Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PHENETHYLAMINE DERIVATIVES, COMPOSITIONS, AND METHODS OF USE
Document Type and Number:
WIPO Patent Application WO/2023/156453
Kind Code:
A1
Abstract:
There are disclosed psychedelic and entactogen compounds, the use of such compounds in the treatment of diseases associated with a serotonin receptor or monoamine transporter, pharmaceutical compositions such as tablet compositions and kits containing the compounds, methods of delivering the compounds in a mist via inhalation, and methods of treating diseases or disorders associated with a serotonin receptor or monoamine transporter, such as inflammation, central nervous system (CNS) disorders or psychological disorders with the compounds of the invention.

Inventors:
NIVOROZHKIN ALEX (US)
HARTSEL JOSHUA A (US)
CANAL CLINTON E (US)
SALITURO FRANCESCO G (US)
MUELLER TINA A (US)
GREENE BRETT J (US)
BELSER ALEX (US)
AVERY KENNETH L (US)
REICHELT AMY CLAIRE (CA)
VARTY GEOFFREY B (US)
PALFREYMAN MICHAEL (US)
Application Number:
PCT/EP2023/053752
Publication Date:
August 24, 2023
Filing Date:
February 15, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CYBIN IRL LTD (IE)
International Classes:
C07C211/27; A61K31/36; A61P25/00; C07C217/08; C07D317/58; C07D327/04
Domestic Patent References:
WO2008016677A22008-02-07
WO2003026559A22003-04-03
WO2002011800A22002-02-14
WO2000051672A12000-09-08
WO2002068029A22002-09-06
WO2002068030A22002-09-06
WO2002068031A22002-09-06
WO2002068032A22002-09-06
WO2003000310A22003-01-03
WO2003020350A12003-03-13
WO2003082393A12003-10-09
WO2003084591A12003-10-16
WO2003090812A22003-11-06
WO2000041755A12000-07-20
Foreign References:
CN112430187A2021-03-02
US4911920A1990-03-27
US5403841A1995-04-04
US5212162A1993-05-18
US4861760A1989-08-29
US5798119A1998-08-25
US5612059A1997-03-18
US5698220A1997-12-16
RU199823U12020-09-21
US7267121B22007-09-11
Other References:
BJORNSTAD K. ET AL: "Development and Clinical Application of an LC-MS-MS Method for Mescaline in Urine", JOURNAL OF ANALYTICAL TOXICOLOGY, vol. 32, no. 3, 1 April 2008 (2008-04-01), US, pages 227 - 231, XP093043580, ISSN: 0146-4760, DOI: 10.1093/jat/32.3.227
REIFF, C. M.RICHMAN, E. E.NEMEROFF, C. B.CARPENTER, L. L.WIDGE, A. S.RODRIGUEZ, C. I.KALIN, N. H.MCDONALD, W. M.: "Psychedelics and Psychedelic-Assisted Psychotherapy", AM J PSYCHIATRY, vol. 777, 2020, pages 391 - 410
JEROME, L.FEDUCCIA, A. A.WANG, J. B.HAMILTON, S.YAZAR-KLOSINSKI, B.EMERSON, A.MITHOEFER, M. C.DOBLIN, R.: "Long-term follow-up outcomes of MDMA-assisted psychotherapy for treatment of PTSD: a longitudinal pooled analysis of six phase 2 trials", PSYCHOPHARMACOLOGY (BERL, vol. 237, 2020, pages 2485 - 2497, XP037188562, DOI: 10.1007/s00213-020-05548-2
GOLDBERG, S. B., PACE, B. T., NICHOLAS, C. R., RAISON, C. L., HUTSON, P. R.: "The experimental effects of psilocybin on symptoms of anxiety and depression: A meta-analysis", PSYCHIATRY RES, vol. 284, 2020, pages 112749, XP086027126, DOI: 10.1016/j.psychres.2020.112749
MORENO, F. A.WIEGAND, C. B.TAITANO, E. K.DELGADO, P. L.: "Safety, tolerability, and efficacy of psilocybin in 9 patients with obsessive-compulsive disorder", Y CLIN PSYCHIATRY, vol. 67, 2006, pages 1735 - 1740
NICHOLS, D. E.: "Psychedelics", PHARMACOL REV, vol. 68, 2016, pages 264 - 355, XP055703032, DOI: 10.1124/pr.115.011478
JOHNSON, M. W., HENDRICKS, P. S., BARRETT, F. S., GRIFFITHS, R. R.: "Classic psychedelics: An integrative review of epidemiology, therapeutics, mystical experience, and brain network function", PHARMACOL THER, vol. 197, 2019, pages 83 - 102, XP085685213, DOI: 10.1016/j.pharmthera.2018.11.010
SEWELL, R. A.HALPERN, J. H.POPE, H. G., JR.: "Response of cluster headache to psilocybin and LSD", NEUROLOGY, vol. 66, 2006, pages 1920 - 1922, XP008145543, DOI: 10.1212/01.wnl.0000219761.05466.43
NICHOLS, D. E.JOHNSON, M. W.NICHOLS, C. D.: "Psychedelics as Medicines: An Emerging New Paradigm", CLIN PHARMACOL THER, vol. 101, 2017, pages 209 - 219, XP055865261, DOI: 10.1002/cpt.557
FLANAGAN, T. W.SEBASTIAN, M. N.BATTAGLIA, D. M.FOSTER, T. P.CORMIER, S. A.NICHOLS, C. D.: "5-HT2 receptor activation alleviates airway inflammation and structural remodeling in a chronic mouse asthma model", LIFE SCI, vol. 236, 2019, pages 116790, XP085873774, DOI: 10.1016/j.lfs.2019.116790
FLANAGAN, T. W.SEBASTIAN, M. N.BATTAGLIA, D. M.FOSTER, T. P.MAILLET, E. L.NICHOLS, C. D.: "Activation of 5-HT2 Receptors Reduces Inflammation in Vascular Tissue and Cholesterol Levels in High-Fat Diet-Fed Apolipoprotein E Knockout Mice", SCI REP, vol. 9, 2019, pages 13444
SEXTON, J. D.NICHOLS, C. D.HENDRICKS, P. S.: "Population Survey Data Informing the Therapeutic Potential of Classic and Novel Phenethylamine, Tryptamine, and Lysergamide Psychedelics", FRONT PSYCHIATRY, vol. 10, 2019, pages 896
FEDUCCIA, A. A.JEROME, L.YAZAR-KLOSINSKI, B.EMERSON, A.MITHOEFER, M. C.DOBLIN, R.: "Breakthrough for Trauma Treatment: Safety and Efficacy of MDMA-Assisted Psychotherapy Compared to Paroxetine and Sertraline", FRONT PSYCHIATRY, vol. 10, 2019, pages 650
CANAL, C. E.: "Handb Exp Pharmacol", vol. 252, 2018, article "Serotonergic Psychedelics: Experimental Approaches for Assessing Mechanisms of Action", pages: 227 - 260
CANAL, C. E.MURNANE, K. S.: "The serotonin 5-HT receptor and the non-addictive nature of classic hallucinogens", Y PSYCHOPHARMACOL, vol. 31, 2017, pages 127 - 143
JAYANTHI, L. D.RAMAMOORTHY, S.: "Regulation of monoamine transporters: influence of psychostimulants and therapeutic antidepressants", AAPS J, vol. 7, 2005, pages E728 - 738, XP035718769, DOI: 10.1208/aapsj070373
SCHENK, S.NEWCOMBE, D.: "Methylenedioxymethamphetamine (MDMA) in Psychiatry: Pros, Cons, and Suggestions", J CLIN PSYCHOPHARMACOL, vol. 38, 2018, pages 632 - 638
GARCIA-ROMEU, A.KERSGAARD, B.ADDY, P. H.: "Clinical applications of hallucinogens: A review", EXP CLIN PSYCHOPHARMACOL, vol. 24, 2016, pages 229 - 268
MORGAN, L.: "MDMA-assisted psychotherapy for people diagnosed with treatment-resistant PTSD: what it is and what it isn't", ANN GEN PSYCHIATRY, vol. 19, 2020, pages 33
HUANG, X.-P.SETOLA, V.YADAV, P. N.ALLEN, J. A.ROGAN, S. C.HANSON, B. J.REVANKAR, C.ROBERS, M.DOUCETTE, C.ROTH, B. L.: "Parallel Functional Activity Profiling Reveals Valvulopathogens Are Potent 5-Hydroxytryptamine(2B) Receptor Agonists: Implications for Drug Safety Assessment", MOLECULAR PHARMACOLOGY, vol. 76, 2009, pages 710 - 722
ROTHMAN, R. B.BAUMANN, M. H.: "Serotonergic drugs and valvular heart disease", EXPERT OPIN DRUG SAF, vol. 8, 2009, pages 317 - 329, XP055418651, DOI: 10.1517/14740330902931524
PARROTT, A. C.: "The potential dangers of using MDMA for psychotherapy", J PSYCHOACTIVE DRUGS, vol. 46, 2014, pages 37 - 43
MEYER, J. S.: "3,4-methylenedioxymethamphetamine (MDMA): current perspectives", SUBST ABUSE REHABIL, vol. 4, 2013, pages 83 - 99
BAYLEN, C. A.ROSENBERG, H.: "A review of the acute subjective effects of MDMA/ecstasy", ADDICTION, vol. 101, 2006, pages 933 - 947, XP071910405, DOI: 10.1111/j.1360-0443.2006.01423.x
ZELLE, R. E., MCCLELLAN, W. J.: "A Simple, High-Yielding Method for the Methylenation of Catechols", TETRAHEDRON LETTERS, vol. 32, 1991, pages 2461 - 2464, XP002072951, DOI: 10.1016/S0040-4039(00)74353-X
BARRETT, F. S.BRADSTREET, M. P.LEOUTSAKOS, J. S.JOHNSON, M. W.GRIFFITHS, R. R.: "The Challenging Experience Questionnaire: Characterization of challenging experiences with psilocybin mushrooms", J PSYCHOPHARMACOL, vol. 30, 2016, pages 1279 - 1295
TUCKER, G. T.LENNARD, M. S.ELLIS, S. W.WOODS, H. F.CHO, A. K.LIN, L. Y.HIRATSUKA, A.SCHMITZ, D. A.CHU, T. Y. Y.: "The demethylenation of methylenedioxymethamphetamine (''ecstasy'') by debrisoquine hydroxylase (CYP2D6", BIOCHEMICAL PHARMACOLOGY, vol. 47, 1994, pages 1151 - 1156, XP025542974, DOI: 10.1016/0006-2952(94)90386-7
SCHMID, Y., VIZELI, P., HYSEK, C.M., PRESTIN, K., MEYER ZU SCHWABEDISSEN, H. E., LIECHTI, M. E.: "CYP2D6 function moderates the pharmacokinetics and pharmacodynamics of 3,4-methylene-dioxymethamphetamine in a controlled study in healthy individuals", PHARMACOGENET GENOMICS, vol. 26, 2016, pages 397 - 401
DE LA TORRE, R.FARRE, M.ROSET, P. N.PIZARRO, N.ABANADES, S.SEGURA, M.SEGURA, J.CAMI, J.: "Human pharmacology of MDMA: pharmacokinetics, metabolism, and disposition", THER DRUG MONIT, vol. 26, 2004, pages 137 - 144
DE LA TORRE, R.FARRE, M.ORTUNO, J.MAS, M.BRENNEISEN, R.ROSET, P. N.SEGURA, J.CAMI, J.: "Non-linear pharmacokinetics of MDMA ('ecstasy') in humans", BRITISH JOURNAL OF CLINICAL PHARMACOLOGY, vol. 49, 2000, pages 104 - 109
FARRE, M.DE LA TORRE, R.MATHUNA, B. O.ROSET, P. N.PEIRO, A. M.TORRENS, M.ORTUNO, J.PUJADAS, M.CAMI, J.: "Repeated doses administration of MDMA in humans: pharmacological effects and pharmacokinetics", PSYCHOPHARMACOLOGY (BERL, vol. 173, 2004, pages 364 - 375
SCHINDLER, C. W.THORNDIKE, E. B.BLOUGH, B. E.TELLA, S. R.GOLDBERG, S. R.BAUMANN, M. H.: "Effects of 3,4-methylenedioxymethamphetamine (MDMA) and its main metabolites on cardiovascular function in conscious rats", BR J PHARMACOL, vol. 171, 2014, pages 83 - 91, XP071095421, DOI: 10.1111/bph.12423
RICKLI, A.LUETHI, D.REINISCH, J.BUCHY, D.HOENER, M. C.LIECHTI, M. E.: "interaction profiles of novel N-2-methoxybenzyl (NBOMe) derivatives of 2,5-dimethoxy-substituted phenethylamines (2C drugs", NEUROPHARMACOLOGY, vol. 99, 2015, pages 546 - 553, XP029301242, DOI: 10.1016/j.neuropharm.2015.08.034
MONTE, A. P.MARONA-LEWICKA, D.PARKER, M. A.WAINSCOTT, D. B.NELSON, D. L.NICHOLS, D. E.: "Dihydrobenzofuran analogues of hallucinogens. 3. Models of 4-substituted (2,5-dimethoxyphenyl)alkylamine derivatives with rigidified methoxy groups", J MED CHEM, vol. 39, 1996, pages 2953 - 2961, XP000887344, DOI: 10.1021/jm960199j
MONTE, A. P.WALDMAN, S. R.MARONA-LEWICKA, D.WAINSCOTT, D. B.NELSON, D. L.SANDERS-BUSH, E.NICHOLS, D. E.: "Dihydrobenzofuran analogues of hallucinogens. 4. Mescaline derivatives", Y MED CHEM, vol. 40, 1997, pages 2997 - 3008, XP000887343, DOI: 10.1021/jm970219x
POTTIE, E.CANNAERT, A.STOVE, C. P.: "In vitro structure-activity relationship determination of 30 psychedelic new psychoactive substances by means of beta-arrestin 2 recruitment to the serotonin 2A receptor", ARCH TOXICOL, 2020
RICKLI, A.MONING, O. D.HOENER, M. C.LIECHTI, M. E.: "Receptor interaction profiles of novel psychoactive tryptamines compared with classic hallucinogens", EUR NEUROPSYCHOPHARMACOL, vol. 26, 2016, pages 1327 - 1337
NICHOLS, D. E.: "Hallucinogens", PHARMACOL THER, vol. 101, 2004, pages 131 - 181, XP055879855, DOI: 10.1016/j.pharmthera.2003.11.002
RICKLI, A.LUETHI, D.REINISCH, J.BUCHY, D.HOENER, M. C.LIECHTI, M. E.: "Receptor interaction profiles of novel N-2-methoxybenzyl (NBOMe) derivatives of 2,5-dimethoxy-substituted phenethylamines (2C drugs", NEUROPHARMACOLOGY, vol. 99, 2015, pages 546 - 553, XP029301242, DOI: 10.1016/j.neuropharm.2015.08.034
BRADEN, M. R.PARRISH, J. C.NAYLOR, J. C.NICHOLS, D. E.: "Molecular interaction of serotonin 5-HT2A receptor residues Phe339(6.51) and Phe340(6.52) with superpotent N-benzyl phenethylamine agonists", MOL PHARMACOL, vol. 70, 2006, pages 1956 - 1964
BUNDGARD, H.: "Design of Prodrugs", 1985, ELSEVIER, pages: 7 - 9,21-24
HIGUCHI, T. ET AL.: "Pro-drugs as Novel Delivery Systems", A.C.S. SYMPOSIUM SERIES, vol. 14
"Bioreversible Carriers in Drug Design", 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS
VANN JONES, S.A.O'KELLY, A.: "Psychedelics as a Treatment for Alzheimer's Disease Dementia", FRONT. SYNAPTIC NEUROSCI., 21 August 2020 (2020-08-21)
KOZLOWSKA, U.NICHOLS, C.WIATR, K.FIGIEL, M.: "From psychiatry to neurology: Psychedelics as prospective therapeutics for neurodegenerative disorders", JOURNAL OF NEUROCHEMISTRY, vol. 00, 2021, pages 1 - 20
GARCIA-ROMEU, A.DARCY, S.JACKSON, H.WHITE, T.ROSENBERG, P.: "Current Topics in Behavioral Neurosciences", 2021, SPRINGER, article "Psychedelics as Novel Therapeutics in Alzheimer's Disease: Rationale and Potential Mechanisms"
MARKOPOULOS AINSERRA ADE GREGORIO DGOBBI G: "Evaluating the Potential Use of Serotonergic Psychedelics in Autism Spectrum Disorder", FRONT PHARMACOL, vol. 12, 2022, pages 749068
SANTUSBAKER, J. CONTROLLED RELEASE, vol. 35, 1995, pages 1 - 21
"Drugs and the Pharmaceutical Science", 2002, MARCEL DEKKER, INC., article "Modified-Release Drug Delivery Technology", pages: 126
VAN WOENSEL M ET AL.: "Formulations for Intranasal Delivery of Pharmacological Agents to Combat Brain Disease: A New Opportunity to Tackle GBM?", CANCERS (BASEL, vol. 5, no. 3, 14 August 2013 (2013-08-14), pages 1020 - 48, XP002778004
BELL, A.: "Drug Delivery Devices Fundamentals and Applications", 1988, DEKKER, article "Intranasal Delivery Devices"
"Remington's Pharmaceutical Sciences", 1975, MACK PUBLISHING CO.
TATSUMI, M ET AL., EUR. J. PHARMACOL., vol. 368, 1999, pages 277 - 283
VERMA ET AL., DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY, vol. 26, 2000, pages 695 - 708
VERMA ET AL., J. CONTROLLED RELEASE, vol. 79, 2002, pages 7 - 27
MULHERON, J.G.CASANAS, S.J.ARTHUR, J.M.GARNOVSKAYA, M.N.GETTYS, T.W.RAYMOND, J.R.: "Human 5-HT1A receptor expressed in insect cells activates endogenous Go-like G protein", J BIOL CHEM, vol. 269, 1994, pages 12954
"Pharmaceutical Pelletization Technology", 1989, MARCEL DEKKER
H. HAMISHEHKAR ET AL.: "The Role of Carrier in Dry Powder Inhaler", RECENT ADVANCES IN NOVEL DRUG CARRIER SYSTEMS, 2012, pages 39 - 66
IIDA, KOTARO ET AL.: "Preparation of dry powder inhalation by surface treatment of lactose carrier particles", CHEMICAL AND PHARMACEUTICAL BULLETIN, vol. 51, no. 1, 2003, pages 1 - 5, XP001143133, DOI: 10.1248/cpb.51.1
MARESH, J. J., RALKO, A. A., SPELTZ, T. E., BURKE, J. L., MURPHY, C. M., GASKELL, Z., GIREL, J. K., TERRANOVA, E., RICHTSCHEIDT, C: "Chemoselective Zinc/HCl Reduction of Halogenated beta-Nitrostyrenes: Synthesis of Halogenated Dopamine Analogues", SYNLETT, vol. 25, 2014, pages 2891 - 2894
SINHABABU, A. K.BORCHARDT, R. T.: "Silica Gel-Assisted Reduction of Nitrostyrenes to 2-Aryl-1-Nitroalkanes with Sodium-Borohydride", TETRAHEDRON LETTERS, vol. 24, 1983, pages 227 - 230
YAMADA, T., KUWATA, M.TAKAKURA, R.MONGUCHI, Y.SAJIKI, H.SAWAMA, Y.: "Organocatalytic Nitroaldol Reaction Associated with Deuterium-Labeling", ADV SYNTH CATAL, vol. 360, 2018, pages 637 - 641
PACHOLCZYK, T ET AL., NATURE, vol. 350, 1991, pages 350 - 354
CHA, J. S.LEE, S. E.LEE, H. S.: "Selective Conversion of Aromatic Nitriles to Aldehydes by Lithium Tris(Dihexylamino)Aluminum Hydride", ORG PREP PROCED INT, vol. 24, 1992, pages 331 - 334
OUK, S.THIEBAUD, S.BORREDON, E.LE GARS, P.: "High performance method for O-methylation of phenol with dimethyl carbonate", APPL CATAL A-GEN, vol. 241, 2003, pages 227 - 233, XP004409349, DOI: 10.1016/S0926-860X(02)00467-2
ALDOUS, F. A.BARRASS, B. C.BREWSTER, K.BUXTON, D. A.GREEN, D. M.PINDER, R. M.RICH, P.SKEELS, M.TUTT, K. J.: "Structure-activity relationships in psychotomimetic phenylalkylamines", J MED CHEM, vol. 17, 1974, pages 1100 - 1111, XP055977130
PEARL, I.A.BEYER, D.L., J. ORG. CHEM., vol. 16, no. 2, 1951, pages 221 - 224
CINELLI, M.A. ET AL., J. MED. CHEM., vol. 60, no. 9, 2017, pages 3958 - 3979
NAGY, J.VERESS, T.: "HPLC Analysis of Hallucinogenic Mushroom Alkaloids (Psilocin and Psilocybin) Applying Hydrophilic Interaction Chromatography (HILIC", J FORENSIC RES, vol. 7, 2016, pages 356
CANAL, C. E.CORDOVA-SINTJAGO, T.LIU, Y.KIM, M. S.MORGAN, D.BOOTH, R. G.: "Molecular pharmacology and ligand docking studies reveal a single amino acid difference between mouse and human serotonin 5-HT receptors that impacts behavioral translation of novel 4-phenyl-2-dimethylaminotetralin ligands", J PHARMACOL EXP THER, vol. 347, 2013, pages 705 - 716
ARMSTRONG, J. L.CASEY, A. B.SARAF, T. S.MUKHERJEE, M.BOOTH, R. G.CANAL, C. E.: "S)-5-(2'-Fluorophenyl)-N,N-dimethyl-1,2,3,4-tetrahydronaphthalen-2-amine, a Serotonin Receptor Modulator, Possesses Anticonvulsant, Prosocial, and Anxiolytic-like Properties in an Fmrl Knockout Mouse Model of Fragile X Syndrome and Autism Spectrum Disorder", ACS PHARMACOL TRANSL SCI, vol. 3, 2020, pages 509 - 523
SARAF, T. S.FELSING, D. E.ARMSTRONG, J. L.BOOTH, R. G.CANAL, C. E.: "Evaluation of lorcaserin as an anticonvulsant in juvenile Fmrl knockout mice", EPILEPSY RES, vol. 175, 2021, pages 106677
KENAKIN, T.: "Theoretical Aspects of GPCR-Ligand Complex Pharmacology", CHEM REV, vol. 117, no. 1, 2017, pages 4 - 20
BRYANT, H.U.NELSON, D.L.BUTTON, D.COLE, H.W.BAEZ, M.B.LUCAITES, V.L.WAINSCOTT, D.B.WHITESITT, C.REEL, J.SIMON, R.: "A novel class of 5-HT2 Areceptor antagonist: aryl aminoguanidines", LIFE SCI, vol. 15, 1996, pages 1259
CHOI, D.S.BIRRAUX, G.LAUNAY, J.M.MAROTEAUX, L.: "The human serotonin 5-HT2B receptor: pharmacological link between 5-HT2 and 5-HT1D receptors", FEBS LETT, vol. 352, 1994, pages 393, XP025600474, DOI: 10.1016/0014-5793(94)00968-6
PRISTUPA, Z.B. ET AL., MOL. PHARMACOL., vol. 45, 1994, pages 125 - 135
PORTER, R.H.P.BENWELL, K.R.LAMB, H.MALCOLM, C.S.ALLEN, N.F.REVELL, D.F.ADAMS, D.R.SHEARDOWN, M.J.: "Functional characterization of agonists at recombinant human 5-HT2a, 5HT2b and 5-HT2c receptors in CHO-K1 cells", BRIT J PHARMACOL, vol. 128, 1999, pages 13, XP002962670, DOI: 10.1038/sj.bjp.0702751
PEROVIC S.MULLER W.E.: "Pharmacological profile of hypericum extract. Effect on serotonin uptake by postsynaptic receptors", ARZNEIMITTELFORSCHUNG, vol. 45, no. 11, 1995, pages 1145 - 8, XP008075764
VERRICO C ET AL.: "MDMA (Ecstasy) and human dopamine, norepinephrine, and serotonin transporters: implications for MDMA-induced neurotoxicity and treatment", PSYCHOPHARMACOLOGY (BERL, 2005, pages 1 - 15
Attorney, Agent or Firm:
MÜLLER-BORÉ & PARTNER PATENTANWÄLTE PARTG MBB (DE)
Download PDF:
Claims:
CLAIMS 1. A compound having a structure of Formula (I): or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, wherein: X1 and X2 are independently hydrogen, deuterium, or a substituted or unsubstituted C1-C6 alkyl; Y1 and Y2 are independently hydrogen, deuterium, or a substituted or unsubstituted C1-C6 alkyl; R2 and R3 are independently hydrogen, deuterium, halogen, a substituted or unsubstituted C1-C6 alkyl, -ORa, or -SRa; R4 and R5 are independently hydrogen, deuterium, halogen, a substituted or unsubstituted C1-C6 alkyl, -ORa, -SRa, or -SeRa; or R4 and R5 together with the atoms attached thereto are optionally joined to form a heterocycloalkyl or heteroaryl; R6 and R7 are independently hydrogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or alternatively R6 and R7 together with the nitrogen atom attached thereto are optionally joined to form a substituted or unsubstituted heterocycloalkyl; each Ra is independently hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl; and wherein at least one of conditions (i)-(iii) are met (i) at least one of X1, X2, Y1, Y2, R2, R3, R4, R5, R6, R7 comprises deuterium, (ii) R4 and R5 together with the atoms attached thereto are joined to form a heterocycloalkyl or heteroaryl comprising deuterium or fluorine, and/or a benzo[d][1,3]oxathiole group, (iii) R4 is -ORa, -SRa, or -SeRa, with Ra in R4 being a C1-C6 alkyl substituted with one or more halogen; and with the proviso that when X1, X2, Y1, and Y2 are each hydrogen or deuterium, both R2 and R5 are not -ORa. 2. The compound of claim 1, wherein at least one of X1, X2, Y1, Y2, R2, R3, R4, R5, R6, R7 comprises deuterium. 3. The compound of claim 1, wherein R4 and R5 together with the atoms attached thereto are joined to form a heterocycloalkyl or heteroaryl comprising deuterium or fluorine, and/or a benzo[d][1,3]oxathiole group. 4. The compound of claim 1, wherein R4 is -ORa, -SRa, or -SeRa, with Ra being a C1-C6 alkyl substituted with one or more halogen. 5. The compound of claim 1, wherein the compound is an agonist of a serotonin 5-HT2 receptor. 6. The compound of claim 1, wherein the compound is an agonist of a serotonin 5-HT2A receptor. 7. The compound of claim 1, having a structure of Formula (II):

or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, wherein: X1 and X2 are independently hydrogen, deuterium, or a substituted or unsubstituted C1-C6 alkyl; Y1 and Y2 are independently hydrogen, deuterium, or a substituted or unsubstituted C1-C6 alkyl; R2 and R3 are independently hydrogen, deuterium, halogen, a substituted or unsubstituted C1-C6 alkyl, -ORa, or -SRa; R6 and R7 are independently hydrogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or alternatively R6 and R7 together with the nitrogen atom attached thereto are optionally joined to form a substituted or unsubstituted heterocycloalkyl; each Ra is independently hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl; A is O or S; Z1 and Z2 are independently hydrogen, deuterium, or fluorine; and when A is O, at least one of X1, X2, Y1, Y2, R2, R3, R6, R7, Z1, Z2 comprises deuterium, and/or at least one of Z1 and Z2 is fluorine. 8. The compound of claim 7, wherein R2 is -ORa.

9. The compound of claim 7, wherein X1 and X2 are hydrogen.

10. The compound of claim 7, wherein X1 and X2 are deuterium.

11. The compound of claim 7, wherein X1 is hydrogen or deuterium, and X2 is a substituted or unsubstituted Ci-Ce alkyl.

12. The compound of claim 7, wherein A is S.

13. The compound of claim 7, wherein A is O.

14. The compound of claim 7, wherein Z1 and Z2 are hydrogen.

15. The compound of claim 7, wherein Z1 and Z2 are deuterium.

16. The compound of claim 7, which is selected from the group consisting of

stereoisomer, or prodrug thereof.

17. The compound of claim 1, having a structure of Formula (III): or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, wherein: X1 and X2 are independently hydrogen, deuterium, or a substituted or unsubstituted C1-C6 alkyl; Y1 and Y2 are independently hydrogen, deuterium, or a substituted or unsubstituted C1-C6 alkyl; R4 is a substituted or unsubstituted C1-C6 alkyl, -ORa, -SRa, or -SeRa; R6 and R7 are independently hydrogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or alternatively R6 and R7 together with the nitrogen atom attached thereto are optionally joined to form a substituted or unsubstituted heterocycloalkyl; each Ra is independently hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl; and wherein at least one of X1, X2, Y1, Y2, R4, R6, R7, Ra comprises deuterium, and/or R4 is -ORa, -SRa, or -SeRa, with Ra in R4 being a C1-C6 alkyl substituted with one or more halogen. 18. The compound of claim 17, wherein R4 is -SMe, -SCD3, -SCF3, -SEt, -Sn-Pr, -SCH2CH2CF3, -SCH2CH2CF2H, -SCH2CH2CFH2, -Me, -CD3, -CF3, -OMe, -OCD3, -OCF3, -OCH2CH2CF3, -OCH2CH2CF2H, -OCH2CH2CFH2 or -Br. 19. The compound of claim 17, wherein each Ra is independently -Me, -CD3, or -CF3. 20. The compound of claim 17, wherein X1 and X2 are hydrogen. 21. The compound of claim 17, wherein X1 and X2 are deuterium. he compound of claim 17, which is selected from the group consisting of

pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof.

23. A pharmaceutical composition, comprising the compound of claim 1 and a pharmaceutically acceptable excipient.

24. The pharmaceutical composition of claim 23, wherein the compound is present in the pharmaceutical composition at a purity of at least 50% by weight based on a total weight of isotopologues of the compound present in the pharmaceutical composition.

25. The pharmaceutical composition of claim 23, wherein any position in the compound having deuterium has a minimum deuterium incorporation of at least 50 atom % at the site of deuteration.

26. The pharmaceutical composition of claim 23, which is substantially free of other isotopologues of the compound.

27. The pharmaceutical composition of claim 23, which is formulated for oral administration.

28. The pharmaceutical composition of claim 23, which is formulated for administration via inhalation.

29. A method of treating a subject with a disease or disorder associated with a serotonin 5- HT2 receptor or a monoamine transporter, the method comprising: administering to the subject a therapeutically effective amount of the compound of claim 1.

30. The method of claim 29, wherein the disease or disorder associated with a serotonin 5- HT2 receptor or a monoamine transporter is a neuropsychiatric disease or an inflammatory disease or disorder.

31. The method of claim 29, wherein the disease or disorder associated with a serotonin 5- HT2 receptor or a monoamine transporter is a central nervous system (CNS) disorder.

32. The method of claim 31, wherein the central nervous system (CNS) disorder is selected from the group consisting of post-traumatic stress disorder (PTSD), major depressive disorder (MDD), treatment-resistant depression (TRD), suicidal ideation, suicidal behavior, major depressive disorder with suicidal ideation or suicidal behavior, non-suicidal self-injury disorder (NSSID), a bipolar disorder and related disorders, cyclothymic disorder, obsessive-compulsive disorder (OCD), generalized anxiety disorder (GAD), social anxiety disorder, a substance use disorder, anorexia nervosa, bulimia nervosa, binge eating disorder, Alzheimer’s disease, persistent symptoms from a SARS-CoV-2 infection (CO VID-19), cluster headache and migraine, attention deficit hyperactivity disorder (ADHD), pain, aphantasia, childhood-onset fluency disorder, major neurocognitive disorder, mild neurocognitive disorder, sexual dysfunction, chronic fatigue syndrome, Lyme disease, and obesity.

33. The method of claim 31, wherein the central nervous system (CNS) disorder is pain.

34. The method of claim 31, wherein the central nervous system (CNS) disorder is sexual dysfunction.

35. The method of claim 29, wherein the disease or disorder associated with a serotonin 5-

HT2 receptor or a monoamine transporter is an autonomic nervous system (ANS) disorder.

36. The method of claim 35, wherein the autonomic nervous system (ANS) disorder is a pulmonary disorder or a cardiovascular disorder.

37. The method of claim 29, wherein the compound is administered orally, sublingually, buccally, topically, via injection, or via inhalation.

38. A single -lay er orally administered tablet composition comprising the compound of claim 1 , and a polymer.

39. The single-layer orally administered tablet composition of claim 38, wherein the composition is adapted for maximum sustained release.

40. The single-layer orally administered tablet composition of claim 38, wherein the tablet composition comprises a combination of (i) a water-insoluble neutrally charged non-ionic matrix; (ii) a polymer carrying one or more negatively charged groups; and (iii) the compound.

41. The single-layer orally administered tablet composition of claim 40, wherein the waterinsoluble neutrally charged non-ionic matrix is selected from a cellulose-based polymer, alone or enhanced by mixing with components selected from the group consisting of starches; waxes; neutral gums; polymethacrylates; PVA; PVA/PVP blends; and mixtures thereof.

42. The single-layer orally administered tablet composition of claim 41, wherein the cellulose-based polymer is hydroxypropyl methylcellulose (HPMC).

43. The single-layer orally administered tablet composition of claim 40, wherein the polymer carrying one or more negatively charged groups is selected from the group consisting of polyacrylic acid, polylactic acid, polyglycolic acid, polymethacrylate carboxylate, a cationexchange resin, a clay, a zeolite, hyaluronic acid, an anionic gum, salts thereof, and mixtures thereof.

44. The single-layer orally administered tablet composition of claim 43, wherein the anionic gum is selected from the group consisting of a naturally occurring material and a semisynthetic material.

45. The single-layer orally administered tablet composition of claim 44, wherein the naturally occurring material is selected from the group consisting of alginic acid, pectin, xanthan gum, carrageenan, locust bean gum, gum arabic, gum karaya, guar gum, and gum tragacanth.

46. The single-layer orally administered tablet composition of claim 44, wherein the semisynthetic material is selected from the group consisting of carboxymethyl-chitin and cellulose gum.

47. The single-layer orally administered tablet composition of claim 38, comprising a therapeutically effective amount of the compound for the treatment of pain.

48. The single-layer orally administered tablet composition of claim 38, comprising a therapeutically effective amount of the compound for the treatment of brain injury.

49. The single-layer orally administered tablet composition of claim 38, comprising a therapeutically effective amount of the compound for the treatment of depression.

50. The single-layer orally administered tablet composition of claim 38, comprising a therapeutically effective amount of the compound for use in treating a disease or disorder associated with a serotonin 5-HT2 receptor or a monoamine transporter.

51. The single-layer orally administered tablet composition of claim 50, wherein the disease or disorder is a central nervous system (CNS) disorder selected from the group consisting of post-traumatic stress disorder (PTSD), major depressive disorder (MDD), treatment-resistant depression (TRD), suicidal ideation, suicidal behavior, major depressive disorder with suicidal ideation or suicidal behavior, non-suicidal self-injury disorder (NSSID), bipolar and related disorders including bipolar I disorder, bipolar II disorder, cyclothymic disorder, obsessive- compulsive disorder (OCD), generalized anxiety disorder (GAD), social anxiety disorder, substance use disorders including alcohol use disorder, opioid use disorder, amphetamine use disorder, nicotine use disorder, and cocaine use disorder, anorexia nervosa, bulimia nervosa, binge eating disorder, Alzheimer’s disease, persistent symptoms from a SARS-CoV-2 infection (CO VID- 19), cluster headache and migraine, attention deficit hyperactivity disorder (ADHD), pain and neuropathic pain, aphantasia, childhood-onset fluency disorder, major neurocognitive disorder, mild neurocognitive disorder, sexual dysfunction, chronic fatigue syndrome, Lyme disease, and obesity.

52. The single-layer orally administered tablet composition of claim 50, wherein the disease or disorder is a condition of the autonomic nervous system (ANS).

53. The single-layer orally administered tablet composition of claim 52, wherein the disease or disorder is a pulmonary disorder.

54. The single-layer orally administered tablet composition of claim 52, wherein the disease or disorder is a cardiovascular disorder.

55. The single-layer orally administered tablet composition of claim 50, wherein the composition achieves a combined concentration of the compound in plasma in the range of 10-500 ng/ml, and maintains this concentration for a duration of release.

56. The single-layer orally administered tablet composition of claim 38, wherein the polymer comprises one or more negatively charged groups.

57. A tablet composition formulated for oral administration comprising the compound of claim 1 , and a polymer.

58. The tablet composition of claim 57, wherein the polymer comprises one or more negatively charged groups.

59. The tablet composition of claim 57, wherein the polymer comprises one or more acid groups.

60. The tablet composition of claim 57, wherein the polymer comprises a water-insoluble neutrally charged non-ionic matrix.

61. The tablet composition of claim 60, wherein the water-insoluble neutrally charged nonionic matrix is selected from a cellulose-based polymer, alone or enhanced by mixing with components selected from the group consisting of starches; waxes; neutral gums; polymethacrylates; PVA; PVA/PVP blends; and mixtures thereof.

62. The tablet composition of claim 61, wherein the cellulose -based polymer is hydroxypropyl methylcellulose (HPMC).

63. A kit for the treatment of a subject comprising 1) a single-layer orally administered tablet composition of claim 38, and 2) instructions for use in the treatment of pain.

64. The kit of claim 63, wherein the polymer comprises one or more negatively charged groups.

65. A kit for the treatment of a subject comprising 1) a single-layer orally administered tablet composition of claim 38, and 2) instructions for use in the treatment of brain injury.

66. The kit of claim 65, wherein the polymer comprises one or more negatively charged groups.

67. A kit for the treatment of a subject comprising 1) a single-layer orally administered tablet composition of claim 38 and 2) instructions for use in the treatment of depression.

68. The kit of claim 67, wherein the polymer comprises one or more negatively charged groups.

69. A kit for the treatment of a subject comprising 1) a single-layer orally administered tablet composition of claim 38 and 2) instructions for use in the treatment of a disease or disorder associated with a serotonin 5-HT2 receptor or a monoamine transporter.

70. The kit of claim 69, wherein the polymer comprises one or more negatively charged groups.

71. A method of delivering a psychedelic drug to a patient in need thereof comprising administering a psychedelic drug dissolved in a liquid phase of a mist via inhalation, wherein the psychedelic drug comprises the compound of claim 1.

72. The method of claim 71, wherein the psychedelic drug is delivered to the patient’s central nervous system.

73. The method of claim 71, wherein the psychedelic drug is delivered with air, oxygen, or a mixture of helium and oxygen.

74. The method of claim 73, wherein the psychedelic drug is delivered with a mixture of helium and oxygen.

75. The method of claim 74, wherein the mixture of helium and oxygen is heated to about 50°C to about 60°C.

76. The method of claim 74, wherein the helium is present in the mixture of helium and oxygen at about 50 to 90% and the oxygen is present in the mixture of helium and oxygen at about 10 to 50%.

77. The method of claim 74, further comprising administering a pretreatment inhalation therapy prior to administration of the mixture of helium and oxygen and the psychedelic drug.

78. The method of claim 77, wherein the pretreatment comprises administering via inhalation a mixture of helium and oxygen heated to about 90°C to about 120°C to the patient.

79. The method of claim 71, further comprising (i) administering via inhalation a mixture of helium and oxygen heated to about 90°C to about 120°C to the patient, and (ii) administering via inhalation to the patient a mist comprising helium and oxygen heated to about 50°C to about 60°C and the psychedelic drug.

80. The method of claim 79, further comprising repeating steps (i) and (ii) at least one time.

81. The method of claims 71, wherein the psychedelic drug is delivered to the patient’s central nervous system with an improvement in drug bioavailability by at least 25% as compared to oral delivery, increased Cmax by at least 25% as compared to oral delivery, reduced Tmax by at least 50% as compared to oral delivery, or a combination thereof.

82. A method of treating a central nervous system (CNS) disorder or psychological disorder comprising administering, via inhalation, a psychedelic drug dissolved in a mist, wherein the psychedelic drug comprises the compound of claim 1.

83. The method of claim 82, wherein the psychedelic drug is delivered with air, oxygen, or a mixture of helium and oxygen.

84. The method of claim 83, wherein the psychedelic drug is delivered with the mixture of helium and oxygen, and the mixture of helium and oxygen is heated to about 50°C to about 60°C prior to administering the psychedelic drug to the patient.

85. The method of claim 82, wherein the CNS disorder is post-traumatic stress disorder (PTSD), major depressive disorder (MDD), treatment-resistant depression (TRD), suicidal ideation, suicidal behavior, major depressive disorder with suicidal ideation or suicidal behavior, non-suicidal self-injury disorder (NSSID), bipolar and related disorders including bipolar I disorder, bipolar II disorder, cyclothymic disorder, obsessive-compulsive disorder (OCD), generalized anxiety disorder (GAD), social anxiety disorder, substance use disorders including alcohol use disorder, opioid use disorder, amphetamine use disorder, nicotine use disorder, and cocaine use disorder, anorexia nervosa, bulimia nervosa, binge eating disorder, Alzheimer’s disease, persistent symptoms from a SARS-CoV-2 infection (COVID-19), cluster headache and migraine, attention deficit hyperactivity disorder (ADHD), pain and neuropathic pain, aphantasia, childhood-onset fluency disorder, major neurocognitive disorder, mild neurocognitive disorder, sexual dysfunction, chronic fatigue syndrome, Lyme disease, or obesity.

86. A method of treating a subject with a disease or disorder associated with a serotonin receptor or a monoamine transporter, the method comprising: administering to the subject transdermally, subcutaneously, or intramuscularly, via an automatic injection device, a therapeutically effective amount of the compound of claim 1.

87. A compound having a structure of Formula (IV): or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, wherein:

X1 is hydrogen or deuterium;

X2 is a substituted or unsubstituted Ci-Ce alkyl;

Y1 and Y2 are independently hydrogen or deuterium;

R3 is hydrogen or deuterium;

R4 is hydrogen, deuterium, halogen, a substituted or unsubstituted Ci-Ce alkyl, a substituted or unsubstituted C3-C10 cycloalkyl, -ORb, -SRb, or -SeRb; R6 and R7 are independently hydrogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or alternatively R6 and R7 together with the nitrogen atom attached thereto are optionally joined to form a substituted or unsubstituted heterocycloalkyl; each Ra is independently a substituted or unsubstituted C1-C6 alkyl; and Rb is hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3- C10 cycloalkyl; with the proviso that at least one of X1, X2, Y1, Y2, R3, R4, R6, R7, and Ra comprises deuterium and/or R4 is -ORb, -SRb, or -SeRb, with Rb in R4 being a C1-C6 alkyl substituted with one or more halogen. 88. The compound of claim 87, which is selected from the group consisting of

pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof.

89. A pharmaceutical composition, comprising the compound of claim 87 and a pharmaceutically acceptable excipient.

90. A method of treating a subject with a disease or disorder associated with a serotonin 5- HT2 receptor or a monoamine transporter, the method comprising: administering to the subject a therapeutically effective amount of the compound of claim 87.

91. The method of claim 90, wherein the disease or disorder associated with a serotonin 5- HT2 receptor or a monoamine transporter is a neuropsychiatric disease or an inflammatory disease or disorder.

92. The method of claim 90, wherein the disease or disorder associated with a serotonin 5- HT2 receptor or a monoamine transporter is a central nervous system (CNS) disorder.

93. The method of claim 92, wherein the central nervous system (CNS) disorder is selected from the group consisting of post-traumatic stress disorder (PTSD), major depressive disorder (MDD), treatment-resistant depression (TRD), suicidal ideation, suicidal behavior, major depressive disorder with suicidal ideation or suicidal behavior, non-suicidal self-injury disorder (NSSID), a bipolar disorder and related disorders, cyclothymic disorder, obsessive-compulsive disorder (OCD), generalized anxiety disorder (GAD), social anxiety disorder, a substance use disorder, anorexia nervosa, bulimia nervosa, binge eating disorder, Alzheimer’s disease, persistent symptoms from a SARS-CoV-2 infection (CO VID-19), cluster headache and migraine, attention deficit hyperactivity disorder (ADHD), pain, aphantasia, childhood-onset fluency disorder, major neurocognitive disorder, mild neurocognitive disorder, sexual dysfunction, chronic fatigue syndrome, Lyme disease, and obesity.

94. A compound having a structure of Formula (V)

or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, wherein: X1 and X2 are independently hydrogen, deuterium, or a substituted or unsubstituted C1-C6 alkyl; Y1 and Y2 are independently hydrogen, deuterium, or a substituted or unsubstituted C1-C6 alkyl; R4 and R5 are independently hydrogen, deuterium, halogen, a substituted or unsubstituted C1-C6 alkyl, -ORa, -SRa, or -SeRa; or R4 and R5 together with the atoms attached thereto are optionally joined to form a heterocycloalkyl or heteroaryl; R6 and R7 are independently hydrogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or alternatively R6 and R7 together with the nitrogen atom attached thereto are optionally joined to form a substituted or unsubstituted heterocycloalkyl; each Ra is independently hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl; and Rb is hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3- C10 cycloalkyl. 95. The compound of claim 94, which is selected from the group consisting of

pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof.

96. A pharmaceutical composition, comprising the compound of claim 94 and a pharmaceutically acceptable excipient.

97. A method of treating a subject with a disease or disorder associated with a serotonin 5- HT2 receptor or a monoamine transporter, the method comprising: administering to the subject a therapeutically effective amount of the compound of claim 94.

98. The method of claim 97, wherein the disease or disorder associated with a serotonin 5- HT2 receptor or a monoamine transporter is a neuropsychiatric disease or an inflammatory disease or disorder.

99. The method of claim 97, wherein the disease or disorder associated with a serotonin 5- HT2 receptor or a monoamine transporter is a central nervous system (CNS) disorder.

100. The method of claim 99, wherein the central nervous system (CNS) disorder is selected from the group consisting of post-traumatic stress disorder (PTSD), major depressive disorder (MDD), treatment-resistant depression (TRD), suicidal ideation, suicidal behavior, major depressive disorder with suicidal ideation or suicidal behavior, non-suicidal self-injury disorder (NSSID), a bipolar disorder and related disorders, cyclothymic disorder, obsessive-compulsive disorder (OCD), generalized anxiety disorder (GAD), social anxiety disorder, a substance use disorder, anorexia nervosa, bulimia nervosa, binge eating disorder, Alzheimer’s disease, persistent symptoms from a SARS-CoV-2 infection (CO VID-19), cluster headache and migraine, attention deficit hyperactivity disorder (ADHD), pain, aphantasia, childhood-onset fluency disorder, major neurocognitive disorder, mild neurocognitive disorder, sexual dysfunction, chronic fatigue syndrome, Lyme disease, and obesity.

Description:
TITLE OF THE INVENTION PHENETHYLAMINE DERIVATIVES, COMPOSITIONS, AND METHODS OF USE CROSS-REFERENCE This application claims priority to U.S. Provisional Application No. 63/268,020, filed on February 15, 2022, and U.S. Provisional Application No.63/268,024, filed on February 15, 2022, each incorporated by reference herein in their entireties. FIELD OF THE INVENTION The present disclosure relates generally to chemical compounds and, in some embodiments, to serotonin 5-HT 2 receptor agonists, and in some embodiments, to serotonin receptor modulators, and in some embodiments to monoamine transporter modulators, and uses in the treatment of diseases associated with a 5-HT2 receptor, and uses in the treatment of diseases associated with a monoamine transporter. BACKGROUND OF THE INVENTION The “background” description provided herein is for the purpose of generally presenting the context of the disclosure. Work of the presently named inventors, to the extent it is described in this background section, as well as aspects of the description which may not otherwise qualify as prior art at the time of filing, are neither expressly or impliedly admitted as prior art against the present invention. There are three, closely related subtypes of serotonin 5-HT 2 receptors (5-HT 2 Rs), 5-HT 2A , 5- HT 2B , and 5-HT 2C , and they are primary targets of classic serotonergic psychedelics, such as lysergic acid diethylamide (LSD), psilocybin, and 2,5-dimethoxy-4-bromoamphetamine (DOB). Classic serotonergic psychedelics and entactogens, which also modulate the activity of the monoamine transporters, including the serotonin transporter (SERT), the norepinephrine transporters (NET), and the dopamine transporter (DAT), have been actively investigated by the research and medical community to alleviate a multitude of central nervous system (CNS) disorders (Reiff, C. M., Richman, E. E., Nemeroff, C. B., Carpenter, L. L., Widge, A. S., Rodriguez, C. I., Kalin, N. H., and McDonald, W. M., 2020, Psychedelics and Psychedelic-Assisted Psychotherapy, Am J Psychiatry 177, 391-410), such as: (i) post-traumatic stress disorder (PTSD)(Jerome, L., Feduccia, A. A., Wang, J. B., Hamilton, S., Yazar- Klosinski, B., Emerson, A., Mithoefer, M. C., and Doblin, R., 2020, Long-term follow-up outcomes of MDMA-assisted psychotherapy for treatment of PTSD: a longitudinal pooled analysis of six phase 2 trials, Psychopharmacology (Berl) 237, 2485-2497), (ii) major depressive disorder (MDD), (iii) treatment-resistant depression (TRD)(Goldberg, S. B., Pace, B. T., Nicholas, C. R., Raison, C. L., and Hutson, P. R., 2020, The experimental effects of psilocybin on symptoms of anxiety and depression: A meta-analysis, Psychiatry Res 284, 112749), (iv) obsessive-compulsive disorder (OCD)(Moreno, F. A., Wiegand, C. B., Taitano, E. K., and Delgado, P. L., 2006, Safety, tolerability, and efficacy of psilocybin in 9 patients with obsessive-compulsive disorder, J Clin Psychiatry 67, 1735-1740), (v) social anxiety disorder (ClinicalTrials.gov, number NCT02008396), (vi) substance use disorders, including but not limited to alcohol use disorder, opioid use disorder, amphetamine use disorder, nicotine use disorder, and cocaine use disorder, (vii) anorexia nervosa, (viii) bulimia nervosa (ClinicalTrials.gov, numbers NCT04454684 and NCT04052568), (ix) Alzheimer’s disease (ClinicalTrials.gov, number NCT04123314), and (x) cluster headache and migraine (Nichols, D. E., 2016, Psychedelics, Pharmacol Rev 68, 264-355; Johnson, M. W., Hendricks, P. S., Barrett, F. S., and Griffiths, R. R., 2019, Classic psychedelics: An integrative review of epidemiology, therapeutics, mystical experience, and brain network function, Pharmacol Ther 197, 83-102; Sewell, R. A., Halpern, J. H., and Pope, H. G., Jr., 2006, Response of cluster headache to psilocybin and LSD, Neurology 66, 1920-1922; ClinicalTrials.gov, number NCT04218539).

These drugs have also been investigated to alleviate conditions of the autonomic nervous system (ANS), including pulmonary disorders (e.g., asthma and chronic obstructive pulmonary disorder (COPD) and cardiovascular disorders (e.g., atherosclerosis), among others (Nichols, D. E., Johnson, M. W., and Nichols, C. D., 2017, Psychedelics as Medicines: An Emerging New Paradigm, Clin Pharmacol Ther 101, 209-219; Flanagan, T. W., Sebastian, M. N., Battaglia, D. M., Foster, T. P., Cormier, S. A., and Nichols, C. D., 2019, 5-HT2 receptor activation alleviates airway inflammation and structural remodeling in a chronic mouse asthma model, Life Sci 236, 116790; Flanagan, T. W., Sebastian, M. N., Battaglia, D. M., Foster, T. P., Maillet, E. L., and Nichols, C. D., 2019, Activation of 5-HT2 Receptors Reduces Inflammation in Vascular Tissue and Cholesterol Levels in High-Fat Diet-Fed Apolipoprotein E Knockout Mice, Sci Rep 9, 13444; Sexton, J. D., Nichols, C. D., and Hendricks, P. S., 2019, Population Survey Data Informing the Therapeutic Potential of Classic and Novel Phenethylamine, Tryptamine, and Lysergamide Psychedelics, Front Psychiatry 10, 896).

Some studies have advanced into Phase III trials, for example the use of 3,4- methylenedioxymethamphetamine (MDMA) for the treatment of PTSD (Feduccia, A. A., Jerome, L., Yazar-Klosinski, B., Emerson, A., Mithoefer, M. C., and Doblin, R., 2019, Breakthrough for Trauma Treatment: Safety and Efficacy of MDMA-Assisted Psychotherapy Compared to Paroxetine and Sertraline, Front Psychiatry 10, 650), and phase 1 trials of 3,4,5-trimethoxyphenethylamine (mescaline) have begun (ClinicalTrials.gov, number NCT04227756).

Mechanistically, the therapeutic effects of psychedelic phenethylamines/amphetamines are thought to be mediated by their interaction with serotonin (5-HT) receptors, particularly 5-HT2A receptors, though other targets, including the 5-HTi receptors (e.g., 5-HTIA, 5-HTIB) may also be involved (Nichols, D. E., 2016, Psychedelics, Pharmacol Rev 68, 264-355; Canal, C. E., 2018, Serotonergic Psychedelics: Experimental Approaches for Assessing Mechanisms of Action, Handb Exp Pharmacol 252, 227-260). A contribution from the 5-HT2C receptor may be responsible for the reported anti- addictive properties of classic psychedelics (Canal, C. E., and Murnane, K. S., 2017, The serotonin 5-HT2C receptor and the non-addictive nature of classic hallucinogens, J Psychopharmacol 31, 127- 143). The effects of entactogen phenethylamines, including MDMA and MDA, are mediated primarily by their interaction with monoamine transporters, particular the serotonin (SERT) and dopamine (DAT) transporters (Jayanthi, L. D., and Ramamoorthy, S., 2005, Regulation of monoamine transporters: influence of psychostimulants and therapeutic antidepressants, AAPS J 7, E728-738).

Safety aspects of psychedelics and entactogens remain a key challenge for clinical applications, with treatment protocols being challenged by the following factors: 1) the relatively slow onset of psychoactive therapeutic benefits; 2) long acting effects, often times requiring full day patient supervision; 3) numerous acute neuropsychiatric and gastrointestinal adverse effects, including anxiety, fear, tachycardia, hypertension, increased body temperature, nausea and vomiting, with many acute adverse effects being attributed to high drug concentrations (“spiking”) in the blood shortly after oral administration; 4) low brain bioavailability (e.g., as observed with mescaline); 5) therapeutic effects requiring high oral doses (e.g., as observed with mescaline and MDMA); and 6) toxicity such as neurotoxicity and cardiotoxicity (Schenk, S., and Newcombe, D., 2018,

Methylenedioxymethamphetamine (MDMA) in Psychiatry: Pros, Cons, and Suggestions, J Clin Psychopharmacol 38, 632-638; Garcia-Romeu, A., Kersgaard, B., and Addy, P. H., 2016, Clinical applications of hallucinogens: A review, Exp Clin Psychopharmacol 24, 229-268; Morgan, L., 2020, MDMA-assisted psychotherapy for people diagnosed with treatment-resistant PTSD: what it is and what it isn't, Ann Gen Psychiatry 19, 33; Schenk, S., and Newcombe, D., 2018, Methylenedioxymethamphetamine (MDMA) in Psychiatry: Pros, Cons, and Suggestions, J Clin Psychopharmacol 38, 632-638; Huang, X.-P., Setola, V., Yadav, P. N., Allen, J. A., Rogan, S. C., Hanson, B. J., Revankar, C., Robers, M., Doucette, C., and Roth, B. L., 2009, Parallel Functional Activity Profiling Reveals Valvulopathogens Are Potent 5-Hydroxytryptamine(2B) Receptor Agonists: Implications for Drug Safety Assessment, Molecular Pharmacology 76, 710-722; Rothman, R. B., and Baumann, M. H., 2009, Serotonergic drugs and valvular heart disease, Expert Opin Drug Saf 8, 317- 329; Parrott, A. C., 2014, The potential dangers of using MDMA for psychotherapy, J Psychoactive Drugs 46, 37-43; Meyer, J. S., 2013, 3,4-methylenedioxymethamphetamine (MDMA): current perspectives, Subst Abuse Rehabil 4, 83-99; Baylen, C. A., and Rosenberg, H., 2006, A review of the acute subjective effects of MDMA/ecstasy, Addiction 101, 933-947; Shulgin, A., and Shulgin, Ann., 1991, Pihkal: a chemical love story, Transform Press, Berkeley, CA; Barrett, F. S., Bradstreet, M. P., Leoutsakos, J. S., Johnson, M. W., and Griffiths, R. R., 2016, The Challenging Experience Questionnaire: Characterization of challenging experiences with psilocybin mushrooms, J Psychopharmacol 30, 1279-1295).

