-yl)propanoic acid

A suspension of (£)-3-(2,2-dideuterio-l,3-benzodioxol-5-yl)prop-2-enoic acid (4.00 g, 20.6 mmol) and palladium on carbon (240 mg, 0.23 mmol, 10wt%) in IMS (40 mL) was stirred at rt under an atmosphere of hydrogen overnight. The mixture was filtered through celite, eluting with IMS (2 x 20 mL) and the filtrate was then concentrated in vacuo to leave 3-(2,2-dideuterio-l,3- benzodioxol-5-yl)propanoic acid (3.90 g, 97% yield) as a solid. The title compound was used in the next step without further purification. LC-MS (LCMS2: Method 2A): Rt 1.19 mins; MS m/z not observed; LC-MS (LCMS2: Method 2B): Rt 0.44 mins; MS m/z 195.0 = [M-HJ’ HNMR (400 MHz, CDCk) 6 9.68 (br. s, 1H), 6.73 (d, J= 7.9 Hz, 1H), 6.70 (d, J= 1.8 Hz, 1H), 6.66 (dd, J= 7.9, 1.8 Hz, 1H), 2.88 (t, J= 7.7 Hz, 2H), 2.64 (t, J= 7.7 Hz, 2H).

Example 7: 2,2-Dideuterio-5,6-dihydrocyclopenta[f|[l,3]benzodioxol-7-on e

Oxalyl chloride (3.78 g, 29.8 mmol, 2.60 mL) was added dropwise over 10 min to a stirred solution of 3-(2,2-dideuterio-l,3-benzodioxol-5-yl)propanoic acid (3.90 g, 19.9 mmol) in DCM (30 mL) at 0 °C under an atmosphere of N2. The mixture was stirred at 0 °C for 10 min, warmed to rt and then stirred at this temperature for 2 h. The mixture was concentrated in vacuo. DCM (30 mL) was added to the residue and then aluminium chloride (3.18 g, 23.9 mmol) was added portionwise over 30 min to the stirred mixture at 0 °C under an atmosphere of N2. The mixture was stirred at 0 °C for 10 min, warmed to rt and then stirred at this temperature for 2 h. The mixture was cooled to 0 °C and then ice cold H2O(60 mL) was added dropwise. The separated aqueous phase was extracted with DCM (2 x 60 mL) and then the combined organic fractions were washed with brine (2 x 60 mL), dried over Na2SO4 and concentrated in vacuo. The residue was purified by column chromatography on silica gel, eluting with 50-60 % EtOAc in petroleum ether, to leave 2,2-dideuterio-5,6-dihydrocyclopenta[f][l,3]benzodioxol-7-on e (2.20 g, 62% yield) as a solid. LC-MS (LCMS2: Method 2A): Rt 1.17 mins; MS m/z 179.1 = [M+H] + ;LC-MS (LCMS2: Method 2B): Rt 1.14 mins; MS m/z 179.1 = [M+H] + ; X H NMR (400 MHz, CDCh) 6 7.11 (s, 1H), 6.84 (s, 1H), 3.04 - 3.00 (m, 2H), 2.70 - 2.66 (m, 2H). The compounds of the following tabulated Examples (Table Ex7) were prepared analogously to

Example 7 from the appropriate carboxylic acid.

Table Ex7 Example 8: (6Z)-2.2-l)ideiiterio-6-hydroxyimino-5//-cyclopent:i|f|| 1.3|benzodioxol-7-one Isopentyl nitrite (947 mg, 8.08 mmol, 1.08 mL) was added dropwise over 5 min to a stirred solution of 2,2-dideuterio-5,6-dihydrocyclopenta[f][l,3]benzodioxol-7-on e (1.20 g, 6.73 mmol) in MeOH (100 mL) at 45 °C under an atmosphere of N2, followed by concentrated HC1 (246 mg, 6.73 mmol, 0.63 mL) which was added dropwise over 1 min. The mixture was cooled to rt and then stirred at this temperature for 1 h. The precipitated solid was collected by filtration, washed with cold MeOH (10 mL) and then dried under vacuum to leave (6Z)-2,2-dideuterio-6- hydroxyimino-5J/-cyclopenta[f][l,3]benzodioxol-7-one (1.00 g, 72% yield) as a solid. The title compound was used in the next step without further purification. LC-MS (LCMS2: Method 2A): Rt 1.07 mins; MS m/z 208.1 = [M+H] + ; LC-MS (LCMS2: Method 2B): Rt 0.92 mins; MS m/z 208.1 = [M+H] + ; 1 H NMR (400 MHz, DMSO-t/e) 8 12.45 (br. s, 1H), 7.17 (s, 1H), 7.14 (s, 1H),

3.65 (s, 2H). The compounds of the following tabulated Examples (Table Ex8) were prepared analogously to Example 8 from the appropriate ketone.