In the case of drugs containing a methylenedioxy ring, such as 3,4- methylenedioxymethamphetamine (MDMA), 3,4-methylenedioxyamphetamine (MDA), 3,4- methylenedioxyphenethylamine (MDPEA), etc., metabolic degradation including O-demethylenation, (primarily mediated by CYP2D6 enzymes) contributes to poor exposure (Tucker, G. T., Lennard, M. S., Ellis, S. W., Woods, H. F., Cho, A. K., Lin, L. Y., Hiratsuka, A., Schmitz, D. A., and Chu, T. Y. Y., 1994, The demethylenation of methylenedioxymethamphetamine (“ecstasy”) by debrisoquine hydroxylase (CYP2D6), Biochemical Pharmacology 47, 1151-1156; Schmid, Y., Vizeli, P., Hysek, C. M., Prestin, K., Meyer Zu Schwabedissen, H. E., and Liechti, M. E., 2016, CYP2D6 function moderates the pharmacokinetics and pharmacodynamics of 3,4-methylene-dioxymethamphetamine in a controlled study in healthy individuals, Pharmacogenet Genomics 26, 397-401). For example, MDPEA is biologically inactive due to extensive first-pass metabolism, while metabolism of drugs such as MDMA follows non-linear pharmacokinetics, which results in disproportionate increases in plasma MDMA concentrations with relatively small increases in dose, contributing to increased toxicity (de la Torre, R., Farre, M., Roset, P. N., Pizarro, N., Abanades, S., Segura, M., Segura, J., and Cami, J., 2004, Human pharmacology of MDMA: pharmacokinetics, metabolism, and disposition, Ther Drug Monit 26, 137- 144; de la Torre, R., Farre, M., Ortuno, J., Mas, M., Brenneisen, R., Roset, P. N., Segura, J., and Cami, J., 2000, Non-linear pharmacokinetics of MDMA ('ecstasy') in humans, British journal of clinical pharmacology 49, 104-109; Farre, M., de la Torre, R., Mathuna, B. O., Roset, P. N., Peiro, A. M., Torrens, M., Ortuno, J., Pujadas, M., and Cami, J., 2004, Repeated doses administration of MDMA in humans: pharmacological effects and pharmacokinetics, Psychopharmacology (Berl) 173, 364-375). Indeed, metabolism of methylenedioxy-containing drugs causes high exposures to toxic metabolites, such as 3,4-dihydroxymethamphetamine in the case of MDMA, a cardiotoxic metabolite (Schindler, C. W., Thorndike, E. B., Blough, B. E., Telia, S. R., Goldberg, S. R., and Baumann, M. H., 2014, Effects of 3,4-methylenedioxymethamphetamine (MDMA) and its main metabolites on cardiovascular function in conscious rats, Br J Pharmacol 171, 83-91).

In the case of amphetamines, e.g., MDMA, 2,4,5-trimethoxyphenylamphetamine (TMA-2), and l-(2,5-dimethoxy-4-(methylthio)phenyl)propan-2-amine (DOT), the lipophilic a-side chain methyl group on the phenethylamine scaffold generally enhances pharmacokinetic and pharmacodynamic properties compared to phenethylamines. Thus, they generally have longer half-lives than phenethylamines. They also have higher intrinsic activity at their targets, e.g., substituted amphetamine psychedelics are full agonists at G protein-coupled receptors (GPCRs), relative to their phenethylamine analogs which are partial agonists (Rickli, A., Luethi, D., Reinisch, J., Buchy, D., Hoener, M. C., and Liechti, M. E., 2015, Receptor interaction profiles of novel N-2-methoxybenzyl (NBOMe) derivatives of 2,5-dimethoxy-substituted phenethylamines (2C drugs), Neuropharmacology 99, 546-553; Monte, A. P., Marona-Lewicka, D., Parker, M. A., Wainscott, D. B., Nelson, D. L., and Nichols, D. E., 1996, Dihydrobenzofuran analogues of hallucinogens. 3. Models of 4-substituted (2,5- dimethoxyphenyl)alkylamine derivatives with rigidified methoxy groups, J Med Chem 39, 2953-2961; Monte, A. P., Waldman, S. R., Marona-Lewicka, D., Wainscott, D. B., Nelson, D. L., Sanders-Bush, E., and Nichols, D. E., 1997, Dihydrobenzofuran analogues of hallucinogens. 4. Mescaline derivatives, J Med Chem 40, 2997-3008; Pottie, E., Cannaert, A., and Stove, C. P., 2020, In vitro structure-activity relationship determination of 30 psychedelic new psychoactive substances by means of beta-arrestin 2 recruitment to the serotonin 2A receptor, Arch Toxicol', Rickli, A., Moning, O. D., Hoener, M. C., and Liechti, M. E., 2016, Receptor interaction profiles of novel psychoactive tryptamines compared with classic hallucinogens, Eur Neuropsychopharmacol 26, 1327-1337). Thus, substituted amphetamines, in general, have relatively higher risk for side effects and toxicity compared to substituted phenethylamines. Further, enantiomeric amphetamines elicit different effects and can be metabolized at different rates, also being sex- and race-dependent and differentiated between poor and fast metabolizers, resulting in even more unpredictable pharmacokinetic outcomes. For example, the (S)- enantiomer of MDMA has been shown to exhibit greater stimulant-like properties, while and the (7?)- enantiomer has been shown to exhibit greater 5-HT2-mediated psychedelic-like properties. The (S)- enantiomer of MDMA is also metabolized more quickly than the (R)-enantiomer resulting in two effects: 1) a lower AUC and plasma half-life of the (5)-compared to the (R)-enantiomer and 2) more persistent bioavailability of the (5)-enantiomer of the active metabolite MDA.

In the case of 3,4,5-substituted phenethylamines such as mescaline found in cacti such as peyote (Lophophora williamsii) and San Pedro (Echinopsis pachanoi), psychedelic effects are produced as a result of 5-HT2A agonism with contributions also from agonism at 5-HT2C and 5-HTIA receptors (Nichols, D. E., 2004, Hallucinogens, Pharmacol Ther 101, 131-181; Rickli, A., Luethi, D., Reinisch, J., Buchy, D., Hoener, M. C., and Liechti, M. E., 2015, Receptor interaction profiles of novel N-2- methoxybenzyl (NBOMe) derivatives of 2,5-dimethoxy-substituted phenethylamines (2C drugs), Neuropharmacology 99, 546-553; Braden, M. R., Parrish, J. C., Naylor, J. C., and Nichols, D. E., 2006, Molecular interaction of serotonin 5-HT2A receptor residues Phe339(6.51) and Phe340(6.52) with superpotent N-benzyl phenethylamine agonists, Mol Pharmacol 70, 1956-1964). Like other members of this class, mescaline induces changes in perception, cognition, emotion, and mood that may underlie its reported neuropsychotherapeutic and psychospiritual benefits (Johnson, M. W ., Hendricks, P. S., Barrett, F. S., and Griffiths, R. R., 2019, Classic psychedelics: An integrative review of epidemiology, therapeutics, mystical experience, and brain network function, Pharmacol Ther 197, 83-102). Mescaline is orally active, however, only at high doses (e.g., -300 mg) owing to low brain bioavailability, has a slow onset of action, and causes nausea (Shulgin, A., and Shulgin, Ann., 1991, Pihkal: a chemical love story, Transform Press, Berkeley, CA).

Clearly, a safe therapeutic window for psychedelics and entactogens, such as those containing a methylenedioxy ring, amphetamines, and 3,4,5-substituted phenethylamines, is very narrow — and it has proven difficult to control drug exposure and maintain drug concentrations in the safe and efficacious range.

SUMMARY OF THE INVENTION

In view of the forgoing, there is a need for novel psychedelic and entactogen compounds which have improved and predictable pharmacokinetic properties — that are shorter acting, bioavailable, are less toxic and cause fewer side effects, and demonstrate enhanced oral activity at lower dosages. There is a further need for efficient, more convenient, and controllable compound formulations that afford no neurologically toxic (e.g., psychotomimetic toxic) plasma concentration.

Accordingly, it is one object of the present invention to provide novel compounds that meet these criteria.

It is another object of the present disclosure to provide novel pharmaceutical compositions which contain the compounds.

It is another object of the present disclosure to provide novel methods of treating a subject with a disease or disorder associated with a serotonin 5-HT2 receptor or with a monoamine transporter, with the compounds.

It is another object of the present disclosure to provide novel tablet compositions, such as singlelayer orally administered tablet compositions, containing the compounds.

It is another object of the present disclosure to provide novel kits containing formulations of the compounds for use in treatment.

It is yet another object of the present disclosure to provide novel methods of delivering the compounds in an aerosol, preferably a mist, via inhalation, such as for the treatment of a central nervous system (CNS) disorder or psychological disorder.

It is yet another object of the invention to provide a novel use of the compounds for treating a subject with a disease or disorder associated with a serotonin 5-HT2 receptor or with a monoamine transporter, such as a central nervous system (CNS) disorder or psychological disorder.

These and other objects, which will become apparent during the following detailed description, have been achieved by the inventors’ discovery of the novel compounds described herein (e.g., a compound of Formula (I) through (V)), including those which maintain preferential binding to G-protein coupled receptors (GPCRs), e.g., 5-HT2 receptors, or monoamine transporters, or 5-HT1 receptors, are bioavailable (e.g., orally bioavailable), have improved exposure (i.e., prevention of high drug concentrations (spiking) observed acutely after administration), and possess advantageous enzymatic degradation profiles which prevent bioactivation into toxic metabolites. As a result, the disclosed compounds have a propensity for reduced side effects, toxicity, and interpatient variability, thereby improving the therapeutic window and enabling practical use in clinical settings. The novel compounds are based on specific molecular modifications which slow or shunt enzymatic degradation at specific sites and/or which introduce metabolic soft spots at other sites—modifications which have been identified only after significant studies. Thus the present invention provides: (1) A compound having a structure of Formula (I): or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, wherein: X 1 and X 2 are independently hydrogen, deuterium, or a substituted or unsubstituted C 1 -C 6 alkyl; Y 1 and Y 2 are independently hydrogen, deuterium, or a substituted or unsubstituted C 1 -C 6 alkyl; R 2 and R 3 are independently hydrogen, deuterium, halogen, a substituted or unsubstituted C 1 -C 6 alkyl, -OR a , -SR a ; R 4 and R 5 are independently hydrogen, deuterium, halogen, a substituted or unsubstituted C 1 -C 6 alkyl, -OR a , -SR a , or -SeR a ; or R 4 and R 5 together with the atoms attached thereto are optionally joined to form a heterocycloalkyl or heteroaryl; R 6 and R 7 are independently hydrogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or alternatively R 6 and R 7 together with the nitrogen atom attached thereto are optionally joined to form a substituted or unsubstituted heterocycloalkyl; each R a is independently hydrogen, deuterium, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl; and wherein at least one of conditions (i)-(iii) are met

(i) at least one of X 1 , X 2 , Y 1 , Y 2 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 comprises deuterium,

(ii) R 4 and R 5 together with the atoms attached thereto are joined to form a heterocycloalkyl or heteroaryl comprising deuterium or fluorine, and/or a benzo[d][l,3]oxathiole group,

(iii) R 4 is -OR a , -SR a , or -SeR a , with R a in R 4 being a Ci-Ce alkyl substituted with one or more halogen; and with the proviso that when X 1 , X 2 , Y 1 , and Y 2 are each hydrogen or deuterium, both R 2 and R 5 are not -OR a .

(2) The compound of (1), wherein at least one of X 1 , X 2 , Y 1 , Y 2 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 comprises deuterium.

(3) The compound of (1) or (2), wherein R 4 and R 5 together with the atoms attached thereto are joined to form a heterocycloalkyl or heteroaryl comprising deuterium or fluorine, and/or a benzo[d][l,3]oxathiole group.

(4) The compound of (1) or (2), wherein R 4 is -OR a , -SR a , or -SeR a , with R a being a Ci-Ce alkyl substituted with one or more halogen.

(5) The compound of any one of (1) to (4), wherein the compound is an agonist of a serotonin 5-HT2 receptor.

(6) The compound of any one of (1) to (5), wherein the compound is an agonist of a serotonin 5 -HT2A receptor.

(7) The compound of any one of (1) to (3) or (5) to (6), having a structure of Formula (II):

or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, wherein:

X 1 and X 2 are independently hydrogen, deuterium, or a substituted or unsubstituted Ci-Ce alkyl; Y 1 and Y 2 are independently hydrogen, deuterium, or a substituted or unsubstituted Ci-Ce alkyl; R 2 and R 3 are independently hydrogen, deuterium, halogen, a substituted or unsubstituted Ci-Ce alkyl, -OR a , or -SR a ;

R 6 and R 7 are independently hydrogen, a substituted or unsubstituted Ci-Ce alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or alternatively R 6 and R 7 together with the nitrogen atom attached thereto are optionally joined to form a substituted or unsubstituted heterocycloalkyl; each R a is independently hydrogen, deuterium, a substituted or unsubstituted Ci-Ce alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl;

A is O or S;

Z 1 and Z 2 are independently hydrogen, deuterium, or fluorine; and when A is O, at least one of X 1 , X 2 , Y 1 , Y 2 , R 2 , R 3 , R 6 , R 7 , Z 1 , Z 2 comprises deuterium, and/or at least one of Z 1 and Z 2 is fluorine.

(8) The compound of (7), wherein R 2 is -OR a .

(9) The compound of (7) or (8), wherein X 1 and X 2 are hydrogen.

(10) The compound of (7) or (8), wherein X 1 and X 2 are deuterium.

(11) The compound of (7) or (8), wherein X 1 is hydrogen or deuterium, and X 2 is a substituted or unsubstituted Ci-Ce alkyl.

(12) The compound of any one of (7) to (11), wherein A is S.

(13) The compound of any one of (7) to (11), wherein A is O.

(14) The compound of any one of (7) to (13), wherein Z 1 and Z 2 are hydrogen.

(15) The compound of any one of (7) to (13), wherein Z 1 and Z 2 are deuterium.

(16) The compound of any one of (7) to (15), which is selected from the group consisting of:

pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof.

(17) The compound of any one of (1) to (2) or (4) to (6), having a structure of Formula (III): or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, wherein:

X 1 and X 2 are independently hydrogen, deuterium, or a substituted or unsubstituted Ci-Ce alkyl;

Y 1 and Y 2 are independently hydrogen, deuterium, or a substituted or unsubstituted Ci-Ce alkyl;

R 4 is a substituted or unsubstituted Ci-Ce alkyl, -OR a , -SR a , or -SeR a ;

R 6 and R 7 are independently hydrogen, a substituted or unsubstituted Ci-Ce alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or alternatively R 6 and R 7 together with the nitrogen atom attached thereto are optionally joined to form a substituted or unsubstituted heterocycloalkyl; each R a is independently hydrogen, deuterium, a substituted or unsubstituted Ci-Ce alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl; and wherein at least one of X 1 , X 2 , Y 1 , Y 2 , R 4 , R 6 , R 7 , R a comprises deuterium, and/or R 4 is -OR a , -SR a , or -SeR a , with R a in R 4 being a Ci-Ce alkyl substituted with one or more halogen.

(18) The compound of (17), wherein R 4 is -SMe, -SCD3, -SCF3, -SEt, -Sn-Pr, -SCH2CH2CF3, -SCH2CH2CF2H, -SCH2CH2CFH2, -Me, -CD3, -CF3, -OMe, -OCD3, -OCF3, -OCH2CH2CF3, -OCH2CH2CF2H, -OCH2CH2CFH2 or -Br.

(19) The compound of (17) or (18), wherein each R a is independently -Me, -CD3, or -CF3.

(20) The compound of any one of (17) to (19), wherein X 1 and X 2 are hydrogen.

(21) The compound of any one of (17) to (19), wherein X 1 and X 2 are deuterium.

(22) The compound of any one of (17) to (21), which is selected from the group consisting of:

pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof.

(23) A pharmaceutical composition, comprising the compound of any one of (1) to (22) and a pharmaceutically acceptable excipient.

(24) The pharmaceutical composition of (23), wherein the compound is present in the pharmaceutical composition at a purity of at least 50% by weight based on a total weight of isotopologues of the compound present in the pharmaceutical composition.

(25) The pharmaceutical composition of (23) or (24), wherein any position in the compound having deuterium has a minimum deuterium incorporation of at least 50 atom % at the site of deuteration.

(26) The pharmaceutical composition of any one of (23) to (25), which is substantially free of other isotopologues of the compound.

(27) The pharmaceutical composition of any one of (23) to (26), which is formulated for oral administration.

(28) The pharmaceutical composition of any one of (23) to (27), which is formulated for administration via inhalation.

(29) A method of treating a subject with a disease or disorder associated with a serotonin 5-HT2 receptor or a monoamine transporter, the method comprising: administering to the subject a therapeutically effective amount of the compound of any one of (1) to (22).

(30) The method of (29), wherein the disease or disorder associated with a serotonin 5-HT2 receptor or a monoamine transporter is a neuropsychiatric disease or an inflammatory disease or disorder.

(31) The method of (29) or (30), wherein the disease or disorder associated with a serotonin 5- HT2 receptor or a monoamine transporter is a central nervous system (CNS) disorder.

(32) The method of (31), wherein the central nervous system (CNS) disorder is selected from the group consisting of post-traumatic stress disorder (PTSD), major depressive disorder (MDD), treatment-resistant depression (TRD), suicidal ideation, suicidal behavior, major depressive disorder with suicidal ideation or suicidal behavior, non-suicidal self-injury disorder (NSSID), a bipolar disorder and related disorders, cyclothymic disorder, obsessive-compulsive disorder (OCD), generalized anxiety disorder (GAD), social anxiety disorder, a substance use disorder, anorexia nervosa, bulimia nervosa, binge eating disorder, Alzheimer’s disease, persistent symptoms from a SARS-CoV-2 infection (COVID-19), cluster headache and migraine, attention deficit hyperactivity disorder (ADHD), pain, aphantasia, childhood-onset fluency disorder, major neurocognitive disorder, mild neurocognitive disorder, sexual dysfunction, chronic fatigue syndrome, Lyme disease, and obesity.

(33) The method of (31) or (32), wherein the central nervous system (CNS) disorder is pain.

(34) The method of (31) or (32), wherein the central nervous system (CNS) disorder is sexual dysfunction.

(35) The method of (29) or (30), wherein the disease or disorder associated with a serotonin 5- HT2 receptor or a monoamine transporter is an autonomic nervous system (ANS) disorder.

(36) The method of (35), wherein the autonomic nervous system (ANS) disorder is a pulmonary disorder or a cardiovascular disorder.

(37) The method of any one of (29) to (36), wherein the compound is administered orally, sublingually, buccally, topically, via injection, or via inhalation. (38) A single-layer orally administered tablet composition comprising the compound of any one of (1) to (22), and a polymer.

(39) The single-layer orally administered tablet composition of (38), wherein the composition is adapted for maximum sustained release.

(40) The single-layer orally administered tablet composition of (38) or (39), wherein the tablet composition comprises a combination of (i) a water-insoluble neutrally charged non-ionic matrix; (ii) a polymer carrying one or more negatively charged groups; and (iii) the compound.

(41) The single-layer orally administered tablet composition of (40), wherein the water-insoluble neutrally charged non-ionic matrix is selected from a cellulose-based polymer, alone or enhanced by mixing with components selected from the group consisting of starches; waxes; neutral gums; poly methacrylates; PVA; PVA/PVP blends; and mixtures thereof.

(42) The single-layer orally administered tablet composition of (41), wherein the cellulose-based polymer is hydroxypropyl methylcellulose (HPMC).

(43) The single-layer orally administered tablet composition of any one of (40) to (42), wherein the polymer carrying one or more negatively charged groups is selected from the group consisting of polyacrylic acid, polylactic acid, polyglycolic acid, polymethacrylate carboxylate, a cation-exchange resin, a clay, a zeolite, hyaluronic acid, an anionic gum, salts thereof, and mixtures thereof.

(44) The single-layer orally administered tablet composition of (43), wherein the anionic gum is selected from the group consisting of a naturally occurring material and a semi-synthetic material.

(45) The single-layer orally administered tablet composition of (44), wherein the naturally occurring material is selected from the group consisting of alginic acid, pectin, xanthan gum, carrageenan, locust bean gum, gum arabic, gum karaya, guar gum, and gum tragacanth.

(46) The single-layer orally administered tablet composition of (44) or (45), wherein the semisynthetic material is selected from the group consisting of carboxymethyl-chitin and cellulose gum.

(47) The single-layer orally administered tablet composition of any one of (38) to (46), comprising a therapeutically effective amount of the compound for the treatment of pain.

(48) The single-layer orally administered tablet composition of any one of (38) to (46), comprising a therapeutically effective amount of the compound for the treatment of brain injury.

(49) The single-layer orally administered tablet composition of any one of (38) to (46), comprising a therapeutically effective amount of the compound for the treatment of depression.

(50) The single-layer orally administered tablet composition of any one of (38) to (46), comprising a therapeutically effective amount of the compound for use in treating a disease or disorder associated with a serotonin 5-HT2 receptor or a monoamine transporter.

(51) The single-layer orally administered tablet composition of (50), wherein the disease or disorder is a central nervous system (CNS) disorder selected from the group consisting of post-traumatic stress disorder (PTSD), major depressive disorder (MDD), treatment-resistant depression (TRD), suicidal ideation, suicidal behavior, major depressive disorder with suicidal ideation or suicidal behavior, non-suicidal self-injury disorder (NSSID), bipolar and related disorders including bipolar I disorder, bipolar II disorder, cyclothymic disorder, obsessive-compulsive disorder (OCD), generalized anxiety disorder (GAD), social anxiety disorder, substance use disorders including alcohol use disorder, opioid use disorder, amphetamine use disorder, nicotine use disorder, and cocaine use disorder, anorexia nervosa, bulimia nervosa, binge eating disorder, Alzheimer’s disease, persistent symptoms from a SARS-CoV-2 infection (COVID-19), cluster headache and migraine, attention deficit hyperactivity disorder (ADHD), pain and neuropathic pain, aphantasia, childhood-onset fluency disorder, major neurocognitive disorder, mild neurocognitive disorder, sexual dysfunction, chronic fatigue syndrome, Lyme disease, and obesity.

(52) The single-layer orally administered tablet composition of (50), wherein the disease or disorder is a condition of the autonomic nervous system (ANS).

(53) The single-layer orally administered tablet composition of (52), wherein the disease or disorder is a pulmonary disorder.

(54) The single-layer orally administered tablet composition of (52), wherein the disease or disorder is a cardiovascular disorder. (55) The single-layer orally administered tablet composition of any one of (50) to (54), wherein the composition achieves a combined concentration of the compound in plasma in the range of 10-500 ng/ml, and maintains this concentration for a duration of release.

(56) The single-layer orally administered tablet composition of any one of (38) to (55), wherein the polymer comprises one or more negatively charged groups.

(57) A tablet composition formulated for oral administration comprising the compound of any one of (1) to (22), and a polymer.

(58) The tablet composition of (57), wherein the polymer comprises one or more negatively charged groups.

(59) The tablet composition of (57) or (58), wherein the polymer comprises one or more acid groups.

(60) The tablet composition of any one of (57) to (59), wherein the polymer comprises a waterinsoluble neutrally charged non-ionic matrix.

(61) The tablet composition of (60), wherein the water-insoluble neutrally charged non-ionic matrix is selected from a cellulose-based polymer, alone or enhanced by mixing with components selected from the group consisting of starches; waxes; neutral gums; poly methacrylates; PVA; PVA/PVP blends; and mixtures thereof.

(62) The tablet composition of (61), wherein the cellulose-based polymer is hydroxypropyl methylcellulose (HPMC).

(63) A kit for the treatment of a subject comprising 1) a single-layer orally administered tablet composition of any one of (38) to (56), and 2) instructions for use in the treatment of pain.

(64) The kit of (63), wherein the polymer comprises one or more negatively charged groups.

(65) A kit for the treatment of a subject comprising 1) a single-layer orally administered tablet composition of any one of (38) to (56), and 2) instructions for use in the treatment of brain injury.

(66) The kit of (65), wherein the polymer comprises one or more negatively charged groups.

(67) A kit for the treatment of a subject comprising 1) a single-layer orally administered tablet composition of any one of (38) to (56), and 2) instructions for use in the treatment of depression.

(68) The kit of (67), wherein the polymer comprises one or more negatively charged groups.

(69) A kit for the treatment of a subject comprising 1) a single-layer orally administered tablet composition of any one of (38) to (56), and 2) instructions for use in the treatment of a disease or disorder associated with a serotonin 5-HT2 receptor or a monoamine transporter.

(70) The kit of (69), wherein the polymer comprises one or more negatively charged groups.

(71) A method of delivering a psychedelic drug to a patient in need thereof comprising administering a psychedelic drug dissolved in a liquid phase of an aerosol (e.g., a mist) via inhalation, wherein the psychedelic drug comprises the compound of any one of (1) to (22).

(72) The method of (71), wherein the psychedelic drug is delivered to the patient’s central nervous system.

(73) The method of (71) or (72), wherein the psychedelic drug is delivered with air, oxygen, or a mixture of helium and oxygen.

(74) The method of any one of (71) to (73), wherein the psychedelic drug is delivered with a mixture of helium and oxygen.

(75) The method of (74), wherein the mixture of helium and oxygen is heated to about 50°C to about 60°C.

(76) The method of (74) or (75), wherein the helium is present in the mixture of helium and oxygen at about 50 to 90% and the oxygen is present in the mixture of helium and oxygen at about 10 to 50%. (77) The method of any one of (74) to (76), further comprising administering a pretreatment inhalation therapy prior to administration of the mixture of helium and oxygen and the psychedelic drug.

(78) The method of (77), wherein the pretreatment comprises administering via inhalation a mixture of helium and oxygen heated to about 90°C to about 120°C to the patient.

(79) The method of any one of (71) to (78), further comprising (i) administering via inhalation a mixture of helium and oxygen heated to about 90°C to about 120°C to the patient, and (ii) administering via inhalation to the patient an aerosol (e.g., a mist) comprising helium and oxygen heated to about 50°C to about 60°C and the psychedelic drug.

(80) The method of (79), further comprising repeating steps (i) and (ii) at least one time.

(81) The method of any one of (71) to (80), wherein the psychedelic drug is delivered to the patient’s central nervous system with an improvement in drug bioavailability by at least 25% as compared to oral delivery, increased Cmax by at least 25% as compared to oral delivery, reduced Tmaxby at least 50% as compared to oral delivery, or a combination thereof.

(82) A method of treating a central nervous system (CNS) disorder or psychological disorder comprising administering, via inhalation, a psychedelic drug dissolved in an aerosol (e.g., a mist), wherein the psychedelic drug comprises the compound of any one of (1) to (22).

(83) The method of (82), wherein the psychedelic drug is delivered with air, oxygen, or a mixture of helium and oxygen.

(84) The method of (83), wherein the psychedelic drug is delivered with the mixture of helium and oxygen, and the mixture of helium and oxygen is heated to about 50°C to about 60°C prior to administering the psychedelic drug to the patient.

(85) The method of any one of (82) to (84), wherein the CNS disorder is post-traumatic stress disorder (PTSD), major depressive disorder (MDD), treatment-resistant depression (TRD), suicidal ideation, suicidal behavior, major depressive disorder with suicidal ideation or suicidal behavior, non- suicidal self-injury disorder (NSSID), bipolar and related disorders including bipolar I disorder, bipolar II disorder, cyclothymic disorder, obsessive-compulsive disorder (OCD), generalized anxiety disorder (GAD), social anxiety disorder, substance use disorders including alcohol use disorder, opioid use disorder, amphetamine use disorder, nicotine use disorder, and cocaine use disorder, anorexia nervosa, bulimia nervosa, binge eating disorder, Alzheimer’s disease, persistent symptoms from a SARS-CoV-2 infection (COVID-19), cluster headache and migraine, attention deficit hyperactivity disorder (ADHD), pain and neuropathic pain, aphantasia, childhood-onset fluency disorder, major neurocognitive disorder, mild neurocognitive disorder, sexual dysfunction, chronic fatigue syndrome, Lyme disease, or obesity. (86) A method of treating a subject with a disease or disorder associated with a serotonin receptor or a monoamine transporter, the method comprising: administering to the subject transdermally, subcutaneously, or intramuscularly, via an automatic injection device, a therapeutically effective amount of the compound of any one of (1) to (22). (87) A compound having a structure of Formula (IV): or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, wherein: X 1 is hydrogen or deuterium; X 2 is a substituted or unsubstituted C 1 -C 6 alkyl; Y 1 and Y 2 are independently hydrogen or deuterium; R 3 is hydrogen or deuterium; R 4 is hydrogen, deuterium, halogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted C3-C10 cycloalkyl, -OR b , -SR b , or -SeR b ; R 6 and R 7 are independently hydrogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or alternatively R 6 and R 7 together with the nitrogen atom attached thereto are optionally joined to form a substituted or unsubstituted heterocycloalkyl; each R a is independently a substituted or unsubstituted C 1 -C 6 alkyl; and R b is hydrogen, deuterium, a substituted or unsubstituted Ci-Ce alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl; with the proviso that at least one of X 1 , X 2 , Y 1 , Y 2 , R 3 , R 4 , R 6 , R 7 , and R a comprises deuterium and/or R 4 is -OR b , -SR b , or -SeR b , with R b in R 4 being a Ci-Ce alkyl substituted with one or more halogen.

(88) The compound of (87), which is selected from the group consisting of

pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof.

(89) A pharmaceutical composition, comprising the compound of (87) and a pharmaceutically acceptable excipient. (90) A method of treating a subject with a disease or disorder associated with a serotonin 5-HT2 receptor or a monoamine transporter, the method comprising: administering to the subject a therapeutically effective amount of the compound of (87).

(91) The method of (90), wherein the disease or disorder associated with a serotonin 5-HT2 receptor or a monoamine transporter is a neuropsychiatric disease or an inflammatory disease or disorder. (92) The method of (90), wherein the disease or disorder associated with a serotonin 5-HT2 receptor or a monoamine transporter is a central nervous system (CNS) disorder. (93) The method of (92), wherein the central nervous system (CNS) disorder is selected from the group consisting of post-traumatic stress disorder (PTSD), major depressive disorder (MDD), treatment-resistant depression (TRD), suicidal ideation, suicidal behavior, major depressive disorder with suicidal ideation or suicidal behavior, non-suicidal self-injury disorder (NSSID), a bipolar disorder and related disorders, cyclothymic disorder, obsessive-compulsive disorder (OCD), generalized anxiety disorder (GAD), social anxiety disorder, a substance use disorder, anorexia nervosa, bulimia nervosa, binge eating disorder, Alzheimer’s disease, persistent symptoms from a SARS-CoV-2 infection (COVID-19), cluster headache and migraine, attention deficit hyperactivity disorder (ADHD), pain, aphantasia, childhood-onset fluency disorder, major neurocognitive disorder, mild neurocognitive disorder, sexual dysfunction, chronic fatigue syndrome, Lyme disease, and obesity. (94) A compound having a structure of Formula (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, wherein: X 1 and X 2 are independently hydrogen, deuterium, or a substituted or unsubstituted C1-C6 alkyl; Y 1 and Y 2 are independently hydrogen, deuterium, or a substituted or unsubstituted C1-C6 alkyl; R 4 and R 5 are independently hydrogen, deuterium, halogen, a substituted or unsubstituted C1-C6 alkyl, -OR a , -SR a , or -SeR a ; or R 4 and R 5 together with the atoms attached thereto are optionally joined to form a heterocycloalkyl or heteroaryl; R 6 and R 7 are independently hydrogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or alternatively R 6 and R 7 together with the nitrogen atom attached thereto are optionally joined to form a substituted or unsubstituted heterocycloalkyl; each R a is independently hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl; and R b is hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl. (95) The compound of (94), which is selected from the group consisting of

pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof.

(96) A pharmaceutical composition, comprising the compound of (94) or (95) and a pharmaceutically acceptable excipient. (97) A method of treating a subject with a disease or disorder associated with a serotonin 5-HT2 receptor or a monoamine transporter, the method comprising: administering to the subject a therapeutically effective amount of the compound of (94) or (95).

(98) The method of (97), wherein the disease or disorder associated with a serotonin 5-HT2 receptor or a monoamine transporter is a neuropsychiatric disease or an inflammatory disease or disorder.

(99) The method of (97), wherein the disease or disorder associated with a serotonin 5-HT2 receptor or a monoamine transporter is a central nervous system (CNS) disorder.

(100) The method of (99), wherein the central nervous system (CNS) disorder is selected from the group consisting of post-traumatic stress disorder (PTSD), major depressive disorder (MDD), treatment-resistant depression (TRD), suicidal ideation, suicidal behavior, major depressive disorder with suicidal ideation or suicidal behavior, non-suicidal self-injury disorder (NSSID), a bipolar disorder and related disorders, cyclothymic disorder, obsessive-compulsive disorder (OCD), generalized anxiety disorder (GAD), social anxiety disorder, a substance use disorder, anorexia nervosa, bulimia nervosa, binge eating disorder, Alzheimer’s disease, persistent symptoms from a SARS-CoV-2 infection (COVID-19), cluster headache and migraine, attention deficit hyperactivity disorder (ADHD), pain, aphantasia, childhood-onset fluency disorder, major neurocognitive disorder, mild neurocognitive disorder, sexual dysfunction, chronic fatigue syndrome, Lyme disease, and obesity.

(101) Use of a compound of any one of (1) to (22) for treating a subject with a disease or disorder, including a disease or disorder associated with a serotonin 5-HT2 receptor or a monoamine transporter.

(102) Use of a compound of (87) or (88) for treating a subject with a disease or disorder, including a disease or disorder associated with a serotonin 5-HT2 receptor or a monoamine transporter.

(103) Use of a compound of (94) or (95) for treating a subject with a disease or disorder, including a disease or disorder associated with a serotonin 5-HT2 receptor or a monoamine transporter.

BRIEF DESCRIPTION OF THE DRAWINGS

The forgoing paragraphs have been provided by way of general introduction and are not intended to limit the scope of the following claims. The described embodiments, together with further advantages, will be best understood by reference to the following detailed description when considered in conjunction with the accompanying drawings, wherein:

Fig. 1 illustrates the synthetic route for making compound 11-11;

Fig. 2 illustrates the synthetic route for making compound 11-14;

Fig. 3 illustrates the synthetic route for making compound 11-15;

Fig. 4 illustrates the synthetic route for making compound 11-16;

Fig. 5 illustrates the synthetic route for making compound 11-17;

Fig. 6 illustrates the synthetic route for making compound 11-20;

Fig. 7 illustrates the synthetic route for making compound 11-21;

Fig. 8 illustrates the synthetic route for making compound 11-58;

Fig. 9 illustrates the synthetic route for making compounds of Formula (II) with an unsubstituted amino ( NH 2 ) group, e.g., compounds IT T, II-4, II-5, II-9, 11-10, 11-12, 11-13, 11-18, 11-19, 11-27, IT- 29, and 11-44;

Fig. 10 illustrates the synthetic route for making compounds of Formula (II) with a methylamino (-NHMe) or methyl-r/3-amino group (-NHCD3), e.g., compounds II-2, II-3, II-6, II-7, II-8, 11-31, IT- 33, 11-35, 11-40, 11-42, 11-45, 11-47, and 11-48;

Fig. 11 illustrates the synthetic route(s) for making compound III-l;

Fig. 12 illustrates the synthetic route(s) for making compound III-2;

Fig. 13 illustrates the synthetic route(s) for making compound III-3;

Fig. 14 illustrates the synthetic route(s) for making compound III-4;

Fig. 15 illustrates the synthetic route for making compound III-5;

Fig. 16 illustrates the synthetic route(s) for making compound III-7;

Fig. 17 illustrates the synthetic route for making compound IV-1;

Fig. 18 illustrates the synthetic route for making compound IV-9, in both (R) and (S) enantiomers;

Fig. 19 illustrates the synthetic route for making compound IV-36;

Fig. 20 illustrates the general procedure for resolution of phenylpropan-2-amine (e.g., amphetamine) enantiomers using fractional crystallization;

Fig. 21 illustrates the synthetic route for making Reference Compound I;

Fig. 22 illustrates the synthetic route for making Reference Compound 2;

Fig. 23 is a graph showing agonist-labeled 5-HT 2 A radioligand ([ 3 H]LSD) competition binding using compound TIT-5; data shown are the average results from four experiments combined (N=12 replicates per concentration for test compound, N=8 for 5-HT); Kd for [ 3 H]LSD was set to 0.78 nM, and data were analyzed using a two-site, fit Ki model (GraphPad Prism 9); the data point for -10 samples is total, specific binding (no compound present);

Fig. 24 is a graph showing agonist-labeled 5-HT2A radioligand ([ 3 H]LSD) competition binding using compound IV-1; data shown are the average results from two experiments combined (N=12 replicates per concentration for test compound, N=4 for 5-HT); Kd for [ 3 H]LSD was set to 0.78 nM, and data were analyzed using a one-site, fit K model (GraphPad Prism 9); the data point for -10 samples is total, specific binding (no compound present); 5-HT data fit better to a two-site model, however, for simplicity the one-site model results are shown;

Fig. 25 is a graph showing agonist-labeled 5-HT2A radioligand ([ 3 H]LSD) competition binding using the R and S enantiomers of compound IV-9; data shown are the average results from five experiments combined (N=15 replicates); Kd for [ 3 H]LSD was set to 0.78 nM, and data were analyzed using a one-site, fit K model (GraphPad Prism 9); the data point for -10 samples is total, specific binding (no compound present); 5-HT data fit better to a two-site model, however, for simplicity the one-site model results are shown; and

Fig. 26 is a graph showing agonist-labeled 5-HT2A radioligand ([ 3 H]LSD) competition binding using the R and S enantiomers of Reference Compound 2; data shown are the average results from four experiments combined (N=12 replicates); Kd for [ 3 H]LSD was set to 0.78 nM, and data were analyzed using a one-site, fit K model (GraphPad Prism 9); the data point for -10 samples is total, specific binding (no compound present); 5-HT data fit better to a two-site model, however, for simplicity the one-site model results are shown.

DETAILED DESCRIPTION

In the following detailed description of the embodiments of the instant disclosure, numerous specific details are set forth in order to provide a thorough understanding of the disclosed embodiments. However, it will be obvious to one skilled in the art that the embodiments of this disclosure may be practiced without these specific details. In other instances, well known methods, procedures, components, and circuits have not been described in detail so as not to unnecessarily obscure aspects of the embodiments of the instant disclosure.

Definitions

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this disclosure belongs.

“Alkyl” refers to monovalent saturated aliphatic hydrocarbyl groups having from 1 to 10 carbon atoms and such as 1 to 6 carbon atoms, or 1 to 5, or 1 to 4, or 1 to 3 carbon atoms. This term includes, by way of example, linear and branched hydrocarbyl groups such as methyl (CH3-), ethyl (CH3CH2-), n-propyl (CH3CH2CH2-), isopropyl ((CH3)2CH-), n-butyl (CH3CH2CH2CH2-), isobutyl ((CH3)2CHCH2-), sec-butyl ((CH3)(CH3CH2)CH-), t-butyl ((CH3)3C-), n-pentyl (CH3CH2CH2CH2CH2- ), and neopentyl ((CH3)3CCH2-). The term “substituted alkyl” refers to an alkyl group as defined herein wherein one or more carbon atoms in the alkyl chain have been optionally replaced with a heteroatom such as -O-, -N-, -S-, -S(O) n - (where n is 0 to 2), -NR- (where R is hydrogen or alkyl) and having from 1 to 10 substituents selected from the group consisting of deuterium, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, oxo, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclyl, heterocyclooxy, hydroxyamino, alkoxyamino, nitro, - SO-alkyl, -SO-aryl, -SO-heteroaryl, -SO2-alkyl, -SO2-aryl, -SO2-heteroaryl, and -NR R ’’ , wherein R and R may be the same or different and are chosen from hydrogen, optionally substituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, aryl, heteroaryl and heterocyclic. “Alkylene” refers to divalent aliphatic hydrocarbyl groups having from 1 to 6, including, for example, 1 to 3 carbon atoms that are either straight-chained or branched, and which are optionally interrupted with one or more groups selected from -O-, -NR 10 -, -NR 10 C(O)-, -C(O)NR 10 - and the like. This term includes, by way of example, methylene (-CH2-), ethylene (-CH 2 CH 2 -), n-propylene (-CH 2 CH 2 CH 2 -), iso-propylene (-CH 2 CH(CH 3 )-), (-C(CH 3 ) 2 CH 2 CH 2 -), (-C(CH 3 ) 2 CH 2 C(O)-), (-C(CH 3 ) 2 CH 2 C(O)NH-), (-CH(CH 3 )CH 2 -), and the like. “Substituted alkylene” refers to an alkylene group having from 1 to 3 hydrogens replaced with substituents as described for carbons in the definition of “substituted” below. The term “alkane” refers to alkyl group and alkylene group, as defined herein. The term “alkylaminoalkyl”, “alkylaminoalkenyl” and “alkylaminoalkynyl” refers to the groups R NHR - where R is alkyl group as defined herein and R is alkylene, alkenylene or alkynylene group as defined herein. The term “alkaryl” or “aralkyl” refers to the groups -alkylene-aryl and -substituted alkylene-aryl where alkylene, substituted alkylene and aryl are defined herein. “Alkoxy” refers to the group –O-alkyl, wherein alkyl is as defined herein. Alkoxy includes, by way of example, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, t-butoxy, sec-butoxy, n-pentoxy, and the like. The term “alkoxy” also refers to the groups alkenyl-O-, cycloalkyl-O-, cycloalkenyl-O-, and alkynyl-O-, where alkenyl, cycloalkyl, cycloalkenyl, and alkynyl are as defined herein. The term “substituted alkoxy” refers to the groups substituted alkyl-O-, substituted alkenyl-O-, substituted cycloalkyl-O-, substituted cycloalkenyl-O-, and substituted alkynyl-O- where substituted alkyl, substituted alkenyl, substituted cycloalkyl, substituted cycloalkenyl and substituted alkynyl are as defined herein. The term “alkoxyamino” refers to the group –NH-alkoxy, wherein alkoxy is defined herein. The term “haloalkoxy” refers to the groups alkyl-O- wherein one or more hydrogen atoms on the alkyl group have been substituted with a halo group and include, by way of examples, groups such as trifluoromethoxy, and the like. The term “haloalkyl” refers to a substituted alkyl group as described above, wherein one or more hydrogen atoms on the alkyl group have been substituted with a halo group. Examples of such groups include, without limitation, fluoroalkyl groups, such as trifluoromethyl, difluoromethyl, trifluoroethyl and the like. The term “alkylalkoxy” refers to the groups -alkylene-O-alkyl, alkylene-O-substituted alkyl, substituted alkylene-O-alkyl, and substituted alkylene-O-substituted alkyl wherein alkyl, substituted alkyl, alkylene and substituted alkylene are as defined herein. The term “alkylthioalkoxy” refers to the group -alkylene-S-alkyl, alkylene-S-substituted alkyl, substituted alkylene-S-alkyl and substituted alkylene-S-substituted alkyl wherein alkyl, substituted alkyl, alkylene and substituted alkylene are as defined herein. “Alkenyl” refers to straight chain or branched hydrocarbyl groups having from 2 to 6 carbon atoms, for example 2 to 4 carbon atoms and having at least 1, for example from 1 to 2 sites of double bond unsaturation. This term includes, by way of example, bi-vinyl, allyl, and but-3-en-1-yl. Included within this term are the cis and trans isomers or mixtures of these isomers. The term “substituted alkenyl” refers to an alkenyl group as defined herein having from 1 to 5 substituents, or from 1 to 3 substituents, selected from alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, oxo, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclyl, heterocyclooxy, hydroxyamino, alkoxyamino, nitro, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SO2-alkyl, -SO2- substituted alkyl, -SO2-aryl and -SO2-heteroaryl. “Alkynyl” refers to straight or branched monovalent hydrocarbyl groups having from 2 to 6 carbon atoms, for example, 2 to 3 carbon atoms and having at least 1 and for example, from 1 to 2 sites The term “substituted alkynyl” refers to an alkynyl group as defined herein having from 1 to 5 substituents, or from 1 to 3 substituents, selected from deuterium, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, oxo, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclyl, heterocyclooxy, hydroxyamino, alkoxyamino, nitro, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SO 2 - alkyl, -SO 2 -substituted alkyl, -SO 2 -aryl, and -SO 2 -heteroaryl. “Alkynyloxy” refers to the group –O-alkynyl, wherein alkynyl is as defined herein. Alkynyloxy includes, by way of example, ethynyloxy, propynyloxy, and the like. “Acyl” refers to the groups H-C(O)-, alkyl-C(O)-, substituted alkyl-C(O)-, alkenyl-C(O)-, substituted alkenyl-C(O)-, alkynyl-C(O)-, substituted alkynyl-C(O)-, cycloalkyl-C(O)-, substituted cycloalkyl-C(O)-, cycloalkenyl-C(O)-, substituted cycloalkenyl-C(O)-, aryl-C(O)-, substituted aryl-C(O)-, heteroaryl-C(O)-, substituted heteroaryl-C(O)-, heterocyclyl-C(O)-, and substituted heterocyclyl-C(O)-, wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein. For example, acyl includes the “acetyl” group CH3C(O) “Acylamino” refers to the groups –NR 20 C(O)alkyl, -NR 20 C(O)substituted alkyl, -NR 20 C(O)cycloalkyl, -NR 20 C(O)substituted cycloalkyl, -NR 20 C(O)cycloalkenyl, -NR 20 C(O)substituted cycloalkenyl, -NR 20 C(O)alkenyl, -NR 20 C(O)substituted alkenyl, -NR 20 C(O)alkynyl, -NR 20 C(O)substituted alkynyl, -NR 20 C(O)aryl, -NR 20 C(O)substituted aryl, -NR 20 C(O)heteroaryl, -NR 20 C(O)substituted heteroaryl, -NR 20 C(O)heterocyclic, and -NR 20 C(O)substituted heterocyclic, wherein R 20 is hydrogen or alkyl and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein. “Aminocarbonyl” or the term “aminoacyl” refers to the group -C(O)NR 21 R 22 , wherein R 21 and R 22 independently are selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic and where R 21 and R 22 are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein.

“Aminocarbonylamino” refers to the group -NR 21 C(O)NR 22 R 23 where R 21 , R 22 , and R 23 are independently selected from hydrogen, alkyl, aryl or cycloalkyl, or where two R groups are joined to form a heterocyclyl group.

The term “alkoxycarbonylamino” refers to the group -NRC(O)OR where each R is independently hydrogen, alkyl, substituted alkyl, aryl, heteroaryl, or heterocyclyl wherein alkyl, substituted alkyl, aryl, heteroaryl, and heterocyclyl are as defined herein.

The term “acyloxy” refers to the groups alkyl-C(O)O-, substituted alkyl-C(O)O-, cycloalkyl- C(O)O-, substituted cycloalkyl-C(O)O-, aryl-C(O)O-, heteroaryl-C(O)O-, and heterocyclyl-C(O)O- wherein alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, heteroaryl, and heterocyclyl are as defined herein.

“Aminosulfonyl” refers to the group -SO2NR 21 R 22 , wherein R 21 and R 22 independently are selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic and where R 21 and R 22 are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group and alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.

“Sulfonylamino” refers to the group -NR 21 SO2R 22 , wherein R 21 and R 22 independently are selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic and where R 21 and R 22 are optionally joined together with the atoms bound thereto to form a heterocyclic or substituted heterocyclic group, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein.

“Aryl” or “Ar” refers to a monovalent aromatic carbocyclic group of from 6 to 18 carbon atoms having a single ring (such as is present in a phenyl group) or a ring system having multiple condensed rings (examples of such aromatic ring systems include naphthyl, anthryl and indanyl) which condensed rings may or may not be aromatic, provided that the point of attachment is through an atom of an aromatic ring. This term includes, by way of example, phenyl and naphthyl. Unless otherwise constrained by the definition for the aryl substituent, such aryl groups can optionally be substituted with from 1 to 5 substituents, or from 1 to 3 substituents, selected from acyloxy, hydroxy, thiol, acyl, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, substituted alkyl, substituted alkoxy, substituted alkenyl, substituted alkynyl, substituted cycloalkyl, substituted cycloalkenyl, amino, substituted amino, aminoacyl, acylamino, alkaryl, aryl, aryloxy, azido, carboxyl, carboxylalkyl, cyano, halogen, nitro, heteroaryl, heteroaryloxy, heterocyclyl, heterocyclooxy, aminoacyloxy, oxyacylamino, thioalkoxy, substituted thioalkoxy, thioaryloxy, thiohetero aryloxy, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO- heteroaryl, -SCh-alkyl, -S Ch-substituted alkyl, -SCh-aryl, -SCh-heteroaryl and trihalomethyl.

“Aryloxy” refers to the group -O-aryl, wherein aryl is as defined herein, including, by way of example, phenoxy, naphthoxy, and the like, including optionally substituted aryl groups as also defined herein.

“Amino” refers to the group -NH2.

The term “substituted amino” refers to the group -NRR where each R is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl, and heterocyclyl provided that at least one R is not hydrogen.

The term “azido” refers to the group -N3.

“Carboxyl,” “carboxy” or “carboxylate” refers to -CO2H or salts thereof.

“Carboxyl ester” or “carboxy ester” or the terms “carboxyalkyl” or “carboxylalkyl” refers to the groups -C(O)O-alkyl, -C(O)O-substituted alkyl, -C(O)O-alkenyl, -C(O)O-substituted alkenyl, - C(O)O-alkynyl, -C(O)O-substituted alkynyl, -C(O)O-aryl, -C(O)O-substituted aryl, -C(O)O-cycloalkyl, -C(O)O-substituted cycloalkyl, -C(O)O-cycloalkenyl, -C(O)O-substituted cycloalkenyl, -C(O)O-heteroaryl, -C(O)O-substituted heteroaryl, -C(O)O-heterocyclic, and -C(O)O-substituted heterocyclic, wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein.

“(Carboxyl ester)oxy” or “carbonate” refers to the groups -O-C(O)O-alkyl, -O-C(O)O-substituted alkyl, -O-C(O)O-alkenyl, -O-C(O)O-substituted alkenyl, -O-C(O)O-alkynyl, -O-C(O)O-substituted alkynyl, -O-C(O)O-aryl, -O-C(O)O-substituted aryl, -O-C(O)O-cycloalkyl, -O-C(O)O-substituted cycloalkyl, -O-C(O)O-cycloalkenyl, -O-C(O)O-substituted cycloalkenyl, -O-C(O)O-heteroaryl, -O-C(O)O-substituted heteroaryl, -O-C(O)O-heterocyclic, and -O-C(O)O-substituted heterocyclic, wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein.

“Cyano” or “nitrile” refers to the group -CN.

“Cycloalkyl” refers to cyclic alkyl groups of from 3 to 10 carbon atoms having single or multiple cyclic rings including fused, bridged, and spiro ring systems. Examples of suitable cycloalkyl groups include, for instance, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl and the like. Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl, and the like, or multiple ring structures such as adamantanyl, and the like.

The term “substituted cycloalkyl” refers to cycloalkyl groups having from 1 to 5 substituents, or from 1 to 3 substituents, selected from deuterium, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, oxo, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclyl, heterocyclooxy, hydroxyamino, alkoxyamino, nitro, -SO-alkyl,

-SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SCh-alkyl, -SCh-substituted alkyl, -SCh-aryl and -SO2- heteroaryl.

“Cycloalkenyl” refers to non-aromatic cyclic alkyl groups of from 3 to 10 carbon atoms having single or multiple rings and having at least one double bond and for example, from 1 to 2 double bonds.

The term “substituted cycloalkenyl” refers to cycloalkenyl groups having from 1 to 5 substituents, or from 1 to 3 substituents, selected from deuterium, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclyl, heterocyclooxy, hydroxyamino, alkoxyamino, nitro, -SO-alkyl,

-SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -S02-alkyl, -S02-substituted alkyl, -S02-aryl and -SO2- heteroaryl.

“Cycloalkynyl” refers to non-aromatic cycloalkyl groups of from 5 to 10 carbon atoms having single or multiple rings and having at least one triple bond.

“Cycloalkoxy” refers to -O-cycloalkyl.

“Cycloalkenyloxy” refers to -O-cycloalkenyl.

“Halo” or “halogen” refers to fluoro, chloro, bromo, and iodo. “Hydroxy” or “hydroxyl” refers to the group -OH.

“Heteroaryl” refers to an aromatic group of from 1 to 15 carbon atoms, such as from 1 to 10 carbon atoms and 1 to 10 heteroatoms selected from the group consisting of oxygen, nitrogen, and sulfur within the ring. Such heteroaryl groups can have a single ring (such as, pyridinyl, imidazolyl or furyl) or multiple condensed rings in a ring system (for example as in groups such as, indolizinyl, quinolinyl, benzofuran, benzimidazolyl or benzo thienyl), wherein at least one ring within the ring system is aromatic and at least one ring within the ring system is aromatic, provided that the point of attachment is through an atom of an aromatic ring. In certain embodiments, the nitrogen and/or sulfur ring atom(s) of the heteroaryl group are optionally oxidized to provide for the N-oxide (N— >0), sulfinyl, or sulfonyl moieties. This term includes, by way of example, pyridinyl, pyrrolyl, indolyl, thiophenyl, and furanyl. Unless otherwise constrained by the definition for the heteroaryl substituent, such heteroaryl groups can be optionally substituted with 1 to 5 substituents, or from 1 to 3 substituents, selected from acyloxy, hydroxy, thiol, acyl, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, substituted alkyl, substituted alkoxy, substituted alkenyl, substituted alkynyl, substituted cycloalkyl, substituted cycloalkenyl, amino, substituted amino, aminoacyl, acylamino, alkaryl, aryl, aryloxy, azido, carboxyl, carboxylalkyl, cyano, halogen, nitro, heteroaryl, heteroaryloxy, heterocyclyl, heterocyclooxy, aminoacyloxy, oxyacylamino, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioheteroaryloxy, -SO- alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SCh-alkyl, -SCh-substituted alkyl, -SCh-aryl, and

-SO2-heteroaryl, and trihalomethyl.

The term “heteroaralkyl” refers to the groups -alkylene-heteroaryl where alkylene and heteroaryl are defined herein. This term includes, by way of example, pyridylmethyl, pyridylethyl, indolylmethyl, and the like.

“Heteroaryloxy” refers to -O-heteroaryl.

“Heterocycle,” “heterocyclic,” “heterocycloalkyl,” and “heterocyclyl” refer to a saturated or unsaturated group having a single ring or multiple condensed rings, including fused bridged and spiro ring systems, and having from 3 to 20 ring atoms, including 1 to 10 hetero atoms. These ring atoms are selected from the group consisting of nitrogen, sulfur, or oxygen, wherein, in fused ring systems, one or more of the rings can be cycloalkyl, aryl, or heteroaryl, provided that the point of attachment is through the non-aromatic ring. In certain embodiments, the nitrogen and/or sulfur atom(s) of the heterocyclic group are optionally oxidized to provide for the N-oxide, -S(O)-, or -SO2- moieties.

Examples of heterocycles and heteroaryls include, but are not limited to, azetidine, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, dihydroindole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthylpyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, phenanthroline, isothiazole, phenazine, isoxazole, phenoxazine, phenothiazine, imidazolidine, imidazoline, piperidine, piperazine, indoline, phthalimide, 1,2,3,4-tetrahydroisoquinoline, 4,5,6,7- tetrahydrobenzo[b]thiophene, thiazole, thiazolidine, thiophene, benzo[b]thiophene, morpholinyl, thiomorpholinyl (also referred to as thiamorpholinyl), 1,1-dioxothiomorpholinyl, piperidinyl, pyrrolidine, tetrahydrofuranyl, benzo[d][1,3]oxathiole, benzo[d][1,3]dioxole, and the like. Unless otherwise constrained by the definition for the heterocyclic substituent, such heterocyclic groups can be optionally substituted with 1 to 5, or from 1 to 3 substituents, selected from deuterium, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, oxo, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclyl, heterocyclooxy, hydroxyamino, alkoxyamino, nitro, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SO2-alkyl, -SO2-substituted alkyl, -SO2-aryl, -SO2-heteroaryl, and fused heterocycle. “Heterocyclyloxy” refers to the group –O-heterocyclyl. The term “heterocyclylthio” refers to the group heterocyclic-S-. The term “heterocyclene” refers to the diradical group formed from a heterocycle, as defined herein. The term “hydroxyamino” refers to the group -NHOH. “Nitro” refers to the group –NO 2 . “Oxo” refers to the atom (=O). “Sulfonyl” refers to the group SO 2 -alkyl, SO 2 -substituted alkyl, SO 2 -alkenyl, SO 2 -substituted alkenyl, SO2-cycloalkyl, SO2-substituted cylcoalkyl, SO2-cycloalkenyl, SO2-substituted cylcoalkenyl, SO2-aryl, SO2-substituted aryl, SO2-heteroaryl, SO2-substituted heteroaryl, SO2-heterocyclic, and SO2- substituted heterocyclic, wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein. Sulfonyl includes, by way of example, methyl-SO2-, phenyl-SO2-, and 4-methylphenyl- SO2-. “Sulfonyloxy” refers to the group –OSO 2 -alkyl, –OSO 2 -substituted alkyl, –OSO 2 -alkenyl, – OSO2-substituted alkenyl, –OSO2-cycloalkyl, –OSO2-substituted cylcoalkyl, –OSO 2 -cycloalkenyl, –OSO 2 -substituted cylcoalkenyl, –OSO 2 -aryl, –OSO 2 -substituted aryl, –OSO2-heteroaryl, –OSO2-substituted heteroaryl, –OSO2-heterocyclic, and –OSO2 substituted heterocyclic, wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein. The term “aminocarbonyloxy” refers to the group -OC(O)NRR where each R is independently hydrogen, alkyl, substituted alkyl, aryl, heteroaryl, or heterocyclic wherein alkyl, substituted alkyl, aryl, heteroaryl and heterocyclic are as defined herein. “Thiol” refers to the group -SH. “Thioxo” or the term “thioketo” refers to the atom (=S). “Alkylthio” or the term “thioalkoxy” refers to the group -S-alkyl, wherein alkyl is as defined herein. In certain embodiments, sulfur may be oxidized to -S(O)-. The sulfoxide may exist as one or more stereoisomers. The term “substituted thioalkoxy” refers to the group -S-substituted alkyl. The term “thioaryloxy” refers to the group aryl-S- wherein the aryl group is as defined herein including optionally substituted aryl groups also defined herein. The term “thioheteroaryloxy” refers to the group heteroaryl-S- wherein the heteroaryl group is as defined herein including optionally substituted aryl groups as also defined herein. The term “thioheterocyclooxy” refers to the group heterocyclyl-S- wherein the heterocyclyl group is as defined herein including optionally substituted heterocyclyl groups as also defined herein. In addition to the disclosure herein, the term “substituted,” when used to modify a specified group or radical, can also mean that one or more hydrogen atoms of the specified group or radical are each, independently of one another, replaced with the same or different substituent groups as defined below. In addition to the groups disclosed with respect to the individual terms herein, substituent groups for substituting for one or more hydrogens (any two hydrogens on a single carbon can be replaced with =O, =NR 70 , =N-OR 70 , =N2 or =S) on saturated carbon atoms in the specified group or radical are, unless otherwise specified, deuterium, -R 60 , halo, =O, -OR 70 , -SR 70 , -NR 80 R 80 , trihalomethyl, -CN, -OCN, - SCN, -NO, -NO2, =N2, -N3, -SO2R 70 , -SO2O M + , -SO2OR 70 , -OSO2R 70 , -OSO2O M + , -OSO2OR 70 , -P(O)(O )2(M + )2, -P(O)(OR 70 )O M + , -P(O)(OR 70 )2, -C(O)R 70 , -C(S)R 70 , -C(NR 70 )R 70 , -C(O)O M + , -C(O)OR 70 , -C(S)OR 70 , -C(O)NR 80 R 80 , -C(NR 70 )NR 80 R 80 , -OC(O)R 70 , -OC(S)R 70 , -OC(O)O-M + , -OC(O)OR 70 , -OC(S)OR 70 , -NR 70 C(O)R 70 , -NR 70 C(S)R 70 , -NR 70 CO 2 M + , -NR 70 CO 2 R 70 , -NR 70 C(S)OR 70 , -NR 70 C(O)NR 80 R 80 , -NR 70 C(NR 70 )R 70 and -NR 70 C(NR 70 )NR 80 R 80 , where R 60 is selected from the group consisting of optionally substituted alkyl, cycloalkyl, heteroalkyl, heterocycloalkylalkyl, cycloalkylalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl, each R 70 is independently hydrogen or R 60 ; each R 80 is independently R 70 or alternatively, two R 80 s, taken together with the nitrogen atom to which they are bonded, form a 5-, 6- or 7-membered heterocycloalkyl which may optionally include from 1 to 4 of the same or different additional heteroatoms selected from the group consisting of O, N and S, of which N may have -H or C1-C3 alkyl substitution; and each M + is a counter ion with a net single positive charge. Each M + may independently be, for example, an alkali ion, such as K + , Na + , Li + ; an ammonium ion, such as + N(R 60 )4; or an alkaline earth ion, such as [Ca 2+ ]o.s, [Mg 2+ ]o.s, or [Ba 2+ ]o.s (“subscript 0.5 means that one of the counter ions for such divalent alkali earth ions can be an ionized form of a compound of the disclosure and the other a typical counter ion such as chloride, or two ionized compounds disclosed herein can serve as counter ions for such divalent alkali earth ions, or a doubly ionized compound of the disclosure can serve as the counter ion for such divalent alkali earth ions). As specific examples, -NR 80 R 80 is meant to include -NH2, -NH-alkyl, A-pyrrolidinyl, A-piperazinyl, 4N- methyl-piperazin-l-yl and A-morpholinyl.

In addition to the disclosure herein, substituent groups for hydrogens on unsaturated carbon atoms in “substituted” alkene, alkyne, aryl and heteroaryl groups are, unless otherwise specified, deuterium, -R 60 , halo, -O M + , -OR 70 , -SR 70 , -S"M + , -NR 80 R 80 , trihalomethyl, -CF 3 , -CN, -OCN, -SCN, -NO, -NO2, -N 3 , -SO2R 70 , -SO 3 "M + , -SO 3 R 70 , -OSO2R 70 , -OSO 3 "M + , -OSO 3 R 70 , -PO 3 2 (M + )2, -P(O)(OR 70 )O-M + , -P(O)(OR 70 )2, -C(O)R 70 , -C(S)R 70 , -C(NR 70 )R 70 , -C0 2 "M + , -CO2R 70 , -C(S)OR 70 , -C(O)NR 80 R 80 , -C(NR 70 )NR 80 R 80 , -OC(O)R 70 , -OC(S)R 70 , -OCO 2 "M + , -OCO2R 70 , -OC(S)OR 70 , -NR 70 C(O)R 70 , -NR 70 C(S)R 70 , -NR 70 CO 2 "M + , -NR 70 CO 2 R 70 ,

-NR 70 C(S)OR 70 , -NR 70 C(O)NR 80 R 80 , -NR 70 C(NR 70 )R 70 and -NR 70 C(NR 70 )NR 80 R 80 , where R 60 , R 70 , R 80 and M + are as previously defined, provided that in case of substituted alkene or alkyne, the substituents are not -O’M + , -OR 70 , -SR 70 , or -S"M + .

In addition to the groups disclosed with respect to the individual terms herein, substituent groups for hydrogens on nitrogen atoms in “substituted” heteroalkyl and cycloheteroalkyl groups are, unless otherwise specified, -R 60 , -O M + , -OR 70 , -SR 70 , -S M + , -NR 80 R 80 , trihalomethyl, -CF 3 , -CN, -NO, -NO2, -S(O) 2 R 70 , -S(O) 2 O’M + , -S(O) 2 OR 70 , -OS(O) 2 R 70 , -OS(O) 2 O’M + , -OS(O) 2 OR 70 , -P(O)(O ) 2 (M + )2, - P(O)(OR 70 )O’M + , -P(O)(OR 70 )(OR 70 ), -C(O)R 70 , -C(S)R 70 , -C(NR 70 )R 70 ,

-C(O)OR 70 , -C(S)OR 70 , -C(O)NR 80 R 80 , -C(NR 70 )NR 80 R 80 , -OC(O)R 70 , -OC(S)R 70 , -OC(O)OR 70 , - OC(S)OR 70 , -NR 70 C(O)R 70 , -NR 70 C(S)R 70 , -NR 70 C(O)OR 70 , -NR 70 C(S)OR 70 ,

-NR 70 C(O)NR 80 R 80 , -NR 70 C(NR 70 )R 70 and -NR 70 C(NR 70 )NR 80 R 80 , where R 60 , R 70 , R 80 and M + are as previously defined.

In addition to the disclosure herein, in a certain embodiment, a group that is substituted has 1, 2, 3, or 4 substituents, 1, 2, or 3 substituents, 1 or 2 substituents, or 1 substituent. It is understood that in all substituted groups defined above, polymers arrived at by defining substituents with further substituents to themselves (e.g., substituted aryl having a substituted aryl group as a substituent which is itself substituted with a substituted aryl group, which is further substituted by a substituted aryl group, etc.) are not intended for inclusion herein. In such cases, the maximum number of such substitutions is three. For example, serial substitutions of substituted aryl groups specifically contemplated herein are limited to substituted aryl-(substituted aryl)-substituted aryl. However, substituent groups defined as e.g., polyethers may contain serial substitution greater than three, e.g., - O-(CH 2 CH 2 O) n -H, where n can be 1, 2, 3, or greater.

Unless indicated otherwise, the nomenclature of substituents that are not explicitly defined herein are arrived at by naming the terminal portion of the functionality followed by the adjacent functionality toward the point of attachment. For example, the substituent “arylalkyloxycarbonyl” refers to the group (aryl)-(alkyl)-O-C(O)-.

As to any of the groups disclosed herein which contain one or more substituents, it is understood, of course, that such groups do not contain any substitution or substitution patterns which are sterically impractical and/or synthetically non-feasible. In addition, the subject compounds include all stereochemical isomers arising from the substitution of these compounds.

As used herein, the term “fatty” describes a compound with a long-chain (linear) hydrophobic portion made up of hydrogen and anywhere from 4 to 26 carbon atoms, which may be fully saturated or partially unsaturated.

When it is defined that a substituent or group “comprise(s) deuterium” or is “comprising deuterium,” it is to be understood that the substituent or group may itself be deuterium, or the substituent or group may contain at least one deuterium substitution in its chemical structure. For example, when substituent “-R” is defined to comprise deuterium, it is to be understood that -R may be -D (-deuterium), or a group such as -CD3 that is consistent with the other requirements set forth of -R.

The phrases “pharmaceutically acceptable,” “physiologically acceptable,” and the like, are employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. When referencing salts, the phrases “pharmaceutically acceptable salt,” “physiologically acceptable salt,” and the like, means a salt which is acceptable for administration to a patient, such as a mammal (salts with counterions having acceptable mammalian safety for a given dosage regime). As is well known in the art, such salts can be derived from pharmaceutically acceptable inorganic or organic bases, by way of example, sodium, potassium, calcium, magnesium, lithium, aluminum, zinc, and ammonium and tetraalkylammonium salts (e.g., salts formed with pharmaceutically acceptable amines such as ammonia, alkylamines, hydroxyalkylamines, lysine, arginine, N- methylglucamine, procaine, etc.), and the like, and when the molecule contains a basic functionality, addition salts with inorganic acids, such as hydrochloride, hydrobromide, sulfate, sulfamate, phosphate, nitrate, perchlorate salts, and the like, and addition salts with organic acids, such as formate, tartrate, besylate, mesylate, acetate, maleate, malonate, oxalate, fumarate, benzoate, salicylate, succinate, oxalate, glycolate, hemi-oxalate, hemi-fumarate, propionate, stearate, lactate, citrate, ascorbate, pamoate, hydroxymaleate, phenylacetate, glutamate, 2-acetoxybenzoate, tosylate, ethanedisulfonate, isethionate salts, and the like. The term “salt thereof’ means a compound formed when a proton of an acid is replaced by a cation, such as a metal cation or an organic cation and the like. Where applicable, the salt is a pharmaceutically acceptable salt, although this is not required for salts of intermediate compounds that are not intended for administration to a patient. By way of example, salts of the present compounds include those wherein the compound is protonated by an inorganic or organic acid to form a cation, with the conjugate base of the inorganic or organic acid as the anionic component of the salt.

“Solvate” refers to a physical association of a compound or salt of the present disclosure with one or more solvent molecules, whether organic, inorganic, or a mixture of both. This physical association includes hydrogen bonding. In certain instances, the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. The solvent molecules in the solvate may be present in a regular arrangement and/or a non-ordered arrangement. The solvate may comprise either a stoichiometric or nonstoichiometric amount of the solvent molecules. “Solvate” encompasses both solution-phase and isolable solvates. Some examples of solvents include, but are not limited to, methanol, ethanol, isopropanol, N,N-dimethylformamide, tetrahydrofuran, dimethylsulfoxide, and water. When the solvent is water, the solvate formed is a hydrate (e.g., monohydrate, dihydrate, etc.). Exemplary solvates thus include, but are not limited to, hydrates, methanolates, ethanolates, isopropanolates, etc. Methods of solvation are generally known in the art.

“Stereoisomer” and “stereoisomers” refer to compounds that have same atomic connectivity but different atomic arrangement in space. Stereoisomers include cis-trans isomers, E and Z isomers, enantiomers, and diastereomers. All forms such as racemates and optically pure stereoisomers of the compounds are contemplated herein. Chemical formulas and compounds which possess at least one stereogenic center, but are drawn without reference to stereochemistry, are intended to encompass both the racemic compound, as well as the separate stereoisomers, e.g., R- and/or S-stereoisomers, each permutation of diastereomers so long as those diastereomers are geometrically feasible, etc.

“Tautomer” refers to alternate forms of a molecule that differ only in electronic bonding of atoms and/or in the position of a proton, such as enol-keto, imine-enamine, and neutral/zwitterionic tautomers, or the tautomeric forms of heteroaryl groups containing a -N=C(H)-NH- ring atom arrangement, such as pyrazoles, imidazoles, benzimidazoles, triazoles, and tetrazoles. Other tautomeric ring atom arrangements are also possible. For example, compounds containing an acid and a base group within the same molecule depicted in neutral form may exist also in a zwitterionic form, as is the case for amino acid/ammonium carboxylate tautomers. A given chemical formula or name shall encompass all tautomeric forms thereof, insofar as they exist.

“Prodrug” is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound described herein. Thus, the term “prodrug” refers to a precursor of a biologically active compound that is pharmaceutically acceptable. A prodrug may be inactive when administered to a subject, e.g., an ester, a phosphate ester, etc. but is converted in vivo to an active compound, for example, by hydrolysis to a free carboxylic acid or free hydroxyl group. The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam). A discussion of prodrugs is provided in Higuchi, T., et al., “Pro-drugs as Novel Delivery Systems,” A.C.S. Symposium Series, Vol. 14, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein. The term “prodrug” is also meant to include any covalently bonded carriers, which release the active compound in vivo when such prodrug is administered to a mammalian subject. Prodrugs of an active compound, as described herein, may be prepared by modifying functional groups present in the active compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent active compound. Prodrugs include compounds wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the active compound is administered to a mammalian subject, cleaves to form a free hydroxyl, free amino or free mercapto group, respectively. Examples of prodrugs include, but are not limited to, ester (e.g., acetate, formate, benzoate, etc.), carbonate, carbamate, and dihydrogen phosphate derivatives of an alcohol, or amide (e.g., acetamide, formamide, benzamide, amides formed from amino acids, etc.), carbamate, etc. derivatives of an amine functional group in the active compound, and the like.

Compounds of the present disclosure may in some cases exist in a crystalline solid form or an amorphous solid form, and as such, these solid forms are contemplated herein. A “crystalline” solid is a type of solid whose fundamental three-dimensional structure contains a highly regular pattern of atoms or molecules — with long range order — forming a crystal lattice, and thus displays sharp characteristic crystalline peak(s) in its X-ray power diffraction (XRPD) pattern. In some instances, crystalline solids can exist in different crystalline forms known as “polymorphs,” which have the same chemical composition, but differ in packing, geometric arrangement, and other descriptive properties of the crystalline solid state. As such, polymorphs may have different solid-state physical properties to affect, for example, the solubility, dissolution rate, bioavailability, chemical and physical stability, flowability, and compressibility, etc. of the compound as well as the safety and efficacy of drug products based on the compound. In the process of preparing a polymorph, further purification, in terms of gross physical purity or optical purity, may be accomplished as well. As used herein, the term “amorphous” refers to a solid material having substantially no long range order in the position of its molecules — the molecules are arranged in a random manner so that there is effectively no well-defined arrangement, e.g., molecular packing, and no long range order. Amorphous solids are generally isotropic, i.e., exhibit similar properties in all directions and do not have definite melting points. For example, an amorphous material is a solid material having substantially no sharp characteristic crystalline peak(s) in its X-ray power diffraction (XRPD) pattern (i.e., is not crystalline as determined by XRPD). Instead, one or several broad peaks (e.g., halos) appear in its XRPD pattern. Broad peaks are characteristic of an amorphous solid. Thus, an “amorphous” subject compound/material is one characterized as having substantially no crystallinity — less than 10% crystallinity, less than 8% crystallinity, less than 6% crystallinity, less than 4% crystallinity, less than 2% crystallinity, less than 1% crystallinity, or 0% crystallinity — i.e., is at least 90%, at least 92%, at least 94%, at least 96%, at least 98%, or 100% amorphous, as determined for example by XRPD. For example, the % crystallinity can in some embodiments be determined by measuring the intensity of one or more peaks in the XRPD diffractogram compared to a reference peak, which may be that of an internal standard. Other characterization techniques, such as modulated differential scanning calorimetry (mDSC) analysis, Fourier transform infrared spectroscopy (FTIR), and other quantitative methods, may also be employed to determine the percent a subject compound/material is amorphous or crystalline, including quantitative methods which provide the above percentages in terms of weight percent.

It will be appreciated that the compounds herein can exist in different salt, solvate, stereoisomer, tautomer, crystalline/amorphous (including polymorphic) forms, and the present disclosure is intended to include all permutations thereof, such as a solvate of a pharmaceutically acceptable salt of a stereoisomer of the subject compound.

A “vapor” is a solid substance in the gas phase at a temperature lower than its critical temperature, meaning that the vapor can be condensed to a liquid by increasing the pressure on it without reducing the temperature.

An “aerosol”, as used herein, is a suspension of fine solid particles or liquid droplets in a gas phase (e.g., air, oxygen, helium, nitrous oxide, and other gases, as well as mixtures thereof). A “mist”, as used herein, is a subset of aerosols, differing from a vapor, and is a dispersion of liquid droplets (liquid phase) suspended in the gas phase (e.g., air, oxygen, helium, and mixtures thereof). The liquid droplets of an aerosol or mist can comprise a drug moiety dissolved in an aqueous liquid, organic solvent, or a mixture thereof. The gas phase of an aerosol or mist can comprise air, oxygen, helium, or other gases, including mixtures thereof. Mists do not comprise solid particulates. Aerosols and mists of the present disclosure can be generated by any suitable methods and devices, examples of which are set forth herein, e.g., through use of an inhaler or nebulizer.

As used herein, the term “inhalation session” describes a dosing event whereby the subject inhales a given dose of drug, irrespective of the number of breadths needed to inhale the given dose. For example, a subject prescribed to take 10 mg of a drug twice a day would undertake two inhalation sessions, each inhalation session providing 10 mg of the drug. The length of time and the number of breaths for each inhalation session would be dependent on factors such as the inhalation device used, the amount of drug that is drawn per breath, the concentration of the drug in the dosage form, the subject’s breathing pattern, etc.

As used herein, the language “release period” describes the time window in which any compound described herein is released from the dosage form (e.g., the matrix) to afford plasma concentrations of compounds described herein. The start time of the release period is defined from the point of administration to a subject, which for oral administration is considered nearly equivalent to entry into the stomach, and initial dissolution by gastric enzymes and acid.

As used herein, the language “maximum sustained release” describes the release window for certain formulations of the present disclosure formulated to increase the release period to a maximum value, which for enteral routes is ultimately limited by the time the gastrointestinal tract naturally excretes all drugs with food.

The language “tamper resistance” is art-recognized to describe aspects of a drug formulation that make it more difficult to use the formulation to abuse the drug moiety of the formulation through extraction for intravenous use, or crushing for freebase use; and therefore reduce the risk for abuse of the drug.

As used herein, the term “steady” describes the stable or steady-state level of a molecule concentration, e.g., concentration of any compound described herein.

As used herein, the term “composition” is equivalent to the term “formulation.”

As used herein, the language “administration event” describes the administration of a given dose to a subject within a short window of time, e.g., less than 10 minutes. An oral administration event may be in the form of administration of, for example, one or more pills within a short window of time.

The term “treating” or “treatment” as used herein means the treating or treatment of a disease or medical condition in a patient, such as a mammal (particularly a human) that includes: ameliorating the disease or medical condition, such as, eliminating or causing regression of the disease or medical condition in a patient; suppressing the disease or medical condition, for example by, slowing or arresting the development of the disease or medical condition in a patient; or alleviating a symptom of the disease or medical condition in a patient. In some embodiments, prophylactic treatment can result in preventing the disease or medical condition from occurring, in a subject.

A “patient” or “subject,” used interchangeably herein, can be any mammal including, for example, a human or a non-human subject. A patient or subject can have a condition to be treated or can be susceptible to a condition to be treated.

As used herein, and unless otherwise specified, the terms “prevent,” “preventing” and “prevention” refer to the prevention of the onset, recurrence or spread of a disease, disorder, or condition, or of one or more symptoms thereof. The terms encompass the inhibition or reduction of a symptom of the particular disease, disorder, or condition. Subjects with familial history of a disease, disorder, or condition, in particular, are candidates for preventive regimens in certain embodiments. In addition, subjects who have a history of recurring symptoms are also potential candidates for the prevention. In this regard, the term “prevention” may be interchangeably used with the term “prophylactic treatment.”

As used herein, and unless otherwise specified, the terms “manage,” “managing” and “management” refer to preventing or slowing the progression, spread or worsening of a disease, disorder, or condition, or of one or more symptoms thereof. Often, the beneficial effects that a subject derives from a prophylactic and/or therapeutic agent do not result in a cure of the disease, disorder, or condition. In this regard, the term “managing” encompasses treating a subject who had suffered from the particular disease, disorder, or condition in an attempt to prevent or minimize the recurrence of the disease, disorder, or condition, or of one or more symptoms thereof.

“Therapeutically effective amount” refers to an amount of a compound sufficient to treat a specified disorder or disease or one or more of its symptoms and/or to prevent the occurrence of the disease or disorder (prophylactically effective amount).

As used herein, and unless otherwise specified, a “prophylactically effective amount” of an active agent, is an amount sufficient to prevent a disease, disorder, or condition, or prevent its recurrence. The term “prophylactically effective amount” can encompass an amount that improves overall prophylaxis or enhances the prophylactic efficacy of another prophylactic agent.

The term “administration schedule” is a plan in which the type, amount, period, procedure, etc. of the drug in the drug treatment are shown in time series, and the dosage, administration method, administration order, administration date, and the like of each drug are indicated. The date specified to be administered is determined before the start of the drug administration. The administration is continued by repeating the course with the set of administration schedules as “courses”. A “continuous” administration schedule means administration every day without interruption during the treatment course. If the administration schedule follows an “intermittent” administration schedule, then days of administration may be followed by “rest days” or days of non-administration of drug within the course. A “drug holiday” indicates that the drug is not administered in a predetermined administration schedule. For example, after undergoing several courses of treatment, a subject may be prescribed a regulated drug holiday as part of the administration schedule, e.g., prior to re -recommencing active treatment.

The language “toxic spikes” is used herein to describe spikes in concentration of any compound described herein that would produce neurological side-effects of sedation or psychotomimetic effects, (e.g., hallucination, dizziness, and nausea), or any unwanted and/or unintended secondary effects caused by the administration of a medicament to an individual resulting in subjective experiences being qualitatively different from those of ordinary consciousness. These experiences can include derealization, depersonalization, hallucinations and/or sensory distortions in the visual, auditory, olfactory, tactile, proprioceptive and/or interoceptive spheres and/or any other perceptual modifications, and/or any other substantial subjective changes in cognition, memory, emotion and consciousness. Such side effects, when unwanted, unintended, and/or severe, can not only have immediate repercussions, but also effect treatment compliance. In particular, side effects may become more pronounced at blood concentration levels of about 250, 300, 400, 500 ng/L or more.

As used herein, and unless otherwise specified, a “neuropsychiatric disease or disorder” is a behavioral or psychological problem associated with a known neurological condition, and typically defined as a cluster of symptoms that co-exist. Examples of neuropsychiatric disorders include, but are not limited to, schizophrenia, cognitive deficits in schizophrenia, attention deficit disorder, attention deficit hyperactivity disorder, bipolar and manic disorders, depression or any combinations thereof.

“Inflammatory conditions or inflammatory disease,” as used herein, refers broadly to chronic or acute inflammatory diseases. Inflammatory conditions and inflammatory diseases, include but are not limited to persistent symptoms from a SARS-CoV-2 infection (COVID-19), e.g. “long covid,” rheumatic diseases (e.g., rheumatoid arthritis, osteoarthritis, psoriatic arthritis) spondyloarthropathies (e.g., ankylosing spondylitis, reactive arthritis, Reiter's syndrome), crystal arthropathies (e.g., gout, pseudogout, calcium pyrophosphate deposition disease), multiple sclerosis, Lyme disease, polymyalgia rheumatica; connective tissue diseases (e.g., systemic lupus erythematosus, systemic sclerosis, polymyositis, dermatomyositis, Sjogren's syndrome); vasculitides (e.g., polyarteritis nodosa, Wegener's granulomatosis, Churg-Strauss syndrome); inflammatory conditions including consequences of trauma or ischaemia, sarcoidosis; vascular diseases including atherosclerotic vascular disease, atherosclerosis, and vascular occlusive disease (e.g., atherosclerosis, ischaemic heart disease, myocardial infarction, stroke, peripheral vascular disease), and vascular stent restenosis; ocular diseases including uveitis, corneal disease, iritis, iridocyclitis, glaucoma, and cataracts.

All diseases and disorders listed herein may be defined as described in the Diagnostic and Statistical Manual of Mental Disorders (DSM-5), published by the American Psychiatric Association, or in International Classification of Diseases (ICD), published by the World Health Organization.

As used herein, and unless specified otherwise, compounds which provide a “psychedelic” effect may also include those compounds which are “entactogenic,” i.e., compounds that produce experiences of emotional communion, oneness, relatedness, emotional openness — that is, empathy or sympathy — as particularly observed and reported for experiences with 3,4-methylenedioxymethamphetamine (MDMA).

As used herein, the term “and/or” includes any and all combinations of one or more of the associated listed items. As used in the description herein and throughout the claims that follow, the meaning of “a”, “an”, and “the” includes plural reference as well as the singular reference unless the context clearly dictates otherwise. The term “about” in association with a numerical value means that the value varies up or down by 5%. For example, for a value of about 100, means 95 to 105 (or any value between 95 and 105).

Compounds

The inventors have identified novel phenethylamine type compounds based on specific molecular modifications that demonstrate preferential binding to G-protein coupled receptors (GPCRs), e.g., 5-HT2 receptors, that are bioavailable (e.g., orally bioavailable), have improved exposure (i.e., prevention of high drug concentrations (spiking) observed acutely after administration), and that possess advantageous enzymatic degradation profiles which prevent bioactivation into toxic metabolites. As a result, the disclosed compounds may have reduced side effects, toxicity, and interpatient variability, thereby improving the therapeutic window and enabling practical use in clinical settings. The novel phenethylamine type compounds are based on specific molecular modifications which slow or shunt enzymatic degradation at specific sites and/or which introduce metabolic soft spots at other sites — modifications which have been identified only after significant studies. Formula (I) Disclosed herein is a compound according to Formula (I): or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, wherein: X 1 and X 2 are independently hydrogen, deuterium, or a substituted or unsubstituted C 1 -C 6 alkyl; Y 1 and Y 2 are independently hydrogen, deuterium, or a substituted or unsubstituted C 1 -C 6 alkyl; R 2 and R 3 are independently hydrogen, deuterium, halogen, a substituted or unsubstituted C 1 -C 6 alkyl, -OR a , or -SR a ; R 4 and R 5 are independently hydrogen, deuterium, halogen, a substituted or unsubstituted C1-C6 alkyl, -OR a , -SR a , or -SeR a ; or R 4 and R 5 together with the atoms attached thereto are optionally joined to form a heterocycloalkyl or heteroaryl; R 6 and R 7 are independently hydrogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or alternatively R 6 and R 7 together with the nitrogen atom attached thereto are optionally joined to form a substituted or unsubstituted heterocycloalkyl; each R a is independently hydrogen, deuterium, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C 3 -C 10 cycloalkyl; and wherein at least one of conditions (i)-(iii) are met (i) at least one of X 1 , X 2 , Y 1 , Y 2 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 comprises deuterium, (ii) R 4 and R 5 together with the atoms attached thereto are joined to form a heterocycloalkyl or heteroaryl comprising deuterium or fluorine, and/or a benzo[d][1,3]oxathiole group, (iii) R 4 is -OR a , -SR a , or -SeR a , with R a in R 4 being a C1-C6 alkyl substituted with one or more halogen; and with the proviso that when X 1 , X 2 , Y 1 , and Y 2 are each hydrogen or deuterium, both R 2 and R 5 are not -OR a . X 1 and X 2 may be the same, or different. In some embodiments, X 1 and X 2 are the same. In some embodiments, X 1 and X 2 are hydrogen. In some embodiments, X 1 and X 2 are deuterium. In some embodiments, X 1 and X 2 are different. In some embodiments, X 1 is hydrogen or deuterium, and X 2 is a substituted or unsubstituted C1-C6 alkyl. In some embodiments, X 2 is an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, and n-propyl, preferably methyl. In some embodiments, X 2 is a substituted C1-C6 alkyl. The alkyl group may contain one, or more than one, substituent. For example, when the alkyl group is a C1 alkyl group (i.e., methyl group), the substituted C 1 alkyl group may be -CDH 2 , -CD 2 H, -CD 3 , -CFH 2 , -CF 2 H, -CF 3 , etc. In some embodiments, one of X 1 and X 2 is deuterium while the other is hydrogen. Y 1 and Y 2 may be the same, or different. In some embodiments, Y 1 and Y 2 are the same. In some embodiments, Y 1 and Y 2 are hydrogen. In some embodiments, Y 1 and Y 2 are deuterium. In some embodiments, Y 1 and Y 2 are different. In some embodiments, one of Y 1 and Y 2 is deuterium while the other is hydrogen. In some embodiments, Y 1 is a substituted or unsubstituted C1-C6 alkyl. In some embodiments, Y 2 is a substituted or unsubstituted C1-C6 alkyl. In some embodiments, R 2 is deuterium. In some embodiments, R 2 is hydrogen. In some embodiments, R 2 is halogen, for example -Br, -F, -Cl, or -I. In some embodiments, R 2 is a an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R 2 is a substituted C1-C6 alkyl. When R 2 is a substituted C1-C6 alkyl, preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl or polyether substituents, etc. The alkyl group may contain one, or more than one, substituent. For example, when the alkyl group is a C 1 alkyl group (i.e., methyl group), the substituted C 1 alkyl group may be - CDH 2 , -CD 2 H, -CD 3 , -CFH 2 , -CF 2 H, -CF 3 , etc. In some embodiments, R 2 is -OR a . In some embodiments, R 2 is -SR a . In some embodiments, R 3 is deuterium. In some embodiments, R 3 is hydrogen. In some embodiments, R 3 is halogen, for example -Br, -F, -Cl, or -I. In some embodiments, R 3 is a an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R 3 is a substituted C1-C6 alkyl. When R 3 is a substituted C1-C6 alkyl, preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl or polyether substituents, etc. The alkyl group may contain one, or more than one, substituent. For example, when the alkyl group is a C1 alkyl group (i.e., methyl group), the substituted C1 alkyl group may be - CDH 2 , -CD 2 H, -CD 3 , -CFH 2 , -CF 2 H, -CF 3 , etc. In some embodiments, R 3 is -OR a . In some embodiments, R 3 is -SR a . In some embodiments, R 4 is deuterium. In some embodiments, R 4 is hydrogen. In some embodiments, R 4 is halogen, for example -Br, -F, -Cl, or -I. In some embodiments, R 4 is an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R 4 is a substituted C1-C6 alkyl. When R 4 is a substituted C1-C6 alkyl, preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl or polyether substituents, cycloalkyl, etc. The alkyl group may contain one, or more than one, substituent. For example, when the alkyl group is a C 1 alkyl group (i.e., methyl group), the substituted C 1 alkyl group may be -CDH 2 , -CD 2 H, -CD 3 , -CFH 2 , -CF 2 H, -CF 3 , etc. In some embodiments, R 4 is -OR a , SR a , or -SeR a , wherein R a in R 4 is hydrogen, deuterium, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl. In some embodiments, R 4 is -OR a . In some embodiments, R 4 is -SR a . In some embodiments, R 4 is -SeR a . In some embodiments, R a in R 4 is hydrogen. In some embodiments, R a in R 4 is deuterium. In some embodiments, R a in R 4 is a substituted or unsubstituted C1-C6 alkyl. In some embodiments, R a in R 4 is an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R a in R 4 is a substituted C1-C6 alkyl. Preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), cyano, polar substituents such as hydroxyl or polyether substituents, cycloalkyl, etc. The C1-C6 alkyl group may contain one, or more than one, substituent. In some embodiments, R a in R 4 is a substituted C 1 alkyl group, examples of which may include, but are not limited to, -CDH 2 , -CD 2 H, -CD 3 , -CFH 2 , - CF 2 H, -CF 3 , and -CH 2 CmB. In some embodiments, R a in R 4 is a substituted C 2 alkyl group, examples of which may include, but are not limited to, -CDHCDH 2 , -CDHCD 2 H, -CD 2 CD 3 , -CH 2 CFH 2 , -CH 2 CF 2 H, -CH 2 CF 3 , and -CH2CH2CmB. In some embodiments, R a in R 4 is not a substituted C2 alkyl group such as a C2 fluoroalkyl group. In some embodiments, R a in R 4 is a substituted C3 alkyl group, examples of which may include, but are not limited to -CH2CH2CF3, -CH2CH2CF2H, -CH2CH2CFH2, -CH2CF2CF2H, and -CH2CH2CH2CmB. In some embodiments, R a in R 4 is a substituted or unsubstituted alkenyl, e.g., a substituted or unsubstituted allyl, butenyl, crotyl, etc. In some embodiments, R a in R 4 is a substituted or unsubstituted alkynyl, e.g., a substituted or unsubstituted acetylenyl, propargyl, homopropargyl, etc. In some embodiments, R a in R 4 is an unsubstituted alkynyl. In some embodiments, in R 4 is -CH 2 CH 2 CH 2 Cm8=. In some embodiments, R a in R 4 is -CH 2 CH 2 CH 2 CH 2 Cm8=. In some embodiments, R a in R 4 is a substituted alkynyl. In some embodiments, R a in R 4 is a substituted propargyl (e.g., -CF2Cm8=$( In some embodiments, R a in R 4 is -CF2CH2Cm8=. In some embodiments, R a in R 4 is -CF2CH2CH2Cm8=. In some embodiments, R a in R 4 is -CF2CH2CH2CH2Cm8=. In some embodiments, R a in R 4 is a substituted or unsubstituted cycloalkyl, for example a substituted or unsubstituted C3-C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted C5-C6 cycloalkyl. In some embodiments, R a in R 4 is an unsubstituted cycloalkyl (e.g., an unsubstituted C 3 -C 10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, R a in R 4 is a substituted cycloalkyl (e.g., a substituted C 3 -C 10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 4 is -SMe, -SCD3, -SCF3, -SCF2H, -SCFH2, -SEt, -Sn-Pr, -SCH2CF3, -SCH2CF2H, -SCH2CFH2, -SCH2CH2CF3, -SCH2CH2CF2H, -SCH2CH2CFH2, -SCH2CF2CF2H, -SCH2Cm8=, -SCm8=, -SCF2Cm8=, -SCH2CH2CH2Cm8=, -SCF2CH2CH28m8=& -Me, -CD3, -CF3, -t-Bu, -C(CD3)3, -OMe, -OCD3, -OCF3, -OCF2H, -OCFH2, -OCH2CF3, -OCH2CF2H, -OCH2CFH2, -OCH2CH2CF3, -OCH2CH2CF2H, -OCH 2 CH 2 CFH 2 , -OCH 2 CF 2 CF 2 H, -OCH 2 Cm8=, -OCm8=, -CH 2 CH 2 CH 2 CH 2 Cm8=, -CF 2 CH 2 CH 2 CH 2 8m8=& '8U& '>& '7[& 'FOAO& 'FO89 3, -SeCF 3, -SeCF 2 H, -SeCFH 2 , -SeEt, -Sen-Pr, -SeCH 2 CF 3 , -SeCH 2 CF 2 H, -SeCH 2 CFH 2 , -SeCH 2 CH 2 CF 3 , -SeCH 2 CH 2 CF 2 H, -SeCH 2 CH 2 CFH 2 , -SeCH 2 CF 2 CF 2 H, -SeCH 2 Cm8=, -SeCm8=, -SeCF 2 Cm8=, -SeCH 2 CH 2 CH 2 8m8=& X[ 'FO8; 2 CH 2 CH 2 8m8=( In some embodiments, R 5 is deuterium. In some embodiments, R 5 is hydrogen. In some embodiments, R 5 is halogen, for example -Br, -F, -Cl, or -I. In some embodiments, R 5 is an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R 5 is a substituted C1-C6 alkyl. When R 5 is a substituted C1-C6 alkyl, preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl or polyether substituents, cycloalkyl, etc. The alkyl group may contain one, or more than one, substituent. For example, when the alkyl group is a C 1 alkyl group (i.e., methyl group), the substituted C1 alkyl group may be -CDH2, -CD 2 H, -CD 3 , -CFH 2 , -CF 2 H, -CF 3 , etc. In some embodiments, R 5 is -OR a , SR a , or -SeR a , wherein R a in R 5 is hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl. In some embodiments, R 5 is -OR a . In some embodiments, R 5 is -SR a . In some embodiments, R 5 is -SeR a . In some embodiments, R a in R 5 is hydrogen. In some embodiments, R a in R 5 is deuterium. In some embodiments, R a in R 5 is a substituted or unsubstituted C1-C6 alkyl. In some embodiments, R a in R 5 is an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R a in R 5 is a substituted C 1 -C 6 alkyl. Preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), cyano, polar substituents such as hydroxyl or polyether substituents, etc. The C 1 -C 6 alkyl group may contain one, or more than one, substituent. In some embodiments, R a in R 5 is a substituted C 1 alkyl group, examples of which may include, but are not limited to, -CDH2, -CD2H, -CD3, -CFH2, -CF2H, - CF3, and -CH28mB( >W \XVO OVLXNSVOW]\& E a in R 5 is a substituted C2 alkyl group, examples of which may include, but are not limited to, -CDHCDH2, -CDHCD2H, -CD2CD3, -CH2CFH2, -CH2CF2H, - CH2CF3, and -CH2CH28mB( >W \XVO OVLXNSVOW]\& E a in R 5 is not a substituted C2 alkyl group such as a C2 fluoroalkyl group. In some embodiments, R a in R 5 is a substituted C3 alkyl group, examples of which may include, but are not limited to -CH2CH2CF3, -CH2CH2CF2H, -CH2CH2CFH2, -CH2CF2CF2H, and -CH2CH2CH2C mB . In some embodiments, R a in R 5 is a substituted or unsubstituted alkenyl, e.g., a substituted or unsubstituted allyl, butenyl, crotyl, etc. In some embodiments, R a in R 5 is a substituted or unsubstituted alkynyl, e.g., a substituted or unsubstituted acetylenyl, propargyl, homopropargyl, etc. In some embodiments, R a in R 5 is an unsubstituted alkynyl. In some embodiments, R a in R 5 is an unsubstituted acetylenyl (-Cm8=$( >W \XVO OVLXNSVOW]\& R a in R 5 is an unsubstituted propargyl (-CH 2 8m8=$( >W \XVO OVLXNSVOW]\& E a in R 5 is -CH 2 CH 2 8m8=( >W \XVO OVLXNSVOW]\& E a in R 5 is -CH2CH2CH2Cm8=. In some embodiments, R a in R 5 is -CH2CH2CH2CH2Cm8=. In some embodiments, R a in R 5 is a substituted alkynyl. In some embodiments, R a in R 5 is a substituted propargyl (e.g., -CF28m8=$( >W \XVO OVLXNSVOW]\& E a in R 5 is -CF2CH28m8=( >W \XVO OVLXNSVOW]\& E a in R 5 is -CF2CH2CH2Cm8=. In some embodiments, R a in R 5 is -CF2CH2CH2CH2Cm8=. In some embodiments, R a in R 5 is a substituted or unsubstituted cycloalkyl, for example a substituted or unsubstituted C3-C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted C5-C6 cycloalkyl. In some embodiments, R a in R 5 is an unsubstituted cycloalkyl (e.g., an unsubstituted C 3 -C 10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, R a in R 5 is a substituted cycloalkyl (e.g., a substituted C 3 -C 10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 5 is -SMe, -SCD3, -SCF3, -SCF2H, -SCFH2, -SEt, -Sn-Pr, -SCH 2 CF 3 , -SCH 2 CF 2 H, -SCH 2 CFH 2 , -SCH 2 CH 2 CF 3 , -SCH 2 CH 2 CF 2 H, -SCH 2 CH 2 CFH 2 , -SCH 2 CF 2 CF 2 H, -SCH 2 Cm8=, -SCm8=, -SCF 2 Cm8=, -SCH 2 CH 2 CH 2 Cm8=, -SCF 2 CH 2 CH 2 8m8=& -Me, -CD 3 , -CF 3 , -t-Bu, -C(CD 3 ) 3 , -OMe, -OCD 3 , -OCF 3 , -OCF 2 H, -OCFH 2 , -OCH 2 CF 3 , -OCH 2 CF 2 H, -OCH 2 CFH 2 , -OCH 2 CH 2 CF 3 , -OCH 2 CH 2 CF 2 H, -OCH 2 CH 2 CFH 2 , -OCH 2 CF 2 CF 2 H, -OCH 2 Cm8=, -OCm8=, -CH 2 CH 2 CH 2 CH 2 Cm8=, -CF2CH2CH2CH28m8=& '8U& '>& '7[& 'FOAO& 'FO893, -SeCF3, -SeCF2H, -SeCFH2, -SeEt, -Sen-Pr, -SeCH2CF3, -SeCH2CF2H, -SeCH2CFH2, -SeCH2CH2CF3, -SeCH2CH2CF2H, -SeCH2CH2CFH2, -SeCH2CF2CF2H, -SeCH2Cm8=, -SeCm8=, -SeCF2Cm8=, -SeCH2CH2CH28m8=& X[ 'FO8;2CH2CH28m8=( In some embodiments, R 4 and R 5 together with the atoms attached thereto are joined to form a heterocycloalkyl or heteroaryl, with specific mention being made to a benzo[d][1,3]oxathiole group or a benzo[d][1,3]dioxole group. In embodiments where R 4 and R 5 together with the atoms attached thereto are joined to form a benzo[d][1,3]oxathiole group or a benzo[d][1,3]dioxole group, either the oxathiole ring or the dioxole ring may be further substituted with substituents as defined herein, e.g., with deuterium substituents, with halogen (e.g., fluorine) substituents, etc. R 6 and R 7 may be the same, or different. In some embodiments, R 6 and R 7 are the same. For example, in some embodiments, both R 6 and R 7 are hydrogen. In some embodiments, R 6 and R 7 are different. For example, in some embodiments, R 6 is hydrogen, and R 7 is a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl. R 6 and R 7 may be, independently, hydrogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl. In some embodiments, R 6 and R 7 may be, independently, hydrogen, an unsubstituted C1-C6 alkyl (e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, and hexyl) or a C 1 -C 6 alkyl substituted with one or more deuterium (e.g., -CDH2, -CD2H, -CD3). In some embodiments, R 6 and/or R 7 is an unsubstituted C 1 -C 6 alkyl, for example, an unsubstituted C1 alkyl, an unsubstituted C2 alkyl, an unsubstituted C3 alkyl, an unsubstituted C4 alkyl, an unsubstituted C5 alkyl, or an unsubstituted C6 alkyl. In some embodiments, R 6 and/or R 7 is an unsubstituted linear C2-C6 alkyl. In some embodiments, R 6 and/or R 7 is an unsubstituted branched C3- C10 alkyl. Examples of an unsubstituted C1-C6 alkyl include, but are not limited to, methyl, ethyl, n- propyl, isopropyl, butyl, isobutyl, t-butyl, n-pentyl, isopentyl, n-hexyl, and isohexyl. In some embodiments, R 6 and/or R 7 is a substituted C1-C6 alkyl, e.g., a substituted C1 alkyl, a substituted C 2 alkyl, a substituted C 3 alkyl, a substituted C 4 alkyl, a substituted C 5 alkyl, or a substituted C 6 alkyl. The alkyl group may contain one, or more than one, substituent. The alkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl, oxo, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), unsubstituted alkenyl, substituted alkenyl, unsubstituted alkynyl, substituted alkynyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. In some embodiments, R 6 and/or R 7 is a C1-C6 alkyl substituted with one or more deuterium atoms, examples of which include, but are not limited to, -CDH2, -CD2H, -CD3, -CD2CD3, and -CD2CD2CD3. In some embodiments, R 6 and/or R 7 is a C1-C6 alkyl substituted with one or more fluorine atoms, i.e., is a fluoroalkyl group. Examples of fluoroalkyl groups include, but are not limited to, -CH2F, -CHF2, -CF3, -CH2CH2F, -CH2CHF2, -CH2CF3, -CH2CH2CH2F, -CH2CH2CHF2, -CH2CH2CF3, -CH2CH2CH2CH2F, -CH 2 CH 2 CH 2 CHF 2 , -CH 2 CH 2 CH 2 CF 3 , -CH 2 CF 2 CHF 2 , -CH 2 CF 2 CF 3 , -CH(CF 3 ) 2 , and -CH(CH 3 )CF 3 . In some embodiments, R 6 and/or R 7 is a C 1 -C 6 alkyl substituted with one or more deuterium atoms and one or more fluorine atoms, examples of which include, but are not limited to, - CD 2 CH 2 F, -CD 2 CHF 2 , -CD 2 CF 3 , -CD 2 CH 2 CH 2 F, -CD 2 CH 2 CHF 2 , -CD 2 CH 2 CF 3 , -CD 2 CD 2 CH 2 , -CD 2 CD 2 CHF 2 , -CD 2 CD 2 CF 3 , -CD 2 CH 2 CH 2 CH 2 F, -CD 2 CH 2 CH 2 CHF 2 , -CD2CH2CH2CF3, -CD2CD2CH2CH2F, -CD2CD2CH2CHF2, -CD2CD2CH2CF3, -CD2CD2CD2CH2F, -CD2CD2CD2CHF2, and -CD2CD2CD2CF3. In some embodiments, R 6 and/or R 7 is a C1-C6 alkyl substituted with a substituted or unsubstituted cycloalkyl. The C1-C6 alkyl may be substituted with, e.g., a substituted or unsubstituted C3-C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted C5-C6 cycloalkyl. In some embodiments, the C1-C6 alkyl is substituted with an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, the C1-C6 alkyl is substituted with a substituted cycloalkyl (e.g., a substituted C3-C10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 6 and/or R 7 is a C1 alkyl substituted with a substituted or unsubstituted cycloalkyl, with particular mention being made to cyclopropylmethyl (-CH 2 C 3 H 5 ). In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted alkenyl, e.g., a substituted or unsubstituted allyl, butenyl, crotyl, etc. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted alkynyl, e.g., a substituted or unsubstituted propargyl. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted cycloalkyl, for example a substituted or unsubstituted C3-C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted C5-C6 cycloalkyl. In some embodiments, R 6 and/or R 7 is an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, R 6 and/or R 7 is a substituted cycloalkyl (e.g., a substituted C3-C10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted heterocycloalkyl. In some embodiments, the unsubstituted or substituted heterocycloalkyl group may be a 3-membered ring, a 4- membered ring, a 5-membered ring, a 6-membered ring, a 7-membered ring, or an 8-membered ring. In some embodiments, R 6 and/or R 7 is an unsubstituted heterocycloalkyl, such as those set forth herein, examples of which include, but are not limited to, aziridine, azetidine, pyrrolidine, isoindole, indole, dihydroindole, indazole, purine, carbazole, carboline, imidazolidine, imidazoline, piperidine, piperazine, indoline, phthalimide, 1,2,3,4-tetrahydroisoquinoline, thiazolidine, morpholine, thiomorpholine, tetrahydrofuran, tetrahydropyran, and 1,3-dioxolane. In some embodiments, R 6 and/or R 7 is a substituted heterocycloalkyl. The substituent(s) may be any recited herein, including, but not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), oxo, and hydroxyl. The heterocycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted aryl. In some embodiments, R 6 and/or R 7 is an unsubstituted aryl, examples of which include, but are not limited to, phenyl and naphthyl. In some embodiments, R 6 and/or R 7 is a substituted aryl. The substituent(s) may be any recited herein, including, but not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The aryl group may contain one, or more than one, substituent. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted heteroaryl. In some embodiments, R 6 and/or R 7 is an unsubstituted heteroaryl, examples of which include, but are not limited to, pyrrolyl, furanyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, imidazolyl, triazolyl, tetrazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, indolyl, benzofuranyl, benzothiophenyl, thiophenyl, benzimidazolyl, quinolinyl, isoquinolinyl, quinazolinyl, quinoxalinyl, and pyrazolyl. In some embodiments, R 6 and/or R 7 is a substituted heteroaryl. The substituent(s) may be any recited herein, including, but not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The heteroaryl group may contain one, or more than one, substituent. In some embodiments, R 6 is hydrogen, and R 7 is a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl. In some embodiments, R 6 is hydrogen, and R 7 is a substituted or unsubstituted C1-C6 alkyl. In some embodiments, R 6 is hydrogen, and R 7 is an unsubstituted C1-C6 alkyl, a C1-C6 alkyl substituted with one or more deuterium atoms, a C1-C6 alkyl substituted with one or more fluorine atoms, or a C1-C6 alkyl substituted with a substituted or unsubstituted cycloalkyl. For example, in some embodiments, R 6 is hydrogen, and R 7 is methyl, ethyl, propyl, -CD3, or cyclopropylmethyl (-CH2C3H5). In some embodiments, R 6 and R 7 together with the nitrogen atom attached thereto are joined to form a substituted or unsubstituted heterocycloalkyl. In some embodiments, R 6 and R 7 together with the nitrogen atom attached thereto are joined to form an unsubstituted heterocycloalkyl. The unsubstituted heterocycloalkyl group may be, e.g., a 3-membered ring, a 4-membered ring, 5-membered ring, a 6- membered ring, a 7-membered ring, an 8-membered ring, etc., which may be optionally fused to other ring(s). The unsubstituted heterocycloalkyl group contains a minimum of one nitrogen ring atom (the nitrogen atom intervening R 6 and R 7 ), and may optionally contain at least one additional hetero-ring atom, which may be one or more of nitrogen, sulfur, or oxygen, for a total of 1, 2, 3, or 4 hetero-ring atoms (at least one of which is a nitrogen ring atom). Examples of unsubstituted heterocyclo alkyl groups formed from joining R 6 and R 7 together with the nitrogen atom attached thereto include, but are not limited to,