Table Ex8

Example 9: 2,2-Dideuterio-6,7-dihydro-5H-cyclopenta[f| [l,3]benzodioxol-6-amine hydrochloride (compound 1) A suspension of (6Z)-2,2-dideuterio-6-hydroxyimino-5J/-cyclopenta[f][l,3]ben zodioxol-7-one (210 mg, 1.01 mmol) and palladium on carbon (100 mg, 0.09 mmol, 10wt%) in acetic acid (2 mL) and sulfuric acid (99 mg, 1.01 mmol, 54 pL) was stirred at rt under an atmosphere of hydrogen for 4 h. The mixture was filtered through the celite, eluting with MeOH (15 mL). The filtrate was purified using an SCX-2 cartridge, eluting with MeOH (15 mL), MeCN (15 mL), MeOH (15 mL) and then 2 M ammonia in MeOH (3 x 15 mL), to leave an oil. The residue was further purified by reverse phase chromatography, eluting with a gradient of 10-40% MeCN in water with 0.1% ammonia, to leave an oil. The residue was dissolved in Et2O (6 mL) and 1,4- di oxane (2 mL) and then 4 M HCI in dioxane (0.5 mL) was added. The precipitated solid was collected by filtration, washed with Et2O (4 x 1 mL), and then the solid was dried under vacuum at 50 °C to leave 2,2-dideuterio-6,7-dihydro-5J/-cyclopenta[f][l,3]benzodioxol -6-amine hydrochloride (20 mg, 9% yield) as a solid. LC-MS (LCMS2: Method 2A): Rt 0.77 mins; MS m/z 180.2 = [M+H] + ; LC-MS (LCMS2: Method 2B): Rt 1.11 mins; MS m/z 180.1 = [M+H] + ; ’H NMR (400 MHz, D 2 O) 8 6.86 (s, 2H), 4.21 - 4.13 (m, 1H), 3.33 (dd, J= 16.7, 7.1 Hz, 2H), 2.94 (dd, J= 16.7, 3.5 Hz, 2H). NH2 and HCI not observed; 2 H NMR (61 MHz, H2O) 8 5.91 (s, 2D). Example 10: 2,2,5,5,6,7,7-Heptadeuteriocyclopenta[f| [l,3]benzodioxol-6-amine hydrochloride (compound 2)

A suspension of (6Z)-2,2,7,7-tetradeuterio-6-hydroxyimino-cyclopenta[f][l,3] benzodioxol-5-one (160 mg, 0.76 mmol) and palladium on carbon (122 mg, 0.11 mmol, 10wt%) in acetic acid-t/4 (4 mL) and sulfuric acid-t 2 (78 mg, 0.76 mmol, 42 pL) was stirred at rt under an atmosphere of deuterium gas overnight. The mixture was filtered through the celite, eluting with MeOH (15 mL). The filtrate was purified using an SCX-2 cartridge, eluting with MeOH (50 mL), MeCN (50 mL), MeOH (50 mL) and then 2 M ammonia in MeOH (2 x 50 mL), to leave an oil. The residue was dissolved in Et20 (6 mL) and 1,4-dioxane (2 mL) and then 4M HC1 in dioxane (0.5 mL) was added. The precipitated solid was collected by filtration, washed with Et2O (3 x 2 mL), and then the solid was dried under vacuum at 50 °C to leave 2, 2, 5, 5, 6,7,7- heptadeuteriocyclopenta[f][l,3]benzodioxol-6-amine hydrochloride (121 mg, 70% yield) as a solid. Spectroscopic data of the title compound showed 90% deuterium incorporation. LC-MS (LCMS2: Method 2A): Rt 0.75 mins; MS m/z 185.1 = [M+H] + ; LC-MS (LCMS2: Method 2B): Rt 1.13 mins; MS m/z 185.1 = [M+H] + ; ’H NMR (400 MHz, D 2 O) 8 6.86 (s, 2H). NH2 and HC1 not observed; 2 H NMR (61 MHz, H2O) 8 5.90 (s, 2D), 4.11 (s, ID), 3.25 (s, 2D), 2.87 (s, 2D). The compounds of the following tabulated Examples (Table ExlO) were prepared analogously to Example 10 from the appropriate oxime.

Table ExlO Example 11: Evaluation of Metabolic Stability in Human Liver Microsomes

Microsomal Assay: Human liver microsomes (20 mg/mL) were obtained. P-nicotinamide adenine dinucleotide phosphate, reduced form (NADPH), magnesium chloride (MgCh), and dimethyl sulfoxide (DMSO) are purchased from Sigma-Aldrich.