In some embodiments, R 6 and R 7 together with the nitrogen atom attached thereto are joined to form a substituted heterocycloalkyl. The substituted heterocycloalkyl group may be, e.g., a 3-membered ring, a 4-membered ring, a 5-membered ring, a 6-membered ring, a 7-membered ring, an 8-membered ring, etc., which may be optionally fused to other ring(s). The substituted heterocycloalkyl group contains a minimum of one nitrogen ring atom (the nitrogen atom intervening R 6 and R 7 ), and may optionally contain additional hetero-ring atoms (e.g., nitrogen, sulfur, or oxygen) for a total of 1, 2, 3, or 4 hetero-ring atoms (at least one of which is a nitrogen ring atom). Examples of the substituted heterocycloalkyl group include, but are not limited to, aziridine, azetidine, pyrrolidine, isoindole, indole, dihydroindole, indazole, purine, carbazole, carboline, imidazolidine, imidazoline, piperidine, piperazine, indoline, 1,2,3,4-tetrahydroisoquinoline, thiazolidine, morpholine, or thiomorpholine, which is substituted with at least one substituent. The substituent(s) may be any recited herein, including, but not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl, oxo, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), unsubstituted alkyl, substituted alkyl, unsubstituted alkenyl, substituted alkenyl, unsubstituted alkynyl, substituted alkynyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The substituted heterocycloalkyl formed from joining R 6 and R 7 together with the nitrogen atom attached thereto contains a heterocycloalkyl group substituted with one, two, three, four, or more substituents. The substituent may be located on a carbon ring atom or on a hetero-ring atom. Examples of substituted heterocycloalkyl groups formed from joining R 6 and R 7 together with the nitrogen atom attached thereto include, but are not limited to, When present in one or more of R 2 to R 5 , each R a may be, independently, hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl. In some embodiments, each R a may be, independently, hydrogen, deuterium, an unsubstituted C 1 -C 6 alkyl (e.g., methyl, ethyl, n- propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl), or a substituted C 1 -C 6 alkyl, with preferred substituents including, but not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl or polyether substituents, etc. In some embodiments, R a is a substituted or unsubstituted C1-C6 alkyl, preferably a C1-C3 alkyl, preferably a substituted or unsubstituted C 1 alkyl, examples of which include, but are not limited to, -CH 3 , -CDH 2 , -CD 2 H, -CD 3 , -CFH2, -CF2H, -CF3. In some embodiments, each R a is -CH3. In some embodiments, each R a is -CD3. In some embodiments, more than one R a is present. In such cases, each R a may be the same, or different. In some embodiments, each R a is the same. In some embodiments, each R a is different, e.g., one R a is - CH3, while another is -CD3. In some embodiments, R a in one or more of R 2 to R 5 is hydrogen. In some embodiments, R a in one or more of R 2 to R 5 is deuterium. In some embodiments, R a in one or more of R 2 to R 5 is a substituted or unsubstituted C1-C6 alkyl. In some embodiments, R a in one or more of R 2 to R 5 is an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n- butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R a in one or more of R 2 to R 5 is a substituted C1-C6 alkyl. Preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), cyano, polar substituents such as hydroxyl or polyether substituents, cycloalkyl, etc. The C1-C6 alkyl group may contain one, or more than one, substituent. In some embodiments, R a in one or more of R 2 to R 5 is a substituted C 1 alkyl group, examples of which may include, but are not limited to, -CDH 2 , -CD 2 H, -CD 3 , -CFH 2 , -CF 2 H, -CF 3 , and -CH 2 8mB( >W \XVO OVLXNSVOW]\& E a in one or more of R 2 to R 5 is a substituted C 2 alkyl group, examples of which may include, but are not limited to, -CDHCDH 2 , -CDHCD 2 H, -CD 2 CD 3 , -CH 2 CFH 2 , -CH 2 CF 2 H, -CH 2 CF 3 , and -CH 2 CH 2 CmB. In some embodiments, R a in one or more of R 2 to R 5 is a substituted C3 alkyl group, examples of which may include, but are not limited to -CH2CH2CF3, -CH2CH2CF2H, -CH2CH2CFH2, -CH2CF2CF2H, and -CH2CH2CH28mB( >W some embodiments, R a in one or more of R 2 to R 5 is a substituted or unsubstituted alkenyl, e.g., a substituted or unsubstituted allyl, butenyl, crotyl, etc. In some embodiments, R a in one or more of R 2 to R 5 is a substituted or unsubstituted alkynyl, e.g., a substituted or unsubstituted acetylenyl, propargyl, homopropargyl, etc. In some embodiments, R a in one or more of R 2 to R 5 is an unsubstituted alkynyl. In some embodiments, R a in one or more of R 2 to R 5 S\ KW ^W\^L\]S]^]ON KMO]bUOWbU #'8m8=$( >W \XVO embodiments, R a in one or more of R 2 to R 5 is an unsubstituted propargyl (-CH2C m8=$( In some embodiments, R a in one or more of R 2 to R 5 is -CH 2 CH 2 Cm8=( In some embodiments, R a in one or more of R 2 to R 5 is -CH 2 CH 2 CH 2 Cm8=. In some embodiments, R a in one or more of R 2 to R 5 is -CH 2 CH 2 CH 2 CH 2 Cm8=. In some embodiments, R a in one or more of R 2 to R 5 is a substituted alkynyl. In some embodiments, R a in one or more of R 2 to R 5 is a substituted propargyl (e.g., -CF 2 8m8=$( >W some embodiments, R a in one or more of R 2 to R 5 is -CF2CH2Cm8=. In some embodiments, R a in one or more of R 2 to R 5 is -CF2CH2CH2Cm8=. In some embodiments, R a in one or more of R 2 to R 5 is -CF2CH2CH2CH28m8=( >W \XVO OVLXNSVOW]\& E a in one or more of R 2 to R 5 is a substituted or unsubstituted cycloalkyl, for example a substituted or unsubstituted C3-C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted C5-C6 cycloalkyl. In some embodiments, R a in one or more of R 2 to R 5 is an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, R a in one or more of R 2 to R 5 is a substituted cycloalkyl (e.g., a substituted C3-C10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In line with the above description for R a , any one or both of R 2 to R 3 may be, independent of each other, -OR a or -SR a , and any one or both of R 4 to R 5 may be, independent of each other, -OR a , -SR a , or -SeR a , examples of which include, but are not limited to, -SMe, -SCD 3, -SCF 3, -SCF 2 H, -SCFH 2 , -SEt, -Sn-Pr, -SCH 2 CF 3 , -SCH 2 CF 2 H, -SCH 2 CFH 2 , -SCH 2 CH 2 CF 3 , -SCH 2 CH 2 CF 2 H, -SCH 2 CH 2 CFH 2 , -SCH 2 CF 2 CF 2 H, -SCH 2 8m8=& 'F8m8=& 'F8; 2 8m8=& -SCH 2 CH 2 CH 2 Cm8=, -SCF 2 CH 2 CH 2 Cm8=, -OMe, -OCD 3 , -OCF 3 , -OCF 2 H, -OCFH 2 , -OCH2CF3, -OCH2CF2H, -OCH2CFH2, -OCH2CH2CF3, -OCH2CH2CF2H, -OCH2CH2CFH2, -OCH2CF2CF2H, -OCH28m8=& 'C8m8=& 'FOAO& 'FO893, -SeCF3, -SeCF2H, -SeCFH2, -SeEt, -Sen-Pr, -SeCH2CF3, -SeCH2CF2H, -SeCH2CFH2, -SeCH2CH2CF3, -SeCH2CH2CF2H, -SeCH2CH2CFH2, -SeCH2CF2CF2H, -SeCH28m8=& 'FO8m8=& -SeCF28m8=& -SeCH2CH2CH28m8=& X[ 'FO8;2CH2CH28m8=( As stated above, any of the above embodiments of the compound of Formula (I) may be provided as long as at least one of conditions (i)-(iii) are met: (i) at least one of X 1 , X 2 , Y 1 , Y 2 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 comprises deuterium, (ii) R 4 and R 5 together with the atoms attached thereto are joined to form a heterocycloalkyl or heteroaryl comprising deuterium or fluorine, and/or a benzo[d][1,3]oxathiole group, (iii) R 4 is -OR a , -SR a , or -SeR a , with R a in R 4 being a C 1 -C 6 alkyl substituted with one or more halogen; and with the proviso that when X 1 , X 2 , Y 1 , and Y 2 are each hydrogen or deuterium, both R 2 and R 5 are not each -OR a . For clarity, compounds in which at least one condition of (i)-(iii) is satisfied, do not require the remaining conditions to be satisfied. For example, compounds in which condition (i) is satisfied do not require conditions (ii) or (iii) to be satisfied. In some embodiments, at least one of X 1 , X 2 , Y 1 , Y 2 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 comprises deuterium. In some embodiments, R 4 and R 5 together with the atoms attached thereto are joined to form a heterocycloalkyl or heteroaryl comprising deuterium or fluorine, and/or a benzo[d][1,3]oxathiole group, which may be optionally substituted e.g., with one or more deuterium and/or one or more halogen (e.g., fluorine). In some embodiments, R 4 is -OR a , -SR a , or -SeR a , with R a in R 4 being a C1-C6 alkyl substituted with one or more halogen (i.e., R 4 is an -O-C1-C6 alkyl group, the alkyl group being substituted with one or more halogen; an -S-C 1 -C 6 alkyl group, the alkyl group being substituted with one or more halogen; or an -Se-C1-C6 alkyl group, the alkyl group being substituted with one or more halogen). Formula (II) In some embodiments, the compound has a structure of Formula (II): or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, wherein: X 1 and X 2 are independently hydrogen, deuterium, or a substituted or unsubstituted C1-C6 alkyl; Y 1 and Y 2 are independently hydrogen, deuterium, or a substituted or unsubstituted C1-C6 alkyl; R 2 and R 3 are independently hydrogen, deuterium, halogen, a substituted or unsubstituted C1-C6 alkyl, -OR a , or -SR a ; R 6 and R 7 are independently hydrogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or alternatively R 6 and R 7 together with the nitrogen atom attached thereto are optionally joined to form a substituted or unsubstituted heterocycloalkyl; each R a is independently hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl; A is O or S; Z 1 and Z 2 are independently hydrogen, deuterium, or fluorine; and when A is O, at least one of X 1 , X 2 , Y 1 , Y 2 , R 2 , R 3 , R 6 , R 7 , Z 1 , Z 2 comprises deuterium, and/or at least one of Z 1 and Z 2 is fluorine. X 1 and X 2 may be the same, or different. In some embodiments, X 1 and X 2 are the same. In some embodiments, X 1 and X 2 are hydrogen. In some embodiments, X 1 and X 2 are deuterium. In some embodiments, X 1 and X 2 are different. In some embodiments, X 1 is hydrogen or deuterium, and X 2 is a substituted or unsubstituted C 1 -C 6 alkyl. In some embodiments, X 2 is an unsubstituted C 1 -C 6 alkyl, examples of which include, but are not limited to, methyl, ethyl, and n-propyl, preferably methyl. In some embodiments, X 2 is a substituted C1-C6 alkyl. The alkyl group may contain one, or more than one, substituent. For example, when the alkyl group is a C1 alkyl group (i.e., methyl group), the substituted C1 alkyl group may be -CDH2, -CD2H, -CD3, -CFH2, -CF2H, -CF3, etc. In some embodiments, one of X 1 and X 2 is deuterium while the other is hydrogen. Y 1 and Y 2 may be the same, or different. In some embodiments, Y 1 and Y 2 are the same. In some embodiments, Y 1 and Y 2 are hydrogen. In some embodiments, Y 1 and Y 2 are deuterium. In some embodiments, Y 1 and Y 2 are different. In some embodiments, one of Y 1 and Y 2 is deuterium while the other is hydrogen. In some embodiments, Y 1 is a substituted or unsubstituted C 1 -C 6 alkyl. In some embodiments, Y 2 is a substituted or unsubstituted C 1 -C 6 alkyl. In some embodiments, R 2 is deuterium. In some embodiments, R 2 is hydrogen. In some embodiments, R 2 is halogen, for example -Br, -F, -Cl, or -I. In some embodiments, R 2 is a an unsubstituted C 1 -C 6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R 2 is a substituted C1-C6 alkyl. When R 2 is a substituted C1-C6 alkyl, preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl or polyether substituents, etc. The alkyl group may contain one, or more than one, substituent. For example, when the alkyl group is a C1 alkyl group (i.e., methyl group), the substituted C1 alkyl group may be - CDH2, -CD2H, -CD3, -CFH2, -CF2H, -CF3, etc. In some embodiments, R 2 is -OR a . In some embodiments, R 2 is -SR a . In some embodiments, R 3 is deuterium. In some embodiments, R 3 is hydrogen. In some embodiments, R 3 is halogen, for example -Br, -F, -Cl, or -I. In some embodiments, R 3 is a an unsubstituted C 1 -C 6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R 3 is a substituted C 1 -C 6 alkyl. When R 3 is a substituted C 1 -C 6 alkyl, preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl or polyether substituents, etc. The alkyl group may contain one, or more than one, substituent. For example, when the alkyl group is a C1 alkyl group (i.e., methyl group), the substituted C1 alkyl group may be - CDH2, -CD2H, -CD3, -CFH2, -CF2H, -CF3, etc. In some embodiments, R 3 is -OR a . In some embodiments, R 3 is -SR a . R 6 and R 7 may be the same, or different. In some embodiments, R 6 and R 7 are the same. For example, in some embodiments, both R 6 and R 7 are hydrogen. In some embodiments, R 6 and R 7 are different. For example, in some embodiments, R 6 is hydrogen, and R 7 is a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl. R 6 and R 7 may be, independently, hydrogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl. In some embodiments, R 6 and R 7 may be, independently, hydrogen, an unsubstituted C1-C6 alkyl (e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, and hexyl) or a C1-C6 alkyl substituted with one or more deuterium (e.g., -CDH2, -CD2H, -CD3). In some embodiments, R 6 and/or R 7 is an unsubstituted C 1 -C 6 alkyl, for example, an unsubstituted C 1 alkyl, an unsubstituted C 2 alkyl, an unsubstituted C 3 alkyl, an unsubstituted C 4 alkyl, an unsubstituted C 5 alkyl, or an unsubstituted C 6 alkyl. In some embodiments, R 6 and/or R 7 is an unsubstituted linear C 2 -C 6 alkyl. In some embodiments, R 6 and/or R 7 is an unsubstituted branched C 3 - C 10 alkyl. Examples of an unsubstituted C 1 -C 6 alkyl include, but are not limited to, methyl, ethyl, n- propyl, isopropyl, butyl, isobutyl, t-butyl, n-pentyl, isopentyl, n-hexyl, and isohexyl. In some embodiments, R 6 and/or R 7 is a substituted C1-C6 alkyl, e.g., a substituted C1 alkyl, a substituted C2 alkyl, a substituted C3 alkyl, a substituted C4 alkyl, a substituted C5 alkyl, or a substituted C6 alkyl. The alkyl group may contain one, or more than one, substituent. The alkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl, oxo, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), unsubstituted alkenyl, substituted alkenyl, unsubstituted alkynyl, substituted alkynyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. In some embodiments, R 6 and/or R 7 is a C 1 -C 6 alkyl substituted with one or more deuterium atoms, examples of which include, but are not limited to, -CDH 2 , -CD 2 H, -CD 3 , -CD 2 CD 3 , and -CD 2 CD 2 CD 3 . In some embodiments, R 6 and/or R 7 is a C 1 -C 6 alkyl substituted with one or more fluorine atoms, i.e., is a fluoroalkyl group. Examples of fluoroalkyl groups include, but are not limited to, -CH2F, -CHF2, -CF3, -CH2CH2F, -CH2CHF2, -CH2CF3, -CH2CH2CH2F, -CH2CH2CHF2, -CH2CH2CF3, -CH2CH2CH2CH2F, -CH2CH2CH2CHF2, -CH2CH2CH2CF3, -CH2CF2CHF2, -CH2CF2CF3, -CH(CF3)2, and -CH(CH3)CF3. In some embodiments, R 6 and/or R 7 is a C1-C6 alkyl substituted with one or more deuterium atoms and one or more fluorine atoms, examples of which include, but are not limited to, - CD2CH2F, -CD2CHF2, -CD2CF3, -CD2CH2CH2F, -CD2CH2CHF2, -CD2CH2CF3, -CD2CD2CH2, -CD2CD2CHF2, -CD2CD2CF3, -CD2CH2CH2CH2F, -CD2CH2CH2CHF2, -CD 2 CH 2 CH 2 CF 3 , -CD 2 CD 2 CH 2 CH 2 F, -CD 2 CD 2 CH 2 CHF 2 , -CD 2 CD 2 CH 2 CF 3 , -CD2CD2CD2CH2F, -CD2CD2CD2CHF2, and -CD2CD2CD2CF3. In some embodiments, R 6 and/or R 7 is a C 1 -C 6 alkyl substituted with a substituted or unsubstituted cycloalkyl. The C1-C6 alkyl may be substituted with, e.g., a substituted or unsubstituted C3-C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted C5-C6 cycloalkyl. In some embodiments, the C1-C6 alkyl is substituted with an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, the C1-C6 alkyl is substituted with a substituted cycloalkyl (e.g., a substituted C3-C10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 6 and/or R 7 is a C1 alkyl substituted with a substituted or unsubstituted cycloalkyl, with particular mention being made to cyclopropylmethyl (-CH2C3H5). In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted alkenyl, e.g., a substituted or unsubstituted allyl, butenyl, crotyl, etc. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted alkynyl, e.g., a substituted or unsubstituted propargyl. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted cycloalkyl, for example a substituted or unsubstituted C 3 -C 10 cycloalkyl, or a substituted or unsubstituted C 4 -C 8 cycloalkyl, or a substituted or unsubstituted C 5 -C 6 cycloalkyl. In some embodiments, R 6 and/or R 7 is an unsubstituted cycloalkyl (e.g., an unsubstituted C 3 -C 10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, R 6 and/or R 7 is a substituted cycloalkyl (e.g., a substituted C 3 -C 10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted heterocycloalkyl. In some embodiments, the unsubstituted or substituted heterocycloalkyl group may be a 3-membered ring, a 4- membered ring, a 5-membered ring, a 6-membered ring, a 7-membered ring, or an 8-membered ring. In some embodiments, R 6 and/or R 7 is an unsubstituted heterocycloalkyl, such as those set forth herein, examples of which include, but are not limited to, aziridine, azetidine, pyrrolidine, isoindole, indole, dihydroindole, indazole, purine, carbazole, carboline, imidazolidine, imidazoline, piperidine, piperazine, indoline, phthalimide, 1,2,3,4-tetrahydroisoquinoline, thiazolidine, morpholine, thiomorpholine, tetrahydrofuran, tetrahydropyran, and 1,3-dioxolane. In some embodiments, R 6 and/or R 7 is a substituted heterocycloalkyl. The substituent(s) may be any recited herein, including, but not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), oxo, and hydroxyl. The heterocycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted aryl. In some embodiments, R 6 and/or R 7 is an unsubstituted aryl, examples of which include, but are not limited to, phenyl and naphthyl. In some embodiments, R 6 and/or R 7 is a substituted aryl. The substituent(s) may be any recited herein, including, but not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The aryl group may contain one, or more than one, substituent. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted heteroaryl. In some embodiments, R 6 and/or R 7 is an unsubstituted heteroaryl, examples of which include, but are not limited to, pyrrolyl, furanyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, imidazolyl, triazolyl, tetrazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, indolyl, benzofuranyl, benzothiophenyl, thiophenyl, benzimidazolyl, quinolinyl, isoquinolinyl, quinazolinyl, quinoxalinyl, and pyrazolyl. In some embodiments, R 6 and/or R 7 is a substituted heteroaryl. The substituent(s) may be any recited herein, including, but not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The heteroaryl group may contain one, or more than one, substituent. In some embodiments, R 6 is hydrogen, and R 7 is a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl. In some embodiments, R 6 is hydrogen, and R 7 is a substituted or unsubstituted C 1 -C 6 alkyl. In some embodiments, R 6 is hydrogen, and R 7 is an unsubstituted C 1 -C 6 alkyl, a C1-C6 alkyl substituted with one or more deuterium atoms, a C1-C6 alkyl substituted with one or more fluorine atoms, or a C 1 -C 6 alkyl substituted with a substituted or unsubstituted cycloalkyl. For example, in some embodiments, R 6 is hydrogen, and R 7 is methyl, ethyl, propyl, -CD3, or cyclopropylmethyl (-CH2C3H5).

In some embodiments, R 6 and R 7 together with the nitrogen atom attached thereto are joined to form a substituted or unsubstituted heterocycloalkyl. In some embodiments, R 6 and R 7 together with the nitrogen atom attached thereto are joined to form an unsubstituted heterocycloalkyl. The unsubstituted heterocycloalkyl group may be, e.g., a 3-membered ring, a 4-membered ring, 5-membered ring, a 6- membered ring, a 7-membered ring, an 8-membered ring, etc., which may be optionally fused to other ring(s). The unsubstituted heterocycloalkyl group contains a minimum of one nitrogen ring atom (the nitrogen atom intervening R 6 and R 7 ), and may optionally contain at least one additional hetero-ring atom, which may be one or more of nitrogen, sulfur, or oxygen, for a total of 1, 2, 3, or 4 hetero-ring atoms (at least one of which is a nitrogen ring atom). Examples of unsubstituted heterocyclo alkyl groups formed from joining R 6 and R 7 together with the nitrogen atom attached thereto include, but are not limited to,

In some embodiments, R 6 and R 7 together with the nitrogen atom attached thereto are joined to form a substituted heterocycloalkyl. The substituted heterocycloalkyl group may be, e.g., a 3-membered ring, a 4-membered ring, a 5-membered ring, a 6-membered ring, a 7-membered ring, an 8-membered ring, etc., which may be optionally fused to other ring(s). The substituted heterocycloalkyl group contains a minimum of one nitrogen ring atom (the nitrogen atom intervening R 6 and R 7 ), and may optionally contain additional hetero-ring atoms (e.g., nitrogen, sulfur, or oxygen) for a total of 1, 2, 3, or 4 hetero-ring atoms (at least one of which is a nitrogen ring atom). Examples of the substituted heterocycloalkyl group include, but are not limited to, aziridine, azetidine, pyrrolidine, isoindole, indole, dihydroindole, indazole, purine, carbazole, carboline, imidazolidine, imidazoline, piperidine, piperazine, indoline, 1,2,3,4-tetrahydroisoquinoline, thiazolidine, morpholine, or thiomorpholine, which is substituted with at least one substituent. The substituent(s) may be any recited herein, including, but not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl, oxo, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), unsubstituted alkyl, substituted alkyl, unsubstituted alkenyl, substituted alkenyl, unsubstituted alkynyl, substituted alkynyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The substituted heterocycloalkyl formed from joining R 6 and R 7 together with the nitrogen atom attached thereto contains a heterocycloalkyl group substituted with one, two, three, four, or more substituents. The substituent may be located on a carbon ring atom or on a hetero-ring atom. Examples of substituted heterocycloalkyl groups formed from joining R 6 and R 7 together with the nitrogen atom attached thereto include, but are not limited to, When present in one or both of R 2 or R 3 , each R a may be, independently, hydrogen, deuterium, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl. In some embodiments, each R a may be, independently, hydrogen, deuterium, an unsubstituted C1-C6 alkyl (e.g., methyl, ethyl, n- propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl), or a substituted C1-C6 alkyl, with preferred substituents including, but not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl or polyether substituents, cycloalkyl, etc. In some embodiments, R a is a substituted or unsubstituted C1-C6 alkyl, preferably a C1-C3 alkyl, preferably a substituted or unsubstituted C1 alkyl, examples of which include, but are not limited to, -CH3, -CDH2, -CD2H, -CD3, -CFH2, -CF2H, -CF3. In some embodiments, each R a is -CH3. In some embodiments, each R a is -CD3. In some embodiments, more than one R a is present. In such cases, each R a may be the same, or different. In some embodiments, each R a is the same. In some embodiments, each R a is different, e.g., one R a is - CH3, while another is -CD3. In some embodiments, R a in one or more of R 2 to R 3 is hydrogen. In some embodiments, R a in one or more of R 2 to R 3 is deuterium. In some embodiments, R a in one or more of R 2 to R 3 is a substituted or unsubstituted C 1 -C 6 alkyl. In some embodiments, R a in one or more of R 2 to R 3 is an unsubstituted C 1 -C 6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n- butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R a in one or more of R 2 to R 3 is a substituted C 1 -C 6 alkyl. Preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), cyano, polar substituents such as hydroxyl or polyether substituents, etc. The C1-C6 alkyl group may contain one, or more than one, substituent. In some embodiments, R a in one or more of R 2 to R 3 is a substituted C1 alkyl group, examples of which may include, but are not limited to, -CDH2, -CD2H, -CD3, -CFH2, -CF2H, -CF3, and -CH28mB( >W \XVO OVLXNSVOW]\& E a in one or more of R 2 to R 3 is a substituted C2 alkyl group, examples of which may include, but are not limited to, -CDHCDH2, -CDHCD2H, -CD2CD3, -CH2CFH2, -CH2CF2H, -CH2CF3, and -CH2CH28mB( >W \XVO OVLXNSVOW]\& E a in one or more of R 2 to R 3 is a substituted C3 alkyl group, examples of which may include, but are not limited to -CH2CH2CF3, -CH 2 CH 2 CF 2 H, -CH 2 CH 2 CFH 2 , -CH 2 CF 2 CF 2 H, and -CH 2 CH 2 CH 2 CmB. In some embodiments, R a in one or more of R 2 to R 3 is a substituted or unsubstituted alkenyl, e.g., a substituted or unsubstituted allyl, butenyl, crotyl, etc. In some embodiments, R a in one or more of R 2 to R 3 is a substituted or unsubstituted alkynyl, e.g., a substituted or unsubstituted acetylenyl, propargyl, homopropargyl, etc. In some embodiments, R a in one or more of R 2 to R 3 is an unsubstituted alkynyl. In some embodiments, R a in one or more of R 2 to R 3 is an unsubstituted acetylenyl (-Cm8=$( >W \XVO OVLXNSVOW]\& R a in one or more of R 2 to R 3 is an unsubstituted propargyl (-CH2Cm8=$( In some embodiments, R a in one or more of R 2 to R 3 is -CH2CH28m8=( >W \XVO OVLXNSVOW]\& E a in one or more of R 2 to R 3 is - CH2CH2CH2Cm8=. In some embodiments, R a in one or more of R 2 to R 3 is -CH2CH2CH2CH2Cm8=. In some embodiments, R a in one or more of R 2 to R 3 is a substituted alkynyl. In some embodiments, R a in one or more of R 2 to R 3 is a substituted propargyl (e.g., -CF28m8=$( >W \XVO OVLXNSVOW]\& E a in one or more of R 2 to R 3 is -CF 2 CH 2 8m8=( >W \XVO OVLXNSVOW]\& E a in one or more of R 2 to R 3 is -CF 2 CH 2 CH 2 8m8=( >W \XVO embodiments, R a in one or more of R 2 to R 3 is -CF2CH2CH2CH28m8=( >W \XVO OVLXNSVOW]\& E a in one or more of R 2 to R 3 is a substituted or unsubstituted cycloalkyl, for example a substituted or unsubstituted C3-C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted Cs-Ce cycloalkyl. In some embodiments, R a in one or more of R 2 to R 3 is an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, R a in one or more of R 2 to R 3 is a substituted cycloalkyl (e.g., a substituted C3-C10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent.

In line with the above description for R a , R 2 and R 3 may be, independent of each other, -OR a or -SR a , examples of which include, but are not limited to, -SMe, -SCD3, -SCF3, -SCF2H, -SCFH2, -SEt, -Sn-Pr, -SCH2CF3, -SCH2CF2H, -SCH2CFH2, -SCH2CH2CF3, -SCH2CH2CF2H, -SCH2CH2CFH2, -SCH2CF2CF2H, -SCH2OCI I, -SC=CI I, -SCF2OCI I, -SCI I2CH2CH2OCI I, -SCF2CH2CH2OCI I, -OMe, -OCD3, -OCF3, -OCF2H, -OCFH2, -OCH2CF3, -OCH2CF2H, -OCH2CFH2, -OCH2CH2CF3, -OCH2CH2CF2H, -OCH2CH2CFH2, -OCH2CF2CF2H, -OCH2OCI I, -OC=CI I,. In some embodiments, R 2 is -OR a or -SR a , while R 3 is hydrogen. In some embodiments, R 3 is -OR a or -SR a , while R 2 is hydrogen.

In some embodiments, A is O (oxygen). In some embodiments, A is S (sulfur).

Z 1 and Z 2 may be the same, or different. In some embodiments, Z 1 and Z 2 are the same. In some embodiments, Z 1 and Z 2 are hydrogen. In some embodiments, Z 1 and Z 2 are deuterium. In some embodiments, Z 1 and Z 2 are fluorine. In some embodiments, Z 1 and Z 2 are different. In some embodiments, one of Z 1 and Z 2 is deuterium while the other is hydrogen.

As stated above, any of the above embodiments of the compound of Formula (II) may be provided as long as when A is O, at least one of X 1 , X 2 , Y 1 , Y 2 , R 2 , R 3 , R 6 , R 7 , Z 1 , Z 2 comprises deuterium, and/or at least one of Z 1 and Z 2 is fluorine.

In some embodiments, the compound, e.g., the compound of Formula (II), is selected from the group consisting of:

pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof.

The compound number, IUPAC name, and substituent listing for the above-identified compounds are provided in Table 1.

Table 1. Exemplary compounds of Formula (II) Table 1 (Continued). Table 1 (Continued). Table 1 (Continued). Table 1 (Continued).

The compounds of Formula (II) may advantageously slow or shunt metabolic degradation that results in the formation of toxic by-products, e.g., O-demethylenation, enabling bioavailable dosing regimens with decreased toxicity and off-target activity. The compounds of formula (II) may also introduce metabolic labile groups, e.g., those compounds with benzo[d][1,3]oxathiole groups, for controlled, consistent exposure, and shortened effects. Formula (III) In some embodiments, the compound has a structure of Formula (III): or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, wherein: X 1 and X 2 are independently hydrogen, deuterium, or a substituted or unsubstituted C1-C6 alkyl; Y 1 and Y 2 are independently hydrogen, deuterium, or a substituted or unsubstituted C1-C6 alkyl; R 4 is a substituted or unsubstituted C1-C6 alkyl, -OR a , -SR a , or -SeR a ; R 6 and R 7 are independently hydrogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or alternatively R 6 and R 7 together with the nitrogen atom attached thereto are optionally joined to form a substituted or unsubstituted heterocycloalkyl; each R a is independently hydrogen, deuterium, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C 3 -C 10 cycloalkyl; and wherein at least one of X 1 , X 2 , Y 1 , Y 2 , R 4 , R 6 , R 7 , R a comprises deuterium, and/or R 4 is -OR a , -SR a , or -SeR a , with R a in R 4 being a C1-C6 alkyl substituted with one or more halogen. X 1 and X 2 may be the same, or different. In some embodiments, X 1 and X 2 are the same. In some embodiments, X 1 and X 2 are hydrogen. In some embodiments, X 1 and X 2 are deuterium. In some embodiments, X 1 and X 2 are different. In some embodiments, X 1 is hydrogen or deuterium, and X 2 is a substituted or unsubstituted C1-C6 alkyl. In some embodiments, X 2 is an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, and n-propyl, preferably methyl. In some embodiments, X 2 is a substituted C1-C6 alkyl. The alkyl group may contain one, or more than one, substituent. For example, when the alkyl group is a C1 alkyl group (i.e., methyl group), the substituted C 1 alkyl group may be -CDH 2 , -CD 2 H, -CD 3 , -CFH 2 , -CF2H, -CF3, etc. In some embodiments, one of X 1 and X 2 is deuterium while the other is hydrogen. Y 1 and Y 2 may be the same, or different. In some embodiments, Y 1 and Y 2 are the same. In some embodiments, Y 1 and Y 2 are hydrogen. In some embodiments, Y 1 and Y 2 are deuterium. In some embodiments, Y 1 and Y 2 are different. In some embodiments, one of Y 1 and Y 2 is deuterium while the other is hydrogen. In some embodiments, Y 1 is a substituted or unsubstituted C1-C6 alkyl. In some embodiments, Y 2 is a substituted or unsubstituted C1-C6 alkyl. In some embodiments, R 4 is a an unsubstituted C 1 -C 6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R 4 is a substituted C 1 -C 6 alkyl. When R 4 is a substituted C 1 -C 6 alkyl, preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl or polyether substituents, cycloalkyl, etc. The alkyl group may contain one, or more than one, substituent. For example, when the alkyl group is a C1 alkyl group (i.e., methyl group), the substituted C1 alkyl group may be -CDH2, -CD2H, -CD3, -CFH2, -CF2H, -CF3, etc. In some embodiments, R 4 is -OR a , SR a , or -SeR a , wherein R a in R 4 is hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl. In some embodiments, R 4 is -OR a . In some embodiments, R 4 is -SR a . In some embodiments, R 4 is -SeR a . In some embodiments, R a in R 4 is hydrogen. In some embodiments, R a in R 4 is deuterium. In some embodiments, R a in R 4 is a substituted or unsubstituted C 1 -C 6 alkyl. In some embodiments, R a in R 4 is an unsubstituted C 1 -C 6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R a in R 4 is a substituted C 1 -C 6 alkyl. Preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), cyano, polar substituents such as hydroxyl or polyether substituents, cycloalkyl, etc. The C 1 -C 6 alkyl group may contain one, or more than one, substituent. In some embodiments, R a in R 4 is a substituted C1 alkyl group, examples of which may include, but are not limited to, -CDH2, -CD2H, -CD3, -CFH2, - CF2H, -CF3, and -CH28mB( >W \XVO OVLXNSVOW]\& E a in R 4 is a substituted C2 alkyl group, examples of which may include, but are not limited to, -CDHCDH2, -CDHCD2H, -CD2CD3, -CH2CFH2, -CH2CF2H, -CH2CF3, and -CH2CH28mB( >W \XVO OVLXNSVOW]\& E a in R 4 is not a substituted C2 alkyl group such as a C2 fluoroalkyl group. In some embodiments, R a in R 4 is a substituted C3 alkyl group, examples of which may include, but are not limited to -CH 2 CH 2 CF 3 , -CH 2 CH 2 CF 2 H, -CH 2 CH 2 CFH 2 , -CH2CF2CF2H, and -CH2CH2CH28mB( >W \XVO OVLXNSVOW]\& E a in R 4 is a substituted or unsubstituted alkenyl, e.g., a substituted or unsubstituted allyl, butenyl, crotyl, etc. In some embodiments, R a in R 4 is a substituted or unsubstituted alkynyl, e.g., a substituted or unsubstituted acetylenyl, propargyl, homopropargyl, etc. In some embodiments, R a in R 4 is an unsubstituted alkynyl. In some embodiments, R a in R 4 is an unsubstituted acetylenyl (-Cm8=$( >W \XVO OVLXNSVOW]\& R a in R 4 is an unsubstituted propargyl (-CH28m8=$( >W \XVO OVLXNSVOW]\& E a in R 4 is -CH2CH28m8=( >W \XVO OVLXNSVOW]\& E a in R 4 is -CH2CH2CH2Cm8=. In some embodiments, R a in R 4 is -CH2CH2CH2CH2Cm8=. In some embodiments, R a in R 4 is a substituted alkynyl. In some embodiments, R a in R 4 is a substituted propargyl (e.g., -CF 2 8m8=$( >W \XVO OVLXNSVOW]\& E a in R 4 is -CF 2 CH 2 8m8=( >W \XVO OVLXNSVOW]\& E a in R 4 is -CF 2 CH 2 CH 2 Cm8=. In some embodiments, R a in R 4 is -CF 2 CH 2 CH 2 CH 2 Cm8=. In some embodiments, R a in R 4 is a substituted or unsubstituted cycloalkyl, for example a substituted or unsubstituted C 3 -C 10 cycloalkyl, or a substituted or unsubstituted C 4 -C 8 cycloalkyl, or a substituted or unsubstituted C 5 -C 6 cycloalkyl. In some embodiments, R a in R 4 is an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, R a in R 4 is a substituted cycloalkyl (e.g., a substituted C3-C10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 4 is -SMe, -SCD 3, -SCF 3, -SCF 2 H, -SCFH 2 , -SEt, -Sn-Pr, -SCH 2 CF 3 , -SCH 2 CF 2 H, -SCH 2 CFH 2 , -SCH 2 CH 2 CF 3 , -SCH 2 CH 2 CF 2 H, -SCH 2 CH 2 CFH 2 , -SCH 2 CF 2 CF 2 H, -SCH 2 Cm8=, -SCm8=, -SCF 2 Cm8=, -SCH 2 CH 2 CH 2 Cm8=, -SCF 2 CH 2 CH 2 Cm8=, -Me, -CD 3 , -CF 3 , -t-Bu, -C(CD 3 ) 3 , -OMe, -OCD 3 , -OCF 3 , -OCF 2 H, -OCFH 2 , -OCH 2 CF 3 , -OCH 2 CF 2 H, -OCH 2 CFH 2 , -OCH 2 CH 2 CF 3 , -OCH 2 CH 2 CF 2 H, -OCH2CH2CFH2, -OCH2CF2CF2H, -OCH2Cm8=, -OCm8=, -SeMe, -SeCD3, -SeCF3, -SeCF2H, -SeCFH2, -SeEt, -Sen-Pr, -SeCH2CF3, -SeCH2CF2H, -SeCH2CFH2, -SeCH2CH2CF3, -SeCH2CH2CF2H, -SeCH2CH2CFH2, -SeCH2CF2CF2H, -SeCH28m8=& 'FO8m8=& 'FO8;28m8=& -SeCH2CH2CH2Cm8=, or -SeCF2CH2CH2Cm8=. In some embodiments, R 4 is -SMe, -SCD3, -SCF3, -SEt, -Sn-Pr, -SCH2CH2CF3, -SCH2CH2CF2H, -SCH2CH2CFH2, -Me, -CD3, -CF3, -OMe, -OCD3, -OCF3, -OCH2CH2CF3, -OCH2CH2CF2H, -OCH2CH2CFH2 or -Br. R 6 and R 7 may be the same, or different. In some embodiments, R 6 and R 7 are the same. For example, in some embodiments, both R 6 and R 7 are hydrogen. In some embodiments, R 6 and R 7 are different. For example, in some embodiments, R 6 is hydrogen, and R 7 is a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl. R 6 and R 7 may be, independently, hydrogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl. In some embodiments, R 6 and R 7 may be, independently, hydrogen, an unsubstituted C1-C6 alkyl (e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, and hexyl) or a C 1 -C 6 alkyl substituted with one or more deuterium (e.g., -CDH 2 , -CD 2 H, -CD 3 ). In some embodiments, R 6 and/or R 7 is an unsubstituted C 1 -C 6 alkyl, for example, an unsubstituted C 1 alkyl, an unsubstituted C 2 alkyl, an unsubstituted C 3 alkyl, an unsubstituted C 4 alkyl, an unsubstituted C 5 alkyl, or an unsubstituted C 6 alkyl. In some embodiments, R 6 and/or R 7 is an unsubstituted linear C2-C6 alkyl. In some embodiments, R 6 and/or R 7 is an unsubstituted branched C3- C10 alkyl. Examples of an unsubstituted C1-C6 alkyl include, but are not limited to, methyl, ethyl, n- propyl, isopropyl, butyl, isobutyl, t-butyl, n-pentyl, isopentyl, n-hexyl, and isohexyl. In some embodiments, R 6 and/or R 7 is a substituted C1-C6 alkyl, e.g., a substituted C1 alkyl, a substituted C2 alkyl, a substituted C3 alkyl, a substituted C4 alkyl, a substituted C5 alkyl, or a substituted C6 alkyl. The alkyl group may contain one, or more than one, substituent. The alkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl, oxo, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), unsubstituted alkenyl, substituted alkenyl, unsubstituted alkynyl, substituted alkynyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. In some embodiments, R 6 and/or R 7 is a C 1 -C 6 alkyl substituted with one or more deuterium atoms, examples of which include, but are not limited to, -CDH2, -CD2H, -CD3, -CD2CD3, and -CD2CD2CD3. In some embodiments, R 6 and/or R 7 is a C1-C6 alkyl substituted with one or more fluorine atoms, i.e., is a fluoroalkyl group. Examples of fluoroalkyl groups include, but are not limited to, -CH2F, -CHF2, -CF3, -CH2CH2F, -CH2CHF2, -CH2CF3, -CH2CH2CH2F, -CH2CH2CHF2, -CH2CH2CF3, -CH2CH2CH2CH2F, -CH2CH2CH2CHF2, -CH2CH2CH2CF3, -CH2CF2CHF2, -CH2CF2CF3, -CH(CF3)2, and -CH(CH3)CF3. In some embodiments, R 6 and/or R 7 is a C1-C6 alkyl substituted with one or more deuterium atoms and one or more fluorine atoms, examples of which include, but are not limited to, - CD 2 CH 2 F, -CD 2 CHF 2 , -CD 2 CF 3 , -CD 2 CH 2 CH 2 F, -CD 2 CH 2 CHF 2 , -CD 2 CH 2 CF 3 , -CD2CD2CH2, -CD2CD2CHF2, -CD2CD2CF3, -CD2CH2CH2CH2F, -CD2CH2CH2CHF2, -CD 2 CH 2 CH 2 CF 3 , -CD 2 CD 2 CH 2 CH 2 F, -CD 2 CD 2 CH 2 CHF 2 , -CD 2 CD 2 CH 2 CF 3 , -CD2CD2CD2CH2F, -CD2CD2CD2CHF2, and -CD2CD2CD2CF3. In some embodiments, R 6 and/or R 7 is a C1-C6 alkyl substituted with a substituted or unsubstituted cycloalkyl. The C1-C6 alkyl may be substituted with, e.g., a substituted or unsubstituted C3-C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted C5-C6 cycloalkyl. In some embodiments, the C1-C6 alkyl is substituted with an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, the C 1 -C 6 alkyl is substituted with a substituted cycloalkyl (e.g., a substituted C 3 -C 10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 6 and/or R 7 is a C1 alkyl substituted with a substituted or unsubstituted cycloalkyl, with particular mention being made to cyclopropylmethyl (-CH2C3H5). In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted alkenyl, e.g., a substituted or unsubstituted allyl, butenyl, crotyl, etc. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted alkynyl, e.g., a substituted or unsubstituted propargyl. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted cycloalkyl, for example a substituted or unsubstituted C 3 -C 10 cycloalkyl, or a substituted or unsubstituted C 4 -C 8 cycloalkyl, or a substituted or unsubstituted C 5 -C 6 cycloalkyl. In some embodiments, R 6 and/or R 7 is an unsubstituted cycloalkyl (e.g., an unsubstituted C 3 -C 10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, R 6 and/or R 7 is a substituted cycloalkyl (e.g., a substituted C3-C10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted heterocycloalkyl. In some embodiments, the unsubstituted or substituted heterocycloalkyl group may be a 3-membered ring, a 4- membered ring, a 5-membered ring, a 6-membered ring, a 7-membered ring, or an 8-membered ring. In some embodiments, R 6 and/or R 7 is an unsubstituted heterocycloalkyl, such as those set forth herein, examples of which include, but are not limited to, aziridine, azetidine, pyrrolidine, isoindole, indole, dihydroindole, indazole, purine, carbazole, carboline, imidazolidine, imidazoline, piperidine, piperazine, indoline, phthalimide, 1,2,3,4-tetrahydroisoquinoline, thiazolidine, morpholine, thiomorpholine, tetrahydrofuran, tetrahydropyran, and 1,3-dioxolane. In some embodiments, R 6 and/or R 7 is a substituted heterocycloalkyl. The substituent(s) may be any recited herein, including, but not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), oxo, and hydroxyl. The heterocycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted aryl. In some embodiments, R 6 and/or R 7 is an unsubstituted aryl, examples of which include, but are not limited to, phenyl and naphthyl. In some embodiments, R 6 and/or R 7 is a substituted aryl. The substituent(s) may be any recited herein, including, but not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The aryl group may contain one, or more than one, substituent. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted heteroaryl. In some embodiments, R 6 and/or R 7 is an unsubstituted heteroaryl, examples of which include, but are not limited to, pyrrolyl, furanyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, imidazolyl, triazolyl, tetrazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, indolyl, benzofuranyl, benzothiophenyl, thiophenyl, benzimidazolyl, quinolinyl, isoquinolinyl, quinazolinyl, quinoxalinyl, and pyrazolyl. In some embodiments, R 6 and/or R 7 is a substituted heteroaryl. The substituent(s) may be any recited herein, including, but not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The heteroaryl group may contain one, or more than one, substituent. In some embodiments, R 6 is hydrogen, and R 7 is a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl. In some embodiments, R 6 is hydrogen, and R 7 is a substituted or unsubstituted C 1 -C 6 alkyl. In some embodiments, R 6 is hydrogen, and R 7 is an unsubstituted C 1 -C 6 alkyl, a C1-C6 alkyl substituted with one or more deuterium atoms, a C1-C6 alkyl substituted with one or more fluorine atoms, or a C1-C6 alkyl substituted with a substituted or unsubstituted cycloalkyl. For example, in some embodiments, R 6 is hydrogen, and R 7 is methyl, ethyl, propyl, -CD3, or cyclopropylmethyl (-CH2C3H5). In some embodiments, R 6 and R 7 together with the nitrogen atom attached thereto are joined to form a substituted or unsubstituted heterocycloalkyl. In some embodiments, R 6 and R 7 together with the nitrogen atom attached thereto are joined to form an unsubstituted heterocycloalkyl. The unsubstituted heterocycloalkyl group may be, e.g., a 3-membered ring, a 4-membered ring, 5-membered ring, a 6- membered ring, a 7-membered ring, an 8-membered ring, etc., which may be optionally fused to other ring(s). The unsubstituted heterocycloalkyl group contains a minimum of one nitrogen ring atom (the nitrogen atom intervening R 6 and R 7 ), and may optionally contain at least one additional hetero-ring atom, which may be one or more of nitrogen, sulfur, or oxygen, for a total of 1, 2, 3, or 4 hetero-ring atoms (at least one of which is a nitrogen ring atom). Examples of unsubstituted heterocycloalkyl groups formed from joining R 6 and R 7 together with the nitrogen atom attached thereto include, but are not limited to, , . In some embodiments, R 6 and R 7 together with the nitrogen atom attached thereto are joined to form a substituted heterocycloalkyl. The substituted heterocycloalkyl group may be, e.g., a 3-membered ring, a 4-membered ring, a 5-membered ring, a 6-membered ring, a 7-membered ring, an 8-membered ring, etc., which may be optionally fused to other ring(s). The substituted heterocycloalkyl group contains a minimum of one nitrogen ring atom (the nitrogen atom intervening R 6 and R 7 ), and may optionally contain additional hetero-ring atoms (e.g., nitrogen, sulfur, or oxygen) for a total of 1, 2, 3, or 4 hetero-ring atoms (at least one of which is a nitrogen ring atom). Examples of the substituted heterocycloalkyl group include, but are not limited to, aziridine, azetidine, pyrrolidine, isoindole, indole, dihydroindole, indazole, purine, carbazole, carboline, imidazolidine, imidazoline, piperidine, piperazine, indoline, 1,2,3,4-tetrahydroisoquinoline, thiazolidine, morpholine, or thiomorpholine, which is substituted with at least one substituent. The substituent(s) may be any recited herein, including, but not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl, oxo, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), unsubstituted alkyl, substituted alkyl, unsubstituted alkenyl, substituted alkenyl, unsubstituted alkynyl, substituted alkynyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The substituted heterocycloalkyl formed from joining R 6 and R 7 together with the nitrogen atom attached thereto contains a heterocycloalkyl group substituted with one, two, three, four, or more substituents. The substituent may be located on a carbon ring atom or on a hetero-ring atom. Examples of substituted heterocycloalkyl groups formed from joining R 6 and R 7 together with the nitrogen atom attached thereto include, but are not limited to, , Each R a may be, independently, hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl. In some embodiments, each R a may be, independently, hydrogen, deuterium, an unsubstituted C1-C6 alkyl (e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec- butyl, t-butyl, n-pentyl, neopentyl, and hexyl), or a substituted C1-C6 alkyl, with preferred substituents including, but not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl or polyether substituents, cycloalkyl, etc. In some embodiments, R a is a substituted or unsubstituted C1-C6 alkyl, preferably a C1-C3 alkyl, preferably a substituted or unsubstituted C1 alkyl, examples of which include, but are not limited to, -CH3, -CDH2, -CD2H, -CD3, -CFH2, -CF2H, -CF3. Each R a may be the same, or different from any other R a present. In some embodiments, each R a is the same. In some embodiments, each R a is -CH3. In some embodiments, each R a is -CD3. In some embodiments, each R a is different. In some embodiments, both R a ’s located at the meta positions of the phenyl group are the same, while any R a present in R 4 may be the same or different from those at the meta positions of the phenyl group. In some embodiments, each R a is independently - Me, -CD 3 , -CF 3 , -Et, -n-Pr, -CH 2 CH 2 CF 3 , -CH 2 CH 2 CF 2 H, or -CH 2 CH 2 CFH 2 . In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is hydrogen. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is deuterium. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is a substituted or unsubstituted C1-C6 alkyl. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is a substituted C1-C6 alkyl. Preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), cyano, polar substituents such as hydroxyl or polyether substituents, cycloalkyl, etc. The C1-C6 alkyl group may contain one, or more than one, substituent. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is a substituted C 1 alkyl group, examples of which may include, but are not limited to, -CDH 2 , -CD 2 H, -CD 3 , -CFH 2 , - CF 2 H, -CF 3 , and -CH 2 CmB. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is a substituted C 2 alkyl group, examples of which may include, but are not limited to, -CDHCDH 2 , -CDHCD 2 H, -CD 2 CD 3 , -CH2CFH2, -CH2CF2H, -CH2CF3, and -CH2CH2CmB. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is a substituted C3 alkyl group, examples of which may include, but are not limited to -CH2CH2CF3, -CH2CH2CF2H, -CH2CH2CFH2, -CH2CF2CF2H, and - CH2CH2CH2CmB. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is a substituted or unsubstituted alkenyl, e.g., a substituted or unsubstituted allyl, butenyl, crotyl, etc. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is a substituted or unsubstituted alkynyl, e.g., a substituted or unsubstituted acetylenyl, propargyl, homopropargyl, etc. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is an unsubstituted alkynyl. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is an unsubstituted acetylenyl (-Cm8=$( >W \XVO OVLXNSVOW]\& R a in one or more of R 4 and the meta positions of the phenyl group is an unsubstituted propargyl (-CH28m8=$( >W \XVO embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is -CH2CH2Cm8=( In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is - CH2CH2CH28m8=( >W \XVO OVLXNSVOW]\& E a in one or more of R 4 and the meta positions of the phenyl group is -CH2CH2CH2CH2Cm8=. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is a substituted alkynyl. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is a substituted propargyl (e.g., -CF 2 8m8=$( >W \XVO OVLXNSVOW]\& E a in one or more of R 4 and the meta positions of the phenyl group is -CF 2 CH 2 Cm8=. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is -CF 2 CH 2 CH 2 8m8=( >W \XVO embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is - CF 2 CH 2 CH 2 CH 2 Cm8=. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is a substituted or unsubstituted cycloalkyl, for example a substituted or unsubstituted C3- C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted C5- C6 cycloalkyl. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, R a in one or more of R 4 and the meta positions of the phenyl group is a substituted cycloalkyl (e.g., a substituted C3-C10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. As stated above, any of the above embodiments of the compound of Formula (III) may be provided as long as at least one of X 1 , X 2 , Y 1 , Y 2 , R 4 , R 6 , R 7 , R a comprises deuterium, and/or R 4 is -OR a , -SR a , or -SeR a , with R a in R 4 being a C1-C6 alkyl substituted with one or more halogen (i.e., R 4 is an -O-C1-C6 alkyl group, the alkyl group being substituted with one or more halogen; an -S-C1-C6 alkyl group, the alkyl group being substituted with one or more halogen; or an -Se-C1-C6 alkyl group, the alkyl group being substituted with one or more halogen). In some embodiments, the compound, e.g., the compound of Formula (III), is selected from the group consisting of:

pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof.

The compound number, IUPAC name, and substituent listing for the above-identified compounds are provided in Table 2. Table 2. Exemplary compounds of Formula (III) Table 2 (Continued). Table 2 (Continued). The compounds of Formula (III) may possess advantageous brain bioavailability, and thus demonstrate enhanced oral activity even at lower dosages. As a result, the compounds of Formula (III) may be suitable for microdosing to achieve durable therapeutic benefits, with decreased toxicity. Formula (IV) In some embodiments, the compound has a structure of Formula (IV): or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, wherein: X 1 is hydrogen or deuterium; X 2 is a substituted or unsubstituted C1-C6 alkyl; Y 1 and Y 2 are independently hydrogen or deuterium; R 3 is hydrogen or deuterium; R 4 is hydrogen, deuterium, halogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted C3-C10 cycloalkyl, -OR b , -SR b , or -SeR b ; R 6 and R 7 are independently hydrogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or alternatively R 6 and R 7 together with the nitrogen atom attached thereto are optionally joined to form a substituted or unsubstituted heterocycloalkyl; each R a is independently a substituted or unsubstituted C1-C6 alkyl; and R b is hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C 3 -C 10 cycloalkyl; with the proviso that at least one of X 1 , X 2 , Y 1 , Y 2 , R 3 , R 4 , R 6 , R 7 , and R a comprises deuterium and/or R 4 is -OR b , -SR b , or -SeR b , with R b in R 4 being a C 1 -C 6 alkyl substituted with one or more halogen. In some embodiments, X 1 is hydrogen. In some embodiments, X 1 is deuterium. In some embodiments, X 2 is an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, and n-propyl, preferably methyl. In some embodiments, X 2 is a substituted C1-C6 alkyl. The alkyl group may contain one, or more than one, substituent. For example, when the alkyl group is a C1 alkyl group (i.e., methyl group), the substituted C1 alkyl group may be -CDH2, -CD2H, -CD3, -CFH2, -CF2H, -CF3, etc. Y 1 and Y 2 may be the same, or different. In some embodiments, Y 1 and Y 2 are the same. In some embodiments, Y 1 and Y 2 are hydrogen. In some embodiments, Y 1 and Y 2 are deuterium. In some embodiments, one of Y 1 and Y 2 is deuterium while the other is hydrogen. In some embodiments, R 3 is deuterium. In some embodiments, R 3 is hydrogen. In some embodiments, R 4 is hydrogen. In some embodiments, R 4 is deuterium. In some embodiments, R 4 is halogen, for example -Br, -F, -Cl, or -I. In some embodiments, R 4 is an unsubstituted C 1 -C 6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R 4 is a substituted C1-C6 alkyl. When R 4 is a substituted C1-C6 alkyl, preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl, alkoxy, or polyether substituents, cycloalkyl, etc. The alkyl group may contain one, or more than one, substituent. For example, when the alkyl group is a C1 alkyl group (i.e., methyl group), the substituted C1 alkyl group may be -CDH2, -CD2H, -CD3, -CFH2, - CF2H, -CF3, etc. In some embodiments, R 4 is a substituted or unsubstituted C3-C10 cycloalkyl. In some embodiments, R 4 is an unsubstituted C 3 -C 10 cycloalkyl, examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cyclooctyl. In some embodiments, R 4 is a substituted C 3 -C 10 cycloalkyl. Preferred substituents may include, but are not limited to, alkyl, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl, alkoxy, or polyether substituents, etc. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 4 is -OR b , SR b , or -SeR b , wherein R b is hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl. In some embodiments, R 4 is -OR b , wherein R b is hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, or a substituted or unsubstituted C 3 -C 10 cycloalkyl, preferably a substituted or unsubstituted C 1 -C 6 alkyl, or a substituted or unsubstituted C 3 -C 10 cycloalkyl, such as those substituted C 1 -C 6 alkyl groups, unsubstituted C 1 -C 6 alkyl groups, substituted C 3 -C 10 cycloalkyl groups, or unsubstituted C 3 -C 10 cycloalkyl groups defined and exemplified herein. In some embodiments, R 4 is -SR b , wherein R b is hydrogen, deuterium, a substituted or unsubstituted C 1 -C 6 alkyl, or a substituted or unsubstituted C 3 -C 10 cycloalkyl, preferably a substituted or unsubstituted C1-C6 alkyl, or a substituted or unsubstituted C3-C10 cycloalkyl, such as those substituted C1-C6 alkyl groups, unsubstituted C1-C6 alkyl groups, substituted C3-C10 cycloalkyl groups, or unsubstituted C3-C10 cycloalkyl groups defined and exemplified herein. In some embodiments, R 4 is -SeR b , wherein R b is hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, or a substituted or unsubstituted C3-C10 cycloalkyl, preferably a substituted or unsubstituted C1-C6 alkyl, or a substituted or unsubstituted C3-C10 cycloalkyl, such as those substituted C1-C6 alkyl groups, unsubstituted C1-C6 alkyl groups, substituted C3-C10 cycloalkyl groups, or unsubstituted C3-C10 cycloalkyl groups defined and exemplified herein. In some embodiments, R b is hydrogen. In some embodiments, R b is deuterium. In some embodiments, R b is a substituted or unsubstituted C 1 -C 6 alkyl. In some embodiments, R b is an unsubstituted C 1 -C 6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R b is a substituted C 1 -C 6 alkyl. Preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), cyano, polar substituents such as hydroxyl or polyether substituents, cycloalkyl etc. The C1-C6 alkyl group may contain one, or more than one, substituent. In some embodiments, R b is a substituted C1 alkyl group, examples of which may include, but are not limited to, -CDH2, -CD2H, - CD3, -CFH2, -CF2H, -CF3, and -CH2CmB. In some embodiments, R b is a substituted C2 alkyl group, examples of which may include, but are not limited to, -CDHCDH2, -CDHCD2H, -CD2CD3, -CH2CFH2, -CH2CF2H, -CH2CF3, and -CH2CH2CmB. In some embodiments, R b is not a substituted C2 alkyl group such as a C2 fluoroalkyl group. In some embodiments, R b is a substituted C 3 alkyl group, examples of which may include, but are not limited to -CH2CH2CF3, -CH2CH2CF2H, -CH2CH2CFH2, -CH 2 CF 2 CF 2 H, and -CH 2 CH 2 CH 2 CmB. In some embodiments, R b is a substituted or unsubstituted alkenyl, e.g., a substituted or unsubstituted allyl, butenyl, crotyl, etc. In some embodiments, R b is a substituted or unsubstituted alkynyl, e.g., a substituted or unsubstituted acetylenyl, propargyl, homopropargyl, etc. In some embodiments, R b is an unsubstituted alkynyl. In some embodiments, R b is KW ^W\^L\]S]^]ON KMO]bUOWbU #'8m8=$( >W \XVO OVLXNSVOW]\& E b is an unsubstituted propargyl (-CH2Cm8=$( In some embodiments, R b is -CH2CH2Cm8=( In some embodiments, R b is -CH2CH2CH2C m8= . In some embodiments, R b is -CH2CH2CH2CH2C m8= . In some embodiments, R b is a substituted alkynyl. In some embodiments, R b is a substituted propargyl (e.g., -CF 2 8m8=$( >W \XVO embodiments, R b is -CF 2 CH 2 Cm8=. In some embodiments, R b is -CF 2 CH 2 CH 2 Cm8=. In some embodiments, R b is -CF 2 CH 2 CH 2 CH 2 8m8=( >W \XVO OVLXNSVOW]\& E b is a substituted or unsubstituted cycloalkyl, for example a substituted or unsubstituted C 3 -C 10 cycloalkyl, or a substituted or unsubstituted C 4 -C 8 cycloalkyl, or a substituted or unsubstituted C 5 -C 6 cycloalkyl. In some embodiments, R b is an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, R b is a substituted cycloalkyl (e.g., a substituted C3- C10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 4 is -SMe, -SCD 3, -SCF 3, -SCF 2 H, -SCFH 2 , -SEt, -Sn-Pr, -SCH 2 CF 3 , -SCH 2 CF 2 H, -SCH 2 CFH 2 , -SCH 2 CH 2 CF 3 , -SCH 2 CH 2 CF 2 H, -SCH 2 CH 2 CFH 2 , -SCH 2 CF 2 CF 2 H, -SCH 2 Cm8=, -SCm8=, -SCF 2 Cm8=, -SCH 2 CH 2 CH 2 Cm8=, -SCF 2 CH 2 CH 2 8m8=& -Me, -CD 3 , -CF 3 , -t-Bu, -C(CD 3 ) 3 , -cyclopentyl, -OMe, -OCD 3 , -OCF 3 , -OCF2H, -OCFH2, -OCH2CF3, -OCH2CF2H, -OCH2CFH2, -OCH2CH2CF3, -OCH2CH2CF2H, -OCH2CH2CFH2, -OCH2CF2CF2H, -OCH2Cm8=, -OCm8=, -CH2CH2CH2CH2Cm8=, -CF2CH2CH2CH28m8=& '8U& '>& '7[& 'FOAO& 'FO893, -SeCF3, -SeCF2H, -SeCFH2, -SeEt, -Sen-Pr, -SeCH2CF3, -SeCH2CF2H, -SeCH2CFH2, -SeCH2CH2CF3, -SeCH2CH2CF2H, -SeCH2CH2CFH2, -SeCH2CF2CF2H, -SeCH28m8=& 'FO8m8=& -SeCF28m8=& -SeCH2CH2CH28m8=& X[ 'FO8;2CH2CH28m8=( >W \XVO OVLXNSVOW]\& E 4 is selected from the group consisting of -SMe, -SCD3, -SCF3, -SCF2H, -SCH2CH2CF3, -SCH2CH2CF2H, -SCH 2 CH 2 CFH 2 , -SEt, -Sn-Pr, -Me, -CD 3 , -CF 3 , -t-Bu, -C(CD 3 ) 3 , -cyclopentyl, -OMe, -OCD 3 , -OCF3, -OCH2CH2CF3, -OCH2CH2CF2H, -OCH2CH2CFH2, -Cl, -I, or -Br. In some embodiments, R 4 is selected from the group consisting of -SMe, -Me, -OCD 3 , -CF 3 , -t-Bu, or -cyclopentyl. In some embodiments, R 4 is selected from the group consisting of -SCF3, -SCF2H, -SCH2CH2CF3, -SCH2CH2CF2H, -SCH2CH2CFH2, -OCH2CH2CF3, -OCH2CH2CF2H, and -OCH2CH2CFH2. In some embodiments, when R 4 is -SCF3, -SCF2H, -SCH2CH2CF3, -SCH2CH2CF2H, -SCH2CH2CFH2, -OCH2CH2CF3, -OCH2CH2CF2H, or -OCH2CH2CFH2, the other substituents (i.e., X 1 , X 2 , Y 1 , Y 2 , R 3 , R 6 , R 7 , and R a ) may, or may not, comprise deuterium. In preferred embodiments, R 4 is - SCF3. R 6 and R 7 may be the same, or different. In some embodiments, R 6 and R 7 are the same. For example, in some embodiments, both R 6 and R 7 are hydrogen. In some embodiments, R 6 and R 7 are different. For example, in some embodiments, R 6 is hydrogen, and R 7 is a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl. R 6 and R 7 may be, independently, hydrogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl. In some embodiments, R 6 and R 7 may be, independently, hydrogen, an unsubstituted C1-C6 alkyl (e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, and hexyl) or a C1-C6 alkyl substituted with one or more deuterium (e.g., -CDH2, -CD2H, -CD3). In preferred embodiments, R 6 and R 7 are hydrogen. In some embodiments, R 6 and/or R 7 is an unsubstituted C1-C6 alkyl, for example, an unsubstituted C 1 alkyl, an unsubstituted C 2 alkyl, an unsubstituted C 3 alkyl, an unsubstituted C 4 alkyl, an unsubstituted C 5 alkyl, or an unsubstituted C 6 alkyl. In some embodiments, R 6 and/or R 7 is an unsubstituted linear C 2 -C 6 alkyl. In some embodiments, R 6 and/or R 7 is an unsubstituted branched C 3 - C 10 alkyl. Examples of an unsubstituted C 1 -C 6 alkyl include, but are not limited to, methyl, ethyl, n- propyl, isopropyl, butyl, isobutyl, t-butyl, n-pentyl, isopentyl, n-hexyl, and isohexyl. In some embodiments, R 6 and/or R 7 is a substituted C1-C6 alkyl, e.g., a substituted C1 alkyl, a substituted C2 alkyl, a substituted C3 alkyl, a substituted C4 alkyl, a substituted C5 alkyl, or a substituted C6 alkyl. The alkyl group may contain one, or more than one, substituent. The alkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl, oxo, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), unsubstituted alkenyl, substituted alkenyl, unsubstituted alkynyl, substituted alkynyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. In some embodiments, R 6 and/or R 7 is a C1-C6 alkyl substituted with one or more deuterium atoms, examples of which include, but are not limited to, -CDH 2 , -CD2H, -CD3, -CD2CD3, and -CD2CD2CD3. In some embodiments, R 6 and/or R 7 is a C1-C6 alkyl substituted with one or more fluorine atoms, i.e., is a fluoroalkyl group. Examples of fluoroalkyl groups include, but are not limited to, -CH2F, -CHF2, -CF3, -CH2CH2F, -CH2CHF2, -CH2CF3, -CH2CH2CH2F, -CH2CH2CHF2, -CH2CH2CF3, -CH2CH2CH2CH2F, -CH2CH2CH2CHF2, -CH2CH2CH2CF3, -CH2CF2CHF2, -CH2CF2CF3, -CH(CF3)2, and -CH(CH3)CF3. In some embodiments, R 6 and/or R 7 is a C 1 -C 6 alkyl substituted with one or more deuterium atoms and one or more fluorine atoms, examples of which include, but are not limited to, -CD 2 CH 2 F, -CD 2 CHF 2 , -CD 2 CF 3 , -CD 2 CH 2 CH 2 F, -CD 2 CH 2 CHF 2 , -CD 2 CH 2 CF 3 , -CD 2 CD 2 CH 2 , -CD 2 CD 2 CHF 2 , -CD 2 CD 2 CF 3 , -CD 2 CH 2 CH 2 CH 2 F, -CD 2 CH 2 CH 2 CHF 2 , -CD 2 CH 2 CH 2 CF 3 , -CD 2 CD 2 CH 2 CH 2 F, -CD 2 CD 2 CH 2 CHF 2 , -CD 2 CD 2 CH 2 CF 3 , -CD 2 CD 2 CD 2 CH 2 F, -CD 2 CD 2 CD 2 CHF 2 , and -CD2CD2CD2CF3. In some embodiments, R 6 and/or R 7 is a C1-C6 alkyl substituted with a substituted or unsubstituted cycloalkyl. The C1-C6 alkyl may be substituted with, e.g., a substituted or unsubstituted C3-C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted C5-C6 cycloalkyl. In some embodiments, the C1-C6 alkyl is substituted with an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, the C1-C6 alkyl is substituted with a substituted cycloalkyl (e.g., a substituted C3-C10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 6 and/or R 7 is a C1 alkyl substituted with a substituted or unsubstituted cycloalkyl, with particular mention being made to cyclopropylmethyl (-CH2C3H5). In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted alkenyl, e.g., a substituted or unsubstituted allyl, butenyl, crotyl, etc. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted alkynyl, e.g., a substituted or unsubstituted propargyl. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted cycloalkyl, for example a substituted or unsubstituted C3-C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted C 5 -C 6 cycloalkyl. In some embodiments, R 6 and/or R 7 is an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, R 6 and/or R 7 is a substituted cycloalkyl (e.g., a substituted C3-C10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent.

In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted heterocycloalkyl. In some embodiments, the unsubstituted or substituted heterocycloalkyl group may be a 3-membered ring, a 4- membered ring, a 5 -membered ring, a 6-membered ring, a 7 -membered ring, or an 8 -membered ring. In some embodiments, R 6 and/or R 7 is an unsubstituted heterocycloalkyl, such as those set forth herein, examples of which include, but are not limited to, aziridine, azetidine, pyrrolidine, isoindole, indole, dihydroindole, indazole, purine, carbazole, carboline, imidazolidine, imidazoline, piperidine, piperazine, indoline, phthalimide, 1,2,3,4-tetrahydroisoquinoline, thiazolidine, morpholine, thiomorpholine, tetrahydrofuran, tetrahydropyran, and 1,3-dioxolane. In some embodiments, R 6 and/or R 7 is a substituted heterocycloalkyl. The substituent(s) may be any recited herein, including, but not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), oxo, and hydroxyl. The heterocycloalkyl group may contain one, or more than one, substituent.

In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted aryl. In some embodiments, R 6 and/or R 7 is an unsubstituted aryl, examples of which include, but are not limited to, phenyl and naphthyl. In some embodiments, R 6 and/or R 7 is a substituted aryl. The substituent(s) may be any recited herein, including, but not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., poly ether groups), halogen (e.g., fluorine), hydroxyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The aryl group may contain one, or more than one, substituent.

In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted heteroaryl. In some embodiments, R 6 and/or R 7 is an unsubstituted heteroaryl, examples of which include, but are not limited to, pyrrolyl, furanyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, imidazolyl, triazolyl, tetrazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, indolyl, benzofuranyl, benzothiophenyl, thiophenyl, benzimidazolyl, quinolinyl, isoquinolinyl, quinazolinyl, quinoxalinyl, and pyrazolyl. In some embodiments, R 6 and/or R 7 is a substituted heteroaryl. The substituent(s) may be any recited herein, including, but not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The heteroaryl group may contain one, or more than one, substituent.

In some embodiments, R 6 is hydrogen, and R 7 is a substituted or unsubstituted Ci-Ce alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl. In some embodiments, R 6 is hydrogen, and R 7 is a substituted or unsubstituted Ci-Ce alkyl. In some embodiments, R 6 is hydrogen, and R 7 is an unsubstituted Ci-Ce alkyl, a Ci-Ce alkyl substituted with one or more deuterium atoms, a Ci-Ce alkyl substituted with one or more fluorine atoms, or a Ci-Ce alkyl substituted with a substituted or unsubstituted cycloalkyl. For example, in some embodiments, R 6 is hydrogen, and R 7 is methyl, ethyl, propyl, -CD3, or cyclopropylmethyl (-CH2C3H5).

In some embodiments, R 6 and R 7 together with the nitrogen atom attached thereto are joined to form a substituted or unsubstituted heterocycloalkyl. In some embodiments, R 6 and R 7 together with the nitrogen atom attached thereto are joined to form an unsubstituted heterocycloalkyl. The unsubstituted heterocycloalkyl group may be, e.g., a 3-membered ring, a 4-membered ring, 5-membered ring, a 6- membered ring, a 7-membered ring, an 8-membered ring, etc., which may be optionally fused to other ring(s). The unsubstituted heterocycloalkyl group contains a minimum of one nitrogen ring atom (the nitrogen atom intervening R 6 and R 7 ), and may optionally contain at least one additional hetero-ring atom, which may be one or more of nitrogen, sulfur, or oxygen, for a total of 1, 2, 3, or 4 hetero-ring atoms (at least one of which is a nitrogen ring atom). Examples of unsubstituted heterocyclo alkyl groups formed from joining R 6 and R 7 together with the nitrogen atom attached thereto include, but are not limited to,

In some embodiments, R 6 and R 7 together with the nitrogen atom attached thereto are joined to form a substituted heterocycloalkyl. The substituted heterocycloalkyl group may be, e.g., a 3-membered ring, a 4-membered ring, a 5-membered ring, a 6-membered ring, a 7-membered ring, an 8-membered ring, etc., which may be optionally fused to other ring(s). The substituted heterocycloalkyl group contains a minimum of one nitrogen ring atom (the nitrogen atom intervening R 6 and R 7 ), and may optionally contain additional hetero-ring atoms (e.g., nitrogen, sulfur, or oxygen) for a total of 1, 2, 3, or 4 hetero-ring atoms (at least one of which is a nitrogen ring atom). Examples of the substituted heterocycloalkyl group include, but are not limited to, aziridine, azetidine, pyrrolidine, isoindole, indole, dihydroindole, indazole, purine, carbazole, carboline, imidazolidine, imidazoline, piperidine, piperazine, indoline, 1,2,3,4-tetrahydroisoquinoline, thiazolidine, morpholine, or thiomorpholine, which is substituted with at least one substituent. The substituent(s) may be any recited herein, including, but not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl, oxo, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), unsubstituted alkyl, substituted alkyl, unsubstituted alkenyl, substituted alkenyl, unsubstituted alkynyl, substituted alkynyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The substituted heterocycloalkyl formed from joining R 6 and R 7 together with the nitrogen atom attached thereto contains a heterocycloalkyl group substituted with one, two, three, four, or more substituents. The substituent may be located on a carbon ring atom or on a hetero-ring atom.

Examples of substituted heterocycloalkyl groups formed from joining R 6 and R 7 together with the nitrogen atom attached thereto include, but are not limited to,

Each R a may be the same, or different. In some embodiments, each R a is the same. Each R a may be, independently, a substituted or unsubstituted Ci-Ce alkyl, preferably a substituted or unsubstituted C1-C3 alkyl, preferably a substituted or unsubstituted Ci alkyl, examples of which include, but are not limited to, -CH3, -CDH2, -CD2H, -CD3, -CFH2, -CF2H, -CF3. In some embodiments, each R a is -CH3. In some embodiments, each R a is -CD3. In some embodiments, each R a is different, e.g., one R a is -CH3, while another is -CD3.

In some embodiments, X 1 is hydrogen or deuterium; X 2 is methyl; Y 1 and Y 2 are each hydrogen or each deuterium; R 3 is hydrogen; each R a is -CH3 or -CD3; R 4 is -SMe, -SCD3, -SCF3, -SCF2H, -SCFH2, -SEt, -Sn-Pr, -SCH2CF3, -SCH2CF2H, -SCH2CFH2, -SCH2CH2CF3, -SCH2CH2CF2H, -SCH2CH2CFH2, -SCH2CF2CF2H, -SCH 2 C=CI I, -SC=CI I, -SCF 2 C=CI I, -SCH2CH2CH2OCI I. -SCF2CH2CH2OCI I. -Me, -CD3, -CF3, -t-Bu, -C(CD 3 ) 3 , -cyclopentyl, -OMe, -OCD3, -OCF3, -OCF 2 H, -OCFH 2 , -OCH2CF3, -OCH2CF2H, -OCH2CFH2, -OCH2CH2CF3, -OCH2CH2CF2H, -OCH2CH2CFH2, -OCH2CF2CF2H, -OCH2OCI I. -OOCI I. -CH2CH2CH2CH2OCI I. -CF2CH2CH2CH2OCI I. -Cl, -I, -Br, -SeMe, -SeCD 3 , -SeCF 3 , -SeCF 2 H, -SeCFH 2 , -SeEt, -Sen-Pr, -SeCH 2 CF 3 , -SeCH 2 CF 2 H, -SeCH 2 CFH 2 , -SeCH 2 CH 2 CF3, -SeCH 2 CH 2 CF 2 H, -SeCH 2 CH 2 CFH 2 , -SeCH 2 CF 2 CF 2 H, -SeCH 2 C=CH, -ScOCI I. -SCCF2OCI I. -SCCH2CH2CH2OCI I. or -SeCF 2 CH 2 CH 2 C=CH, preferably -SMe, -SCD3, -SCF3, -SCF 2 H, -SCH2CH2CF3, -SCH2CH2CF2H, -SCH2CH2CFH2, -SEt, -Sn-Pr, -Me, -CD 3 , -CF3, -t-Bu, -C(CD 3 )3, -cyclopentyl, -OMe, -OCD3, -OCF3, -OCH2CH2CF3, -OCH2CH2CF2H, -OCH2CH2CFH2, -Cl, -I, or -Br; and R 6 and R 7 are hydrogen.

As stated above, any of the above embodiments of the compound of Formula (IV) may be provided as long as at least one of X 1 , X 2 , Y 1 , Y 2 , R 3 , R 4 , R 6 , R 7 , and R a comprises deuterium and/or R 4 is -OR b , -SR b , or -SeR b , with R b in R 4 being a C1-C6 alkyl substituted with one or more halogen. In some embodiments, at least one of X 1 , X 2 , Y 1 , Y 2 , R 3 , R 4 , R 6 , R 7 , and R a comprises deuterium. In some embodiments, R 4 is -OR b , -SR b , or -SR b , with R b in R 4 being a C1-C6 alkyl substituted with one or more halogen (i.e., R 4 is an -O-C1-C6 alkyl group, the alkyl group being substituted with one or more halogen; an -S-C1-C6 alkyl group, the alkyl group being substituted with one or more halogen; or an - Se-C 1 -C 6 alkyl group, the alkyl group being substituted with one or more halogen). In some embodiments, the compound, e.g., the compound of Formula (IV), is selected from the group consisting of:

pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof.

The compound number, IUPAC name, and substituent listing for the above-identified compounds are provided in Table 3. Table 3. Exemplary compounds of Formula (IV) Table 3 (Continued). Table 3 (Continued). Table 3 (Continued).

The compounds of Formula (IV) may possess enhanced pharmacokinetic properties with less drug spiking and less accumulation of toxic metabolites, thereby reducing early onset adverse effects (e.g., anxiety and nausea) and permitting lower dosing regimens which decrease neurotoxicity and cardiovascular adverse events (including tachycardia, hypertension and valvular heart disease) associated with chronic administration. Further, the compounds disclosed herein (e.g., compounds of Formula (IV)) may normalize the different biological effects and metabolic rates between enantiomeric partners to achieve more predictable pharmacokinetic outcomes and a reduction in inter-patient variability.

Formula (V)

In some embodiments, the compound has a structure of Formula (V): or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, wherein: X 1 and X 2 are independently hydrogen, deuterium, or a substituted or unsubstituted C1-C6 alkyl; Y 1 and Y 2 are independently hydrogen, deuterium, or a substituted or unsubstituted C1-C6 alkyl; R 4 and R 5 are independently hydrogen, deuterium, halogen, a substituted or unsubstituted C1-C6 alkyl, -OR a , -SR a , or -SeR a ; or R 4 and R 5 together with the atoms attached thereto are optionally joined to form a heterocycloalkyl or heteroaryl; R 6 and R 7 are independently hydrogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or alternatively R 6 and R 7 together with the nitrogen atom attached thereto are optionally joined to form a substituted or unsubstituted heterocycloalkyl; each R a is independently hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl; and R b is hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl. X 1 and X 2 may be the same, or different. In some embodiments, X 1 and X 2 are the same. In some embodiments, X 1 and X 2 are hydrogen. In some embodiments, X 1 and X 2 are deuterium. In some embodiments, X 1 and X 2 are different. In some embodiments, X 1 is hydrogen or deuterium, and X 2 is a substituted or unsubstituted C 1 -C 6 alkyl. In some embodiments, X 2 is an unsubstituted C 1 -C 6 alkyl, examples of which include, but are not limited to, methyl, ethyl, and n-propyl, preferably methyl. In some embodiments, X 2 is a substituted C 1 -C 6 alkyl. The alkyl group may contain one, or more than one, substituent. For example, when the alkyl group is a C1 alkyl group (i.e., methyl group), the substituted C1 alkyl group may be -CDH2, -CD2H, -CD3, -CFH2, -CF2H, -CF3, etc. In some embodiments, one of X 1 and X 2 is deuterium while the other is hydrogen. Y 1 and Y 2 may be the same, or different. In some embodiments, Y 1 and Y 2 are the same. In some embodiments, Y 1 and Y 2 are hydrogen. In some embodiments, Y 1 and Y 2 are deuterium. In some embodiments, Y 1 and Y 2 are different. In some embodiments, one of Y 1 and Y 2 is deuterium while the other is hydrogen. In some embodiments, Y 1 is a substituted or unsubstituted C1-C6 alkyl. In some embodiments, Y 2 is a substituted or unsubstituted C 1 -C 6 alkyl. In some embodiments, R 4 is deuterium. In some embodiments, R 4 is hydrogen. In some embodiments, R 4 is halogen, for example -Br, -F, -Cl, or -I. In some embodiments, R 4 is an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R 4 is a substituted C1-C6 alkyl. When R 4 is a substituted C1-C6 alkyl, preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl or polyether substituents, cycloalkyl, etc. The alkyl group may contain one, or more than one, substituent. For example, when the alkyl group is a C1 alkyl group (i.e., methyl group), the substituted C 1 alkyl group may be -CDH 2 , -CD 2 H, -CD 3 , -CFH 2 , -CF 2 H, -CF 3 , etc. In some embodiments, R 4 is -OR a , SR a , or -SeR a , wherein R a in R 4 is hydrogen, deuterium, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C 3 -C 10 cycloalkyl. In some embodiments, R 4 is -OR a . In some embodiments, R 4 is -SR a . In some embodiments, R 4 is -SeR a . In some embodiments, R a in R 4 is hydrogen. In some embodiments, R a in R 4 is deuterium. In some embodiments, R a in R 4 is a substituted or unsubstituted C1-C6 alkyl. In some embodiments, R a in R 4 is an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R a in R 4 is a substituted C1-C6 alkyl. Preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), cyano, polar substituents such as hydroxyl or polyether substituents, cycloalkyl, etc. The C1-C6 alkyl group may contain one, or more than one, substituent. In some embodiments, R a in R 4 is a substituted C 1 alkyl group, examples of which may include, but are not limited to, -CDH 2 , -CD 2 H, -CD 3 , -CFH 2 , - CF 2 H, -CF 3 , and -CH 2 8mB( >W \XVO OVLXNSVOW]\& E a in R 4 is a substituted C 2 alkyl group, examples of which may include, but are not limited to, -CDHCDH 2 , -CDHCD 2 H, -CD 2 CD 3 , -CH 2 CFH 2 , -CH 2 CF 2 H, -CH 2 CF 3 , and -CH 2 CH 2 8mB( >W \XVO OVLXNSVOW]\& E a in R 4 is not a substituted C 2 alkyl group such as a C 2 fluoroalkyl group. In some embodiments, R a in R 4 is a substituted C3 alkyl group, examples of which may include, but are not limited to -CH2CH2CF3, -CH2CH2CF2H, -CH2CH2CFH2, -CH2CF2CF2H, and -CH2CH2CH28mB( >W \XVO OVLXNSVOW]\& E a in R 4 is a substituted or unsubstituted alkenyl, e.g., a substituted or unsubstituted allyl, butenyl, crotyl, etc. In some embodiments, R a in R 4 is a substituted or unsubstituted alkynyl, e.g., a substituted or unsubstituted acetylenyl, propargyl, homopropargyl, etc. In some embodiments, R a in R 4 is an unsubstituted alkynyl. In some embodiments, R a in R 4 is an unsubstituted acetylenyl (-Cm8=$( >W \XVO OVLXNSVOW]\& R a in R 4 is an unsubstituted propargyl (-CH 2 8m8=$( >W \XVO OVLXNSVOW]\& E a in R 4 is -CH 2 CH 2 8m8=( >W \XVO OVLXNSVOW]\& E a in R 4 is -CH2CH2CH28m8=( >W \XVO OVLXNSVOW]\& E a in R 4 is -CH2CH2CH2CH28m8=( >W \XVO embodiments, R a in R 4 is a substituted alkynyl. In some embodiments, R a in R 4 is a substituted propargyl (e.g., -CF28m8=$( >W \XVO OVLXNSVOW]\& E a in R 4 is -CF2CH28m8=( >W \XVO OVLXNSVOW]\& E a in R 4 is -CF2CH2CH2Cm8=. In some embodiments, R a in R 4 is -CF2CH2CH2CH2Cm8=. In some embodiments, R a in R 4 is a substituted or unsubstituted cycloalkyl, for example a substituted or unsubstituted C3-C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted C5-C6 cycloalkyl. In some embodiments, R a in R 4 is an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, R a in R 4 is a substituted cycloalkyl (e.g., a substituted C 3 -C 10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 4 is -SMe, -SCD3, -SCF3, -SCF2H, -SCFH2, -SEt, -Sn-Pr, -SCH2CF3, -SCH2CF2H, -SCH2CFH2, -SCH2CH2CF3, -SCH2CH2CF2H, -SCH2CH2CFH2, -SCH2CF2CF2H, -SCH28m8=& 'F8m8=& -SCF28m8=& -SCH2CH2CH28m8=& -SCF2CH2CH2Cm8=, -Me, -CD3, -CF3, -t-Bu, -C(CD3)3, -OMe, -OCD3, -OCF3, -OCF2H, -OCFH2, -OCH2CF3, -OCH2CF2H, -OCH2CFH2, -OCH2CH2CF3, -OCH2CH2CF2H, -OCH2CH2CFH2, -OCH2CF2CF2H, -OCH2C m8= , -OC m8= , -CH2CH2CH2CH2C m8= , -CF 2 CH 2 CH 2 CH 2 Cm8=, -Cl, -I, -Br, -SeMe, -SeCD 3, -SeCF 3, -SeCF 2 H, -SeCFH 2 , -SeEt, -Sen-Pr, -SeCH 2 CF 3 , -SeCH 2 CF 2 H, -SeCH 2 CFH 2 , -SeCH 2 CH 2 CF 3 , -SeCH 2 CH 2 CF 2 H, -SeCH 2 CH 2 CFH 2 , -SeCH 2 CF 2 CF 2 H, -SeCH 2 Cm8=, -SeCm8=, -SeCF 2 Cm8=, -SeCH 2 CH 2 CH 2 Cm8=, or -SeCF 2 CH 2 CH 2 Cm8=. In some embodiments, R 5 is deuterium. In some embodiments, R 5 is hydrogen. In some embodiments, R 5 is halogen, for example -Br, -F, -Cl, or -I. In some embodiments, R 5 is an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R 5 is a substituted C1-C6 alkyl. When R 5 is a substituted C1-C6 alkyl, preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl or polyether substituents, cycloalkyl, etc. The alkyl group may contain one, or more than one, substituent. For example, when the alkyl group is a C1 alkyl group (i.e., methyl group), the substituted C 1 alkyl group may be -CDH 2 , -CD2H, -CD3, -CFH2, -CF2H, -CF3, etc. In some embodiments, R 5 is -OR a , SR a , or -SeR a , wherein R a in R 5 is hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl. In some embodiments, R 5 is -OR a . In some embodiments, R 5 is -SR a . In some embodiments, R 5 is -SeR a . In some embodiments, R a in R 5 is hydrogen. In some embodiments, R a in R 5 is deuterium. In some embodiments, R a in R 5 is a substituted or unsubstituted C1-C6 alkyl. In some embodiments, R a in R 5 is an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R a in R 5 is a substituted C 1 -C 6 alkyl. Preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), cyano, polar substituents such as hydroxyl or polyether substituents, etc. The C 1 -C 6 alkyl group may contain one, or more than one, substituent. In some embodiments, R a in R 5 is a substituted C 1 alkyl group, examples of which may include, but are not limited to, -CDH 2 , -CD 2 H, -CD 3 , -CFH 2 , -CF 2 H, - CF3, and -CH28mB( >W \XVO OVLXNSVOW]\& E a in R 5 is a substituted C2 alkyl group, examples of which may include, but are not limited to, -CDHCDH2, -CDHCD2H, -CD2CD3, -CH2CFH2, -CH2CF2H, - CH2CF3, and -CH2CH28mB( >W \XVO OVLXNSVOW]\& E a in R 5 is not a substituted C2 alkyl group such as a C2 fluoroalkyl group. In some embodiments, R a in R 5 is a substituted C3 alkyl group, examples of which may include, but are not limited to -CH2CH2CF3, -CH2CH2CF2H, -CH2CH2CFH2, -CH2CF2CF2H, and -CH2CH2CH28mB( >W \XVO OVLXNSVOW]\& E a in R 5 is a substituted or unsubstituted alkenyl, e.g., a substituted or unsubstituted allyl, butenyl, crotyl, etc. In some embodiments, R a in R 5 is a substituted or unsubstituted alkynyl, e.g., a substituted or unsubstituted acetylenyl, propargyl, homopropargyl, etc. In some embodiments, R a in R 5 is an unsubstituted alkynyl. In some embodiments, R a in R 5 is an unsubstituted acetylenyl (-Cm8=$( >W some embodiments, R a in R 5 is an unsubstituted propargyl (-CH 2 Cm8=$( In some embodiments, R a in R 5 is -CH 2 CH 2 Cm8=( In some embodiments, R a in R 5 is -CH 2 CH 2 CH 2 8m8=( >W \XVO OVLXNSVOW]\& E a in R 5 is -CH 2 CH 2 CH 2 CH 2 8m8=( >W \XVO embodiments, R a in R 5 is a substituted alkynyl. In some embodiments, R a in R 5 is a substituted propargyl (e.g., -CF2Cm8=$( In some embodiments, R a in R 5 is -CF2CH2Cm8=. In some embodiments, R a in R 5 is -CF2CH2CH28m8=( >W \XVO OVLXNSVOW]\& E a in R 5 is -CF2CH2CH2CH28m8=( >W \XVO embodiments, R a in R 5 is a substituted or unsubstituted cycloalkyl, for example a substituted or unsubstituted C3-C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted C5-C6 cycloalkyl. In some embodiments, R a in R 5 is an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, R a in R 5 is a substituted cycloalkyl (e.g., a substituted C3-C10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 5 is -SMe, -SCD3, -SCF3, -SCF2H, -SCFH2, -SEt, -Sn-Pr, -SCH2CF3, -SCH2CF2H, -SCH2CFH2, -SCH2CH2CF3, -SCH2CH2CF2H, -SCH2CH2CFH2, -SCH 2 CF 2 CF 2 H, -SCH 2 8m8=& 'F8m8=& -SCF 2 8m8=& -SCH 2 CH 2 CH 2 8m8=& -SCF 2 CH 2 CH 2 Cm8=, -Me, -CD 3 , -CF 3 , -t-Bu, -C(CD 3 ) 3 , -OMe, -OCD 3 , -OCF 3 , -OCF 2 H, -OCFH 2 , -OCH 2 CF 3 , -OCH 2 CF 2 H, -OCH 2 CFH 2 , -OCH 2 CH 2 CF 3 , -OCH 2 CH 2 CF 2 H, -OCH 2 CH 2 CFH 2 , -OCH 2 CF 2 CF 2 H, -OCH 2 8m8=& 'C8m8=& '8= 2 CH 2 CH 2 CH 2 8m8=& -CF 2 CH 2 CH 2 CH 2 Cm8=, -Cl, -I, -Br, -SeMe, -SeCD 3, -SeCF 3, -SeCF 2 H, -SeCFH 2 , -SeEt, -Sen-Pr, -SeCH2CF3, -SeCH2CF2H, -SeCH2CFH2, -SeCH2CH2CF3, -SeCH2CH2CF2H, -SeCH2CH2CFH2, -SeCH2CF2CF2H, -SeCH28m8=& 'FO8m8=& -SeCF28m8=& -SeCH2CH2CH2Cm8=, or -SeCF2CH2CH2Cm8=. In some embodiments, R 4 and R 5 together with the atoms attached thereto are joined to form a heterocycloalkyl or heteroaryl, with specific mention being made to a benzo[d][1,3]oxathiole group or a benzo[d][1,3]dioxole group. In embodiments where R 4 and R 5 together with the atoms attached thereto are joined to form a benzo[d][1,3]oxathiole group or a benzo[d][1,3]dioxole group, either the oxathiole ring or the dioxole ring may be further substituted with substituents as defined herein, e.g., with deuterium substituents, with halogen (e.g., fluorine) substituents, etc. R 6 and R 7 may be the same, or different. In some embodiments, R 6 and R 7 are the same. For example, in some embodiments, both R 6 and R 7 are hydrogen. In some embodiments, R 6 and R 7 are different. For example, in some embodiments, R 6 is hydrogen, and R 7 is a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl. R 6 and R 7 may be, independently, hydrogen, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl. In some embodiments, R 6 and R 7 may be, independently, hydrogen, an unsubstituted C1-C6 alkyl (e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, and hexyl) or a C1-C6 alkyl substituted with one or more deuterium (e.g., -CDH 2 , -CD 2 H, -CD 3 ). In some embodiments, R 6 and/or R 7 is an unsubstituted C1-C6 alkyl, for example, an unsubstituted C 1 alkyl, an unsubstituted C 2 alkyl, an unsubstituted C 3 alkyl, an unsubstituted C 4 alkyl, an unsubstituted C5 alkyl, or an unsubstituted C6 alkyl. In some embodiments, R 6 and/or R 7 is an unsubstituted linear C2-C6 alkyl. In some embodiments, R 6 and/or R 7 is an unsubstituted branched C3- C10 alkyl. Examples of an unsubstituted C1-C6 alkyl include, but are not limited to, methyl, ethyl, n- propyl, isopropyl, butyl, isobutyl, t-butyl, n-pentyl, isopentyl, n-hexyl, and isohexyl. In some embodiments, R 6 and/or R 7 is a substituted C1-C6 alkyl, e.g., a substituted C1 alkyl, a substituted C2 alkyl, a substituted C3 alkyl, a substituted C4 alkyl, a substituted C5 alkyl, or a substituted C 6 alkyl. The alkyl group may contain one, or more than one, substituent. The alkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl, oxo, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), unsubstituted alkenyl, substituted alkenyl, unsubstituted alkynyl, substituted alkynyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. In some embodiments, R 6 and/or R 7 is a C1-C6 alkyl substituted with one or more deuterium atoms, examples of which include, but are not limited to, -CDH2, -CD2H, -CD3, -CD2CD3, and -CD2CD2CD3. In some embodiments, R 6 and/or R 7 is a C1-C6 alkyl substituted with one or more fluorine atoms, i.e., is a fluoroalkyl group. Examples of fluoroalkyl groups include, but are not limited to, -CH2F, -CHF2, -CF3, -CH2CH2F, -CH2CHF2, -CH2CF3, -CH2CH2CH2F, -CH2CH2CHF2, -CH2CH2CF3, -CH2CH2CH2CH2F, -CH2CH2CH2CHF2, -CH2CH2CH2CF3, -CH2CF2CHF2, -CH2CF2CF3, -CH(CF3)2, and -CH(CH 3 )CF 3 . In some embodiments, R 6 and/or R 7 is a C 1 -C 6 alkyl substituted with one or more deuterium atoms and one or more fluorine atoms, examples of which include, but are not limited to, - CD 2 CH 2 F, -CD 2 CHF 2 , -CD 2 CF 3 , -CD 2 CH 2 CH 2 F, -CD 2 CH 2 CHF 2 , -CD 2 CH 2 CF 3 , -CD 2 CD 2 CH 2 , -CD 2 CD 2 CHF 2 , -CD 2 CD 2 CF 3 , -CD 2 CH 2 CH 2 CH 2 F, -CD 2 CH 2 CH 2 CHF 2 , -CD 2 CH 2 CH 2 CF 3 , -CD 2 CD 2 CH 2 CH 2 F, -CD 2 CD 2 CH 2 CHF 2 , -CD 2 CD 2 CH 2 CF 3 , -CD2CD2CD2CH2F, -CD2CD2CD2CHF2, and -CD2CD2CD2CF3. In some embodiments, R 6 and/or R 7 is a C1-C6 alkyl substituted with a substituted or unsubstituted cycloalkyl. The C1-C6 alkyl may be substituted with, e.g., a substituted or unsubstituted C3-C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted C5-C6 cycloalkyl. In some embodiments, the C1-C6 alkyl is substituted with an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, the C 1 -C 6 alkyl is substituted with a substituted cycloalkyl (e.g., a substituted C 3 -C 10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 6 and/or R 7 is a C1 alkyl substituted with a substituted or unsubstituted cycloalkyl, with particular mention being made to cyclopropylmethyl (-CH2C3H5). In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted alkenyl, e.g., a substituted or unsubstituted allyl, butenyl, crotyl, etc. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted alkynyl, e.g., a substituted or unsubstituted propargyl. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted cycloalkyl, for example a substituted or unsubstituted C3-C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted C5-C6 cycloalkyl. In some embodiments, R 6 and/or R 7 is an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, R 6 and/or R 7 is a substituted cycloalkyl (e.g., a substituted C3-C10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted heterocycloalkyl. In some embodiments, the unsubstituted or substituted heterocycloalkyl group may be a 3-membered ring, a 4- membered ring, a 5-membered ring, a 6-membered ring, a 7-membered ring, or an 8-membered ring. In some embodiments, R 6 and/or R 7 is an unsubstituted heterocycloalkyl, such as those set forth herein, examples of which include, but are not limited to, aziridine, azetidine, pyrrolidine, isoindole, indole, dihydroindole, indazole, purine, carbazole, carboline, imidazolidine, imidazoline, piperidine, piperazine, indoline, phthalimide, 1,2,3,4-tetrahydroisoquinoline, thiazolidine, morpholine, thiomorpholine, tetrahydrofuran, tetrahydropyran, and 1,3-dioxolane. In some embodiments, R 6 and/or R 7 is a substituted heterocycloalkyl. The substituent(s) may be any recited herein, including, but not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), oxo, and hydroxyl. The heterocycloalkyl group may contain one, or more than one, substituent. In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted aryl. In some embodiments, R 6 and/or R 7 is an unsubstituted aryl, examples of which include, but are not limited to, phenyl and naphthyl. In some embodiments, R 6 and/or R 7 is a substituted aryl. The substituent(s) may be any recited herein, including, but not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The aryl group may contain one, or more than one, substituent.

In some embodiments, R 6 and/or R 7 is a substituted or unsubstituted heteroaryl. In some embodiments, R 6 and/or R 7 is an unsubstituted heteroaryl, examples of which include, but are not limited to, pyrrolyl, furanyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, imidazolyl, triazolyl, tetrazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, indolyl, benzofuranyl, benzothiophenyl, thiophenyl, benzimidazolyl, quinolinyl, isoquinolinyl, quinazolinyl, quinoxalinyl, and pyrazolyl. In some embodiments, R 6 and/or R 7 is a substituted heteroaryl. The substituent(s) may be any recited herein, including, but not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The heteroaryl group may contain one, or more than one, substituent.

In some embodiments, R 6 is hydrogen, and R 7 is a substituted or unsubstituted Ci-Ce alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl. In some embodiments, R 6 is hydrogen, and R 7 is a substituted or unsubstituted Ci-Ce alkyl. In some embodiments, R 6 is hydrogen, and R 7 is an unsubstituted Ci-Ce alkyl, a Ci-C 6 alkyl substituted with one or more deuterium atoms, a Ci-Ce alkyl substituted with one or more fluorine atoms, or a Ci-Ce alkyl substituted with a substituted or unsubstituted cycloalkyl. For example, in some embodiments, R 6 is hydrogen, and R 7 is methyl, ethyl, propyl, -CD3, or cyclopropylmethyl (-CH2C3H5).

In some embodiments, R 6 and R 7 together with the nitrogen atom attached thereto are joined to form a substituted or unsubstituted heterocycloalkyl. In some embodiments, R 6 and R 7 together with the nitrogen atom attached thereto are joined to form an unsubstituted heterocycloalkyl. The unsubstituted heterocycloalkyl group may be, e.g., a 3-membered ring, a 4-membered ring, 5-membered ring, a 6- membered ring, a 7-membered ring, an 8-membered ring, etc., which may be optionally fused to other ring(s). The unsubstituted heterocycloalkyl group contains a minimum of one nitrogen ring atom (the nitrogen atom intervening R 6 and R 7 ), and may optionally contain at least one additional hetero-ring atom, which may be one or more of nitrogen, sulfur, or oxygen, for a total of 1, 2, 3, or 4 hetero-ring atoms (at least one of which is a nitrogen ring atom). Examples of unsubstituted heterocyclo alkyl groups formed from joining R 6 and R 7 together with the nitrogen atom attached thereto include, but are not limited to,

In some embodiments, R 6 and R 7 together with the nitrogen atom attached thereto are joined to form a substituted heterocycloalkyl. The substituted heterocycloalkyl group may be, e.g., a 3-membered ring, a 4-membered ring, a 5-membered ring, a 6-membered ring, a 7-membered ring, an 8-membered ring, etc., which may be optionally fused to other ring(s). The substituted heterocycloalkyl group contains a minimum of one nitrogen ring atom (the nitrogen atom intervening R 6 and R 7 ), and may optionally contain additional hetero-ring atoms (e.g., nitrogen, sulfur, or oxygen) for a total of 1, 2, 3, or 4 hetero-ring atoms (at least one of which is a nitrogen ring atom). Examples of the substituted heterocycloalkyl group include, but are not limited to, aziridine, azetidine, pyrrolidine, isoindole, indole, dihydroindole, indazole, purine, carbazole, carboline, imidazolidine, imidazoline, piperidine, piperazine, indoline, 1,2,3,4-tetrahydroisoquinoline, thiazolidine, morpholine, or thiomorpholine, which is substituted with at least one substituent. The substituent(s) may be any recited herein, including, but not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl, oxo, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), unsubstituted alkyl, substituted alkyl, unsubstituted alkenyl, substituted alkenyl, unsubstituted alkynyl, substituted alkynyl, unsubstituted cycloalkyl, substituted cycloalkyl, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The substituted heterocycloalkyl formed from joining R 6 and R 7 together with the nitrogen atom attached thereto contains a heterocycloalkyl group substituted with one, two, three, four, or more substituents. The substituent may be located on a carbon ring atom or on a hetero-ring atom. Examples of substituted heterocycloalkyl groups formed from joining R 6 and R 7 together with the nitrogen atom attached thereto include, but are not limited to, When present in one or both of R 4 and R 5 , each R a may be, independently, hydrogen, deuterium, a substituted or unsubstituted C1-C6 alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted alkynyl, or a substituted or unsubstituted C3-C10 cycloalkyl. In some embodiments, each R a may be, independently, hydrogen, deuterium, an unsubstituted C1-C6 alkyl (e.g., methyl, ethyl, n- propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl), or a substituted C 1 -C 6 alkyl, with preferred substituents including, but not limited to, deuterium, halogen (e.g., fluorine), polar substituents such as hydroxyl or polyether substituents, etc. In some embodiments, R a is a substituted or unsubstituted C 1 -C 6 alkyl, preferably a C 1 -C 3 alkyl, preferably a substituted or unsubstituted C1 alkyl, examples of which include, but are not limited to, -CH3, -CDH2, -CD2H, -CD3, -CFH 2 , -CF 2 H, -CF 3 . In some embodiments, each R a is -CH 3 . In some embodiments, each R a is -CD 3 . In some embodiments, R a is present in both R 4 and R 5 . In such cases, each R a may be the same, or different. In some embodiments, each R a is the same. In some embodiments, each R a is different, e.g., one R a is -CH3, while another is -CD3. In some embodiments, R a in one or both of R 4 and R 5 is hydrogen. In some embodiments, R a in one or both of R 4 and R 5 is deuterium. In some embodiments, R a in one or both of R 4 and R 5 is a substituted or unsubstituted C1-C6 alkyl. In some embodiments, R a in one or both of R 4 and R 5 is an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R a in one or both of R 4 and R 5 is a substituted C1-C6 alkyl. Preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), cyano, polar substituents such as hydroxyl or polyether substituents, cycloalkyl, etc. The C1-C6 alkyl group may contain one, or more than one, substituent. In some embodiments, R a in one or both of R 4 and R 5 is a substituted C1 alkyl group, examples of which may include, but are not limited to, -CDH 2 , -CD 2 H, -CD 3 , -CFH 2 , -CF 2 H, -CF 3 , and -CH 2 CmB. In some embodiments, R a in one or both of R 4 and R 5 is a substituted C 2 alkyl group, examples of which may include, but are not limited to, -CDHCDH 2 , -CDHCD 2 H, -CD 2 CD 3 , -CH 2 CFH 2 , -CH 2 CF 2 H, -CH 2 CF 3 , and -CH 2 CH 2 8mB( >W \XVO OVLXNSVOW]\& R a in one or both of R 4 and R 5 is a substituted C 3 alkyl group, examples of which may include, but are not limited to -CH2CH2CF3, -CH2CH2CF2H, -CH2CH2CFH2, -CH2CF2CF2H, and -CH2CH2CH28mB( >W some embodiments, R a in one or both of R 4 and R 5 is a substituted or unsubstituted alkenyl, e.g., a substituted or unsubstituted allyl, butenyl, crotyl, etc. In some embodiments, R a in one or both of R 4 and R 5 is a substituted or unsubstituted alkynyl, e.g., a substituted or unsubstituted acetylenyl, propargyl, homopropargyl, etc. In some embodiments, R a in one or both of R 4 and R 5 is an unsubstituted alkynyl. In some embodiments, R a in one or both of R 4 and R 5 is an unsubstituted acetylenyl (-Cm8=$( >W \XVO embodiments, R a in one or both of R 4 and R 5 is an unsubstituted propargyl (-CH28m8=$( >W \XVO embodiments, R a in one or both of R 4 and R 5 is -CH2CH2C m8=( In some embodiments, R a in one or both of R 4 and R 5 is -CH 2 CH 2 CH 2 8m8=( >W \XVO OVLXNSVOW]\& E a in one or both of R 4 and R 5 is -CH 2 CH 2 CH 2 CH 2 Cm8=. In some embodiments, R a in one or both of R 4 and R 5 is a substituted alkynyl. In some embodiments, R a in one or both of R 4 and R 5 is a substituted propargyl (e.g., -CF 2 Cm8=$( In some embodiments, R a in one or both of R 4 and R 5 is -CF2CH2Cm8=. In some embodiments, R a in one or both of R 4 and R 5 is -CF2CH2CH2Cm8=. In some embodiments, R a in one or both of R 4 and R 5 is -CF2CH2CH2CH2Cm8=. In some embodiments, R a in one or both of R 4 and R 5 is a substituted or unsubstituted cycloalkyl, for example a substituted or unsubstituted C3-C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted C5-C6 cycloalkyl. In some embodiments, R a in one or both of R 4 and R 5 is an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, R a in one or both of R 4 and R 5 is a substituted cycloalkyl (e.g., a substituted C 3 -C 10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In line with the above description for R a , and any one or both of R 4 to R 5 may be, independent of each other, -OR a , -SR a , or -SeR a , examples of which include, but are not limited to, -SMe, -SCD3, -SCF3, -SCF2H, -SCFH2, -SEt, -Sn-Pr, -SCH2CF3, -SCH2CF2H, -SCH 2 CFH 2 , -SCH 2 CH 2 CF 3 , -SCH 2 CH 2 CF 2 H, -SCH 2 CH 2 CFH 2 , -SCH 2 CF 2 CF 2 H, -SCH 2 8m8=& -SCm8=, -SCF 2 Cm8=, -SCH 2 CH 2 CH 2 Cm8=, -SCF 2 CH 2 CH 2 Cm8=, -OMe, -OCD 3 , -OCF 3 , -OCF 2 H, -OCFH 2 , -OCH 2 CF 3 , -OCH 2 CF 2 H, -OCH 2 CFH 2 , -OCH 2 CH 2 CF 3 , -OCH 2 CH 2 CF 2 H, -OCH 2 CH 2 CFH 2 , -OCH 2 CF 2 CF 2 H, -OCH 2 8m8=& 'C8m8=& 'FOAO& 'FO89 3, -SeCF 3, -SeCF 2 H, -SeCFH 2 , -SeEt, -Sen-Pr, -SeCH 2 CF 3 , -SeCH 2 CF 2 H, -SeCH 2 CFH 2 , -SeCH 2 CH 2 CF 3 , -SeCH2CH2CF2H, -SeCH2CH2CFH2, -SeCH2CF2CF2H, -SeCH28m8=& 'FO8m8=& 'FO8;28m8=& -SeCH2CH2CH28m8=& X[ 'FO8;2CH2CH28m8=( In some embodiments, R b is hydrogen. In some embodiments, R b is deuterium. In some embodiments, R b is a substituted or unsubstituted C1-C6 alkyl. In some embodiments, R b is an unsubstituted C1-C6 alkyl, examples of which include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, and hexyl. In some embodiments, R b is a substituted C1-C6 alkyl. Preferred substituents may include, but are not limited to, deuterium, halogen (e.g., fluorine), cyano, polar substituents such as hydroxyl or polyether substituents, cycloalkyl etc. The C 1 -C 6 alkyl group may contain one, or more than one, substituent. In some embodiments, R b is a substituted C 1 alkyl group, examples of which may include, but are not limited to, -CDH 2 , -CD 2 H, - CD 3 , -CFH 2 , -CF 2 H, -CF 3 , and -CH 2 CmB. In some embodiments, R b is a substituted C 2 alkyl group, examples of which may include, but are not limited to, -CDHCDH 2 , -CDHCD 2 H, -CD 2 CD 3 , -CH 2 CFH 2 , -CH 2 CF 2 H, -CH 2 CF 3 , and -CH2CH2CmB. In some embodiments, R b is not a substituted C2 alkyl group such as a C2 fluoroalkyl group. In some embodiments, R b is a substituted C3 alkyl group, examples of which may include, but are not limited to -CH2CH2CF3, -CH2CH2CF2H, -CH2CH2CFH2, -CH2CF2CF2H, and -CH2CH2CH2CmB. In some embodiments, R b is a substituted or unsubstituted alkenyl, e.g., a substituted or unsubstituted allyl, butenyl, crotyl, etc. In some embodiments, R b is a substituted or unsubstituted alkynyl, e.g., a substituted or unsubstituted acetylenyl, propargyl, homopropargyl, etc. In some embodiments, R b is an unsubstituted alkynyl. In some embodiments, R b is KW ^W\^L\]S]^]ON KMO]bUOWbU #'8m8=$( >W \XVO OVLXNSVOW]\& E b is an unsubstituted propargyl (-CH28m8=$( >W \XVO OVLXNSVOW]\& E b is -CH2CH28m8=( >W \XVO OVLXNSVOW]\& E b is -CH 2 CH 2 CH 2 Cm8=. In some embodiments, R b is -CH 2 CH 2 CH 2 CH 2 Cm8=. In some embodiments, R b is a substituted alkynyl. In some embodiments, R b is a substituted propargyl (e.g., -CF28m8=$( >W \XVO embodiments, R b is -CF2CH2Cm8=. In some embodiments, R b is -CF2CH2CH2Cm8=. In some embodiments, R b is -CF2CH2CH2CH2Cm8=. In some embodiments, R b is a substituted or unsubstituted cycloalkyl, for example a substituted or unsubstituted C3-C10 cycloalkyl, or a substituted or unsubstituted C4-C8 cycloalkyl, or a substituted or unsubstituted C5-C6 cycloalkyl. In some embodiments, R b is an unsubstituted cycloalkyl (e.g., an unsubstituted C3-C10 cycloalkyl), examples of which may include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, R b is a substituted cycloalkyl (e.g., a substituted C 3 - C 10 cycloalkyl). The cycloalkyl group may be substituted with any one or more substituents recited herein, examples of those substituents may include, but are not limited to, deuterium, unsubstituted alkyl, substituted alkyl, unsubstituted alkoxy, substituted alkoxy (e.g., polyether groups), halogen (e.g., fluorine), hydroxyl, oxo, unsubstituted heterocycloalkyl, substituted heterocycloalkyl, unsubstituted aryl, substituted aryl, unsubstituted heteroaryl, and substituted heteroaryl. The cycloalkyl group may contain one, or more than one, substituent. In some embodiments, at least one of X 1 , X 2 , Y 1 , Y 2 , R 4 , R 5 , R 7 , R a , and R b comprises deuterium. In some embodiments, R 4 is -OR a , -SR a , or -SR a , with R a in R 4 being an unsubstituted C1-C6 alkyl (i.e., R 4 is an -O-C1-C6 alkyl group; an -S-C1-C6 alkyl group; or an -Se-C1-C6 alkyl group). In some embodiments, R 4 is -OR a , -SR a , or -SR a , with R a in R 4 being a C1-C6 alkyl substituted with one or more halogen (i.e., R 4 is an -O-C1-C6 alkyl group, the alkyl group being substituted with one or more halogen; an -S-C 1 -C 6 alkyl group, the alkyl group being substituted with one or more halogen; or an -Se-C 1 -C 6 alkyl group, the alkyl group being substituted with one or more halogen). In some embodiments, R 5 is -OR a , -SR a , or -SR a , with R a in R 5 being an unsubstituted C 1 -C 6 alkyl (i.e., R 4 is an -O-C 1 -C 6 alkyl group; an -S-C 1 -C 6 alkyl group; or an -Se-C 1 -C 6 alkyl group). In some embodiments, R 5 is -OR a , -SR a , or -SR a , with R a in R 5 being a C 1 -C 6 alkyl substituted with one or more halogen (i.e., R 4 is an -O-C 1 -C 6 alkyl group, the alkyl group being substituted with one or more halogen; an -S-C1-C6 alkyl group, the alkyl group being substituted with one or more halogen; or an -Se-C1-C6 alkyl group, the alkyl group being substituted with one or more halogen). In some embodiments, R b comprises deuterium. In some embodiments, R b is a C1-C6 alkyl substituted with one or more deuterium. In some embodiments, R b is a C1-C6 alkyl substituted with one or more halogen (e.g., -CF3 or -CF2H). In some embodiments, the compound, e.g., the compound of Formula (V), is selected from the group consisting of: pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof.