Determination of Metabolic Stability: 7.5 mM stock solutions of test compounds of the above structural formula (e.g., of an embodiment or aspect of embodiment thereof described herein), or pharmaceutically acceptable salt thereof, are prepared in DMSO. The 7.5 mM stock solutions are diluted to 12.5-50 pM in acetonitrile (ACN). The 20 mg/mL human liver microsomes are diluted to 0.625 mg/mL in 0.1 M potassium phosphate buffer, pH 7.4, containing 3 mM MgCh. The diluted microsomes are added to wells of a 96-well deep-well polypropylene plate in triplicate. A 10 pL aliquot of the 12.5-50 pM test compound is added to the microsomes and the mixture is pre-warmed for 10 minutes. Reactions are initiated by addition of pre-warmed NADPH solution. The final reaction volume is 0.5 mL and contains 4.0 mg/mL human liver microsomes, 0.25 pM test compound, and 2 mM NADPH in 0.1 M potassium phosphate buffer, pH 7.4, and 3 mM MgCh. The reaction mixtures are incubated at 37 °C, and 50 pL aliquots are removed at 0, 5, 10, 20, and 30 minutes and added to shallow-well 96-well plates which contain 50 pL of ice-cold ACN (acetonitrile) with internal standard to stop the reactions. The plates are stored at 4 °C for 20 minutes after which 100 pL of water is added to the wells of the plate before centrifugation to pellet precipitated proteins. Supernatants are transferred to another 96-well plate and analyzed for amounts of parent remaining by LC-MS/MS using an Applied Bio-systems API 4000 mass spectrometer. The same procedure is followed for the non-labeled, such as the non-deuterated counterpart of the compound and the positive control, 7- ethoxycoumarin (1 pM). Testing is done in triplicate.

Data analysis: The in vitro T/ 2 s for test compounds are calculated from the slopes of the linear regression of % parent remaining (In) vs incubation time relationship. in vitro T/ 2 = 0.693/k k = -[slope of linear regression of % parent remaining (In) vs incubation time] The apparent intrinsic clearance is calculated using the following equation:

CLint (mL/min/kg) = (0.693 / in vitro T) (Incubation Volume / mg of microsomes) (45 mg microsomes / gram of liver) (20 gm of liver / kg b.w.) Data analysis is performed using Microsoft Excel Software. In these experiments, values equal to or more than a 15% increase in half-life are considered to be a significant difference if the apparent intrinsic clearance ratio (isotopically enriched compound/ MDAI) is >1.15 or <0.85, then there is considered to be significant differentiation.

Table Exl 1. Metabolic stability in human liver microsomes of representative deuterated compounds

* Average of n = 2 experiments

** Average of n = 2 experiments; 30- and 45-minute time points excluded because verapamil is metabolized very fast and is completely gone by 30 min. Thus, the 30 min and 45 min timepoints are not able to be measured.

Based on the results in Table Exl 1, Compound 6,7,7-Trideuterio-5Z7- cyclopenta[f][l,3]benzodioxol-6-amine hydrochloride (compound 3) and Compound 2,2,6,7,7- Pentadeuterio-5Z7-cyclopenta[f][l,3]benzodioxol-6-amine hydrochloride (compound 4) exhibit significantly lower in vitro clearance than MDAI HC1 salt (reference compound). It should be noted that 6,7,7-Trideuterio-5Z7-cyclopenta[f][l,3]benzodioxol-6-amine hydrochloride (compound 3) and 2,2,6,7,7-Pentadeuterio-5Z7-cyclopenta[f][l,3]benzodioxol-6- amine hydrochloride (compound 4) were tested as racemic mixtures. Each compound can be separated into its respective enantiomers. Compounds 1 and 2 also exhibit differences in half-life and intrinsic clearance compared to MDAI HC1 (reference compound).

Example 12: Oral Bioavailability in Rats - Pharmacokinetics of test articles following a single intravenous or oral administration in rats: A pharmacokinetic (PK) study is performed in three male Sprague-Dawley (SD) rats following intravenous (IV) and oral (PO) administration of MDAI, or test deuterated MDAI, at 1 mg/kg (IV) and 10 (PO) mg/kg. Test compounds, or MDAI, are measured in plasma.

A detailed description of the in vivo methods:

Rat Strain

Rats used in these studies are specific pathogen free. The strain of rats is Sprague Dawley. Male rats are 175 - 225g on receipt and are allowed to acclimatise for 5-7 days. Animal Housing Rats are group housed in sterilised individual ventilated cages that expose the animals at all times to HEPA filtered sterile air. Animals will have free access to food and water (sterile) and will have sterile aspen chip bedding (at least once weekly). The room temperature is 22°C +/- 1°C, with a relative humidity of 60% and maximum background noise of 56dB. Rats are exposed to 12 hour light/dark cycles.

Treatment

Test article is diluted 10% v/v DMSO, 40% v/v PEG-400, 50% v/v Water. The test articles are administered in a dose volume of 2mL/kg for intravenous (IV) and 5mL/kg (PO) for oral routes of administration. Single IV/PO dose pharmacokinetics study in rats

Each test article is administered as a single IV bolus (via a lateral tail-vein) or a single oral gavage in cohorts of 3 rats per route. Following dose administrations, a lOOpL whole blood sample (EDTA) is collected via the tail-vein at time-points described in Table 1. The blood is centrifuged to separate plasma. Approximately 40pL of plasma is dispensed per time-point, per rat, in a 96 well plate and frozen until analysis. Bioanalysis is carried out on plasma samples.

Table Exl2: Single IV and oral dose pharmacokinetics profile of test articles in rat plasma

Dose formulation Samples Dose formulation samples are diluted in two steps with 50:50 (v/v) methanol/water to an appropriate concentration, then diluted 10:90 (v/v) with control matrix to match to the calibration standard in plasma.

Sample Extraction procedure Calibration and QC standards, incurred samples, blank matrix and dose formulation samples are extracted by protein precipitation, via the addition of a bespoke acetonitrile (ACN)- based Internal Standard (IS) solution, containing several compounds and including Metoprolol and Rosuvastatin, both of which are monitored for during analysis. Following centrifugation, a 40 pL aliquot of supernatant is diluted by the addition of 80 pL water. The prepared sample extracts are analysed by LC-MS/MS.

Example of Bioanalytical Method and Assay Information Document:

1 According to the plate layout, aliquot to wells in 0.8 mL 96-well plate (Abgene). 30 pL for Calibration, QC standards, blanks and dose formulation check.

2 Prepare Calibration and QC standards according to the assay information. Dilute dose formulation according to the assay information. Aliquot incurred samples according to the plate layout & assay information.

3 Add 90 pL of ACN internal standard and vortex mix for 5 minutes at 850 rpm

4 Centrifuge at nominally 4000 rpm for 10 minutes

6 Transfer 40 pL of supernatant into a new 0.8 mL Abgene plate.

6 Add 80 pL of water to all transferred supernatant.

7 Vortex mix for 30 seconds at 1400 rpm

8 Analyse immediately by LC-MS/MS or store at +4°C until analysis.

Example 13 Effect of MDAI (0.15, 0.31, 0.625, 1.25 and 2.5 mg/kg ip) and chlordiazepoxide on rat behavior in an elevated zero maze test Background

The rat zero-maze model is a refined alternative to the plus-maze, the most widely used animal model of anxiety, and consists of an elevated annular platform, divided equally into four quadrants. Two opposite quadrants are enclosed by Perspex walls on both the inner and the outer edges of the platform, while the remaining two opposite quadrants are open being enclosed only by a Perspex “lip”. Animals will show a preference for the closed areas, and avoidance of the open sections is assumed to stem from a rodent’s natural aversion to open, exposed spaces. The primary index of anxiety, which reflect changes in open arm preference, is the percentage of time spent on the open areas. A reduction in the amount of activity on the open areas is considered to reflect an increase in anxiety while an increase in time in the open arms is indicative of an anxiolytic effect. Other, ethologically-based behaviors are also assessed as indices of anxiety including total open arm entries, total line crossings, and the frequency of exploratory head dipping over the edge of the platform (HDIPS).

Shepherd, JK; Grewel, SS; Fletcher, A; Bill, DJ; Dourish, CT (1994) Behavioural and pharmacological validation of the elevated “zero-maze” as an animal model of anxiety. Psychopharmacol., 116:56-64.

Animals

Male Sprague-Dawley 200-250 g (Envigo UK) rats were used. Animals were group- housed (5 per cage; cage size: 40 x 40 x 20 cm) in a temperature-controlled environment (22±2°C), under a 12 h light-dark cycle (lights on: 08:00 hours) for one week prior to testing. Food and water were freely available. Number of animals per group =5. Animals were moved into the experimental room 16-24 hours before testing.

Apparatus

The elevated 0-maze comprises a black Perspex annular platform (105 cm diameter, 10 cm width) elevated to 65 cm above ground level, divided equally into four quadrants. Two opposite quadrants are enclosed by clear red Perspex walls (27 cm high) on both the inner and outer edges of the platform, while the remaining two opposite quadrants are surrounded only by a Perspex "lip" (1 cm high) which serves as a tactile guide to animals on these open areas.

Procedure

Subjects were weighed and tail marked before being injected. After a specified pretreatment time, subjects were placed in a closed quadrant and a 5-min test period were recorded on videotape for subsequent analysis. The maze was cleaned with 5% methanol/water solution and dried thoroughly between test sessions. Behavioural measures comprise percentage time spent on the open areas (%TO). Animals are scored as being in the open area when all four paws were in an open quadrant and in the closed area only when all four paws have passed over the open-closed divide. All testing were carried out between 9.00 and 16.00 hours.

Formulation:

IP: MDAI (HC1 salt with 82.94% free base content) was formulated in Vehicle 1 (Saline) for injection to concentrations of 0.03, 0.062, 0.125 and 0.5 mg/mL to provide doses of 0.15, 0.31, 0.625, 1.25 and 2.5 mg/kg when administered ip in 5 mL/kg dosing volumes.

Chlordiazepoxide was formulated in Vehicle 1 (saline) to a concentration of 1.2 mg/mL to provide a dose of 6 mg/kg when administered ip in 5 mL/kg dosing volumes.

Protocol: Effect of administration of MDAI and chlordiazepoxide on behavior in a rat flmaze study

35 male Sprague-Dawley rats in treatment groups of 2 or 5, was intraperitoneally dosed with either Vehicle 1 (saline) or MDAI at l of 5 dose levels (0.15, 0.31, 0.625, 1.25 and 2.5) or chlordiazepoxide (6 mg/kg) in 5 mL/kg injection volumes. Thirty min later at T=0, rats were individually placed in a closed arm of the zero-maze and behavior assessed by a “blind” observer using remote video monitoring over the subsequent 5 min. The animal was then removed and the maze carefully wiped with 5% methanol/water solution before the next test was begun. Three rats from the vehicle group, 3 rats from the 2.5mg/kg MDAI group, and 3 rats from the CDP group were tested on a separate day from the other rats.