The compounds of Formula (I) through (V) may contain a stereogenic center. In such cases, the compounds may exist as different stereoisomeric forms, even though Formulas (I) through (V), and compounds belonging thereto, are drawn without reference to stereochemistry. Accordingly, the present disclosure includes all possible stereoisomers and includes not only racemic compounds but the individual enantiomers (enantiomerically pure compounds) and their non-racemic mixtures as well.

When a compound is desired as a single enantiomer, such may be obtained by stereospecific synthesis, by resolution of the final product or any convenient intermediate, or by chiral chromatographic methods as each are known in the art. Resolution of the final product, an intermediate, or a starting material may be performed by any suitable method known in the art. In some embodiments, the compounds described herein, e.g., a compound of Formula (I) through

(V), are racemic. In some embodiments, the compounds described herein, e.g., a compound of Formula (I) through (V), are enantiomerically pure. In some embodiments, the compounds described herein, e.g., a compound of Formula (I) through (V) are non-stereogenic (achiral). In some embodiments, the compound is an agonist of a serotonin 5-HT2 receptor. In some embodiments, the compound can be an agonist of a serotonin 5-HT2A receptor. In some embodiments, the compound is a modulator of a monoamine transporter. In some embodiments, the compound is a modulator of a serotonin transporter (SERT). In some embodiments, the compound is a modulator of a norepinephrine transporter (NET). In some embodiments, the compound is a modulator of a dopamine transporter (DAT). In some embodiments, administration of the compound elicits psychedelic effects. In some embodiments, administration of the compound does not elicit psychedelic effects (e.g., at dosages that would classically produce psychedelic effects). In some embodiments, administration of the compound elicits entactogenic effects.

Also disclosed herein is a pharmaceutically acceptable salt of the compounds of the present disclosure, e.g., a compound of Formula (I) through (V). The acid used to form the pharmaceutically acceptable salt of the compound of Formula (I) through (V) may be a monoacid, a diacid, a triacid, a tetraacid, or may contain a higher number of acid groups. The acid groups may be, e.g., a carboxylic acid, a sulfonic acid, a phosphonic acid, or other acidic moieties containing at least one replaceable hydrogen atom. Examples of acids for use in the preparation of the pharmaceutically acceptable (acid addition) salts disclosed herein include, but are not limited to, acetic acid, 2,2-dichloroacetic acid, phenylacetic acid, acylated amino acids, alginic acid, ascorbic acid, L-aspartic acid, sulfonic acids (e.g., benzenesulfonic acid, camphorsulfonic acid, (+)-(lS)-camphor-10-sulfonic acid, ethane- 1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, methanesulfonic acid, naphthalene-2-sulfonic acid, naphthalene- 1,5-disulfonic acid, p-toluenesulfonic acid, ethanedisulfonic acid, etc.), benzoic acids (e.g., benzoic acid, 4-acetamidobenzoic acid, 2-acetoxybenzoic acid, salicylic acid, 4-amino-salicylic acid, gentisic acid, etc.), boric acid, (+)-camphoric acid, cinnamic acid, citric acid, cyclamic acid, cyclohexanesulfamic acid, dodecylsulfuric acid, formic acid, fumaric acid, galactaric acid, glucoheptonic acid, D-gluconic acid, D-glucuronic acid, L-glutamic acid, a-oxo-glutaric acid, glycolic acid, hippuric acid, hydrobromic acid, hydrochloric acid, hydroiodic acid, (+)-L-lactic acid, (-)-D-lactic acid, (±)-DL-lactic acid, lactobionic acid, maleic acid, malic acid, (-)-L-malic acid, (+)-D-malic acid, hydroxymaleic acid, malonic acid, (±)-DL-mandelic acid, isethionic acid, l-hydroxy-2-naphthoic acid, nicotinic acid, nitric acid, orotic acid, oxalic acid, pamoic acid, perchloric acid, phosphoric acid, L- pyroglutamic acid, saccharic acid, succinic acid, sulfuric acid, sulfamic acid, tannic acid, tartaric acids (e.g., DL-tartaric acid, (+)-L-tartaric acid, (-)-D-tartaric acid), thiocyanic acid, propionic acid, valeric acid, and fatty acids (including fatty mono- and di- acids, e.g., adipic (hexandioic) acid, lauric (dodecanoic) acid, linoleic acid, myristic (tetradecanoic) acid, capric (decanoic) acid, stearic (octadecanoic) acid, oleic acid, caprylic (octanoic) acid, palmitic (hexadecenoic) acid, sebacic acid, undecylenic acid, caproic acid, etc.). In some embodiments, the pharmaceutically acceptable salt of the compound of Formula (I) through (V) is a fatty acid salt. The fatty acid used to make the fatty acid salt of the compound of Formula (I) through (V) may be a fatty monoacid or a fatty diacid, and may contain a fatty hydrocarbon portion made up of hydrogen and anywhere from 4, from 6, from 8, from 10, from 12, from 14, from 16, and up to 26, up to 24, up to 22, up to 20, up to 18 carbon atoms, which may be fully saturated or partially unsaturated. In some embodiments, the pharmaceutically acceptable salt of the compound of Formula (I) through (V) is an adipate salt, a laurate salt, a linoleate salt, a myristate salt, a caprate salt, a stearate salt, an oleate salt, a caprylate salt, a palmitate salt, a sebacate salt, an undecylenate salt, or a caproate salt of the compound of Formula (I) through (V).

Methods for preparing pharmaceutically acceptable salt forms of pharmaceutical compounds are known by those of ordinary skill in the art. In some embodiments, the method includes:

(a) suspending the compound of Formula (I) through (V) in a solvent or mixture of solvents;

(b) contacting an acid with the compound of Formula (I) through (V) to provide a mixture;

(c) optionally heating the mixture;

(d) optionally cooling the mixture; and

(e) isolating the salt.

Various solvents may be used in the disclosed methods, including one or more protic solvents, one or more aprotic solvents, or mixtures thereof. In some embodiments, the solvent(s) used in the method of preparing the salt is/are a protic solvent(s). In some embodiments, the solvent used in the method of preparing the salt is selected from the group consisting of methanol, ethanol, propanol, isopropanol (IP A), butanol, 2-butanol, acetone, butanone, dioxanes (1,4-dioxane), water, tetrahydrofuran (THF), acetonitrile (MeCN), ether solvents (e.g., t-butylmethyl ether (TBME)), hexane, heptane, octane, and combinations thereof. In some embodiments, the solvent is ethanol. In some embodiments, the solvent is 1,4-dioxane. In some embodiments, the solvent is acetonitrile. In some embodiments, the solvent is tetrahydrofuran.

Suitable acids for use in the preparation of pharmaceutically acceptable acid addition salts may include those described heretofore. The acid may be an inorganic acid such as hydrochloric acid, or an organic acid, with organic acids being preferred. In some embodiments, the acid is an organic acid selected from the group consisting of ascorbic acid, citric acid, fumaric acid, maleic acid, malonic acid, (-)-L-malic acid, (+)-L-tartaric acid, methanesulfonic acid, benzenesulfonic acid, toluenesulfonic acid, benzoic acid, salicylic acid, succinic acid, oxalic acid, D-glucuronic acid, glutaric acid salt, and acetic acid. In some embodiments, the acid is an organic acid selected from the group consisting of benzenesulfonic acid, (+)-L-tartaric acid, fumaric acid, acetic acid, citric acid, malonic acid, succinic acid, oxalic acid, benzoic acid, and salicylic acid. In some embodiments, the acid is a fatty acid, such as adipic (hexandioic) acid, lauric (dodecanoic) acid, linoleic acid, myristic (tetradecanoic) acid, capric (decanoic) acid, stearic (octadecanoic) acid, oleic acid, caprylic (octanoic) acid, palmitic (hexadecenoic) acid, sebacic acid, undecylenic acid, caproic acid, etc., with particular mention being made to adipic (hexandioic) acid, lauric (dodecanoic) acid, linoleic acid, myristic (tetradecanoic) acid, capric (decanoic) acid, stearic (octadecanoic) acid, oleic acid, and caprylic (octanoic) acid.

In some embodiments, a stoichiometric (or superstoichiometric) quantity of the acid is contacted with the compound of Formula (I) through (V). In some embodiments, a sub-stoichiometric (e.g., 0.5 molar equivalents) quantity of the acid is contacted with the compound of Formula (I) through (V). The use of sub- stoichiometric quantities of the acid may be desirable when, for example, the acid contains at least two acidic protons (e.g., two or more carboxylic acid groups) and the target salt is a hemi-acid salt.

In some embodiments, the mixture is heated, e.g., refluxed, prior to cooling.

In some embodiments, the mixture is cooled and the salt is precipitated out of the solution. In some embodiments, the salt is precipitated out of solution in crystalline form. In some embodiments, the salt is precipitated out of solution in amorphous form.

Isolation of the salt may be performed by various well-known isolation techniques, such as filtration, decantation, and the like. In some embodiments, the isolating step includes filtering the mixture.

After isolation, additional crystallization and/or recrystallization steps may also optionally be performed, if desired, for example to increase purity, crystallinity, etc.

In some embodiments, compounds of the present disclosure, e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof, is in the form of a solvate. Examples of solvate forms include, but are not limited to, hydrates, methanolates, ethanolates, isopropanolates, etc., with hydrates and ethanolates being preferred. The solvate may be formed from stoichiometric or nonstoichiometric quantities of solvent molecules. Solvates of the compounds herein may be in the form of isolable solvates. In one non-limiting example, as a hydrate, the compound may be a monohydrate, a dihydrate, etc. Solvates of the compounds herein also include solution-phase forms. Thus, in some embodiments, the present disclosure provides solution-phase compositions of the compounds of the present disclosure, or any pharmaceutically acceptable salts thereof, which are in solvated form, preferably fully solvated form.

In some embodiments, the compound of the present disclosure, e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, is provided in crystalline form, e.g., as determined by XRPD. Accordingly, pharmaceutical compositions may be prepared from a compound of Formula (I) through (V), in crystalline form including in one or more polymorphic forms, and may be used for treatment as set forth herein. Crystalline forms may be advantageous in terms of stability and providing well-defined physical properties, which is desirable for pharmaceutical preparation and administration.

In some embodiments, the compound of the present disclosure, e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, is provided in amorphous form, e.g., as determined by XRPD. Accordingly, pharmaceutical compositions may be prepared from a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, in one or more amorphic forms, and may be used for treatment as set forth herein. Amorphous forms typically possess higher aqueous solubility and rates of dissolution compared to their crystalline counterparts, and thus may be well suited for quick acting dosage forms adapted to rapidly release the active agent, such as for orodispersible dosage forms (ODxs), immediate release (IR) dosage forms, and the like.

Compounds of the present disclosure, e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, may generally be prepared according to, or analogous to, the synthetic routes exemplified herein. Other synthetic routes may also be used according to techniques and procedures known to those of ordinary skill in the art.

Therapeutic applications and methods

Also disclosed herein is a method of treating a subject with a disease or disorder comprising administering to the subject a therapeutically effective amount of a compound as disclosed herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof). In some embodiments, the disease or disorder is associated with a serotonin 5-HT2 receptor. In some embodiments, the disease or disorder is associated with a monoamine transporter.

The dosage and frequency (single or multiple doses) of the compounds administered herein can vary depending upon a variety of factors, including, but not limited to, the compound to be administered; the disease/condition being treated; route of administration; size, age, sex, health, body weight, body mass index, and diet of the subject; nature and extent of symptoms of the disease being treated; presence of other diseases or other health-related problems; kind of concurrent treatment; and complications from any disease or treatment regimen. Other therapeutic regimens or agents can be used in conjunction with the methods and compounds disclosed herein.

Therapeutically effective amounts for use in humans may be determined from animal models. For example, a dose for humans can be formulated to achieve a concentration that has been found to be effective in animals. The dosage in humans can be adjusted by monitoring response to the treatment and adjusting the dosage upwards (e.g., up-titration) or downwards (e.g., down-titration). Dosages may be varied depending upon the requirements of the subject and the compound being employed. The dose administered to a subject, in the context of the pharmaceutical compositions presented herein, should be sufficient to effect a beneficial therapeutic response in the subject over time. The size of the dose also will be determined by the existence, nature, and extent of any adverse side effects. Generally, treatment is initiated with smaller dosages, which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under circumstances is reached.

Dosage amounts and intervals can be adjusted individually to provide levels of the administered compounds effective for the particular clinical indication being treated. This will provide a therapeutic regimen that is commensurate with the severity of the individual’s disease state.

Routes of administration may include oral routes (e.g., enteral/gastric delivery, intraoral administration such buccal, lingual, and sublingual routes), parenteral routes (e.g., intravenous, intradermal, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial, and subcutaneous administration), topical routes (e.g., conjuctival, intracorneal, intraocular, ophthalmic, auricular, transdermal, nasal (e.g., intranasal), vaginal, uretheral, respiratory, and rectal administration), inhalation, or others sufficient to affect a beneficial therapeutic response.

Administration may follow a continuous administration schedule, or an intermittent administration schedule. The administration schedule may be varied depending on the active ingredient(s) employed, the condition being treated, the administration route, etc. For example, administration of a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, may be performed once a day (QD), or in divided dosages throughout the day, such as 2-times a day (BID), 3-times a day (TID), 4-times a day (QID), or more. In some embodiments administration may be performed nightly (QHS). In some embodiments, administration is performed as needed (PRN). Administration may also be performed on a weekly basis, e.g., once a week, twice a week, three times a week, four times a week, every other week, every two weeks, etc., or less. The administration schedule may also designate a defined number of treatments per treatment course, for example, the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, may be administered 1 time, 2 times, 3 times, 4 times, 5 times, 6 times, 7 times, or 8 times per treatment course. Other administration schedules may also be deemed appropriate using sound medical judgement.

The dosing can be continuous (7 days of administration in a week) or intermittent, for example, depending on the pharmacokinetics and a particular subject’s clearance/accumulation of the drug. If intermittently, the schedule may be, for example, 4 days of administration and 3 days off (rest days) in a week or any other intermittent dosing schedule deemed appropriate using sound medical judgement. For example, intermittent dosing may involve administration of a single dose within a treatment course. The dosing whether continuous or intermittent is continued for a particular treatment course, typically at least a 28-day cycle (1 month), which can be repeated with or without a drug holiday. Longer or shorter courses can also be used such as 14 days, 18 days, 21 days, 24 days, 35 days, 42 days, 48 days, or longer, or any range therebetween. The course may be repeated without a drug holiday or with a drug holiday depending upon the subject. Other schedules are possible depending upon the presence or absence of adverse events, response to the treatment, patient convenience, and the like.

Utilizing the teachings provided herein, an effective prophylactic or therapeutic treatment regimen can be planned that does not cause substantial toxicity and yet is entirely effective to treat the clinical symptoms demonstrated by the particular patient. This planning should involve the careful choice of active compound by considering factors such as compound potency, relative bioavailability, patient body weight, presence and severity of adverse side effects, preferred mode of administration, and the toxicity profile of the selected agent.

A therapeutically effective dose of the compounds of the present disclosure may vary depending on the variety of factors described above, but is typically that which provides the compound of Formula (I) through (V) in an amount of about 0.00001 mg to about 10 mg per kilogram body weight of the subject, or any range in between, e.g., about 0.00001 mg/kg, about 0.00005 mg/kg, about 0.0001 mg/kg, about 0.0005 mg/kg, about 0.001 mg/kg, about 0.005 mg/kg, about 0.01 mg/kg, about 0.05 mg/kg, about

0.1 mg/kg, about 0.2 mg/kg, about 0.3 mg/kg, about 0.4 mg/kg, about 0.5 mg/kg, about 0.6 mg/kg, about

0.7 mg/kg, about 0.8 mg/kg, about 0.9 mg/kg, about 1.0 mg/kg, about 2.0 mg/kg, about 3.0 mg/kg, about

4.0 mg/kg, about 5.0 mg/kg, about 6.0 mg/kg, about 7.0 mg/kg, about 8.0 mg/kg, about 9.0 mg/kg, about

10.0 mg/kg of the compound of Formula (I) through (V) (on an active basis).

The compounds of the present disclosure (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof) may be administered at a psychedelic dose. Psychedelic dosing, by mouth or otherwise, may in some embodiments range from about 0.3 mg/kg, about 0.35 mg/kg, about 0.4 mg/kg, about 0.45 mg/kg, about 0.5 mg/kg, and up to about 5 mg/kg, about 4 mg/kg, about 3 mg/kg, about 2 mg/kg, about 1 mg/kg, about 0.95 mg/kg, about 0.9 mg/kg, about 0.85 mg/kg, about 0.8 mg/kg, about 0.75 mg/kg, about 0.7 mg/kg, about 0.65 mg/kg, about 0.6 mg/kg, about 0.55 mg/kg of the compound of Formula (I) through (V) (on an active basis). Higher dosing may also be used in some embodiments, as described above. In some embodiments, psychedelic doses are administered once by mouth or otherwise, with the possibility of repeat doses at least one week apart. In some instances, no more than 5 doses are given in any one course of treatment. Courses can be repeated as necessary, with or without a drug holiday. Such acute treatment regimens may be accompanied by psychotherapy, before, during, and/or after the psychedelic dose. These treatments are appropriate for a variety of mental health disorders disclosed herein, examples of which include, but are not limited to, major depressive disorder (MDD), therapy resistant depression (TRD), anxiety disorders, and substance use disorders (e.g., alcohol use disorder, opioid use disorder, amphetamine use disorder, nicotine use disorder, smoking, and cocaine use disorder).

The compounds of the present disclosure (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), may be administered at subpsychoactive (yet still potentially serotonergic) concentrations to achieve durable therapeutic benefits, with decreased toxicity, and may thus be suitable for microdosing. Sub-psychedelic dosing, by mouth or otherwise, may in some embodiments range from about 0.00001 mg/kg, about 0.00005 mg/kg, about 0.0001 mg/kg, about 0.0005 mg/kg, about 0.001 mg/kg, about 0.005 mg/kg, about 0.006 mg/kg, about 0.008 mg/kg, about 0.009 mg/kg, about 0.01 mg/kg, and up to about 0.3 mg/kg, about 0.25 mg/kg, about 0.2 mg/kg, about 0.15 mg/kg, about 0.1 mg/kg, about 0.083 mg/kg, about 0.08 mg/kg, about 0.075 mg/kg, about 0.07 mg/kg, about 0.06 mg/kg, about 0.05 mg/kg, about 0.04 mg/kg, about 0.03 mg/kg, about 0.02 mg/kg of the compound of Formula (I) through (V) (on an active basis). Typically, subpsychedelic doses are administered orally up to every day, for a treatment course (e.g., 1 month). However, there is no limitation on the number of doses at sub-psychedelic doses — dosing can be less frequent or more frequent as deemed appropriate. Courses can be repeated as necessary, with or without a drug holiday.

Sub-psychedelic dosing can also be carried out, for example, by transdermal delivery, subcutaneous administration, etc., via modified, controlled, slow, or extended release dosage forms, including, but not limited to, depot dosage forms, implants, patches, and pumps, which can be optionally remotely controlled. Here, doses would achieve similar blood levels as low oral dosing, but would nevertheless be sub-psychedelic.

Sub-psychedelic doses can be used, e.g., for the chronic treatment or maintenance of a variety of diseases or disorders disclosed herein, examples of which include, but are not limited to, depression (e.g., MDD), inflammation, pain, and neuroinflammation.

The compounds of the present disclosure (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), may be used for a maintenance regimen. As used herein, a “maintenance regimen” generally refers to the administration of the compounds of the present disclosure (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof) following achievement of a target dose, e.g., following completion of an up-titration regimen, and/or following a positive clinical response, e.g., improvement of the patient's condition, either to the same drug or to a different drug. In some embodiments, the patient is administered a first drug for a therapeutic regimen and a second drug for a maintenance regimen, wherein the first and second drugs are different. For example, the patient may be administered a therapeutic regimen of a first drug which is not a compound of the present disclosure (e.g., the first drug is a serotonergic psychedelic such as LSD, psilocybin, MDMA, dimethyltryptamine, etc., or a non-psychedelic drug), followed by a compound of the present disclosure (as the second drug) in a maintenance regimen. In another example, a different compound of the present disclosure is used for the therapeutic regimen (first drug) than is used for the maintenance regimen (second drug). In some embodiments, the patient is administered the same compound of the present disclosure for both a therapeutic regimen and a maintenance regimen. In any case, the maintenance dose of the compounds of the present disclosure may be used to ‘maintain’ the therapeutic response and/or to prevent occurrences of relapse. When the same compound of the present disclosure is used for both the original therapeutic regimen and for the maintenance regimen, the maintenance dose of the compound may be at or below the therapeutic dose. In some embodiments, the maintenance dose is a psychedelic dose. In some embodiments, the maintenance dose is a sub-psychedelic dose. Generally, dosing is carried out daily or intermittently for the maintenance regimen, however, maintenance regimens can also be carried out continuously, for example, over several days, weeks, months, or years. Moreover, the maintenance dose may be given to a patient over a long period of time, even chronically.

The subjects treated herein may have a disease or disorder associated with a serotonin 5-HT2 receptor. The subjects treated herein may have a disease or disorder associated with a monoamine transporter.

In some embodiments, the disease or disorder is a neuropsychiatric disease or disorder or an inflammatory disease or disorder. In some embodiments, the neuropsychiatric disease or disorder is not schizophrenia or cognitive deficits in schizophrenia.

In some embodiments, the disease or disorder is a central nervous system (CNS) disorder, including, but not limited to, major depressive disorder (MDD), treatment-resistant depression (TRD), post-traumatic stress disorder (PTSD), bipolar and related disorders (including, but not limited to, bipolar I disorder, bipolar II disorder, cyclothymic disorder), obsessive-compulsive disorder (OCD), generalized anxiety disorder (GAD), social anxiety disorder, substance use disorders (including, but not limited to, alcohol use disorder, opioid use disorder, amphetamine use disorder, nicotine use disorder, smoking, and cocaine use disorder), eating disorders (including, but not limited to anorexia nervosa, bulimia nervosa, binge-eating disorder, etc.), Alzheimer’s disease, cluster headache and migraine, attention deficit hyperactivity disorder (ADHD), pain and neuropathic pain, aphantasia, childhood-onset fluency disorder, major neurocognitive disorder, mild neurocognitive disorder, suicidal ideation, suicidal behavior, major depressive disorder with suicidal ideation or suicidal behavior, melancholic depression, atypical depression, dysthymia, non-suicidal self-injury disorder (NSSID), chronic fatigue syndrome, Lyme disease, gambling disorder, paraphilic disorders (including, but not limited to, pedophilic disorder, exhibitionistic disorder, voyeuristic disorder, fetishistic disorder, sexual masochism or sadism disorder, and transvestic disorder, etc.), sexual dysfunction (e.g., low libido, hypoactive sexual desire disorder (HSDD), etc.), peripheral neuropathy, obesity, and persistent symptoms from a SARS- CoV-2 infection (COVID-19), e.g. “long covid.”

In some embodiments, the methods provided herein are used to treat a subject with a depressive disorder. As used herein, the terms “depressive disorder” or “depression” refers to a group of disorders characterized by low mood that can affect a person’s thoughts, behavior, feelings, and sense of wellbeing lasting for a period of time. In some embodiments, the depressive disorder disrupts the physical and psychological functions of a person. In some embodiments, the depressive disorder causes a physical symptom such as weight loss, aches or pains, headaches, cramps, or digestive problems. In some embodiments, the depressive disorder causes a psychological symptom such as persistent sadness, anxiety, feelings of hopelessness and irritability, feelings of guilt, worthlessness, or helplessness, loss of interest or pleasure in hobbies and activities, difficulty concentrating, remembering, or making decisions. In some embodiments, the depressive disorder is major depressive disorder (MDD), atypical depression, bipolar disorder, catatonic depression, depressive disorder due to a medical condition, postpartum depression, premenstrual dysphoric disorder, seasonal affective disorder, or treatmentresistant depression (TRD).

In some embodiments, the disease or disorder is major depressive disorder (MDD). As used herein, the term “major depressive disorder” refers to a condition characterized by a time period of low mood that is present across most situations. Major depressive disorder is often accompanied by low self- esteem, loss of interest in normally enjoyable activities, low energy, and pain without a clear cause. In some instances, major depressive order is characterized by symptoms of depression lasting at least two weeks. In some instances, an individual experiences periods of depression separated by years. In some instances, an individual experiences symptoms of depression that are nearly always present. Major depressive disorder can negatively affect a person’s personal, work, or school life, as well as sleeping, eating habits, and general health. Approximately 2-7% of adults with major depressive disorder commit suicide, and up to 60% of people who commit suicide had major depressive disorder or another related mood disorder. Dysthymia is a subtype of major depressive disorder consisting of the same cognitive and physical problems as major depressive disorder with less severe but longer-lasting symptoms. Exemplary symptoms of a major depressive disorder include, but are not limited to, feelings of sadness, tearfulness, emptiness or hopelessness, angry outbursts, irritability or frustration, even over small matters, loss of interest or pleasure in most or all normal activities, sleep disturbances, including insomnia or sleeping too much, tiredness and lack of energy, reduced appetite, weight loss or gain, anxiety, agitation or restlessness, slowed thinking, speaking, or body movements, feelings of worthlessness or guilt, fixating on past failures or self-blame, trouble thinking, concentrating, making decisions, and remembering things, frequent thoughts of death, suicidal thoughts, suicide attempts, or suicide, and unexplained physical problems, such as back pain or headaches.

As used herein, the term “atypical depression” refers to a condition wherein an individual shows signs of mood reactivity (i.e., mood brightens in response to actual or potential positive events), significant weight gain, increase in appetite, hypersomnia, heavy, leaden feelings in arms or legs, and/or long-standing pattern of interpersonal rejection sensitivity that results in significant social or occupational impairment. Exemplary symptoms of atypical depression include, but are not limited to, daily sadness or depressed mood, loss of enjoyment in things that were once pleasurable, major changes in weight (gain or loss) or appetite, insomnia or excessive sleep almost every day, a state of physical restlessness or being rundown that is noticeable by others, daily fatigue or loss of energy, feelings of hopelessness, worthlessness, or excessive guilt almost every day, problems with concentration or making decisions almost every day, recurring thoughts of death or suicide, suicide plan, or suicide attempt.

As used herein, the term “bipolar disorder” refers to a condition that causes an individual to experience unusual shifts in mood, energy, activity levels, and the ability to carry out day-to day tasks. Individuals with bipolar disorder experience periods of unusually intense emotion, changes in sleep patterns and activity levels, and unusual behaviors. These distinct periods are called “mood episodes.” Mood episodes are drastically different from the moods and behaviors that are typical for the person. Exemplary symptoms of mania, excessive behavior, include, but are not limited to, abnormally upbeat, jumpy, or wired behavior; increased activity, energy, or agitation, exaggerated sense of well-being and self-confidence, decreased need for sleep, unusual talkativeness, racing thoughts, distractibility, and poor decision-making-for example, going on buying sprees, taking sexual risks, or making foolish investments. Exemplary symptoms of depressive episodes or low mood, include, but are not limited to, depressed mood, such as feelings of sadness, emptiness, hopelessness, or tearfulness; marked loss of interest or feeling no pleasure in all-or almost all-activities, significant weight loss, weight gain, or decrease or increase in appetite, insomnia or hypersomnia (excessive sleeping or excessive sleepiness), restlessness or slowed behavior, fatigue or loss of energy, feelings of worthlessness or excessive or inappropriate guilt, decreased ability to think or concentrate, or indecisiveness, and thinking about, planning or attempting suicide. Bipolar disorder includes bipolar I disorder, bipolar II disorder, and cyclothymic disorder. Bipolar I disorder is defined by manic episodes that last at least 7 days or by severe manic symptoms that require hospitalization. A subject with bipolar I disorder may also experience depressive episodes typically lasting at least 2 weeks. Episodes of depression with mixed features, i.e., depressive and manic symptoms at the same time, are also possible. Bipolar II disorder is characterized by a pattern of depressive and hypomanic episodes, but not severe manic episodes typical of bipolar I disorder. Cyclothymic disorder (also referred to as cyclothymia) is characterized by periods of hypomanic symptoms (elevated mood and euphoria) and depressive symptoms lasting over a period of at least 2 years. The mood fluctuations are not sufficient in number, severity, or duration to meet the full criteria for a hypomanic or depressive episode.

As used herein, the term “catatonic depression” refers to a condition causing an individual to remain speechless and motionless for an extended period. Exemplary symptoms of catatonic depression include, but are not limited to, feelings of sadness, which can occur daily, a loss of interest in most activities, sudden weight gain or loss, a change in appetite, trouble falling asleep, trouble getting out of bed, feelings of restlessness, irritability, feelings of worthlessness, feelings of guilt, fatigue, difficulty concentrating, difficulty thinking, difficulty making decisions, thoughts of suicide or death, and/or a suicide attempt.

As used herein, the term “depressive disorder due to a medical condition” refers to a condition wherein an individual experiences depressive symptoms caused by another illness. Examples of medical conditions known to cause a depressive disorder include, but are not limited to, HIV/AIDS, diabetes, arthritis, strokes, brain disorders such as Parkinson's disease, Huntington's disease, multiple sclerosis, and Alzheimer's disease, metabolic conditions (e.g., vitamin B12 deficiency), autoimmune conditions (e.g., lupus and rheumatoid arthritis), viral or other infections (hepatitis, mononucleosis, herpes), back pain, and cancer (e.g., pancreatic cancer).

As used herein, the term “postpartum depression” refers to a condition as the result of childbirth and hormonal changes, psychological adjustment to parenthood, and/or fatigue. Postpartum depression is often associated with women, but men can also suffer from postpartum depression as well. Exemplary symptoms of postpartum depression include, but are not limited to, feelings of sadness, hopeless, emptiness, or overwhelmed; crying more often than usual or for no apparent reason; worrying or feeling overly anxious; feeling moody, irritable, or restless; oversleeping, or being unable to sleep even when the baby is asleep; having trouble concentrating, remembering details, and making decisions; experiencing anger or rage; losing interest in activities that are usually enjoyable; suffering from physical aches and pains, including frequent headaches, stomach problems, and muscle pain; eating too little or too much; withdrawing from or avoiding friends and family; having trouble bonding or forming an emotional attachment with the baby; persistently doubting his or ability to care for the baby; and thinking about harming themselves or the baby. As used herein, the term “premenstrual dysphoric disorder” refers to a condition wherein an individual expresses mood lability, irritability, dysphoria, and anxiety symptoms that occur repeatedly during the premenstrual phase of the cycle and remit around the onset of menses or shortly thereafter. Exemplary symptoms of premenstrual dysphoric disorder includes, but are not limited to, lability (e.g., mood swings), irritability or anger, depressed mood, anxiety and tension, decreased interest in usual activities, difficulty in concentration, lethargy and lack of energy, change in appetite (e.g., overeating or specific food cravings), hypersomnia or insomnia, feeling overwhelmed or out of control, physical symptoms (e.g., breast tenderness or swelling, joint or muscle pain, a sensation of 'bloating' and weight gain), self-deprecating thoughts, feelings of being keyed up or on edge, decreased interest in usual activities (e.g., work, school, friends, hobbies), subjective difficulty in concentration, and easy fatigability.

As used herein, the term “seasonal affective disorder” refers to a condition wherein an individual experiences mood changes based on the time of the year. In some instances, an individual experiences low mood, low energy, or other depressive symptoms during the fall and/or winter season. In some instances, an individual experiences low mood, low energy, or other depressive symptoms during the spring and/or summer season. Exemplary symptoms of seasonal affective disorder include, but are not limited to, feeling depressed most of the day or nearly every day, losing interest in activities once found enjoyable, having low energy, having problems with sleeping, experiencing changes in appetite or weight, feeling sluggish or agitated, having difficulty concentrating, feeling hopeless, worthless, or guilty, and having frequent thoughts of death or suicide.

In some embodiments, a depressive disorder comprises a medical diagnosis based on the criteria and classification from Diagnostic and Statistical Manual of Mental Disorders, 5th Ed. In some embodiments, a depressive disorder comprises a medical diagnosis based on an independent medical evaluation.

In some embodiments, the methods described herein are provided to a subject with depression that is resistant to treatment. In some embodiments, the subject has been diagnosed with treatmentresistant depression (TRD). The term “treatment -resistant depression” refers to a kind of depression that does not respond or is resistant to at least one or more treatment attempts of adequate dose and duration. In some embodiments, the subject with treatment-resistant depression has failed to respond to 1 treatment attempt, 2 treatment attempts, 3 treatment attempts, 4 treatment attempts, 5 treatment attempts, or more. In some embodiments, the subject with treatment-resistant depression has been diagnosed with major depressive disorder and has failed to respond to 3 or more treatment attempts. In some embodiments, the subject with treatment resistant depression has been diagnosed with bipolar disorder and has failed to respond to 1 treatment attempt. In some embodiments, the methods provided herein reduce at least one sign or symptom of a depressive disorder. In some embodiments, the methods provided herein reduce at least one sign or symptom of a depressive disorder by between about 5 % and about 100 %, for example, about 5 %, about 10 %, about 15 %, about 20 %, about 25 %, about 30 %, about 35 %, about 40 %, about 45 %, about 50 %, about 55 %, about 60 %, about 65 %, about 70 %, about 75 %, about 80 %, about 85 %, about 90 %, about 95 %, or about 100 %, or more, compared to prior to treatment.

In some embodiments, the disease or disorder is an anxiety disorder. As used herein, the term “anxiety disorder” refers to a state of apprehension, uncertainty, and/or fear resulting from the anticipation of an event and/or situation. Anxiety disorders cause physiological and psychological signs or symptoms. Non-limiting examples of physiological symptoms include muscle tension, heart palpitations, sweating, dizziness, shortness of breath, tachycardia, tremor, fatigue, worry, irritability, and disturbed sleep. Non-limiting examples of psychological symptoms include fear of dying, fear of embarrassment or humiliation, fear of an event occurring, etc. Anxiety disorders also impair a subject’s cognition, information processing, stress levels, and immune response. In some embodiments, the methods disclosed herein treat chronic anxiety disorders. As used herein, a “chronic” anxiety disorder is recurring. Examples of anxiety disorders include, but are not limited to, generalized anxiety disorder (GAD), social anxiety disorder, panic disorder, panic attack, a phobia-related disorder (e.g., phobias related to flying, heights, specific animals such as spiders/dogs/snakes, receiving injections, blood, etc., agoraphobia), separation anxiety disorder, selective mutism, anxiety due to a medical condition, post- traumatic stress disorder (PTSD), obsessive-compulsive disorder (OCD), substance-induced anxiety disorder, etc.

In some embodiments, the subject in need thereof develops an anxiety disorder after experiencing the effects of a disease. The effects of a disease include diagnosis of an individual with said disease, diagnosis of an individual’s loved ones with said disease, social isolation due to said disease, quarantine from said disease, or social distancing as a result of said disease. In some embodiments, an individual is quarantined to prevent the spread of the disease. In some embodiments, the disease is COVID-19, SARS, or MERS. In some embodiments, a subject develops an anxiety disorder after job loss, loss of housing, or fear of not finding employment.

In some embodiments, the disease or disorder is generalized anxiety disorder (GAD). Generalized anxiety disorder is characterized by excessive anxiety and worry, fatigue, restlessness, increased muscle aches or soreness, impaired concentration, irritability, and/or difficulty sleeping. In some embodiments, a subject with generalized anxiety disorder does not have associated panic attacks. In some embodiments, the subject has generalized anxiety disorder with depression. In some embodiments, the methods herein are provided to a subject with generalized anxiety disorder also having symptoms of depression. In some embodiments, after treating the symptom is reduced compared to prior to treating by about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%.

In some embodiments, the disease or disorder is social anxiety disorder. As used herein, “social anxiety disorder” is a marked fear or anxiety about one or more social situations in which the individual is exposed to possible scrutiny by others. Non-limiting examples of situations which induce social anxiety include social interactions (e.g., having a conversation, meeting unfamiliar people), being observed (e.g., eating or drinking), and performing in front of others (e.g., giving a speech). In some embodiments, the social anxiety disorder is restricted to speaking or performing in public. In some embodiments, treating according to the methods of the disclosure reduces or ameliorates a symptom of social anxiety disorder. In some embodiments, after treating the symptom is reduced compared to prior to treating by about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%.

In some embodiments, the disease or disorder is a compulsive disorder, such as obsessive- compulsive disorder (OCD), body-focused repetitive behavior, hoarding disorder, gambling disorder, compulsive buying, compulsive internet use, compulsive video gaming, compulsive sexual behavior, compulsive eating, compulsive exercise, body dysmorphic disorder, hoarding disorder, dermatillomania, trichotillomania, excoriation, substance-induced obsessive compulsive and related disorder, or an obsessive-compulsive disorder due to another medical condition, etc., or a combination thereof. In some embodiments, the disease or disorder is obsessive-compulsive disorder (OCD).

In some embodiments, at least one sign or symptom of an anxiety disorder is improved following the administration of a compound as disclosed herein. In some embodiments, a sign or symptom of an anxiety disorder is measured according to a diary assessment, an assessment by a clinician or caregiver, or a clinical scale. In some embodiments, treatment causes a demonstrated improvement in one or more of the following: State-Trait Anxiety Inventory (STAI), Beck Anxiety Inventory (BAI), Hospital Anxiety and Depression Scale (HADS), Generalized Anxiety Disorder questionnaire-IV (GADQ- IV), Hamilton Anxiety Rating Scale (HARS), Leibowitz Social Anxiety Scale (LSAS), Overall Anxiety Severity and Impairment Scale (OASIS), Hospital Anxiety and Depression Scale (HADS), Patient Health Questionnaire 4 (PHQ- 4), Social Phobia Inventory (SPIN), Brief Trauma Questionnaire (BTQ), Combat Exposure Scale (CES), Mississippi Scale for Combat-Related PTSD (M-PTSD), Posttraumatic Maladaptive Beliefs Scale (PMBS), Perceived Threat Scale (DRRI-2 Section: G), PTSD Symptom Scale-Interview for DSM-5 (PSS-I-5), Structured Interview for PTSD (SI- PTSD), Davidson Trauma Scale (DTS), Impact of Event Scale-Revised (IES-R), Posttraumatic Diagnostic Scale (PDS-5), Potential Stressful Events Interview (PSEI), Stressful Life Events Screening Questionnaire (SLESQ), Spielberger’s Trait and Anxiety, Generalized Anxiety Dis- order 7-Item Scale, The Psychiatric Institute Trichotillomania Scale (PITS), The MGH Hairpulling Scale (MGH-HPS), The NIMH Trichotillomania Severity Scale (NIMH-TSS), The NIMH Trichotillomania Impairment Scale (NIMH- TIS), The Clinical Global Impression (CGI), the Brief Social Phobia Scale (BSPS), The Panic Attack Questionnaire (PAQ), Panic Disorder Severity Scale, Florida Obsessive-Compulsive Inventory (FOCI), The Leyton Obsessional Inventory Survey Form, The Vancouver Obsessional Compulsive Inventory (VOCI), The Schedule of Compulsions, Obsessions, and Pathological Impulses (SCOPI), Padua Inventory-Revised (PI-R), Quality of Life (QoL), The Clinical Global Improvement (CGI) scale, The Yale-Brown Obsessive-Compulsive Scale (Y-BOCS), The Yale-Brown Obsessive-Compulsive Scale Second Edition (Y-BOCS-II), The Dimensional Yale-Brown Obsessive-Compulsive Scale (DY-BOCS), The National Institute of Mental Health- Global Obsessive-Compulsive Scale (NIMH-GOCS), The Yale- Brown Obsessive-Compulsive Scale Self-Report (Y-BOCS-SR), The Obsessive-Compulsive Inventory-Re- vised (OCI-R), and the Dimensional Obsessive-Compulsive Scale (DOCS), or a combination thereof. In some embodiments, treating according to the methods of the disclosure results in an improvement in an anxiety disorder compared to pre-treatment of about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%, according to any one of the diary assessments, assessments by a clinical or caregiver, or clinical scales, described herein or known in the art.

In some embodiments, the disease or disorder is attention deficit disorder (ADD). ADD is most commonly diagnosed in children under the age of 16 who have 6 or more symptoms of inattention (5 or more for older teenagers) for at least 6 consecutive months, but no signs of hyperactivity/impulsivity. The symptoms of inattention include, but are not limited to, trouble paying attention, avoids long mental tasks such as homework, trouble staying on task, disorganized or forgetful, doesn’t appear to listen when spoken to, doesn’t pay close attention to details. Loses things often, makes careless mistakes, and struggles to follow through with instructions. In some embodiments, the disease or disorder is attention deficit hyperactivity disorder (ADHD). ADHD is marked by an ongoing pattern of inattention and/or hyperactivity-impulsivity. Hyperactivity-impulsivity symptoms may often include, but are not limited to, fidgeting or squirming while seated, leaving their seats in situations where staying seated is expected, running, dashing, or climbing around at inappropriate times, being unable to engage in hobbies quietly, being constantly in motion, talking excessively, answering questions before they are fully asked, having difficulty waiting for one’s turn, and interrupting or intruding on others during conversations or activities.

In some embodiments, the disease or disorder is a headache disorder. As used herein, the term “headache disorder” refers to a disorder characterized by recurrent headaches. Headache disorders include migraine, tension-type headache, cluster headache, and chronic daily headache syndrome.

In some embodiments, a method of treating cluster headaches in a subject in need thereof is disclosed herein. In some embodiments, at least one sign or symptom of cluster headache is improved following treatment. In some embodiments, the sign or symptom of cluster headache is measured according to a diary assessment, a physical or psychological assessment by clinician, an imaging test, or a neurological examination. Cluster headache is a primary headache disorder and belongs to the trigeminal autonomic cephalalgias. The definition of cluster headaches is a unilateral headache with at least one autonomic symptom ipsilateral to the headache. Attacks are characterized by severe unilateral pain predominantly in the first division of the trigeminal nerve-the fifth cranial nerve whose primary function is to provide sensory and motor innervation to the face. Attacks are also associated with prominent unilateral cranial autonomic symptoms and subjects often experience agitation and restlessness during attacks. In some embodiments, a subject with cluster headaches also experiences nausea and/or vomiting. In some embodiments, a subject with cluster headaches experiences unilateral pain, excessive tearing, facial flushing, a droopy eyelid, a constricted pupil, eye redness, swelling under or around one or both eyes, sensitivity to light, nausea, agitation, and restlessness.

In some embodiments, a method of treating migraines in a subject in need thereof is disclosed herein. A migraine is a moderate to severe headache that affects one half or both sides of the head, is pulsating in nature, and last from 2 to 72 hours. Symptoms of migraine include headache, nausea, sensitivity to light, sensitivity to sound, sensitivity to smell, dizziness, difficulty speaking, vertigo, vomiting, seizure, distorted vision, fatigue, or loss of appetite. Some subjects also experience a prodromal phase, occurring hours or days before the headache, and/or a postdromal phase following headache resolution. Prodromal and postdromal symptoms include hyperactivity, hypoactivity, depression, cravings for particular foods, repetitive yawning, fatigue and neck stiffness and/or pain. In some embodiments, the migraine is a migraine without aura, a migraine with aura, a chronic migraine, an abdominal migraine, a basilar migraine, a menstrual migraine, an ophthalmoplegic migraine, an ocular migraine, an ophthalmic migraine, or a hemiplegic migraine. In some embodiments, the migraine is a migraine without aura. A migraine without aura involves a migraine headache that is not accompanied by a headache. In some embodiments, the migraine is a migraine with aura. A migraine with aura is primarily characterized by the transient focal neurological symptoms that usually precede or sometimes accompany the headache. Less commonly, an aura can occur without a headache, or with a non-migraine headache. In some embodiments, the migraine is a hemiplegic migraine. A hemiplegic migraine is a migraine with aura and accompanying motor weakness. In some embodiments, the hemiplegic migraine is a familial hemiplegic migraine or a sporadic hemiplegic migraine. In some embodiments, the migraine is a basilar migraine. A subject with a basilar migraine has a migraine headache and an aura accompanied by difficulty speaking, world spinning, ringing in ears, or a number of other brainstem-related symptoms, not including motor weakness. In some embodiments, the migraine is a menstrual migraine. A menstrual migraine occurs just before and during menstruation. In some embodiments, the subject has an abdominal migraine. Abdominal migraines are often experienced by children. Abdominal migraines are not headaches, but instead stomach aches. In some embodiments, a subject with abdominal migraines develops migraine headaches. In some embodiments, the subject has an ophthalmic migraine also called an “ocular migraine.” Subjects with ocular migraines experience vision or blindness in one eye for a short time with or after a migraine headache. In some embodiments, a subject has an ophthalmoplegic migraine. Ophthalmoplegic migraines are recurrent attacks of migraine headaches associated with paresis of one or more ocular cranial nerves. In some embodiments, the subject in need of treatment experiences chronic migraines. As defined herein, a subject with chronic migraines has more than fifteen headache days per month. In some embodiments, the subject in need of treatment experiences episodic migraines. As defined herein, a subject with episodic migraines has less than fifteen headache days per month.

In some embodiments, a method of treating chronic daily headache syndrome (CDHS) in a subject in need thereof is disclosed herein. A subject with CDHS has a headache for more than four hours on more than 15 days per month. Some subjects experience these headaches for a period of six months or longer. CHDS affects 4% of the general population. Chronic migraine, chronic tension-type headaches, new daily persistent headache, and medication overuse headaches account for the vast majority of chronic daily headaches.

In some embodiments, after treating according to the methods of the disclosure, the frequency of headaches and/or related symptoms decreases by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%, compared to prior to said treating.

In some embodiments, after treating according to the methods of the disclosure, the length of a headache attack decreases by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%, compared to prior to said treating. In some embodiments, at least one sign or symptom of headache disorder is improved following administration of a compound disclosed herein. In some embodiments, a sign or symptom of a headache disorder is measured according to a diary assessment, an assessment by a clinician or caregiver, or a clinical scale. In some embodiments, treatment of the present disclosure causes a demonstrated improvement in one or more of the following: the Visual Analog Scale, Numeric Rating Scale, the Short Form Health Survey, Profile of Mood States, the Pittsburgh Sleep Quality Index, the Major Depression Inventory, the Perceived Stress Scale, the 5-Level EuroQoL-5D, the Headache Impact Test; the ID- migraine; the 3-item screener; the Minnesota Multiphasic Personality Inventory; the Hospital Anxiety and Depression Scale (HADS), the 50 Beck Depression Inventory (BDI; both the original BD151 and the second edition, BDI-1152), the 9-item Patient Health Questionnaire (PHQ- 9), the Migraine Disability Assessment Questionnaire (MI- DAS), the Migraine-Specific Quality of Life Questionnaire version 2.1 (MSQ v2.1), the European Quality of Life-5 Dimensions (EQ-5D), the Short-form 36 (SF- 36), or a combination thereof. In some embodiments, treating according to the methods of the disclosure results in an improvement in a headache disorder compared to pre-treatment of about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%, according to any one of the diary assessments, assessments by a clinical or caregiver, or clinical scales, described herein or known in the art. In some embodiments, the sign or symptom of the headache disorder is measured according to a diary assessment, a physical or psychological assessment by clinician, an imaging test, an electroencephalogram, a blood test, a neurological examination, or combination thereof. In some embodiments, the blood test evaluates blood chemistry and/or vitamins.

In some embodiments, the disease or disorder is a substance use disorder. Substance addictions which can be treated using the methods herein include addictions to addictive substances/agents such as recreational drugs and addictive medications. Examples of addictive substances/agents include, but are not limited to, alcohol, e.g., ethyl alcohol, gamma hydroxybutyrate (GHB), caffeine, nicotine, cannabis (marijuana) and cannabis derivatives, opiates and other morphine-like opioid agonists such as heroin, phencyclidine and phencyclidine-like compounds, sedative hypnotics such as benzodiazepines, methaqualone, mecloqualone, etaqualone and barbiturates and psychostimulants such as cocaine, amphetamines and amphetamine-related drugs such as dextroamphetamine and methylamphetamine. Examples of addictive medications include, e.g., benzodiazepines, barbiturates, and pain medications including alfentanil, allylprodine, alphaprodine, anileridine benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, cyclazocine, desomorphine, dextromoramide, dezocine, diampromide, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene fentanyl, heroin, hydrocodone, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levallorphan, levorphanol, levophenacylmorphan, lofenitanil, meperidine, meptazinol, metazocine, methadone, metopon, morphine, myrophine, nalbuphine, narceine, nicomorphine, norlevorphanol, normethadone, nalorphine, normorphine, norpipanone, opium, oxycodone, OXYCONTIN®, oxymorphone, papaveretum, pentazocine, phenadoxone, phenomorphan, phenazocine, phenoperidine, piminodine, piritramide, propheptazine, promedol, properidine, propiram, propoxyphene sufentanil, tramadol, and tilidine. In some embodiments, the disease or disorder is alcohol use disorder (AUD). In some embodiments, the disease or disorder is nicotine use (e.g., smoking) disorder, and the therapy is used for e.g., smoking cessation.

In some embodiments, the disclosure provides for the management of sexual dysfunction, which may include, but is not limited to, sexual desire disorders, for example, decreased libido; sexual arousal disorders, for example, those causing lack of desire, lack of arousal, pain during intercourse, and orgasm disorders such as anorgasmia; and erectile dysfunction; particularly sexual dysfunction disorders stemming from psychological factors.

In some embodiments, the disease or disorder is an eating disorder. As used herein, the term “eating disorder” refers to any of a range of psychological disorders characterized by abnormal or disturbed eating habits. Non-limiting examples of eating disorders include pica, anorexia nervosa, bulimia nervosa, rumination disorder, avoidant/restrictive food intake disorder, binge-eating disorder, other specified feeding or eating disorder, unspecified feeding or eating disorder, or combinations thereof. In some embodiments, the eating disorder is pica, anorexia nervosa, bulimia nervosa, rumination disorder, avoidant/restrictive food intake disorder, binge-eating disorder, or combinations thereof. In some embodiments, the methods disclosed herein treat chronic eating disorders. As used herein, a “chronic” eating disorder is recurring. In some embodiments, at least one sign or symptom of an eating disorder is improved following administration of a compound disclosed herein. In some embodiments, a sign or symptom of an eating disorder is measured according to a diary assessment, an assessment by a clinician or caregiver, or a clinical scale. Non-limiting examples of clinical scales, diary assessments, and assessments by a clinician or caregiver include: the Mini International Neuropsychiatric Interview (MINI), the McLean Screening Instrument for Borderline Personality Disorder (MSI-BPD), the Eating Disorder Examination (EDE), the Eating Disorder Questionnaire (EDE-Q), the Eating Disorder Examination Questionnaire Short Form (EDE-QS), the Physical Appearance State and Trait Anxiety Scale-State and Trait version (PASTAS), Spielberger State-Trait Anxiety Inventory (STAI), Eating Disorder Readiness Ruler (ED-RR), Visual Analogue Rating Scales (VAS), the Montgomery-Asberg Depression Rating Scale (MADRS), Yale-Brown Cornell Eating Disorder Scale (YBC-EDS), Yale-Brown Cornell Eating Disorder Scale Self Report (YBC-EDS-SRQ), the Body Image State Scale (BISS), Clinical impairment assessment (CIA) questionnaire, the Eating Disorder Inventory (EDI) (e.g. version 3: EDI-3), the Five Dimension Altered States of Consciousness Questionnaire (5D-ASC), the Columbia-Suicide Severity Rating Scale (C-SSRS), the Life Changes Inventory (LCI), and combinations thereof. In some embodiments, treating according to the methods of the disclosure results in an improvement in an eating disorder compared to pre-treatment of about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%, according to any one of the diary assessments, assessments by a clinical or caregiver, or clinical scales, described herein or known in the art.

In some embodiments, the disease or disorder is multiple sclerosis (MS). MS is a chronic, inflammatory disease of unknown etiology that involves an immune-mediated attack on the central nervous system. Myelin and the oligodendrocytes that form myelin appear to be the primary targets of the inflammatory attack, although the axons themselves are also damaged. MS disease activity can be monitored by cranial scans, including magnetic resonance imaging (MRI) of the brain, accumulation of disability, as well as rate and severity of relapses. The diagnosis of clinically definite MS as determined by the Poser criteria requires at least two neurological events suggesting demyelination in the CNS separated in time and in location. Various MS disease stages and/or types are described in Multiple Sclerosis Therapeutics (Duntiz, 1999). Among them, relapsing-remitting multiple sclerosis (RRMS) is the most common form at the time of initial diagnosis. Many subjects with RRMS have an initial relapsing-remitting course for 5-15 years, which then advances into the secondary progressive MS (SPMS) disease course. Relapses result from inflammation and demyelination, whereas restoration of nerve conduction and remission is accompanied by resolution of inflammation, redistribution of sodium channels on demyelinated axons and remyelination. In some embodiments, the multiple sclerosis is relapsing multiple sclerosis. In some embodiments, the relapsing multiple sclerosis is relapsingremitting multiple sclerosis. In some embodiments, the methods herein reduce a symptom of multiple sclerosis in the subject. In some embodiments, the symptom is a MRI-monitored multiple sclerosis disease activity, relapse rate, accumulation of physical disability, frequency of relapses, decreased tune to confirmed disease progression, decreased time to confirmed relapse, frequency of clinical exacerbation, brain atrophy, neuronal dysfunction, neuronal injury, neuronal degeneration, neuronal apoptosis, risk for confirmed progression, deterioration of visual function, fatigue, impaired mobility, cognitive impairment, reduction of brain volume, abnormalities observed in whole Brain MTR histogram, deterioration in general health status, functional status, quality of life, and/or symptom severity on work. In some embodiments, the methods herein decrease or inhibit reduction of brain volume. In some embodiments, brain volume is measured by percent brain volume change (PBVC). In some embodiments, the methods herein increase time to confirmed disease progression. In some embodiments, time to confirmed disease progression is increased by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, for example at least 20-60%. In some embodiments, the methods herein decrease abnormalities observed in whole Brain MTR histogram. In some embodiments, the accumulation of physical disability is measured by Kurtzke Expanded Disability Status Scale (EDSS) score. In some embodiments, the accumulation of physical disability is assessed by the time to confirmed disease progression as measured by Kurtzke Expanded Disability Status Scale (EDSS) score.