Table Exl3: Synopsis of testing schedule MDAI and chlordiazepoxide in the rat elevated zero maze model of anxiety.

Statistical analysis

Data was analysed with Statistica software (StatSoft USA version 10.0). All data was expressed as means + SEM. Data was analyzed by 1 way ANOVA and Dunnett’s or Newman- Keuls test. Statistical significance in all analyses was assumed when P < 0.05.

Discussion:

CDP showed a significant increase in the percentage of time in the open arms (%TO) but only showed a trend towards increased total open arm entries, the total line crossings, and the frequency of HDIPS which showed that the study was underpowered to detect an effect. An increase in total open arm entries, total line crossings, and the frequency of HDIPS all indicates an anxiolytic effect. We found that MDAI dosed at 0.625mg/kg showed a numerical increase in %TO that was greater than vehicle or any other dose of MDAI (FIG.l). This indicates that this middle dose of MDAI was anxiolytic. This also shows that doses of MDAI that are too low or too high induce an anxiogenic effect as we saw the opposite effect on %TO with these doses. This trend towards an anxiolytic effect with the middle dose of MDAI was also seen with total open arm entries with only the middle dose showing an increase vs placebo that trended towards significance. The higher and lower doses all trended towards an anxiogenic effect on that measure (FIG. 2). This was further confirmed with total line crossings (FIG. 3) and frequency of HDIPS (FIG. 4). These data show that MDAI has anxiolytic effects in a specific dose range. Additionally, we found the surprising finding that low doses of MDAI are anxiogenic which means that patients who take MDAI would need to be titrated up to the effective dose. There are several critical implications of this finding. The first is that patients treated with any form of MDAI must receive a dose high enough to reach the anxiolytic threshold since lower doses may cause anxiety as a side effect and result in worsening of the disorder being treated. This could have especially severe implications for anxiety disorders or depressive disorders including post-traumatic stress disorder, generalized anxiety disorder, panic disorder, major depressive disorder, or treatment resistant depression. All of these indications are associated with an increased level of anxiety as a major symptom. In these cases, a drug-induced increase in anxiety due to improper dosing of MDAI could have severe side effects on patients and worsen their underlying disorder. The data presented herein show that patients treated MDAI must be especially careful to titrate up to the therapeutic dose to avoid the anxiogenic effects and to reach the anxiolytic effect level. The data show that in some embodiments a Risk Evaluation and Mitigation Strategy (REMS) program should be utilized so that patients treated with MDAI should undergo an initial dose titration to determine the effective range specific to that patient. This dose titrating protocol would decrease the side effects related to underdosing MDAI.

The data also inform Phase 2 and Phase 3 clinical trial design. Clinical trials for neurological and psychiatric disorders often include one or more low dose arms to show a dose dependent effect of the full dose on the disease of interest. However, this data shows that MDAI should only be dosed at the full effective dose and a low dose arm should not be included as a comparator as this may lead to harmful side effects on the patients. This data shows that studies of MDAI should only use inactive matched placebo or a different standard of care therapeutic as a control. In clinical trials MDAI should only be dosed at its effective dose range to avoid harmful side effects to the patients. This would be especially critical in clinical studies of anxiety disorders or depression including post-traumatic stress disorder, generalized anxiety disorder, panic disorder, major depressive disorder, or treatment resistant depression where increased anxiety could worsen the underlying disorder and lead to potentially devastating effects on the patients.

Example 14: MDAI Dose Titration Risk Evaluation and Mitigation Strategy (REMS) Protocol

General Information on MDAI Treatment Session

Initial MDAI dosing and subsequent dosing adjustments must be done under the supervision of a qualified healthcare professional in a clinic or inpatient setting. The patient must remain under supervision of the healthcare professional for at least 6 hours and up to approximately 24 hours after the final MDAI dose adjustment. The patient will be assessed periodically during the session for anxiety and other effects of MDAI. Dose adjustments within a MDAI treatment session will be based on changes from baseline levels of anxiety. Postdose anxiety measurement timing and duration of observation after dosing are shown here (h tips : //psv chon autwiki . org/wiki/MD Al) :

Duration of effects of MDAI

MDAI dosing is shown here: MDAI dosages

Predose Assessment

The patient’s baseline level of anxiety will be measured and recorded.

Initial MDAI Dosing The patient will receive an initial single oral dose of MDAI in the range of approximately

100 mg - 200 mg based on oral doses reported as producing moderate effects (https ://psychonautwiki . org/wiki/MD Al).

Postdose Assessment

Change from baseline anxiety level will be measured at approximately .75 to 2 hours after dosing based on reported time to achieve peak effects

(https ://psychonautwiki . org/wiki/MD Al).

MDAI Dose Adjustment MDAI effects have been maintained by taking a larger initial dose followed by smaller doses (30 mg to 100 mg p.o.). Re-dose of one-third to one-half the initial dose usually prolongs duration (https://psychonautwiki.org/wiki/MDAI). Accordingly, the dose of MDAI will be adjusted based on change from baseline in anxiety as follows:

MDAI dose adjustment

MDAI Discontinuation

The patient will be observed for at least 6 hours after final MDAI dose is administered.

The patient may be confined to the inpatient unit for prolonged observation up to approximately 24 hours after last MDAI dose if indicated based on persistent effects.

Anxiety that appears after the final MDAI titration dose is administered can be managed with an appropriate anxiolytic agent. If this is necessary, the patient must remain under observation and undergo periodic reassessment until the supervising healthcare professional determines the patient can be discharged from care.

Example 15: A double-blind, randomized, placebo-controlled clinical trial of MDAI- assisted psychotherapy in PTSD

A multicenter, randomized, double-blind, placebo-controlled trial is conducted to assess the efficacy and safety of MDAI-assisted psychotherapy versus psychotherapy with placebo control in participants diagnosed with at least moderate post-traumatic stress disorder (PTSD). Rationale

PTSD is a debilitating and often times chronic disorder associated with profound mental, physical, occupational, and functional impairment. PTSD can develop due to exposure to a traumatic event or persistent or recurring threats to an individual. Studies indicate that approximately 10% of individuals exposed to a traumatic event eventually go on to be diagnosed with PTSD (American Psychiatric Association. Diagnostic and statistical manual of mental disorders, 5 th edition, 2013). PTSD is a complex psychiatric disorder characterized by symptom heterogeneity including avoidance of trauma-related material, emotional blunting and distancing, hyper-vigilance, hyper-arousal, persistent negative alterations in mood, persistent alterations in cognition, disturbing thoughts, disruptions in sleep and/or dreams, and physical or mental distress. Symptoms can be severe and long lasting. Although this symptom heterogeneity may suggest a wide spectrum of separate disturbances, emotional dysregulation is considered to be a core component of this disorder. Particularly germane to the pathogenesis and progression of PTSD, emotional dysregulation in affected individuals is believed to give rise to observable and measurable features such as presence of hypervigilance and attend onal biases, enhanced startle response, hyper-arousal, apathetic feeling or emotional numbness, irritability, enhanced memories associated with traumatic events, difficulty in discerning danger versus safety, a generalization of fear, and avoidance of reminders of trauma. Emotional dysregulation may be defined and also measured by elevated emotional reactivity based on abnormal detection or appraisal of emotional triggers involving bottom-up sensory detection and neuronal processing. Biochemical alterations found in individuals diagnosed with PTSD suggest abnormalities in the hypothalamic-pituitary-adrenal (HP A) axis. The HPA axis is known to regulate reactions to stress and controls significant aspects of the neuroendocrine system impacting many homeostatic systems in the body. In a typical flight-or-flight response in a healthy individual, catecholamine and cortisol levels detected in urine rise after exposure to a stressor. In PTSD, many individuals show a low secretion of cortisol and high secretion of catecholamine in response to a stressor indicating a change in catecholamine to cortisol ratio in the urine. More evidence that the HPA axis is impacted in PTSD is found in elevated levels of catecholamines and corticotropinreleasing factor in the brain of many affected individuals.

The initiation and/or maintenance of emotional dysregulation in PTSD may be due to abnormalities in top-down control of emotional responses indicating that cognitive influences and higher order representations may impinge on information and emotional processing. Certainly, some aspects of abnormalities in neuronal processing in PTSD occur either implicitly (e.g., unconsciously) or explicitly (e.g., consciously) indicating involvement of distinct cognitive processes. Exaggerated responses in the amygdala and insular cortex have been demonstrated in meta-analyses in PTSD pathology, as have decreases in activity in other brain regions including the anterior cingulate cortex and aspects the prefrontal cortex including the ventromedial prefrontal cortex. In addition to changes in patterns of neuronal activity in individuals with PTSD, several neuroanatomical changes in PTSD have also been demonstrated. A reduction of total brain volume, intracranial volume, and the volumes in regions such as the hippocampus (particularly localized to the CA3 and dentate gyrus regions), insular cortex, and anterior cingulate cortex have been indicated in occurring in some individuals with PTSD through meta- analyses of structural MRI studies. Animal studies have shown that severe chronic stress leads to atrophy of apical dendrites in the CA3 region of the hippocampus, reduced hippocampus neurogenesis, and elevated granule cell death in the dentate gyrus due to elevated levels of glucocorticoids (Gould E. and Tanapat. (1999). Stress and hippocampal neurogenesis. Biol. Psychiatry 46, 1472-1479.) Connections between brain areas such as the amygdala, hippocampus, prefrontal cortex, and hypothalamus can facilitate activation of the HPA axis to illustrate interactions between brain regions with structural changes and affected biochemical regulatory systems in PTSD.