In some embodiments, the disease or disorder is a disease or disorder characterized by, or otherwise associated with, neuroinflammation. Compounds and compositions of the present disclosure may provide cognitive benefits to subject’s suffering from neurological and neurodegenerative diseases such as Alzheimer’s disease and other dementia subtypes, Parkinson’s disease, amyotrophc lateral sclerosis (ALS), and others where neuroinflammation is a hallmark of disease pathophysiology and progression. For example, emerging psychedelic research/clinical evidence indicates that psychedelics may be useful as disease-modifying treatments in subjects suffering from neurodegenerative diseases such as Alzheimer’s disease and other forms of dementia. See Vann Jones, S.A. and O’Kelly, A. “Psychedelics as a Treatment for Alzheimer’s Disease Dementia” Front. Synaptic Neurosci., 21, August 2020; Kozlowska, U., Nichols, C., Wiatr, K., and Figiel, M. (2021), “From psychiatry to neurology: Psychedelics as prospective therapeutics for neurodegenerative disorders” Journal of Neurochemistry, 00, 1- 20; Garcia-Romeu, A., Darcy, S., Jackson, H., White, T., Rosenberg, P. (2021), “Psychedelics as Novel Therapeutics in Alzheimer’s Disease: Rationale and Potential Mechanisms” In: Current Topics in Behavioral Neurosciences. Springer, Berlin, Heidelberg. For example, psychedelics are thought to stimulate neurogenesis, provoke neuroplastic changes, and to reduce neuroinflammation. Thus, in some embodiments, the compounds of the present disclosure (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof) are used for the treatment of neurological and neurodegenerative disorders such as Alzheimer’ s disease, dementia subtypes, Parkinson’s disease, and amyotrophc lateral sclerosis (AES), where neuroinflammation is associated with disease pathogenesis. In some embodiments, the compounds of the present disclosure are used for the treatment of Alzheimer’s disease. In some embodiments, the compounds of the present disclosure are used for the treatment of dementia. In some embodiments, the compounds of the present disclosure are used for the treatment of Parkinson’s disease. In some embodiments, the compounds of the present disclosure are used for the treatment of amyotrophc lateral sclerosis (ALS). As described above, such treatment may stimulate neurogenesis, provoke neuroplastic changes, and/or provide neuroinflammatory benefits (e.g., reduced neuroinflammation compared to prior to the beginning of treatment), and as a result, may slow or prevent disease progression, slow or reverse brain atrophy, and reduce symptoms associated therewith (e.g., memory loss in the case of Alzheimer’s and related dementia disorders). While not limited thereto, pharmaceutical compositions adapted for oral and/or extended-release dosing are appropriate for such treatment methods, with sub-psychedelic dosing being preferred. In some embodiments, treating according to the methods of the disclosure results in an improvement in cognition in subject’s suffering from a neurological or neurodegenerative disease compared to pre-treatment of about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%, according to any one of a diary assessments, assessments by a clinical or caregiver, or clinical scales, described herein or known in the art.

Further, many of the behavioral issues associated with chronic and/or life-threatening illnesses, including neurodegenerative disorders such as Alzheimer’s disease, may benefit from treatment with the compounds disclosed herein. Indeed, depression, anxiety, or stress can be common among patients who have chronic and/or life-threatening illnesses such as Alzheimer's disease, autoimmune diseases (e.g., systemic lupus erythematosus, rheumatoid arthritis, and psoriasis), cancer, coronary heart disease, diabetes, epilepsy, HIV/AIDS, hypothyroidism, multiple sclerosis, Parkinson’s disease, and stroke. For example, depression is common in Alzheimer’s disease as a consequence of the disease, as well as being a risk factor for the disease itself. Symptoms of depression, anxiety, or stress can occur after diagnosis with the disease or illness. Patients that have depression, anxiety, or stress concurrent with another medical disease or illness can have more severe symptoms of both illnesses and symptoms of depression, anxiety, or stress can continue even as a patient’s physical health improves. Compounds described herein can be used to treat depression, anxiety, and/or stress associated with a chronic or life-threatening disease or illness.

Accordingly, in some embodiments, the methods herein are used to treat symptoms, e.g., depression, anxiety, and/or stress, associated with a chronic and/or life-threatening disease or disorder, including neurological and neurodegenerative diseases. In some embodiments, the methods provided herein reduce at least one sign or symptom of a neurological and/or neurodegenerative disease. In some embodiments, the methods provided herein reduce at least one sign or symptom of a neurological and/or neurodegenerative disease (e.g., depression, anxiety, and/or stress) by between about 5 % and about 100 %, for example, about 5 %, about 10 %, about 15 %, about 20 %, about 25 %, about 30 %, about 35 %, about 40 %, about 45 %, about 50 %, about 55 %, about 60 %, about 65 %, about 70 %, about 75 %, about 80 %, about 85 %, about 90 %, about 95 %, or about 100 %, or more, compared to prior to treatment, e.g., according to any one of the diary assessments, assessments by a clinical or caregiver, or clinical scales, described herein or known in the art. In some embodiments, the disease or disorder is Alzheimer’s disease. In some embodiments, the methods herein are used for the treatment of depression, anxiety, and/or stress associated with Alzheimer’s disease. In some embodiments, the disease or disorder is Parkinson’s disease. In some embodiments, the methods herein are used for the treatment of depression, anxiety, and/or stress associated with Parkinson’s disease. In some embodiments, the disease or disorder is amyotrophc lateral sclerosis (ALS). In some embodiments, the methods herein are used for the treatment of depression, anxiety, and/or stress associated with amyotrophc lateral sclerosis (ALS). In some embodiments, the disease or disorder is cancer related depression and anxiety. As discussed above, oral and/or extended- release dosing is appropriate for such applications, particularly when blood concentrations of active ingredient (e.g., a compound of Formula (I) through (V)) are kept below the psychedelic threshold.

In some embodiments, the compounds and compositions disclosed herein are used for treatment of brain injury, including traumatic brain injury (TBI). TBI is an injury to the brain caused by an external force, and can be classified based on severity, ranging from mild traumatic brain injury (mTBI/concussion) to severe traumatic brain injury. TBI can also be categorized by mechanism, as either a closed or penetrating head injury, or other features such as whether it is occurring in a specific location or over a widespread area. TBI can result in physical, cognitive, social, emotional and behavioral symptoms, which may be treated herein. Some of the imaging techniques used for diagnosis and recovery markers include computed tomography (CT) and magnetic resonance imaging (MRIs).

In some embodiments, the disease or disorder is a neurological and developmental disorder such as autism spectrum disorder, including Asperger’s syndrome. For example, Asperger’s syndrome is a subtype of autism spectrum disorder that is treatable with anxiety drugs. Subjects with autism spectrum disorder may present with various signs and symptoms, including, but not limited to, a preference for non-social stimuli, aberrant non-verbal social behaviors, decreased attention to social stimuli, irritability, anxiety (e.g., generalized anxiety and social anxiety in particular), and depression. In some embodiments, the autism spectrum disorder comprises a medical diagnosis based on the criteria and classification from Diagnostic and Statistical Manual of Mental Disorders, 5th Ed (DSM-5). Current evidence supports the use of psychedelics for ameliorating behavior atypicalities of autism spectrum disorder, including reduced social behavior, anxiety, and depression (see Markopoulos A, Inserra A, De Gregorio D, Gobbi G. Evaluating the Potential Use of Serotonergic Psychedelics in Autism Spectrum Disorder. Front Pharmacol. 2022; 12:749068). The signs and symptoms of autism spectrum disorder may be treated with the methods herein.

In some embodiments, the disease or disorder is a genetic condition that causes learning disabilities and cognitive impairment. An example of such a genetic condition is fragile X syndrome, caused by changes in the gene Fragile X Messenger Ribonucleoprotein 1 (FMRI), which can cause mild to moderate intellectual disabilities in most males and about one-third of affected females. Fragile X syndrome and autism spectrum disorder are closely associated because the FMRI gene is a leading genetic cause of autism spectrum disorder (see Markopoulos A, Inserra A, De Gregorio D, Gobbi G. Evaluating the Potential Use of Serotonergic Psychedelics in Autism Spectrum Disorder. Front Pharmacol. 2022;12:749068). Subjects with fragile X syndrome may display anxiety, hyperactive behavior (e.g., fidgeting and impulsive actions), attention deficit disorder, mood and aggression abnormalities, poor recognition memory, and/or features of autism spectrum disorder, and these signs and symptoms may be treated with the methods herein. Clinical trials with psychedelics for the treatment of fragile X syndrome and autism spectrum disorder are currently ongoing (ClinicalTrials.gov, number NCT04869930).

In some embodiments, the disease or disorder is mental distress, e.g., mental distress in frontline healthcare workers.

In some embodiments, the compounds and compositions disclosed herein are used for treatment of tic disorders, including Tourette’s Syndrome, which is also variously referred to as Tourette Syndrome, Tourette’s Disorder, Gilles de la Tourette syndrome (GTS), or simply Tourette’s or TS. The tic disorder may also be a pediatric autoimmune disorder associated with streptococcal infection (PANDAS), a transient tic disorder, a chronic tic disorder, or a tic disorder not otherwise specified (NOS). Tic disorders are defined in the Diagnostic and Statistical Manual of Mental Disorders (DSM) based on type (motor or phonic) and duration of tics (sudden, rapid, nonrhythmic movements), or similarly by the World Health Organization (ICD-10 codes). Tics are involuntary or semi-voluntary, sudden, brief, intermittent, repetitive movements (motor) or sounds (phonic) that are classified as simple or complex. Simple tics, for example, eye blinking or facial grimacing, are relatively easy to camouflage and may go largely unnoticed. Complex tics, such as body contortions, self-injurious behavior, obscene gestures, or shouting of socially inappropriate word or phrases, can appear to be purposeful actions and are particularly distressing. Transient tic disorders are generally characterized by multiple motor and/or phonic tics that occur for at least four weeks but less than 12 months. Chronic tic disorders are generally characterized by either single or multiple motor or phonic tics, but not both, which are present for more than a year. Tourette's Syndrome is a neurodevelopment disorder diagnosed when both motor and phonic tics are present (although not necessarily concurrently) for more than one year. Thus, Tourette’s syndrome (TS) is a chronic neuropsychiatric disorder characterized by the presence of fluctuating motor and phonic tics. The typical age of onset is between five and seven years. Affected children may become the target of teasing by peers, which in turn can result in low self-esteem, social isolation, poor school performance, depression and anxiety. In addition to causing social embarrassment, sudden, forceful tics can be painful, and violent head and neck tics have been reported to cause secondary neurologic deficits, such as compressive cervical myelopathy. Tourette's Syndrome patients are also at increased risk for obsessive-compulsive disorder (OCD), depression, and attention-deficit- hyperactivity disorder (ADHD). Tic disorder NOS is diagnosed when tics are present but do not meet the criteria for any specific tic disorder. The present compounds and compositions can also be administered for the treatment of tics induced as a side effect of a medication; tics associated with autism; and Tourettism (the presence of Tourette-like symptoms in the absence of Tourette's Syndrome (e.g., as a result of another disease or condition, such as a sporadic, genetic, or neurodegenerative disorder)).

In some embodiments, the disclosure provides for the management of different kinds of pain, including but not limited to cancer pain, e.g., refractory cancer pain; neuropathic pain; postoperative pain; opioid-induced hyperalgesia and opioid-related tolerance; neurologic pain; postoperative/post- surgical pain; complex regional pain syndrome (CRPS); shock; limb amputation; severe chemical or thermal burn injury; sprains, ligament tears, fractures, wounds and other tissue injuries; dental surgery, procedures and maladies; labor and delivery; during physical therapy; radiation poisoning; acquired immunodeficiency syndrome (AIDS); epidural (or peridural) fibrosis; orthopedic pain; back pain; failed back surgery and failed laminectomy; sciatica; painful sickle cell crisis; arthritis; autoimmune disease; intractable bladder pain; pain associated with certain viruses, e.g., shingles pain or herpes pain; acute nausea, e.g., pain that may be causing the nausea or the abdominal pain that frequently accompanies sever nausea; migraine, e.g., with aura; and other conditions including depression (e.g., acute depression or chronic depression), depression along with pain, alcohol dependence, acute agitation, refractory asthma, acute asthma (e.g., unrelated pain conditions can induce asthma), epilepsy, acute brain injury and stroke, Alzheimer's disease and other disorders. The pain may be persistent or chronic pain that lasts for weeks to years, in some cases even though the injury or illness that caused the pain has healed or gone away, and in some cases despite previous medication and/or treatment. In addition, the disclosure includes the treatment/management of any combination of these types of pain or conditions.

In some embodiments, the pain treated/managed is acute breakthrough pain or pain related to wind-up that can occur in a chronic pain condition. In some embodiments of the disclosure, the pain treated/managed is cancer pain, e.g., refractory cancer pain. In some embodiments of the disclosure, the pain treated/managed is post-surgical pain. In some embodiments of the disclosure, the pain treated/managed is orthopedic pain. In some embodiments of the disclosure, the pain treated/managed is back pain. In some embodiments of the disclosure, the pain treated/managed is neuropathic pain. In some embodiments of the disclosure, the pain treated/managed is dental pain. In some embodiments of the disclosure, the condition treated/managed is depression. In some embodiments of the disclosure, the pain treated/managed is chronic pain in opioid-tolerant patients. In some embodiments, the disease or disorder is arthritis. Types of arthritis include osteoarthritis, rheumatoid arthritis, childhood arthritis, fibromyalgia, gout, and lupus. In some embodiments, the disease or disorder is osteoarthritis. In some embodiments, the disease or disorder is rheumatoid arthritis. In some embodiments, the disease or disorder is childhood arthritis. In some embodiments, the disease or disorder is gout. In some embodiments, the disease or disorder is lupus. In some embodiments, the disease or disorder is fibromyalgia. Fibromyalgia is a disorder characterized by widespread musculoskeletal pain accompanied by fatigue, sleep, memory and mood issues. Fibromyalgia is believed to amplify painful sensations by affecting brain and spinal cord processes involving painful and nonpainful signaling. Symptoms often begin after an event, such as physical trauma, surgery, infection or significant psychological stress. In other cases, symptoms gradually accumulate over time with no single triggering event. Women are more likely to develop fibromyalgia than are men. Many people who have fibromyalgia also have tension headaches, temporomandibular joint (TMJ) disorders, irritable bowel syndrome, anxiety and depression.

In some embodiments, the disease or disorder is inflammatory bowel disease (IBD). IBD is a term for two conditions, Crohn’s disease and ulcerative colitis, that are characterized by chronic inflammation of the gastrointestinal (GI) tract, with such prolonged inflammation resulting in damage to the GI tract. Subjects suffering from IBD may experience persistent diarrhea, abdominal pain, rectal bleeding/bloody stools, weight loss, and fatigue. IBD may be diagnosed, and treatment may be monitored, using one or more of endoscopy, colonoscopy, contrast radiography, MRI, computed tomography (CT), stool samples, and blood tests, known by those of ordinary skill in the art.

In some embodiments, the disease or disorder includes conditions of the autonomic nervous system (ANS). In these embodiments, it may be preferable to use compounds of the present disclosure which are peripherally-restricted.

In some embodiments, the disease or disorder includes pulmonary disorders including asthma and chronic obstructive pulmonary disorder (COPD).

In some embodiments, the disease or disorder includes cardiovascular disorders including atherosclerosis.

In some embodiments, the disease or disorder is a sleep disorder such as narcolepsy, insomnia, nightmare disorder, sleep apnea, central sleep apnea, obstructive sleep apnea, hypopnea, sleep-related hypoventilation, restless legs syndrome, and jet lag. In some embodiments, the disease or disorder is narcolepsy.

In some embodiments, the disclosure relates to a method of treating a disease or condition by modulating the serotonin system (e.g., serotonin receptors, the serotonin transporter), wherein the method comprises administering an effective amount of any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof) to a subject in need thereof. In some embodiments, the disease or condition is selected from: levodopa-induced dyskinesia; dementia (e.g., Alzheimer's dementia), tinnitus, treatment resistant depression (TRD), major depressive disorder, neuropathic pain, agitation resulting from or associated with Alzheimer's disease, pseudobulbar effect, autism, Bulbar function, generalized anxiety disorder, schizophrenia, diabetic neuropathy, acute pain, depression, bipolar depression, suicidality, neuropathic pain, or post-traumatic stress disorder (PTSD). In some embodiments, the disease or condition is a psychiatric or mental disorder (e.g., schizophrenia, mood disorder, substance induced psychosis, major depressive disorder (MDD), bipolar disorder, bipolar depression (BDep), post-traumatic stress disorder (PTSD), suicidal ideation, anxiety, obsessive compulsive disorder (OCD), and treatment-resistant depression (TRD)). In other embodiments, the disease or condition is a neurological disorder (e.g., Huntington's disease (HD), Alzheimer's disease (AD), or systemic lupus erythematosus (SLE)).

In some embodiments, the disclosure relates to a method of treating an ocular disease, such as uveitis, corneal disease, iritis, iridocyclitis, glaucoma, and cataracts, by administering ophthalmically a therapeutically effective amount of any of the compounds described herein (e.g., any of the compounds described herein (e.g., a compound of Formula (I) through (V)) to a subject in need thereof. For example, compounds herein may be administered in the form of solutions, suspensions, ointments, emulsions, gel-forming solutions, powders for solutions, gels, ocular inserts, and implants. In some embodiments, the compounds are administered in the form of an eye drop formulation.

For example, in some embodiments, the disclosure provides a method of treating a subject with any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), comprising the step of administering to a subject an orally administered tablet composition, e.g., matrix composition, of the disclosure comprising any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), such that the subject is treated.

The administering physician can provide a method of treatment that is prophylactic or therapeutic by adjusting the amount and timing of any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), on the basis of observations of one or more symptoms of the disorder or condition being treated. In some embodiments of the disclosure, the subject is a mammal. In some embodiments of the disclosure, the mammal is a human.

In some embodiments, the disclosure provides a method of continuous oral administration of any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof). Any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), may be formulated into a tablet composition, e.g., singlelayer tablet, that provides a steady release of a therapeutically effective concentration of the compound over a complete release period without neurologically toxic spikes, e.g., no sedative or psychotomimetic toxic spikes in plasma concentration of any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof). The tablet composition may be orally administered to a subject, such that a continuous therapeutically effective concentration of any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), is provided to the subject.

Compounds of the present disclosure may possess advantageous metabolic degradation profiles which prevent high drug concentrations observed acutely after administration, while also enhancing brain levels of the active compound, so that in some embodiments the therapeutic doses may be reduced. As a result, the compounds may be suitable for microdosing to achieve durable therapeutic benefits, with decreased toxicity, e.g., toxicity associated with activation of 5-HT2B receptors associated with valvular heart disease (Rothman, R. B., and Baumann, M. H., 2009, Serotonergic drugs and valvular heart disease, Expert Opin Drug Saf8, 317-329).

In some embodiments, the compounds/compositions of the disclosure may be used as a standalone therapy. In some embodiments, the compounds/compositions of the disclosure may be used as an adjuvant/combination therapy. In some embodiments, the subject with a disorder is administered the compound/composition of the present disclosure and at least one additional therapy and/or therapeutic. In some embodiments, administration of an additional therapy and/or therapeutic is prior to administration of the compound/composition of the present disclosure. In some embodiments, administration of an additional therapy and/or therapeutic is after administration of the compound/composition of the present disclosure. In some embodiments, administration of an additional therapy and/or therapeutic is concurrent with administration of the compound/composition of the present disclosure. In some embodiments, the additional therapy is an antidepressant, an anticonvulsant, lisdexamfetamine dimesylate, an antipsychotic, an anxiolytic, an anti-inflammatory drug, a benzodiazepine, an analgesic drug, a cardiovascular drug, an opioid antagonist, or combinations thereof.

In some embodiments, the additional therapy is a benzodiazepine. In some embodiments, the benzodiazepine is diazepam or alprazolam.

In some embodiments, the additional therapy is a N-methyl-D-aspartate (NMDA) receptor antagonist. In some embodiments, the NMDA receptor antagonist is ketamine. In some embodiments, the NMDA receptor antagonist is nitrous oxide.

In some embodiments, the additional therapy is an antidepressant. In some embodiments, an antidepressant indirectly affects a neurotransmitter receptor, e.g., via interactions affecting the reactivity of other molecules at a neurotransmitter receptor. In some embodiments, an antidepressant is an agonist. In some embodiments, an antidepressant is an antagonist. In some embodiments, an antidepressant acts (either directly or indirectly) at more than one type of neurotransmitter receptor. In some embodiments, an antidepressant is chosen from buproprion, citalopram, duloxetine, escitalopram, fluoxetine, fluvoxamine, milnacipran, mirtazapine, paroxetine, reboxetine, sertraline, and venlafaxine.

In some embodiments, the antidepressant is a tricyclic antidepressant (“TCA”), selective serotonin reuptake inhibitor (“SSRI”), serotonin and noradrenaline reuptake inhibitor (“SNRI”), dopamine reuptake inhibitor (“DRI”), noradrenaline reuptake Monoamine oxidase inhibitor (“MAOI”), including inhibitor (“NRU”), dopamine, serotonin and noradrenaline reuptake inhibitor (“DSNRI”), a reversible inhibitor of monoamine oxidase type A (RIMA), or combination thereof. In some embodiments, the antidepressant is a TCA. In some embodiments, the TCA is imipramine or clomipramine. In some embodiments, the antidepressant is an SRI. In some embodiments, the SSRI is escitalopram, paroxetine, sertraline, fluvoxamine, fluoxetine, or combinations thereof. In some embodiments, the SNRI is venlafaxine. In some embodiments, the additional therapy is pregabalin.

In some embodiments, the additional therapeutic is an anticonvulsant. In some embodiments, the anticonvulsant is gabapentin, carbamazepine, ethosuximide, lamotrigin, felbamate, topiramate, zonisamide, tiagabine, oxcarbazepine, levetiracetam, divalproex sodium, phenytoin, fosphenytoin. In some embodiments, the anticonvulsant is topiramate.

In some embodiments, the additional therapeutic is an antipsychotic. In some embodiments, the antipsychotic is a pheno thiazine, butryophenone, thioxanthene, clozapine, risperidone, olanzapine, or sertindole, quetiapine, aripiprazole, zotepine, perospirone, a neurokinin-3 antagonist, such as osanetant and talnetant, rimonabant, or a combination thereof.

In some embodiments, the additional therapeutic is an anti-inflammatory drug. In some embodiments, the anti-inflammatory drug is a nonsteroidal anti-inflammatory drugs (NSAIDS), steroid, acetaminophen (COX-3 inhibitors), 5-lipoxygenase inhibitor, leukotriene receptor antagonist, leukotriene A4 hydrolase inhibitor, angiotensin converting enzyme antagonist, beta blocker, antihistaminic, histamine 2 receptor antagonist, phosphodiesterase-4 antagonist, cytokine antagonist, CD44 antagonist, antineoplastic agent, 3-hydroxy-3-methylglutaryl coenzyme A inhibitor (statins), estrogen, androgen, antiplatelet agent, antidepressant, Helicobacter pylori inhibitors, proton pump inhibitor, thiazolidinedione, dual-action compounds, or combination thereof.

In some embodiments, the additional therapeutic is an anti-anxiolytic. In some embodiments, an anxiolytic is chosen from alprazolam, an alpha blocker, an antihistamine, a barbiturate, a beta blocker, bromazepam, a carbamate, chlordiazepoxide, clonazepam, clorazepate, diazepam, flurazepam, lorazepam, an opioid, oxazepam, temazepam, or triazolam.

In some embodiments, the pharmaceutical compositions of the disclosure are administered in combination with an opioid, which may be used for example to reduce pain. In some embodiments, the pharmaceutical compositions of the present disclosure serve the purpose of an opioid-sparing medication, i.e., to reduce the amount of opioids necessary to treat a patient.

In some embodiments, the additional therapy is an opioid antagonist. Non-limiting examples of opioid antagonists include naloxone, naltrexone, nalmefene, nalorphine, nalrphine dinicotinate, levallrphan, samidorphan, nalodeine, alvimopan, methylnaltrexone, naloxegol, 6-naltrexol, axelopran, bevenopran, methylsamidorphan, naldemedine, buprenorphine, decozine, butorphanol, levorphanol, nalbuphine, pentazocine, and phenazocine.

In some embodiments, the additional therapy is a cardiovascular drug. Non-limiting examples of cardiovascular drugs include digoxin or (3p,5p,12p)-3-[(O-2,6-dideoxy-p-£)-ribo-hexopyranosyl- (1— >4)-<9-2,6-dideoxy-p-Z)-ribo-hexopyranosyl-(l— >4)-2,6-dideoxy-p-Z)-ribohexopyranosyl) oxy]-

12,14-dihydroxy-card-20(22)-enolide, lisinopril, captopril, ramipril, trandolapril, benazepril, cilazapril, enalapril, moexipril, perindopril, quinapril, fludrocortisone, enalaprilate, quinapril, perindopril, apixaban, dabigatran, edoxaban, heparin, rivaroxaban, warfarin, aspirin, clopidogrel, dipyridamole, prasugrel, ticagrelor, azilsartan, candesartan, eprosartan, irbesartan, losartan, olmesartan, telmisartan, valsartanscaubitril, acebutolol, atenolol, betaxolol, bisoprolol, metoprolol, nadolol, propranolol, sotalol, amlodipine, diltiazem, felodipine, nifedipine, nimodipine, nisolidipine, verapamil, statins, nicotinic acids, diuretics, vasodilators, and combinations thereof.

In some embodiments, the subject is administered at least one therapy. Non-limiting examples of therapies include transcranial magnetic stimulation, cognitive behavioral therapy, interpersonal psychotherapy, dialectical behavior therapy, mindfulness techniques, or acceptance, commitment therapy, or combinations thereof.

Pharmaceutical compositions

Also disclosed herein is a pharmaceutical composition comprising a compound as disclosed herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof) and a pharmaceutically acceptable excipient. The pharmaceutical compositions may contain one, or more than one, compound of the present disclosure.

The pharmaceutical composition may comprise a single compound of Formula (I) through (V) or a mixture of compounds of Formula (I) through (V). The pharmaceutical composition may be formed from an isotopologue mixture of the disclosed compounds. In some embodiments, a subject compound of Formula (I) through (V) may be present in the pharmaceutical composition at a purity of at least 20% by weight, at least 30% by weight, at least 40% by weight, at least 50% by weight, at least 60% by weight, at least 70% by weight, at least 80% by weight, at least 90% by weight, at least 95% by weight, at least 99% by weight, based on a total weight of isotopologues of the compound of Formula (I) through (V) present in the pharmaceutical composition. In some embodiments, the composition comprises a subject compound of Formula (I) through (V), and is substantially free of other isotopologues of the subject compound, in either free base or salt form, e.g., the composition has less than 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 or 0.5 mole percent of other isotopologues of the subject compound.

In some embodiments, any position in the compound having deuterium has a minimum deuterium incorporation that is greater than that found naturally occurring in hydrogen (about 0.016 atom %). In some embodiments, any position in the compound having deuterium has a minimum deuterium incorporation of at least 10 atom %, at least 20 atom %, at least 25 atom %, at least 30 atom %, at least 40 atom %, at least 45 atom %, at least 50 atom %, at least 60 atom %, at least 70 atom %, at least 80 atom %, at least 90 atom %, at least 95 atom %, at least 99 atom % at the site of deuteration.

In some embodiments, the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, is chemically pure, for example has a chemical purity of greater than 90%, 92%, 94%, 96%, 97%, 98%, or 99% by liquid chromatography (e.g., UPLC or HPLC). In some embodiments, the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, has no single impurity of greater than 1%, greater than 0.5%, greater than 0.4%, greater than 0.3%, or greater than 0.2%, measured by liquid chromatography (e.g., UPLC or HPLC). In some embodiments, the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, has a chemical purity of greater than 97 area %, greater than 98 area %, or greater than 99 area % by liquid chromatography (e.g., UPLC or HPLC). In some embodiments, the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, has no single impurity greater than 1 area %, greater than 0.5 area %, greater than 0.4 area %, greater than 0.3 area %, or greater than 0.2 area % as measured by liquid chromatography (e.g., UPLC or HPLC).

The pharmaceutical composition may be formulated with an enantiomerically pure compound of the present disclosure, e.g., a compound of Formula (I) through (V), or a racemic mixture of the compounds. As described herein, a racemic compound of Formula (I) through (V) may contain about 50% of the R- and S -stereoisomers based on a molar ratio (about 48 to about 52 mol %, or about a 1:1 ratio)) of one of the isomers. In some embodiments, a composition, medicament, or method of treatment may involve combining separately produced compounds of the R- and S -stereoisomers in an approximately equal molar ratio (about 48 to 52%). In some embodiments, a medicament or pharmaceutical composition may contain a mixture of separate compounds of the R- and S- stereoisomers in different ratios. In some embodiments, the pharmaceutical composition contains an excess (greater than 50%) of the R-enantiomer. Suitable molar ratios of R/S may be from about 1.5:1, 2:1, 3:1, 4:1, 5:1, 10:1, or higher. In some embodiments, a pharmaceutical composition may contain an excess of the S-enantiomer, with the ratios provided for R/S reversed. Other suitable amounts of R/S may be selected. For example, the R-enantiomer may be present in amounts of at least about 55% to 100%, or at least 65%, at least 75%, at least 80%, at least 85%, at least 90%, about 95%, about 98%, or 100%. In other embodiments, the S-enantiomer may be present in a higher percentage, e.g., in amounts of at least about 55% to 100%, or at least 65%, at least 75%, at least 80%, at least 85%, at least 90%, about 95%, about 98%, or 100%. Ratios between all these exemplary embodiments as well as greater than and less than them while still within the disclosure, all are included. Compositions may contain a mixture of the racemate and a separate compound of Formula (I) through (V), in free base and/or in salt form.

The pharmaceutical composition may be formulated with one or more polymorphs of the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, including crystalline and/or amorphous forms (e.g., polymorphs) of the compounds or salts thereof.

A pharmaceutical composition can be in unit dosage form. In such form, the pharmaceutical composition is subdivided into unit doses containing appropriate quantities of the active ingredient. 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. Pharmaceutical compositions may be generally provided herein which comprise about 0.001 to about 1000 mg, about 1 to about 500 mg, about 2 to about 100 mg, about 0.001 mg, about 0.01 mg, about 0.1 mg, about 1 mg, about 2 mg, about 3 mg, about 5 mg, about 10 mg, about 20 mg, about 30 mg, about 40 mg, about 50 mg, about 75 mg, about 100 mg, about 150 mg, about 200 mg, about 300 mg, about 400 mg, about 500 mg of one or more compounds as disclosed herein (on active basis). The quantity of compound(s) (e.g., compound(s) of Formula (I) through (V)) (on active basis) in a unit dose preparation may be varied or adjusted within the above ranges as deemed appropriate using sound medical judgment, according to the particular application, administration route, potency of the active ingredient, etc. The composition can, if desired, also contain other compatible therapeutic agents/active ingredients.

In some embodiments, the pharmaceutical composition comprises at least 0.1% by weight, at least 0.5% by weight, at least 1% by weight, at least 5% by weight, at least 10% by weight, at least 15% by weight, at least 20% by weight, at least 25% by weight, at least 30% by weight, at least 35% by weight, at least 40% by weight, at least 45% by weight, at least 50% by weight, and up to 99.9% by weight, up to 99.5% by weight, up to 99% by weight, up to 98% by weight, up to 97% by weight, up to 95% by weight, up to 90% by weight, up to 85% by weight, up to 80% by weight, up to 75% by weight, up to 70% by weight, up to 65% by weight, up to 60% by weight, up to 55% by weight of the compound of Formula (I) through (V), based on a total weight of the pharmaceutical composition.

The term “excipient” refers to a diluent, adjuvant, vehicle, or carrier with which a compound of the present disclosure is formulated for administration to a mammal. “Pharmaceutically acceptable excipients” may be those diluents, adjuvants, vehicles, or carriers approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in mammals, such as humans. Such pharmaceutically acceptable excipients can be solids, semi-solids, or liquids. Examples of solid or semi-solid pharmaceutically acceptable excipients include, but are not limited to, magnesium carbonate, magnesium stearate, talc, keratin, sugar, lactose, pectin, dextrin, fructose, starch, starch paste, gum acacia, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, colloidal silica, urea, and the like. Examples of liquid pharmaceutically acceptable excipients include, but are not limited to, water, saline, and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. In addition, auxiliary, stabilizing, solubilizing, disintegrating, thickening, lubricating, flavoring, buffering, coloring agents, sweetening agents, and other pharmaceutical additives may be included in the disclosed compositions, for example those set forth hereinafter.

The pharmaceutical compositions disclosed herein may be administered at once, or multiple times at intervals of time. It is understood that the precise dosage and duration of treatment may vary with the age, weight, and condition of the patient being treated, and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test or diagnostic data. It is further understood that for any particular individual, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the formulations.

In the case wherein the patient's condition does not improve, upon the doctor's discretion the compounds may be administered chronically, that is, for an extended period of time, including throughout the duration of the patient's life in order to ameliorate or otherwise control or limit the symptoms of the patient's disease or condition.

In the case wherein the patient's status does improve, upon the doctor's discretion the compounds may be given continuously or temporarily suspended for a certain length of time (i.e., a “drug holiday”).

Once improvement of the patient's conditions has occurred, a maintenance dose is administered if desired or necessary. Subsequently, the dosage or the frequency of administration, or both, can be reduced, as a function of the symptoms, to a level at which the improved disorder is retained. Patients can, however, require intermittent treatment on a long-term basis upon any recurrence of symptoms.

Pharmaceutical compositions can take the form of capsules, tablets, pills, pellets, lozenges, powders, granules, syrups, elixirs, solutions, suspensions, emulsions, suppositories, or sustained-release formulations thereof, or any other form suitable for administration to a mammal. Administration of the subject compounds may be systemic or local. In some instances, the pharmaceutical compositions are formulated for administration in accordance with routine procedures as a pharmaceutical composition adapted for oral, intravenous, intradermal, transdermal, or inhalation administration, or other routes of administration as set forth herein, to humans. Examples of suitable pharmaceutical excipients and methods for formulation thereof are described in Remington: The Science and Practice of Pharmacy, Alfonso R. Gennaro ed., Mack Publishing Co. Easton, Pa., 19th ed., 1995, Chapters 86, 87, 88, 91, and 92, incorporated herein by reference. The choice of excipient will be determined in part by the particular compound, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of the subject pharmaceutical compositions. Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, dispersible granules, and the like. A solid excipient may be one or more substance that may also act as diluents, flavoring agents, binders, preservatives, disintegrating agents, an encapsulating material, etc. Preparations include the formulation of the active compound with encapsulating material as a carrier providing a capsule in which the active component with or without other carriers, is surrounded by a carrier, which is thus in association with it. In powders, the excipient may be a finely divided solid in a mixture with the finely divided active ingredient. In tablets, the active ingredient may be mixed with the excipient(s) having the necessary binding properties in suitable proportions and compacted in the shape and size desired. Liquid form preparations include solutions and emulsions, for example, water, water/propylene glycol solutions, or organic solvents. For parenteral injection, liquid preparations can be formulated in solution in aqueous polyethylene glycol solution. When administered to a mammal, the compounds and compositions of the present disclosure and pharmaceutically acceptable excipients may be sterile. In some instances, an aqueous medium is employed as a vehicle e.g., when the subject compound is administered intravenously, intradermally, or via inhalation, such as water, saline solutions, and aqueous dextrose and glycerol solutions.

In some embodiments, administration to a mammal will result in systemic release of a compound of the present disclosure (for example, into the bloodstream). Routes of administration may include oral routes (e.g., enteral/gastric delivery, intraoral administration such buccal, lingual, and sublingual routes); topical administration (e.g., conjuctival, intracorneal, intraocular, ophthalmic, auricular, transdermal, nasal (e.g., intranasal), vaginal, uretheral, respiratory, and rectal administration); administration by inhalation via, for example a nebulizer or inhaler; and parenteral routes (e.g., intravenous, intradermal, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial, and subcutaneous administration, including those routes using an automatic injection device). In some embodiments, the pharmaceutical composition herein is formulated for oral administration. In some embodiments, the pharmaceutical composition herein is formulated for administration via inhalation. In some embodiments, the pharmaceutical composition herein is formulated for administration via injection, e.g. intravenously, intramuscularly, intracutaneously, subcutaneously, intraduodenally, or intraperitoneally. In some embodiments, the pharmaceutical composition herein is formulated for topical administration, e.g., as a cream or in the form of a skin patch for transdermal administration. In some embodiments, the compounds described herein may be administered via an automatic injection device.

As described below, the pharmaceutical compositions of the present disclosure may be specially formulated for administration in solid, semi-solid, or liquid form, including those adapted for the following:

A. Oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, pills, cachets, lozenges, films, or capsules, e.g., those targeted for buccal, sublingual, and systemic absorption, boluses, powders, granules, syrups, pastes for application to the tongue;

B. Parenteral administration, for example, by subcutaneous, intradermal, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained release formulation;

C. Topical application/transdermal administration, for example, as a cream, ointment, or a controlled release patch or spray applied to the skin, or orifices and/or mucosal surfaces such as intravaginally or intrarectally, for example, as a pessary, cream or foam;

D. Modified release dosage forms, including delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated-, fast-, targeted-, programmed-release, and gastric retention dosage forms, such modified release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy, supra; Modified-Release Drug Delivery Technology, Rathbone et al., Eds., Drugs and the Pharmaceutical Science, Marcel Dekker, Inc.: New York, N.Y., 2002; Vol. 126); and

E. Inhalation administration, for example as an aerosol, preferably a mist. Tamper resistant dosage forms/packaging of any of the disclosed pharmaceutical compositions are contemplated.

A. Oral Administration

The pharmaceutical compositions disclosed herein may be provided in solid, semisolid, or liquid dosage forms for oral administration. As used herein, oral administration includes gastric (enteral) delivery, for example whereby the medication is taken by mouth and swallowed, as well as intraoral administration such as through the mucosal linings of the oral cavity, e.g., buccal, lingual, and sublingual administration. Suitable oral dosage forms include, but are not limited to, tablets, capsules, caplets, pills, troches, lozenges, pastilles, cachets, gelcaps, caps, pellets, orodispersible dosage forms (e.g., orally disintegrating tablets), sublingual tablets, buccal tablets, medicated chewing gum, granules, bulk powders, effervescent or non-effervescent powders or granules, solutions, boluses, emulsions, suspensions, solutions, wafers, films, sprinkles, elixirs, and syrups. In addition to the active ingredient(s), the pharmaceutical compositions may contain one or more pharmaceutically acceptable excipients (e.g., carriers or vehicles), including, but not limited to, binders, fillers, diluents, disintegrants, wetting agents, lubricants, glidants, coloring agents, dye-migration inhibitors, sweetening agents, preservatives, antioxidants, lyoprotectants, stabilizing agents, solubilizing agents, complexing agents, and flavoring agents. In some embodiments, the pharmaceutical composition contains from about 1% to about 95% by weight, 5% to about 70% by weight, or from about 10% to about 60% by weight, or from about 20% to about 50% by weight, or from about 30% to about 40% by weight of the active ingredient (e.g., a compound of Formula (I) through (V)).

In some embodiments, pharmaceutical compositions of the present disclosure may be in orodispersible dosage forms (ODxs), including orally disintegrating tablets (ODTs) (also sometimes referred to as fast disintegrating tablets, orodispersible tablets, or fast dispersible tablets) or orodispersible films (ODFs) (or wafers). Such dosage forms allow for pre-gastric absorption of the compounds herein, e.g., when administered intraorally through the mucosal linings of the oral cavity, e.g., buccal, lingual, and sublingual administration, for increased bioavailability and faster onset compared to oral administration through the gastrointestinal tract.

Orodispersible dosage forms can be prepared by different techniques, such as freeze drying (lyophilization), molding, spray drying, mass extrusion or compressing. Preferably, the orodispersible dosage forms are prepared by lyophilization. In some embodiments, the orodispersible dosage forms disintegrate in less than about 90 seconds, in less than about 60 seconds, in less than about 30 seconds, in less than about 20, in less than about 10 seconds, in less than about 5 seconds, or in less than about 2 seconds after being received in the oral cavity. In some embodiments, the orodispersible dosage forms dissolve in less than about 90 seconds, in less than about 60 seconds, or in less than about 30 seconds after being received in the oral cavity. In some embodiments, the orodispersible dosage forms disperse in less than about 90 seconds, in less than about 60 seconds, in less than about 30 seconds, in less than about 20, in less than about 10 seconds, in less than about 5 seconds, or in less than about 2 seconds after being received in the oral cavity. In some embodiments, the pharmaceutical compositions are in the form of orodispersible dosage forms, such as oral disintegrating tablets (ODTs), having a disintegration time according to the United States Phamacopeia (USP) disintegration test <701 > of not more than about 30 seconds, not more than about 20, not more than about 10 seconds, not more than about 5 seconds, not more than about 2 seconds. Orodispersible dosage forms having longer disintegration times according to the United States Phamacopeia (USP) disintegration test <701 >, such as when adapted for extended release, for example 2 minutes, 3 minutes, 4 minutes, 5 minutes, 10 minutes, 15 minutes, 20 minutes, 25 minutes, 30 minutes, 45 minutes, 60 minutes, or any range therebetween, or longer, are also contemplated.

In some embodiments, the pharmaceutical compositions are in the form of lyophilized orodispersible dosage forms, such as lyopholized ODTs. In some embodiments, the lyophilized orodispersible dosage forms (e.g., lyophilized ODTs) are created by creating a porous matrix by subliming the water from pre-frozen aqueous formulation of the drug containing matrix- forming agents and other excipients such as those set forth herein, e.g., one or more lyoprotectants, preservatives, antioxidants, stabilizing agents, solubilizing agents, flavoring agents, etc. In some embodiments, the orodispersible dosage form comprises two component frameworks of a lyophilized matrix system that work together to ensure the development of a successful formulation. In some embodiments, the first component is a water-soluble polymer such as gelatin, dextran, alginate, and maltodextrin. This component maintains the shape and provides mechanical strength to the dosage form (binder). In some embodiments, the second constituent is a matrix-supporting/disintegration-enhancing agent such as sucrose, lactose, mannitol, xylitol, microcrystalline cellulose, calcium diphosphate, and/or starch, which acts by cementing the porous framework, provided by the water-soluble polymer and accelerates the disintegration of the orodispersible dosage form. In some embodiments, the lyophilized orodispersible dosage form (e.g., lyophilized ODT) includes gelatin and mannitol. In some embodiments, the lyophilized orodispersible dosage form (e.g., lyophilized ODT) includes gelatin, mannitol, and one or more of a lyoprotectant, a preservative, an antioxidant, a stabilizing agent, a solubilizing agent, a flavoring agent, etc., with particular mention being made to citric acid. A non-limiting example of an ODT formulation is Zydis® orally dispersible tablets (available from Catalent). In some embodiments, the ODT formulation (e.g., Zydis® orally dispersible tablets) includes one or more water-soluble polymers, such as gelatin, one or more matrix materials, fillers, or diluents such as mannitol, a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, and optionally a lyoprotectant, a preservative, an antioxidant, a stabilizing agent, a solubilizing agent, and/or a flavoring agent. In some embodiments, the ODT formulation (e.g., Zydis® orally dispersible tablets) includes gelatin, mannitol, a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, and an organic acid, nonlimiting examples of which are citric acid and/or tartaric acid, or any suitable organic acid set forth herein.

In some embodiments, the pharmaceutical composition is in the form of lyophilized orodispersible film (ODF) (or wafer). In some embodiments, the pharmaceutical compositions are in the form of lyophilized ODFs protected for the long-term storage by a specialty packaging excluding moisture, oxygen, and light. In some embodiments, the lyophilized ODFs are created by creating a porous matrix by subliming the water from pre-frozen aqueous formulation of the drug containing matrix-forming agents and other excipients such as those set forth herein, e.g., one or more of a lyoprotectant, a preservative, an antioxidant, a stabilizing agent, a solubilizing agent, a flavoring agent, etc. In some embodiments, the lyophilized ODF includes a thin water-soluble film matrix. In some embodiments, the ODFs comprise two component frameworks of a lyophilized matrix system that work together to ensure the development of a successful formulation. In some embodiments, the first component is a water-soluble polymer such as gelatin, dextran, alginate, and maltodextrin. This component maintains the shape and provides mechanical strength to the film/wafer (binder). In some embodiments, the second constituent is matrix-supporting/disintegration-enhancing agents such as sucrose and mannitol, which acts by cementing the porous framework, provided by the water-soluble polymer and accelerates the disintegration of the wafer. In some embodiments, the lyophilized ODFs include gelatin and mannitol. In some embodiments, the lyophilized ODFs include gelatin, mannitol, and one or more of a lyoprotectant, a preservative, an antioxidant, a stabilizing agent, a solubilizing agent, a flavoring agent, etc., with particular mention being made to citric acid.

In some embodiments, the ODF (or wafer) can comprise a monolayer, bilayer, or trilayer. In some embodiments, the monolayer ODF contains an active agent and one or more pharmaceutically acceptable excipients (e.g., carrier or excipients). In some embodiments, the bilayer ODF contains one or more excipients, such as a solubilizing agent, in a first layer and an active agent in the second layer. This configuration allows the active agent to be stored separately from the excipients and can increase the stability of the active agent and optionally increase the shelf life of the composition compared to the case where the excipients and the active agent were contained in a single layer. For tri-layer ODFs, each of the layers may be different or two of the layers, such as the upper and lower layers, may have substantially the same composition. In some embodiments, the lower and upper layers surround a core layer containing the active agent. In some embodiments, the lower and upper layers may contain one or more excipients, such as a solubilizing agent. In some embodiments, the lower and upper layers have the same composition. Alternatively, the lower and upper layers may contain different excipients or different amounts of the same excipient. The core layer typically contains the active agent, optionally with one or more excipients.

In some embodiments, in addition to the active ingredient(s), the pharmaceutical compositions in orodispersible dosage forms (ODxs) may contain one or more pharmaceutically acceptable excipients (e.g., carriers or vehicles). For example, in some embodiments, pharmaceutical compositions in orodispersible dosage forms include one or more of pharmaceutically acceptable a lyoprotectant, a preservative, an antioxidant, a stabilizing agent, a solubilizing agent, a flavoring agent, etc.

Examples of pharmaceutically acceptable lyoprotectants include, but are not limited to, disaccharides such as sucrose and trehalose, anionic polymers such as sulfobutylether-p-cyclodextrin (SBECD) and hyaluronic acid, and hydroxylated cyclodextrins.

Examples of pharmaceutically acceptable preservatives include, but are not limited to, glycerin, methyl and propylparaben, benzoic acid, sodium benzoate and alcohol.

Examples of pharmaceutically acceptable antioxidants, which may act to further enhance stability of the composition, include: (1) water soluble antioxidants, such as ascorbic acid, cysteine or salts thereof (cysteine hydrochloride), sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.

Examples of pharmaceutically acceptable stabilizing agents include, but are not limited to, fatty acids, fatty alcohols, alcohols, long chain fatty acid esters, long chain ethers, hydrophilic derivatives of fatty acids, polyvinyl pyrrolidones, polyvinyl ethers, polyvinyl alcohols, hydrocarbons, hydrophobic polymers, moisture-absorbing polymers, glycerol, methionine, monothioglycerol, ascorbic acid, citric acid, polysorbate, arginine, cyclodextrins, microcrystalline cellulose, modified celluloses (e.g., carboxymethylcellulose, sodium salt), sorbitol, and cellulose gel.

Examples of pharmaceutically acceptable solubilizing agents (or dissolution aids) include, but are not limited to, citric acid, hydroxypropylcellulose, hydroxypropylmethylcellulose, sodium stearyl fumarate, methacrylic acid copolymer LD, methylcellulose, sodium lauryl sulfate, polyoxyl 40 stearate, purified shellac, sodium dehydroacetate, fumaric acid, DL-malic acid, L-ascorbyl stearate, L- asparagine acid, adipic acid, aminoalkyl methacrylate copolymer E, propylene glycol alginate, casein, casein sodium, a carboxyvinyl polymer, carboxymethylethylcellulose, powdered agar, guar gum, succinic acid, copolyvidone, cellulose acetate phthalate, tartaric acid, dioctylsodium sulfosuccinate, zein, powdered skim milk, sorbitan trioleate, lactic acid, aluminum lactate, ascorbyl palmitate, hydroxy ethylmethylcellulose, hydroxypropylmethylcelluloseacetate succinate, polyoxyethylene (105) polyoxypropylene (5) glycol, polyoxyethylene hydrogenated castor oil 60, polyoxyl 35 castor oil, poly (sodium 4-styrenesulfonate), polyvinylacetaldiethylamino acetate, polyvinyl alcohol, maleic acid, methacrylic acid copolymer S, lauromacrogol, sulfuric acid, aluminum sulfate, phosphoric acid, calcium dihydrogen phosphate, sodium dodecylbenzenesulfonate, a vinyl pyrrolidone-vinyl acetate copolymer, sodium lauroyl sarcosinate, acetyl tryptophan, sodium methyl sulfate, sodium ethyl sulfate, sodium butyl sulfate, sodium octyl sulfate, sodium decyl sulfate, sodium tetradecyl sulfate, sodium hexadecyl sulfate, and sodium octadecyl sulfate. Of these, in some embodiments, such as in ODT formulation, citric acid is preferred.

Flavoring agents include natural flavors extracted from plants, such as fruits, and synthetic blends of compounds which produce a pleasant taste sensation or taste masking effect. Examples of flavoring agents include, but are not limited to, aspartame, saccharin (as sodium, potassium or calcium saccharin), cyclamate (as a sodium, potassium or calcium salt), sucralose, acesulfame-K, thaumatin, neohisperidin, dihydrochalcone, ammoniated glycyrrhizin, dextrose, maltodextrin, fructose, levulose, sucrose, glucose, wild orange peel, citric acid, tartaric acid, oil of wintergreen, oil of peppermint, methyl salicylate, oil of spearmint, oil of sassafras, oil of clove, cinnamon, anethole, menthol, thymol, eugenol, eucalyptol, lemon, lime, and lemon-lime.

Cyclodextrins such as a-cyclodextrin, p-cyclodextrin, y-cyclodextrin, methyl-p-cyclodextrin, hydroxyethyl p-cyclodextrin, hydroxypropyl-P-cyclodextrin, hydroxypropyl y-cyclodextrin, sulfated P- cyclodextrin, sulfated a-cyclodextrin, sulfobutyl ether p-cyclodextrin, or other solubilized derivatives can also be advantageously used to enhance delivery of compositions described herein.

Pharmaceutical compositions adapted for oral administration, e.g., general tablets including single-layer tablets, compressed tablets, coated tablets, etc. may be formulated with various excipients such as those set forth herein. Examples of suitable excipients may include, but are not limited to, binders, fillers, diluents, disintegrants, wetting agents, lubricants, glidants, coloring agents, dyemigration inhibitors, sweetening agents, preservatives, antioxidants, stabilizing agents, solubilizing agents, and flavoring agents.

Binders or granulators impart cohesiveness to a tablet to ensure the tablet remains intact after compression. Suitable binders or granulators include, but are not limited to, starches, such as corn starch, potato starch, and pre-gelatinized starch (e.g., STARCH 1500); gelatin; sugars, such as sucrose, glucose, dextrose, molasses, and lactose; natural and synthetic gums, such as acacia, alginic acid, alginates, extract of Irish moss, Panwar gum, ghatti gum, mucilage of isabgol husks, carboxymethylcellulose, methylcellulose, polyvinylpyrrolidone (PVP), Veegum, larch arabogalactan, powdered tragacanth, and guar gum; celluloses, such as ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose, methyl cellulose, hydroxyethylcellulose (HEC), hydroxypropylcellulose (HPC), hydroxypropyl methyl cellulose (HPMC); microcrystalline celluloses, such as AVICEL-PH-101, AVICEL-PH-103, AVICEL RC-581, AVICEL-PH-105 (FMC Corp., Marcus Hook, Pa.); and mixtures thereof. Suitable fillers include, but are not limited to, talc, calcium carbonate, microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof. The binder or filler may be present, e.g., from about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 99% by weight in the pharmaceutical compositions disclosed herein, or any range therebetween.

Suitable diluents include, but are not limited to, dicalcium phosphate, calcium sulfate, lactose, sorbitol, sucrose, inositol, cellulose, kaolin, mannitol, sodium chloride, dry starch, and powdered sugar. Certain diluents, such as mannitol, lactose, sorbitol, sucrose, and inositol, when present in sufficient quantity, can impart properties to some compressed tablets that permit disintegration in the mouth by chewing. Such compressed tablets can be used as chewable tablets.

Suitable disintegrants include, but are not limited to, agar; bentonite; celluloses, such as methylcellulose and carboxymethylcellulose; wood products; natural sponge; cation-exchange resins; alginic acid; gums, such as guar gum and Veegum HV; citrus pulp; cross-linked celluloses, such as croscarmellose; cross-linked polymers, such as crospovidone; cross-linked starches; calcium carbonate; microcrystalline cellulose, such as sodium starch glycolate; polacrilin potassium; starches, such as com starch, potato starch, tapioca starch, and pre-gelatinized starch; clays; aligns; and mixtures thereof. The amount of disintegrant in the pharmaceutical compositions disclosed herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art. The pharmaceutical compositions disclosed herein may contain, e.g., from about 0.5 to about 15% or from about 1 to about 5% by weight of a disintegrant.

Suitable lubricants include, but are not limited to, calcium stearate; magnesium stearate; mineral oil; light mineral oil; glycerin; sorbitol; mannitol; glycols, such as glycerol behenate and polyethylene glycol (PEG); stearic acid; sodium lauryl sulfate; sodium stearyl fumarate; talc; hydrogenated vegetable oil, including peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil; zinc stearate; ethyl oleate; ethyl laureate; agar; starch; lycopodium; silica or silica gels, such as AEROSIL® 200 (W.R. Grace Co., Baltimore, Md.) and CAB-O-SIL® (Cabot Co. of Boston, Mass.); and mixtures thereof. The pharmaceutical compositions disclosed herein may contain, e.g., about 0.1 to about 5% by weight of a lubricant. Suitable glidants include colloidal silicon dioxide, CAB-O-SIL® (Cabot Co. of Boston, Mass.), and asbestos-free talc.

Coloring agents include any of the approved, certified, water soluble FD&C dyes, and water insoluble FD&C dyes suspended on alumina hydrate, and color lakes and mixtures thereof. A color lake is the combination by adsorption of a water-soluble dye to a hydrous oxide of a heavy metal, resulting in an insoluble form of the dye.

Sweetening agents include sucrose, lactose, mannitol, syrups, glycerin, and artificial sweeteners, such as saccharin and aspartame.

Suitable emulsifying agents include gelatin, acacia, tragacanth, bentonite, and surfactants, such as polyoxyethylene sorbitan monooleate (TWEEN® 20), polyoxyethylene sorbitan monooleate 80 (TWEEN® 80), and triethanolamine oleate.

Suspending and dispersing agents include sodium carboxymethylcellulose, pectin, tragacanth, Veegum, acacia, sodium carbomethylcellulose, hydroxypropyl methylcellulose, and polyvinylpyrolidone.

Preservatives include glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol.

Wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate, and polyoxyethylene lauryl ether.

Solvents include glycerin, sorbitol, ethyl alcohol, and syrup. Examples of non-aqueous liquids utilized in emulsions include mineral oil and cottonseed oil. Organic acids include citric and tartaric acid. Sources of carbon dioxide include sodium bicarbonate and sodium carbonate.

It should be understood that many excipients (carriers, vehicles, etc.) may serve several functions, even within the same formulation. Particular mention is made to pharmaceutical compositions herein containing citric acid, which may play multiple roles as a stabilizing agent, as a solubilizing agent to provide fast dissolution of the active for rapid onset, etc., particularly for dosage forms adapted for rapid onset and a shorter duration of drug action, such as orodispersible dosage forms (e.g., ODTs and ODFs).

The tablet dosage forms may be prepared from the active ingredient in powdered, crystalline, or granular forms, alone or in combination with one or more excipients described herein, including binders, disintegrants, controlled-release polymers, lubricants, diluents, and/or colorants. Flavoring and sweetening agents are especially useful in the formation of chewable tablets and lozenges.