MDAI is a synthetic analog of the psychedelic phenethylamine class of compounds known to act as a mixed reuptake inhibitor/releasing agent of serotonin and administration of MDAI can produce acute modulations of neurotransmission. MDAI administration also has indirect effects on neurohormone release. MDAI can function as a psychoplastogen promoting neuronal growth, modulating neuronal connectivity, and regulating neuronal plasticity through longer term neuronal changes. The combined neurobiol ogical effects of MDAI administration on individuals reduce fear of emotional injury or distress, enhance introspection and communication, and increase empathetic feelings and compassion. Additionally, MDAI may serve to enhance fear extinction. These combined effects may yield acute and longer-term productive psychological states to enhance behavioral or cognitive-behavioral therapies. MDAI administration may enhance neuronal function at the biochemical and cellular levels to generate or restore favorable neural network pathways and connectivity to increase behavioral or cognitive-behavioral therapy productiveness.

Study Design

This multicenter, randomized, double-blind, placebo-controlled trial is conducted at various sites in the United States with IRB approval from each study site. A flexible dose of MDAI hydrochloride salt or placebo, followed by a supplemental half-dose unless contraindicated by patient’s previous response or medical history, is also administered during the Treatment Period with psychotherapy in at least 3 blinded monthly Experimental Sessions. The Supplemental Dose extends the duration of drug effects on the participants during an Experimental Session. An optional Risk Evaluation and Mitigation Strategy (REMS) Protocol may be implemented for MDAI and placebo-groups. The Treatment Period lasts for approximately 12 weeks. During the Treatment Period, each Experimental Session is followed by three Intervening Sessions of non-drug psychotherapy. Each Experimental Session involves an overnight stay. The Primary Outcome measure, the change in Clinician Administered PTSD Scale for DSM-5 (CAPS-5), is determined by a blinded Independent Rater (IR) pool multiple times throughout the study. The study consists of separate periods for each participant. Initially, prospective participants undergo a Screening Period involving an initial eligibility assessment, a medical history intake, informed consent, and enrollment of eligible participants. Next, a Preparation Period is undertaken for enrolled participants involving medication tapering and clinical baseline assessments to confirm each participant meets enrollment criteria. As part of the Preparation Period, a detailed assessment of co-morbidities to PTSD is recorded. Participants may remain on prescribed courses of selective serotonin reuptake inhibitor (SSRI) or serotonin and norepinephrine reuptake inhibitor (SNRI) treatment. Dosages and/or frequency of administration of a prescribed SSRI or SNRI may be adjusted to fit within study parameters. Participants may be required to taper a prescribed course of medication in order to maintain eligibility within the study. The Treatment Period consists of three monthly Experimental Sessions and associated Intervening Sessions of integrative behavioral psychotherapy. The Treatment Period lasts approximately 12 weeks. Following the Treatment Period is a Follow-up Period and Study Conclusion. During the Follow-up Period and Study Conclusion, participants complete 4 weeks with no study visits, followed by a Study Conclusion visit.

The Treatment Period schedule follows the Screening Period and the Preparatory Period

[0001] The Follow-up Period schedule and Study Conclusion follow the Screening Period and the Treatment Period.

Dose Selection

This study compares the effects of three blinded Experimental Sessions of psychotherapy in combination with flexible doses of MDAI or placebo administered as described below. Non- drug preparatory and intervening psychotherapy sessions are also included. Patient’s weight is determined for dosage calculation. Initial dose is 100 mg unless this will result in a dosage of less than 1.5 mg/kg of patient weight. Initial dose thereby adjusted upward in 50 mg increments to deliver the lowest dose possible of at least 1.5 mg/kg of patient weight. Initial dose for Experimental Session 2 and 3 is cumulative dose calculated by adding the initial dose plus REMS protocol dose used the previous Experimental Session for each patient.

Randomization and masking Randomization occurs prior to the initiation of Experimental Session 1. Each participant is provided the next randomized number in a sequence by a blinded study monitor. Participants are then randomized, according to a computer-generated randomization schedule, 1 : 1 : 1 : 1 to MDAI or placebo. The randomization schedule is prepared and implemented by an independent statistician. Participants, clinicians, and study teams are blinded to treatment allocation. MDAI treatment groups may be subjected to anxiogenic effects due to underdosing of participants. As such, an optional dose titration schedule (REMS protocol) exists for MDAI treatment groups if a participant displays no change or a significant worsening of assessed anxiety symptomatology. Participants are assessed for general well-being and anxiety by a medical practitioner about 0.75 hours after the first dose is administered. Assessments performed may include general assessments of physical and mental well-being, a structured clinical interview for DSM-5 (SCID- 5) module Al, and/or a STAI assessment and may continue throughout the period of overnight observation.

Subjects then undergo three Intervening Sessions with the first session the morning after the initial dose administration. MDAI treatment group or placebo group participants qualifying with a significant worsening of assessed anxiety symptomatology would undergo a placebo dose titration administration. Subjects would then undergo three Intervening Sessions with the first session the morning after the placebo dose titration administration. The pharmacist at each site, who prepares the treatments according to the randomization schedule, and an unblinded monitor, who performs drug accountability during the study, are unblinded. No other study personnel are unblinded until after formal locking of the study database. In the event of a medical emergency, the pharmacist is to reveal actual treatment contents to the primary investigator, who is to alert the Sponsor of the emergency. If the participant or study center personnel are unblinded, the subject is to be removed from the study.

Outcomes

The primary objective of this study is to evaluate the efficacy and safety of MDAI treatment combined with psychotherapy to treat moderate to severe PTSD compared to identical psychotherapy combined with placebo treatment. Treatment outcomes are determined based on a change in CAPS-5 Total Severity.

Several secondary objectives are designed for this study. One is an evaluation of clinician-rated functional impairment of MDAI treatment combined with psychotherapy to treat moderate to severe PTSD compared to identical psychotherapy combined with placebo treatment. Treatment outcomes are determined based on a change in SDS. Another secondary objective of this study is to evaluate clinician-rated depression of MDAI treatment combined with psychotherapy to treat moderate to severe PTSD compared to identical psychotherapy combined with placebo treatment. Identical study parameters are in place as for the clinicianrated functional impairment assessment except that treatment outcomes are determined based on a change in HAM-D. An additional secondary objective of this study is to evaluate sleep assessments of MDAI treatment combined with psychotherapy to treat moderate to severe PTSD compared to identical psychotherapy combined with placebo treatment. Identical study parameters are in place as for the clinician-rated functional impairment assessment except that treatment outcomes are determined based on a change in ESS. Co-morbidities present in participants with a strong positive response to MDAI treatment are correlated. Co-morbidities present in participants with weak-to-no positive response to MDAI treatment are correlated. Changes to presence or severity of co-morbidities from the Preparation Period to the Study Conclusion are recorded to determine if MDAI treatment combined with psychotherapy in moderate to severe PTSD subjects affects co-morbid phenotypes not falling under the constellation of PTSD symptoms.

Participant Populations

Participants are recruited through referrals by other treatment providers or through print or internet advertisements. The Sponsor monitors demographics of individuals assessed for enrollment to encourage diversity and an unbiased representation of the total PTSD population. Participants must be 18 years of age or older, have a confirmed diagnosis of at least moderate PTSD according to PCL-5 at the Screening Period. Medical history intake must indicate a presence of PTSD symptoms for at least 6 months prior to the Screening Period. Participants may be enrolled in the study while remaining on a treatment regimen involving SSRI or SNRI treatment prescribed for PTSD. In some cases, enrolled participants currently taking an SSRI, an SNRI, or another medication are tapered off these medications and stabilized prior to baseline assessments. Participants with a confirmed personality disorder diagnosis are excluded from this study. Participants must be in good general physical health without one or more severe chronic conditions that could affect the safety or tolerability of MDAI treatment.

Statistical Analysis The change from baseline in CAPS-5, SDS, HAM-D, and ESS in participants is analyzed using a mixed effects model for repeated measures (MMRM) to obtain covariance parameter estimates. The model includes treatment center, treatment subtype, baseline assessments, assessment time point, and time point-by-treatment as explanatory variables. Treatment center is treated as a random effect; all other explanatory variables are treated as fixed effects. Modelbased point estimates (e.g., least squares means, 95% confidence intervals, and p-values) are reported for each time point. With a sample size of 50 participants per treatment group, this study has 90% power to detect a significant treatment effect, using a two-sided test with an alpha value of 0.05. Additional participants may be enrolled with conditional power analysis conducted at a group-unblinded interim analysis time point for efficacy when 200 participants are enrolled and at least 60% of the blinded participants (N=120) have completed a final CAPS-5 assessment and reached Study Conclusion.

Results

The results may indicate that the primary objective is achieved. At the point of Study Conclusion, MDAI-treated participants may demonstrate a significant mean reduction in CAPS- 5 assessment compared to the placebo group. Significant improvements in CAPS-5 assessments may be observed for MDAI-treated participants at time points of Intervening Session 1C, Intervening Session 2C, Intervening Session 3C and Study Conclusion, compared to placebo- treated controls. The results may indicate that the secondary objectives of this study are also achieved. At the point of Study Conclusion MDAI-treated participants may demonstrate a significant improvement in clinician-rated functional impairment score as measured by SDS compared to placebo-treated controls. At the point of Study Conclusion, MDAI-treated participants may demonstrate a significant improvement depression as measured by HAM-D compared to placebo-treated controls. At the point of Study Conclusion, MDAI-treated participants may demonstrate a significant improvement in lessening daytime sleepiness as measured by ESS. At the point of Study Conclusion MDAI-treated participants may demonstrate a significant improvement in clinician-rated functional impairment score, in depression, and in lessening daytime sleepiness compared to placebo-treated controls without a significant increase in adverse anxiogenic incidents.

In view of the many possible embodiments to which the principles of the disclosed invention may be applied, it should be recognized that the illustrated embodiments are only preferred examples of the invention and should not be taken as limiting the scope of the invention. Rather, the scope of the invention is defined by the following claims. We therefore claim as our invention all that comes within the scope and spirit of these claims.