The pharmaceutical compositions herein may be in the form of compressed tablets, tablet triturates, chewable lozenges, rapidly dissolving tablets, multiple compressed tablets, or enteric-coating tablets, sugar-coated, or film-coated tablets. Enteric-coated tablets are compressed tablets coated with substances that resist the action of stomach acid but dissolve or disintegrate in the intestine, thus protecting the active ingredients from the acidic environment of the stomach. Enteric-coatings include, but are not limited to, fatty acids, fats, phenylsalicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalates. In some embodiments, the enteric coatings protect the dosage form from the acidic environment of the stomach and maintain an extended-release profile. Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which may be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation. Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material. Film coatings include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coating imparts the same general characteristics as sugar coating. Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press-coated or dry-coated tablets. In some embodiments, the solid oral dosage form (e.g., a single-layer tablet or caplet) is coated with one or more protective layers of inactive pharmaceutically acceptable excipients to form a modified-release formulation, e.g., to ensure steady release of the active ingredient from the matrix and avoid concentration bursts at the early release time points.

The pharmaceutical composition may be orally administered to a subject, such that a continuous therapeutically effective concentration of any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), is provided to the subject. In this regard, the disclosure provides novel and inventive formulations for oral administration comprising, e.g., optimal matrices discovered for the long-term steady release of any of the compounds of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, with reduced sedative and psychotomimetic side effects.

In some embodiments, the pharmaceutical composition (e.g., a tablet composition formulated for oral administration such as a single-layer tablet composition), comprises any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), and a polymer. In some embodiments, the pharmaceutical composition includes: (i) a water-insoluble neutrally charged non-ionic matrix; and (ii) a polymer carrying one or more negatively charged groups.

In some embodiments, the tablet composition is a modified-release tablet adapted for sustained release and preferably maximum sustained release. In some embodiments, the release period of any of the compounds described herein (e.g., a compound of Formula (I) through (V)), in the formulations of the disclosure is greater than 4 hours, greater than 6 hours, greater than 8 hours, greater than 10 hours, greater than 12 hours, greater than 16 hours, greater than 20 hours, greater than 24 hours, greater than 28 hours, greater than 32 hours, greater than 36 hours, greater than 48 hours.

In some embodiments, the tablet composition is adapted for tamper resistance. In some embodiments, the tablet composition comprises polyethylene oxide (PEO), e.g., MW about 2,000 to about 7,000 KDa, optionally in combination with HPMC. In some embodiments, the tablet composition may further comprise polyethylene glycol (PEG), e.g., PEG 8K. In some embodiments, the tablet composition may further comprise a polymer carrying one or more negatively charged groups, e.g., polyacrylic acid. In some embodiments, the tablet composition comprising PEO is further subjected to heating/annealing, e.g., extrusion conditions.

In some embodiments, the pharmaceutical composition comprises a combination of (i) a waterinsoluble neutrally charged non-ionic matrix; (ii) a polymer carrying one or more negatively charged groups; and (iii) any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof.

In some embodiments, the polymer carrying one or more negatively charged groups is selected from the group consisting of polyacrylic acid, polylactic acid, polyglycolic acid, polymethacrylate carboxylates, cation-exchange resins, clays, zeolites, hyaluronic acid, anionic gums, salts thereof, and mixtures thereof. In some embodiments, the anionic gum is selected from the group consisting of naturally occurring materials and semi-synthetic materials. In some embodiments, the naturally occurring material is selected from the group consisting of alginic acid, pectin, xanthan gum, carrageenan, locust bean gum, gum arabic, gum karaya, guar gum, and gum tragacanth. In some embodiments, the semi-synthetic material is selected from the group consisting of carboxymethyl-chitin and cellulose gum.

Moreover, without wishing to be bound by theory, in some embodiments, the role of the polymer carrying one or more negatively charged groups, e.g., moieties of acidic nature as in those of the acidic polymers described herein, surprisingly offers significant retention of any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), in the matrix. In some embodiments, this negative charge may be created in situ, for example, based on release of a proton due to pKa and under certain pH conditions or through electrostatic interaction/creation of negative charge. Further noting that acidic polymers may be the salts of the corresponding weak acids that will be the related protonated acids in the stomach; which, and without wishing to be bound by theory, will neutralize the charge and may reduce the interactions of any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), with the matrix. In addition, the release matrix may be further complemented by other inactive pharmaceutical ingredients to aid in preparation of the appropriate solid dose form such as fillers, disintegrants, flow improving agents, lubricants, colorants, taste maskers.

In some embodiments, the water-insoluble neutrally charged non-ionic matrix is selected from cellulose-based polymers such as HPMC, alone or enhanced by mixing with components selected from the group consisting of starches; waxes; neutral gums; poly methacrylates; PVA; PVA/PVP blends; and mixtures thereof.

In some embodiments, the cellulose-based polymer is hydroxypropyl methylcellulose (HPMC). In some embodiments, the tablet composition comprises about 10 to 70%, 20 to 60%, or 30 to 50% hydroxypropyl methylcellulose by weight, about 10 to 30%, or about 15 to 20% starch by weight, or any combination thereof.

In some embodiments, the disclosure provides a method of formulating any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), to ensure the steady release of a therapeutically effective concentration of any of the compounds from an oral tablet without neurologically toxic spikes, e.g., sedative or psychotomimetic toxic spikes, in plasma concentration of any of the compounds. In some embodiments, the method comprises the step of combining (i) a water-insoluble neutrally charged nonionic matrix; (ii) a polymer carrying one or more negatively charged groups; and (iii) any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), to produce an orally administered tablet composition, e.g., single-layer. In some embodiments, the method comprises the step of combining (i) polyethylene oxide (PEO), e.g., MW about 2,000 to about 7,000 KDa, with HPMC, and (ii) any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), to produce an orally administered tablet composition, e.g., single-layer. In some embodiments, the method comprises the step of combining polyethylene oxide (PEO) with HPMC, and any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), the tablet composition may further comprise a polymer carrying one or more negatively charged groups, e.g., polyacrylic acid and/or may be further subjected to heating/annealing, e.g., extrusion conditions.

Disclosed herein are pharmaceutical compositions in modified release dosage forms, which comprise a compound as disclosed herein and one or more release controlling excipients or carriers as described herein. Suitable modified release dosage excipients include, but are not limited to, hydrophilic or hydrophobic matrix devices, water-soluble separating layer coatings, enteric coatings, osmotic devices, multiparticulate devices, and combinations thereof. The pharmaceutical compositions may also comprise non-release controlling excipients or carriers.

In some embodiments, provided is a modified release oral formulation. In some embodiments, the oral pharmaceutical composition is for low dose maintenance therapy that can be constructed using the compounds described herein, capitalizing on the ability of the phenethylamine-type compounds described herein to bind with anionic polymers.

In some embodiments, the pharmaceutical composition contains a compound of the present disclosure, which is an orally active, peripherally -restricted, 5-HT2 agonist, for the treatment of autonomic nervous system disorders, including pulmonary disorders (e.g., asthma) and cardiovascular disorders (e.g., atherosclerosis).

Further disclosed herein are pharmaceutical compositions in enteric coated dosage forms, which comprise a compound as disclosed herein and one or more release controlling excipients or carriers for use in an enteric coated dosage form. The pharmaceutical compositions may also comprise non-release controlling excipients or carriers.

Further disclosed herein are pharmaceutical compositions in effervescent dosage forms, which comprise a compound as disclosed herein and one or more release controlling excipients or carriers for use in an effervescent dosage form. The pharmaceutical compositions may also comprise non-release controlling excipients or carriers. Effervescent means that the dosage form, when mixed with liquid, including water, juice, saliva, etc., evolves a gas. In general, the effervescent dosage forms of the present disclosure comprise an organic acid and a source of carbon dioxide, referred to herein as an “effervescent couple.” Such effervescent dosage forms effervesce (evolve gas) through chemical reaction between the organic acid and the source of carbon dioxide, which takes place upon exposure to an aqueous environment, such as upon placement in water, juice, or other drinkable fluid, or from the aqueous environment in the oral cavity, such as saliva in the mouth. Specifically, the reaction between the organic acid and the source of carbon dioxide produces carbon dioxide gas upon contact with an aqueous medium such as water, juice, or saliva. While use of disintegrants are optional, effervescent dosage forms do not require a disintegrant as the evolution of the gas in situ facilitates the disintegration process.

For clarity, an “effervescent couple” refers to at least one organic acid and at least one source of carbon dioxide being contained in a dosage form, regardless of assembly — for example, the organic acid and the source of carbon dioxide can be admixed (as powders), layered on top of one another, agglomerated or otherwise “glued” together in granular form, or held separately from one another such as in separate layers within the dosage form. Further, the term “couple” in this context is not meant to be limited to only an organic acid and a source of carbon dioxide and is open to the inclusion of other materials unless specified otherwise; for example, effervescent agglomerates/granules made from bringing together (or “gluing”) an organic acid and a source of carbon dioxide may include other vehicles including binders (the “glue”) and the effervescent agglomerates/granules may nonetheless be referred to as an effervescent couple.

The organic acid may be a monoacid, a diacid, a triacid, a tetraacid, or may contain a higher number of acid groups. One organic acid or mixtures of organic acids may be used. In addition to an acid group(s) (e.g., one or more carboxylic acid moieties), the organic acid may also contain one or more hydroxyl functionalities as part of its structure (i.e., the organic acid may be a hydroxy acid). In some embodiments, the organic acid is an a-hydroxy acid. In some embodiments, the organic acid is a P-hydroxy acid. In some embodiments, the organic acid is a y-hydroxy acid. Examples of hydroxy acids include, but are not limited to, glycolic acid, lactic acid, citric acid, tartaric acid, and malic acid. In some embodiments, the organic acid is citric acid and/or tartaric acid. In some embodiments, the organic acid is citric acid. In some embodiments, the organic acid is tartaric acid. In some embodiments, the organic acid is an enedioic acid, examples of which may include, but are not limited to, fumaric acid and maleic acid. In some embodiments, the organic acid is fumaric acid. In some embodiments, the organic acid is maleic acid. Mixtures and/or hydrates of the disclosed organic acid may also be used in the disclosed pharmaceutical compositions. In some embodiments, the organic acid is not a sulfonic acid (e.g., benzenesulfonic acid, camphorsulfonic acid, (+)-(lS)-camphor-10-sulfonic acid, ethane- 1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, methanesulfonic acid, naphthalene-2-sulfonic acid, naphthalene- 1,5-disulfonic acid, p-toluenesulfonic acid, ethanedisulfonic acid, etc.). In some embodiments, the organic acid is not a benzoic acid (e.g., benzoic acid, 4-acetamidobenzoic acid, 2- acetoxybenzoic acid, salicylic acid, 4-amino-salicylic acid, gentisic acid, etc.).

The source of carbon dioxide may include, but is not limited to, sodium bicarbonate, sodium carbonate, potassium carbonate, potassium bicarbonate, magnesium carbonate, calcium carbonate, and sesquicarbonate. The source of carbon dioxide can be used singly, or in combination. In some embodiments, the source of carbon dioxide is sodium bicarbonate. In some embodiments, the source of carbon dioxide is sodium carbonate. In some embodiments, the source of carbon dioxide is potassium carbonate. In some embodiments, the source of carbon dioxide is potassium bicarbonate. However, reactants which evolve oxygen or other gases besides carbon dioxide, and which are safe for human consumption, are also contemplated for use in the disclosed effervescent dosage forms, in addition to or in lieu of the source of carbon dioxide. While not wishing to be bound by theory, it is believed that the effervescence can help quickly break up the dosage form, and in some routes of administration such as intraoral routes, can help reduce the perception of grittiness by providing a distracting sensory experience of effervescence. In some embodiments, the effervescent dosage form is to be reconstituted in a drinkable fluid such as water or juice, thereby forming an oral liquid dosage form (e.g., solution), prior to consumption. In some embodiments, the effervescent dosage form is to be placed in the oral cavity, where contact with the aqueous environment (saliva) causes disintegration/dissolution of the dosage form along with effervescence. Here, the contents of the effervescent dosage form may be converted into a liquid or semi-solid dosage form, such as a solution, syrup, or paste upon mixing with the saliva, and subsequently swallowed. Alternatively, the effervescent dosage form may be an intraoral dosage form, e.g., a buccal, lingual, or sublingual dosage form, whereby placement in the aqueous environment (saliva) of the oral cavity causes disintegration/dissolution of the dosage form along with effervescence, and pre-gastric absorption of the contents through the oral mucosa. Such pre-gastric absorption may provide for increased bioavailability and faster onset compared to oral administration through the gastrointestinal tract. In some embodiments, the effervescent dosage form is a sublingual dosage form to be disintegrated/dissolved under the tongue, whereby the contents (e.g., the compounds of the present disclosure) are absorbed through the mucous membrane beneath the tongue where they enter venous circulation. In some embodiments, the effervescent dosage form is a buccal dosage form to be disintegrated/dissolved in the buccal cavity, whereby the contents (e.g., the compounds of the present disclosure) are absorbed through the oral mucosa lining the mouth where they enter venous circulation. Effervescent dosage forms may be advantageous for the treatment of pediatric/adolescent patients or patients that have general difficulty swallowing traditional dosage forms such as general tablets or capsules, since effervescent dosage forms can be reconstituted into easy to swallow liquid or semi-solid dosage forms or taken intraorally.

When adapted for intraoral administration, it may be beneficial to formulate the effervescent dosage form with a bioadhesive agent, in addition to the effervescent couple. “Bioadhesive agents” are substances which promote adhesion or adherence to a biological surface, such as mucous membranes. For example, bioadhesive agents are themselves capable of adhering to a biological surface when placed in contact with that surface (e.g., mucous membrane) in order to enable compositions of the disclosure to adhere to that surface, which promotes more efficient transfer of the contents from the dosage form to the biological surface. A variety of polymers known in the art can be used as bioadhesive agents, for example polymeric substances, preferably with an average (weight average) molecular weight above 5,000 g/mol. It is preferred that such polymeric materials are capable of rapid swelling when placed in contact with an aqueous medium such a water or saliva, and/or are substantially insoluble in water at room temperature and atmospheric pressure. Examples of suitable bioadhesive agents include, but are not limited to, cyclodextrin, cellulose derivatives such as hydroxypropylmethyl cellulose (HPMC), hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), methyl cellulose, ethyl hydroxyethyl cellulose, carboxymethyl cellulose, modified cellulose gum and sodium carboxymethyl cellulose (NaCMC); starch derivatives such as moderately cross-linked starch, modified starch and sodium starch glycolate; acrylic polymers such as carbomer and its derivatives (polycarbophyl, Carbopol®, etc.); polyvinylpyrrolidone (PVP); polyethylene oxide (PEO); chitosan (poly-(D-glucosamine)); natural polymers such as gelatin, sodium alginate, pectin; scleroglucan; xanthan gum; guar gum; poly co- (methylvinyl ether/maleic anhydride); and crosscarmellose (e.g. crosscarmellose sodium). Such polymers may be crosslinked. Combinations of two or more bioadhesive agents can also be used.

An effervescent couple can be coated with a pharmaceutically acceptable excipient, e.g., with a binder, a protective coating such as a solvent protective coating, an enteric coating, an anti-caking agent, and/or a pH modifier to prevent premature reaction, e.g., with air, moisture, and/or other ingredients contained in the pharmaceutical composition. Each component of the effervescent couple, e.g., the organic acid and/or the source of carbon dioxide, can also individually be coated with a pharmaceutically acceptable excipient, e.g., with a binder, a protective coating such as a solvent protective coating, an enteric coating, an anti-caking agent, and/or a pH modifier to prevent premature reaction, e.g., with air, moisture, and/or other ingredients contained in the pharmaceutical composition. The effervescent couple can also be mixed with previously lyophilized particles, such as one or more pharmaceutically active ingredients coated with a solvent protective or enteric coating.

The effervescent dosage form may be prepared by methods known to those skilled in the art, including, but not limited to, slugging, direct compression, roller compaction, dry or wet granulation, fusion granulation, melt-granulation, vacuum granulation, and fluid bed spray granulation, any of which may be optionally followed by compression/tableting.

The pharmaceutical compositions disclosed herein may be formulated as non-effervescent or effervescent granules and powders. The non-effervescent or effervescent granules and powders may be reconstituted into a liquid dosage form, or alternatively, compressed to form tablet dosage forms which are either non-effervescent or effervescent, respectively. Pharmaceutically acceptable excipients used in the non-effervescent or effervescent granules or powders may include, but are not limited to, binders, granulators, fillers, diluents, sweetening agent, wetting agents, stabilizing agents, solubilizing agents, anti-caking agents, pH modifiers, or any other pharmaceutical vehicle described herein. In some embodiments, the pharmaceutically acceptable excipient comprises an organic acid, such as glycolic acid, lactic acid, citric acid, tartaric acid, malic acid, fumaric acid, and/or maleic acid.

Pharmaceutically acceptable excipients used in the effervescent granules or powders include an effervescent couple, i.e., an organic acid and a source of carbon dioxide. Effervescent powders may be produced by blending or admixing the organic acid and the source of carbon dioxide (the effervescent couple) and optionally any other desired pharmaceutically acceptable excipient. Effervescent granules may be produced by physically adhering or “gluing” the effervescent couple (the organic acid and the source of carbon dioxide) together using an edible or pharmaceutically acceptable binder such as polyvinylpyrrolidone, polyvinyl alcohol, L-leucine, polyethylene glycol, gum arabic, or the like, including combinations thereof. These types of granules are made by processes generically known as “wet granulation.” Granulating solvents such as ethanol and/or isopropyl alcohol are often used to aid this type of granulation process. Since the effervescent couple is physically bound together in the granule, the gas generating reaction is usually quite vigorous, leading to rapid dissolution times. Another type of “wet granulation” product that is specific to effervescent products is known as “fusion” type granules. These granules are formed by reacting the organic acid and source of carbon dioxide with a small amount of water (or sometimes a hydrous alcohol granulating solvent, such as various commercial grades of ethanol or isopropyl alcohol) in a highly controlled way. Since the effervescent reaction generates carbon dioxide, fusion granules tend to be quite porous, which decreases their density and also their dissolution time. Accordingly, effervescent granules prepared by wet granulation or fusion type processes may be desirable for making orodispersible dosage forms (ODxs) or other dosage forms where quick dissolving/disintegrating properties are sought. Effervescent tablet dosage forms prepared through tableting, e.g., compression, of effervescent granules or powders are also included in the present disclosure.

Additionally disclosed are pharmaceutical compositions in a dosage form that has an instant releasing component and at least one delayed releasing component, and is capable of giving a discontinuous release of the compound in the form of at least two consecutive pulses separated in time from about 0.1 up to about 24 hours (e.g., about 0.1, 0.5, 1, 2, 4, 6, 8, 10, 12, 14, 16, 18, 10, 22, or 24 hours). The pharmaceutical compositions comprise a compound as disclosed herein and one or more release controlling and non-release controlling excipients or carriers, such as those excipients or carriers suitable for a disruptable semipermeable membrane and as swellable substances.

Disclosed herein also are pharmaceutical compositions in a dosage form for oral administration to a subject, which comprise a compound disclosed herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof) and one or more pharmaceutically acceptable excipients enclosed in an intermediate reactive layer comprising a gastric juice-resistant polymeric layered material partially neutralized with alkali and having cation exchange capacity and a gastric juice-resistant outer layer.

The dosage form may be an immediate release (IR) dosage form, examples of which include, but are not limited to, an immediate release (IR) tablets or an immediate release (IR) capsule. In addition to the active ingredient (e.g., a compound of Formula (I) through (V)), dosage forms adapted for immediate release may include one or more pharmaceutically acceptable excipients which readily disperse, dissolve, or otherwise breakdown in the gastric environment so as not to delay or prolong dissolution/absorption of the active ingredient(s). Examples of pharmaceutically acceptable excipients for immediate release dosage forms include, but are not limited to, one or more binders/granulators, matrix materials, fillers, diluents, disintegrants, dispersing agents, solubilizing agents, lubricants, and/or performance modifiers. In some embodiments, the immediate release (IR) dosage form is an immediate release (IR) tablet comprising one or more of microcrystalline cellulose, sodium carboxymethylcellulose, magnesium stearate, mannitol, crospovidone, and sodium stearyl fumarate. In some embodiments, the immediate release (IR) dosage form comprises microcrystalline cellulose, sodium carboxymethylcellulose, and magnesium stearate. In some embodiments, the immediate release (IR) dosage form comprises mannitol, crospovidone, and sodium stearyl fumarate.

The pharmaceutical compositions disclosed herein may be disclosed as soft or hard capsules, which can be made from gelatin, methylcellulose, starch, or calcium alginate. The hard gelatin capsule, also known as dry-filled capsule (DFC) or powder in capsule (PIC), consists of two sections, one slipping over the other, thus completely enclosing the active ingredient. The soft elastic capsule (SEC) is a soft, globular shell, such as a gelatin shell, which is plasticized by the addition of glycerin, sorbitol, or a similar polyol. The soft gelatin shells may contain a preservative to prevent the growth of microorganisms. Suitable preservatives are those as described herein, including methyl- and propyl-parabens, and sorbic acid. The liquid, semisolid, and solid dosage forms disclosed herein may be encapsulated in a capsule. Suitable liquid and semisolid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils, or triglycerides. The capsules may also be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient.

In some embodiments, the pharmaceutical compositions are in the form of immediate-release capsules for oral administration, and may further comprise cellulose, iron oxides, lactose, magnesium stearate, and sodium starch glycolate.

In some embodiments, the pharmaceutical compositions are in the form of delayed-release capsules for oral administration, and may further comprise cellulose, ethylcellulose, gelatin, hypromellose, iron oxide, and titanium dioxide.

In some embodiments, the pharmaceutical compositions are in the form of enteric coated delayed-release tablets for oral administration, and may further comprise carnauba wax, crospovidone, diacetylated monoglycerides, ethylcellulose, hydroxypropyl cellulose, hypromellose phthalate, magnesium stearate, mannitol, sodium hydroxide, sodium stearyl fumarate, talc, titanium dioxide, and yellow ferric oxide.

In some embodiments, the pharmaceutical compositions are in the form of enteric coated delayed-release tablets for oral administration, and may further comprise calcium stearate, crospovidone, hydroxypropyl methylcellulose, iron oxide, mannitol, methacrylic acid copolymer, polysorbate 80, povidone, propylene glycol, sodium carbonate, sodium lauryl sulfate, titanium dioxide, and triethyl citrate.

The formulations of the disclosure comprise orally administered pharmaceutical compositions, such as tablet, capsule, caplets, gelcap and cap compositions, which may include uncoated tablets or coated tablets, caplets and caps (including film-coated, sugar-coated tablets, and gastro-resistant/enteric- coated tablets). The oral pharmaceutical compositions for oral use may include the active ingredients, e.g., any of the compounds described herein (e.g., a compound of Formula (I) through (V)), mixed with pharmaceutically acceptable inactive excipients such as diluents, disintegrating agents, binding agents, lubricating agents, powder flow improving agent, wetting agents, sweetening agents, flavoring agents, coloring agents and preservatives. Moreover, oral pharmaceutical compositions of the disclosure are solid dosage forms intended for oral administration, e.g., obtained by dry granulation with single or multiple compressions of powders or granules. In some embodiments, the oral pharmaceutical compositions may be obtained by using wet granulation techniques. In some embodiments, the oral pharmaceutical compositions may be obtained by molding, heating/annealing, or extrusion techniques.

In some embodiments, the oral tablets are right circular solid cylinders, the end surfaces of which are flat or convex, and the edges of which may be beveled. In some embodiments, the surfaces are convex. In addition, they may have lines or break-marks (scoring), symbols or other markings.

In some embodiments, the break-mark(s) is/are intended to permit accurate subdivision of the tablet in order to provide doses of less than one tablet. In some embodiments, the tablet compositions comprise one or more excipients such as diluents, binders, disintegrating agents, glidants, lubricants, substances capable of modifying the behavior of the dosage forms and the active ingredient(s) in the gastrointestinal tract, coloring matter authorized by the appropriate national or regional authority and flavoring substances. When such excipients are used it is necessary to ensure that they do not adversely affect the stability, dissolution rate, bioavailability, safety or efficacy of the active ingredient(s); there must be no incompatibility between any of the components of the dosage form.

Coated tablets are tablets covered with one or more layers of mixtures of substances such as natural or synthetic resins, polymers, gums, fillers, sugars, plasticizers, polyols, waxes, coloring matters authorized by the appropriate national or regional authority, and flavoring substances. Such coating materials do not contain any active ingredient, e.g., any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof). The tablets may be coated for a variety of reasons such as protection of the active ingredients from burst release from the matrix, air, moisture or light, masking of unpleasant tastes and odors or improvement of appearance. The substance used for coating may be applied as a solution or suspension.

In some embodiments, the manufacturing processes for the oral pharmaceutical compositions, e.g., tablets, meet the requirements of good manufacturing practices (GMP). In some embodiments, one or more measures are taken in the manufacture of oral pharmaceutical compositions selected from the following: ensure that mixing with excipients is carried out in a manner that ensures homogeneity; ensure that the oral pharmaceutical compositions possess a suitable mechanical strength to avoid crumbling or breaking on subsequent processing, e.g., coating, storage and distribution; minimize the degradation of the active ingredient; minimize the risk of microbial contamination; minimize the risk of cross-contamination. In addition, in the manufacture of scored tablets (tablets bearing a break-mark or marks) for which subdivision is intended in order to provide doses of less than one tablet measures are taken to: ensure the effectiveness of break-marks with respect to the uniformity of mass or content, as appropriate, of the subdivided parts so that the patient receives the intended dose.

When used for daily dosing, a suitable therapeutically effective dose will generally be in the range of about 0.00001 to about 10 mg per kilogram body weight of the subject per day, or about 0.01 to about 10 mg per kilogram body weight of the subject per day, or in the range of about 0.1 to about 5 mg per kilogram body weight per day, or in the range of about 0.5 to about 3 mg per kilogram body weight per day, or in the range of about 1 to about 2 mg per kilogram body weight per day. Additional 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, Maack Publishing Co, Easton Pa. (“Remington's”). After a pharmaceutical composition has been formulated in an acceptable excipient, it can be placed in an appropriate container and labeled for treatment of an indicated condition. For administration of the formulations comprising any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), such labeling would include, e.g., instructions concerning the amount, frequency, method of administration, treatment regimen and indications.

The pharmaceutical compositions disclosed herein may be disclosed in liquid and semisolid dosage forms, including emulsions, solutions, suspensions, elixirs, and syrups. An emulsion is a two- phase system, in which one liquid is dispersed in the form of small globules throughout another liquid, which can be oil-in-water or water-in-oil. Emulsions may include a pharmaceutically acceptable nonaqueous liquids or solvent, emulsifying agent, and preservative. Suspensions may include a pharmaceutically acceptable suspending agent and preservative. Aqueous alcoholic solutions may include a pharmaceutically acceptable acetal, such as a di(lower alkyl) acetal of a lower alkyl aldehyde (the term “lower” means an alkyl having between 1 and 6 carbon atoms), e.g., acetaldehyde diethyl acetal; and a water-miscible solvent having one or more hydroxyl groups, such as propylene glycol and ethanol. Elixirs are clear, sweetened, and hydroalcoholic solutions. Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may also contain a preservative. For a liquid dosage form, for example, a solution in a polyethylene glycol may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be measured conveniently for administration.

Other useful liquid and semisolid dosage forms include, but are not limited to, those containing the active ingredient(s) disclosed herein (e.g. , a compound of Formula (I) through (V)), and a dialkylated mono- or poly-alkylene glycol, including, 1 ,2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol- 750-dimethyl ether, wherein 350, 550, and 750 refer to the approximate average molecular weight of the polyethylene glycol. These formulations may further comprise one or more antioxidants, such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxy coumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates. In some embodiments, examples of pharmaceutically acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alphatocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.

Cyclodextrins such as a-cyclodextrin, p-cyclodextrin, y-cyclodextrin, methyl-p-cyclodextrin, hydroxyethyl p-cyclodextrin, hydroxypropyl-P-cyclodextrin, hydroxypropyl y-cyclodextrin, sulfated P- cyclodextrin, sulfated a-cyclodextrin, sulfobutyl ether p-cyclodextrin, or other solubilized derivatives can also be advantageously used to enhance delivery of compositions described herein.

The pharmaceutical compositions disclosed herein for oral administration may be also disclosed in the forms of liposomes, micelles, microspheres, or nanosystems.

Eiquid dosage forms such as aqueous solutions suitable for oral use can be prepared by dissolving the active ingredient (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), in pharmaceutically acceptable aqueous medium (e.g., water) and optionally adding a suitable excipient(s) such as coloring agents, flavoring agents, stabilizing agents, and thickening agents, as desired. Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active ingredient in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, and other well- known suspending agents. Also included are solid form preparations that are intended to be converted, shortly before use, to liquid dosage forms, including those intended for oral administration. Such liquid dosage forms include solutions, suspensions, and emulsions. These preparations may contain, in addition to the active ingredient, pharmaceutically acceptable excipients such as coloring agents, flavoring agents, stabilizing agents, buffering agents (buffers), artificial and natural sweeteners, dispersants, thickening agents, solubilizing agents, and the like. The pharmaceutical compositions disclosed herein may be disclosed as non-effervescent or effervescent, granules, tablets, and powders, to be reconstituted into a liquid dosage form prior to use. For example, oral liquid dosage forms may be prepared by reconstituting a solid dosage form disclosed herein into a pharmaceutically acceptable aqueous medium such as water, juice, or other drinkable fluid prior to use.

Pharmaceutically acceptable carriers and excipients used in the non-effervescent granules or powders may include diluents, sweeteners, and wetting agents. Pharmaceutically acceptable carriers and excipients used in the effervescent granules or powders may include organic acids and a source of carbon dioxide.

Coloring and flavoring agents can be used in any of the disclosed dosage forms.

The pharmaceutical compositions disclosed herein may be co-formulated with other active ingredients which do not impair the desired therapeutic action, or with substances that supplement the desired action, such as hydrocortisone.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for the treatment of pain. In some embodiments, the pain treated is cancer pain, e.g., refractory cancer pain. In some embodiments, the pain treated is post-surgical pain. In some embodiments, the pain treated is orthopedic pain. In some embodiments, the pain treated is back pain. In some embodiments, the pain treated is neuropathic pain. In some embodiments, the pain treated is dental pain. In some embodiments, the pain treated is chronic pain. In some embodiments, the pain treated is chronic pain in opioid-tolerant patients. In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating acute pain (e.g., acute trauma pain).

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for the treatment of depression.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for the treatment of brain injury.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for the treatment of stroke.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating migraine, e.g., with aura.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating refractory asthma.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating alcohol dependence.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating post traumatic stress disorder (PTSD).

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating depression (e.g., treatment resistant depression (TRD) or bipolar depression).

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating major depressive disorder (MDD).

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating anxiety (e.g., generalized anxiety disorder). In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating comorbidities such as generalized anxiety disorder with depression.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating schizophrenia.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating bipolar disorder.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating suicidality or suicidal ideation.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating autism.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating diabetic neuropathy.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating neuropathic pain.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating levodopa- induced dyskinesia.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating or modulating a pseudobulbar effect or Bulbar function.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating Alzheimer's disease or conditions associated with Alzheimer's disease (e.g., Alzheimer's dementia or Alzheimer's agitation).

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating tinnitus.

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating inflammation, e.g., inflammation associated with persistent symptoms from a SARS-CoV-2 infection (COVID-19), e.g. “long covid.”

In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating a disease or disorder associated with a serotonin 5-HT2 receptor. In some embodiments, the oral dosage form (e.g., tablet composition) comprises a therapeutically effective amount of any of the compounds described herein for use in treating a disease or disorder associated with a serotonin receptor or a monoamine transporter.

In some embodiments, the disease or disorder is selected from the group consisting of central nervous system (CNS) disorders, including post-traumatic stress disorder (PTSD), major depressive disorder (MDD), treatment-resistant depression (TRD), suicidal ideation, suicidal behavior, major depressive disorder with suicidal ideation or suicidal behavior, non-suicidal self-injury disorder (NSSID), bipolar and related disorders including bipolar I disorder, bipolar II disorder, cyclothymic disorder, obsessive-compulsive disorder (OCD), generalized anxiety disorder (GAD), social anxiety disorder, substance use disorders including alcohol use disorder, opioid use disorder, amphetamine use disorder, nicotine use disorder, and cocaine use disorder, anorexia nervosa, bulimia nervosa, binge eating disorder, Alzheimer’s disease, cluster headache and migraine, attention deficit hyperactivity disorder (ADHD), pain and neuropathic pain, aphantasia, childhood-onset fluency disorder, major neurocognitive disorder, mild neurocognitive disorder, sexual dysfunction, chronic fatigue syndrome, Lyme disease, and obesity.

In some embodiments, the disease or disorder includes conditions of the autonomic nervous system (ANS).

In some embodiments, the disease or disorder includes pulmonary disorders including asthma and chronic obstructive pulmonary disorder (COPD).

In some embodiments, the disease or disorder includes cardiovascular disorders including atherosclerosis.

In some embodiments, the oral dosage form (e.g., tablet composition) is utilized as a 2-times a day (BID), 3-times a day (TID) or 4-times a day (QID) application. In some embodiments, the oral dosage form (e.g., tablet composition) is utilized as a once a day (QD) application. In some embodiments, the oral dosage form (e.g., tablet composition) is utilized as a nightly (QHS) application. In some embodiments the oral dosage form (e.g., tablet composition) is utilized as an as needed (PRN) application.

In some embodiments, when administered in unit dosage form, the oral administration event may comprise one single unit dose (e.g., pill) or multiple unit doses (e.g., multiple pills) which together sum to the desired dosage.

B. Parenteral Administration

The pharmaceutical compositions disclosed herein may be administered parenterally by injection, infusion, perfusion, or implantation, for local or systemic administration. Parenteral administration, as used herein, includes, but is not limited to, intravenous, intradermal, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial, and subcutaneous administration.

The pharmaceutical compositions disclosed herein may be formulated in any dosage forms that are suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems, and solid forms suitable for solutions or suspensions in liquid prior to injection. Such dosage forms can be prepared according to conventional methods known to those skilled in the art of pharmaceutical science (see, Remington: The Science and Practice of Pharmacy, supra).

In some embodiments, the pharmaceutical composition is in the form of an injectable (liquid) dosage form (e.g., for intravenous, intramuscular, subcutaneous, etc. administration). In some embodiments, injectable (liquid) dosage forms (e.g., for intravenous, intramuscular, subcutaneous, etc. administration) are prepared by reconstituting a solid dosage form disclosed herein into a pharmaceutically acceptable liquid medium such as water, saline solutions, viscous aqueous solutions/suspensions, water-miscible vehicles (e.g., organic solvents such as N-methyl-2-pyrrolidone), etc. prior to use.

The pharmaceutical compositions intended for parenteral administration may include one or more pharmaceutically acceptable excipients, including, but not limited to, aqueous vehicles, water- miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, cryoprotectants, lyoprotectants, thickening agents, pH adjusting agents, and inert gases.

Suitable aqueous vehicles include, but are not limited to, water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringers injection, isotonic dextrose injection, sterile water injection, dextrose and lactated Ringers injection. Non-aqueous vehicles include, but are not limited to, fixed oils of vegetable origin, castor oil, corn oil, cottonseed oil, olive oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oils, hydrogenated soybean oil, and medium-chain triglycerides of coconut oil, and palm seed oil. Water-miscible vehicles include, but are not limited to, ethanol, 1,3 -butanediol, liquid polyethylene glycol (e.g., polyethylene glycol 300 and polyethylene glycol 400), propylene glycol, glycerin, N-methyl-2-pyrrolidone, dimethylacetamide, and dimethylsulfoxide.

Suitable antimicrobial agents or preservatives include, but are not limited to, phenols, cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzates, thimerosal, benzalkonium chloride, benzethonium chloride, methyl- and propyl-parabens, and sorbic acid. Suitable isotonic agents include, but are not limited to, sodium chloride, glycerin, and dextrose. Suitable buffering agents include, but are not limited to, phosphate, borate, sulfate, and citrate buffers. Suitable antioxidants are those as described herein, including bisulfite and sodium metabisulfite. Suitable local anesthetics include, but are not limited to, procaine hydrochloride. Suitable suspending and dispersing agents are those as described herein, including sodium carboxymethylcelluose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone. Suitable emulsifying agents include those described herein, including polyoxyethylene sorbitan monolaurate, polyoxyethylene sorbitan monooleate 80, and triethanolamine oleate. Suitable sequestering or chelating agents include, but are not limited to, EDTA. Suitable pH adjusting agents include, but are not limited to, sodium hydroxide, hydrochloric acid, citric acid, and lactic acid. Suitable complexing agents include, but are not limited to, cyclodextrins, including a-cyclodextrin, p-cyclodextrin, methyl-p-cyclodextrin, hydroxypropyl-3-cyclodextrin/hydroxypropyl- P-cyclodextrin, sulfobutylether-p-cyclodextrin, and sulfobutylether 7-O-cyclodextrin (CAPTISOL®, CyDex, Lenexa, Kans.). Suitable thickening or viscosity building agents include, for example, polyvinyl alcohol, polyvinyl pyrrolidone, methyl cellulose, hydroxypropyl methylcellulose, hydroxyethyl cellulose, carboxymethyl cellulose (e.g., sodium carboxymethyl cellulose), hydroxypropyl cellulose, chondroitin sulfate and salts thereof, hyaluronic acid and salts thereof, including crosslinked variations of any of the forgoing, and combinations of the foregoing.

In some embodiments, the pharmaceutical composition is in an injectable (liquid) dosage form. In some embodiments, the injectable (liquid) dosage form comprises a compound of the present disclosure, an aqueous vehicle (e.g., isotonic saline), a buffering agent (e.g., a citric acid buffer), optionally a pH adjusting agent (e.g., sodium hydroxide), and optionally an isotonic agent. In some embodiments, the injectable (liquid) dosage form comprises a compound of the present disclosure, an aqueous vehicle (e.g., isotonic saline), and a pH adjusting agent (e.g., sodium hydroxide), wherein the injectable (liquid) dosage form is formulated without a buffering agent (e.g., a citric acid buffer). In some embodiments, the injectable (liquid) dosage form is prepared by reconstituting a solid dosage form comprising a compound of the present disclosure, into an aqueous vehicle such as isotonic saline. Reconstitution of the solid dosage form, e.g., crystalline form of a compound of the present disclosure, can be performed immediately prior to use.

The pharmaceutical compositions disclosed herein may be formulated for single or multiple dosage administration. The single dosage formulations are packaged in an ampule, a vial, or a syringe. The multiple dosage parenteral formulations must contain an antimicrobial agent at bacteriostatic or fungistatic concentrations. All parenteral formulations must be sterile, as known and practiced in the art. The pharmaceutical composition may be intended for intravenous use. The pharmaceutically acceptable excipient can include buffers to adjust the pH to a desirable range for intravenous use. Many buffers including salts of inorganic acids such as phosphate, borate, and sulfate are known.

In some embodiments, the pharmaceutical compositions are disclosed as ready-to-use sterile solutions. In some embodiments, the pharmaceutical compositions are disclosed as sterile dry soluble products, including lyophilized powders and hypodermic tablets, to be reconstituted with an excipient (e.g., vehicle) prior to use. In some embodiments, the pharmaceutical compositions are disclosed as ready-to-use sterile suspensions. In some embodiments, the pharmaceutical compositions are disclosed as sterile dry insoluble products to be reconstituted with an excipient (e.g., vehicle) prior to use. In some embodiments, the pharmaceutical compositions are disclosed as ready-to-use sterile emulsions.

The pharmaceutical compositions may be formulated as a suspension, solid, semi-solid, or thixotropic liquid, for administration as an implanted depot or to generate a depot-like effect.

In some embodiments, the pharmaceutical compositions disclosed herein are dispersed in a solid inner matrix, which is surrounded by an outer polymeric membrane that is insoluble in body fluids but allows the active ingredient in the pharmaceutical compositions to diffuse through. Fatty acid salts of the compounds of Formula (I) through (V) may be well-suited for such dosage forms. Suitable inner matrixes include polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinylacetate copolymers, silicone rubbers, poly dimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers, such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinylalcohol, and crosslinked partially hydrolyzed polyvinyl acetate. Suitable outer polymeric membranes include polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinylacetate copolymers, silicone rubbers, polydimethyl siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride, vinylchloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer.

In some embodiments, the pharmaceutical composition is in the form of a viscous aqueous solution/suspension for injection to provide a slow/sustained absorption or depot-like effect. Here, pharmaceutical excipients which build viscosity may be used, such as thickening or viscosity building agents including, but not limited to, polyvinyl alcohol, polyvinyl pyrrolidone, methyl cellulose, hydroxypropyl methylcellulose, hydroxyethyl cellulose, carboxymethyl cellulose (e.g., sodium carboxymethyl cellulose), hydroxypropyl cellulose, chondroitin sulfate and salts thereof, hyaluronic acid and salts thereof, and combinations of the foregoing. In some embodiments, the pharmaceutically acceptable excipient comprises sodium carboxymethyl cellulose, hyaluronic acid and salts thereof, or a combination thereof. Such viscous aqueous solution/suspension dosage forms may be particularly well suited for subcutaneous or intramuscular administration, where the active ingredient can be slowly released from the injection site and absorbed over sustained periods, generating a depot-like release effect. Further, crosslinked versions of any of the forgoing may be utilized. The rate of release of the active ingredient can be controlled through the extent of cross-linking of any of the thickening or viscosity building agents described herein, or by controlling the rate that any of the forgoing are crosslinked through use, amount, or type of crosslinking agent employed. For example, a slow/sustained absorption or depot-like effect can be achieved through use or formation of a crosslinked hyaluronic acid at the injection site. In some embodiments, administration of a viscous aqueous solution/suspension dosage form, e.g., via subcutaneous or intramuscular injection, provides a release period of about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, or any range therebetween, or longer.

In some embodiments, the pharmaceutical composition is formulated with a pharmaceutically acceptable salt of a compound of Formula (I) through (V) with poor aqueous solubility (e.g., a water solubility at 22°C of less than 5 mg/mL, less than 4 mg/mL, less than 3 mg/mL, less than 2 mg/mL, less than 1 mg/mL, less than 0.5 mg/mL, less than 0.1 mg/mL), such as a fatty acid salt of a compound of Formula (I) through (V). Examples of fatty acid salt forms include, but are not limited to, those formed by contacting a compound of Formula (I) through (V) with adipic (hexandioic) acid, lauric (dodecanoic) acid, linoleic acid, myristic (tetradecanoic) acid, capric (decanoic) acid, stearic (octadecanoic) acid, oleic acid, caprylic (octanoic) acid, palmitic (hexadecenoic) acid, sebacic acid, undecylenic acid, or caproic acid. Such pharmaceutical compositions may be particularly well suited for subcutaneous or intramuscular administration, where the active ingredient can slowly solubilize and be slowly released from the injection site and absorbed over sustained periods, generating a depot-like release effect. These “slow release” salts may be optionally formulated with thickening or viscosity building agents, e.g., in viscous aqueous solution/suspension formulations. In some embodiments, administration of a pharmaceutical composition formulated with a pharmaceutically acceptable salt of a compound of Formula (I) through (V) with poor aqueous solubility, e.g., via subcutaneous or intramuscular injection, provides a release period of about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, or any range therebetween, or longer.

C. Topical Administration

The pharmaceutical compositions disclosed herein may be administered topically to the skin, orifices, or mucosa. The effects may be local or systemic. Topical administration, as described herein, includes, but is not limited to, conjuctival, intracorneal, intraocular, ophthalmic, auricular, transdermal, nasal (e.g., intranasal), vaginal, uretheral, respiratory, and rectal administration.

The pharmaceutical compositions disclosed herein may be formulated in any dosage form that is suitable for topical administration for local or systemic effect, including emulsions, solutions, suspensions, creams, gels, hydrogels, ointments, powders or dusting powders, dressings, elixirs, lotions, suspensions, tinctures, pastes, foams, films, aerosols, irrigations, sprays, suppositories, bandages, dermal patches, and inhalants. The topical formulation of the pharmaceutical compositions disclosed herein may contain the active ingredient(s) which may be mixed under sterile conditions with a pharmaceutically acceptable excipient, e.g., with any preservatives, buffers, absorption enhancers, propellants which may be required. Liposomes, micelles, microspheres, nanosystems, and mixtures thereof, may also be used.

Pharmaceutically acceptable excipients suitable for use in the topical formulations disclosed herein include, but are not limited to, aqueous vehicles, water-miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, penetration enhancers, cryoprotectants, lyoprotectants, thickening agents, and inert gases.

The ointments, pastes, creams and gels may contain, in addition to an active ingredient(s), excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.

Powders and sprays can contain, in addition to an active ingredient(s), excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays, such as those used for (intra)nasal administration, can additionally contain customary propellants, such as fluorohydrocarbons, chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.

The pharmaceutical compositions may additionally include components to provide sustained release and/or comfort. Such components include high molecular weight, anionic mucomimetic polymers, gelling polysaccharides, and finely-divided drug carrier substrates. These components are discussed in greater detail in U.S. Pat. Nos. 4,911,920; 5,403,841; 5,212,162; and 4,861,760, the entire contents of these patents are incorporated herein by reference in their entirety.

Transdermal delivery devices (e.g., patches) may be used. Such dosage forms have the added advantage of providing controlled delivery of active ingredient(s) to the body. That is, the compounds of the present disclosure (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof) can be administered via a transdermal patch at a steady state concentration, whereby the active ingredient(s) is gradually administered over time, thus avoiding drug spiking and adverse events/toxicity associated therewith.

Transdermal patch dosage forms herein may be formulated with various amounts of the active ingredient(s), depending on the disease/condition being treated, the active ingredient(s) employed, the permeation and size of the transdermal delivery device, the release time period, etc. For example, when formulated with a compound of Formula (I) through (V), a unit dose preparation may be varied or adjusted e.g., from 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, to 200 mg, 175 mg, 150 mg, 125 mg, 100 mg, 95 mg, 90 mg, 85 mg, 80 mg, 75 mg, 70 mg, 65 mg, 60 mg, 55 mg of the compound of Formula (I) through (V) (active basis) or otherwise as deemed appropriate using sound medical judgment, according to the particular application and the potency of compound.

Transdermal patches formulated with the disclosed compounds may be suitable for microdosing or sub-psychedelic (also referred to herein as sub-psychoactive) dosing, to achieve durable therapeutic benefits, with decreased toxicity. In some embodiments, the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, is administered via a transdermal patch at sub-psychoactive (yet still potentially serotonergic concentrations) concentrations, for example, over an extended period such as over a 8, 24, 48, 72, 84, 96, or 168 hour time period.

In addition to the active ingredient(s), and any optional pharmaceutically acceptable excipient(s), the transdermal patch may also include one or more of a pressure sensitive adhesive layer, a backing, and a release liner, as is known to those of ordinary skill in the art.

Transdermal patch dosage forms can be made by dissolving or dispersing the compounds herein in the proper medium. In some embodiments, the compounds of the present disclosure may be dissolved/dispersed directly into a polymer matrix forming the pressure sensitive adhesive layer. Such transdermal patches are called drug-in-adhesive (DIA) patches. Preferred DIA patch forms are those in which the active ingredient(s) is distributed uniformly throughout the pressure sensitive adhesive polymer matrix. In some embodiments, the active ingredient(s) may be provided in a layer containing the active ingredient(s) plus a polymer matrix which is separate from the pressure sensitive adhesive layer. In any case, the compounds of the present disclosure may optionally be formulated with suitable excipient(s) such as carrier agents, permeation agents/absorption enhancers, humectants/crystallization inhibitors, etc. to increase the flux across the skin.

Examples of carrier agents may include, but are not limited to, C8-C22 fatty acids, such as oleic acid, undecanoic acid, valeric acid, heptanoic acid, pelargonic acid, capric acid, lauric acid, and eicosapentaenoic acid; C8-C22 fatty alcohols such as octanol, nonanol, oleyl alcohol, decyl alcohol and lauryl alcohol; lower alkyl esters of C8-C22 fatty acids such as ethyl oleate, isopropyl myristate, butyl stearate, and methyl laurate; di(lower)alkyl esters of C6-C22 diacids such as diisopropyl adipate; monoglycerides of C8-C22 fatty acids such as glyceryl monolaurate; tetrahydrofurfuryl alcohol polyethylene glycol ether; polyethylene glycol, propylene glycol; 2-(2-ethoxyethoxy)ethanol; diethylene glycol monomethyl ether; alkylaryl ethers of polyethylene oxide; polyethylene oxide monomethyl ethers; polyethylene oxide dimethyl ethers; glycerol; ethyl acetate; acetoacetic ester; N- alkylpyrrolidone; cyclodextrins, such as a-cyclodextrin, p-cyclodextrin, y-cyclodextrin, or derivatives Examples of permeation agents/absorption enhancers include, but are not limited to, sulfoxides, such as dodecylmethylsulfoxide, octyl methyl sulfoxide, nonyl methyl sulfoxide, decyl methyl sulfoxide, undecyl methyl sulfoxide, 2-hydroxydecyl methyl sulfoxide, 2-hydroxy-undecyl methyl sulfoxide, 2-hydroxydodecyl methyl sulfoxide, and the like; surfactant-lecithin organogel (PLO), such as those formed from an aqueous phase with one or more of poloxamers, CARBOPOL and PEMULEN, a lipid phase formed from one or more of isopropyl palmitate and PPG-2 myristyl ether propionate, and lecithin; fatty acids, esters, and alcohols, such as oleyloleate and oleyl alcohol; keto acids such as levulinic acid; glycols and glycol ethers, such as diethylene glycol monoethyl ether; including mixtures thereof. Examples of humectants/crystallization inhibitors include, but are not limited to, polyvinyl pyrrolidone-co-vinyl acetate, HPMC, polymethacrylate, and mixtures thereof. The pressure sensitive adhesive layer may be formed from polymers including, but not limited to, acrylics (polyacrylates including alkyl acrylics), polyvinyl acetates, natural and synthetic rubbers (e.g., polyisobutylene), ethylenevinylacetate copolymers, polysiloxanes, polyurethanes, plasticized polyether block amide copolymers, plasticized styrene-butadiene rubber block copolymers, and mixtures thereof. The pressure-sensitive adhesive layer used in the transdermal patch of the present disclosure may be formed from an acrylic polymer pressure-sensitive adhesive, preferably an acrylic copolymer pressure sensitive adhesive. The acrylic copolymer pressure sensitive adhesive may be obtained by copolymerization of one or more alkyl (meth)acrylates (e.g., 2-ethylhexyl acrylate); aryl (meth)acrylates; arylalkyl (meth)acrylate; and (meth)acrylates with functional groups such as hydroxyalkyl (meth)acrylates (e.g., hydroxyethyl acrylate, 2-hydroxypropyl acrylate, 3-hydroxypropyl acrylate, 4-hydroxybutyl acrylate, 2-hydroxyethyl methacrylate, 2-hydroxypropyl methacrylate, 3- hydroxypropyl methacrylate, and 4-hydroxybutyl methacrylate), carboxylic acid containing (meth)acrylates (e.g., acrylic acid), and alkoxy (meth)acrylates (e.g., methoxyethyl acrylate); optionally with one or more copolymerizable monomers (e.g., vinylpyrrolidone, vinyl acetate, etc.). Specific examples of acrylic pressure-sensitive adhesives may include, but are not limited to, DURO-TAK products (Henkel) such as DURO-TAK 87-900A, DURO-TAK 87-9301, DURO-TAK 87-4098, DURO-TAK 87-2074, DURO-TAK 87-235A, DURO-TAK 87-2510, DURO-TAK 87-2287, DURO- TAK 87-4287, DURO-TAK 87-2516, DURO-TAK 387-2052, and DURO-TAK 87-2677. The backing used in the transdermal patch of the present disclosure may include flexible backings such as films, nonwoven fabrics, Japanese papers, cotton fabrics, knitted fabrics, woven fabrics, and laminated composite bodies of a nonwoven fabric and a film. Such a backing is preferably composed of a soft material that can be in close contact with a skin and can follow skin movement and of a material that can suppress skin rash and other discomforts following prolonged use of the patch. Examples of the backing materials include, but are not limited to, polyethylene, polypropylene, polyethylene terephthalate, polybutylene terephthalate, polyethylene naphthalate, polystyrene, nylon, cotton, acetate rayon, rayon, a rayon/polyethylene terephthalate composite body, polyacrylonitrile, polyvinyl alcohol, acrylic polyurethane, ester polyurethane, ether polyurethane, a styrene-isoprene- styrene copolymer, a styrene-butadiene-styrene copolymer, a styrene-ethylene-propylene-styrene copolymer, styrene -butadiene rubber, an ethylene- vinyl acetate copolymer, or cellophane, for example. Preferred backings do not adsorb or release the active ingredient(s). In order to suppress the adsorption and release of the active ingredient(s), to improve transdermal absorbability of the active ingredient(s), and to suppress skin rash and other discomforts, the backing preferably includes one or more layers composed of the material above and has a water vapor permeability. Specific examples of backings may include, but are not limited to, 3M COTRAN products such as 3M COTRAN ethylene vinyl acetate membrane film 9702, 3M COTRAN ethylene vinyl acetate membrane film 9716, 3M COTRAN polyethylene membrane film 9720, 3M COTRAN ethylene vinyl acetate membrane film 9728, and the like.

The release liner used in the transdermal patch of the present disclosure may include, but is not limited to, a polyester film having one side or both sides treated with a release coating, a polyethylene laminated high-quality paper treated with a release coating, and a glassine paper treated with a release coating. The release coating may be a fluoropolymer, a silicone, a fluorosilicone, or any other release coating known to those of ordinary skill in the art. The release liner may have an uneven surface in order to easily take out the transdermal patch from a package. Examples of release liners may include, but are not limited to SCOTCHPAK products from 3M such as 3M SCOTCHPAK 9744, 3M SCOTCHPAK 9755, 3M SCOTCHPAK 9709, and 3M SCOTCHPAK 1022.

Other layers such as abuse deterrent layers formulated with one or more irritants (e.g., sodium lauryl sulfate, poloxamer, sorbitan monoesters, glyceryl monooleates, spices, etc.), may also be employed.

Methods disclosed herein using a transdermal patch dosage form provide for systemic delivery of small doses of active ingredient(s), preferably over extended periods of time such as up to 168 hour time periods, for example from 2 to 96 hours, or 4 to 72 hours, or 8 to 24 hours, or 10 to 18 hours, or 12 to 14 hours. In particular, the compound of Formula (I) through (V) can be delivered in small, steady, and consistent doses such that deleterious or undesirable side-effects can be avoided. In some embodiments, the compound of Formula (I) through (V) is administered transdermally at subpsychoactive (yet still potentially serotonergic concentrations) concentrations. An exemplary drug-in-adhesive (DIA) patch formulation may comprise 5 to 30 wt.% of a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, 30 to 70 wt.% pressure sensitive adhesive (e.g., DURO-TAK 387-2052, DURO-TAK 87-2677, and DURO-TAK 87-4098), 1 to 10 wt.% permeation agents/absorption enhancers (e.g., oleyloleate, oleyl alcohol, levulinic acid, diethylene glycol monoethyl ether, etc.), and 5 to 35 wt.% crystallization inhibitor (e.g., polyvinyl pyrrolidone-co-vinyl acetate, HPMC, polymethacrylate, etc.), each based on a total weight of the DIA patch formulation, though it should be understood that many variations are possible in light of the teachings herein.

Therefore, provided herein are methods of treating a disease or disorder, including those associated with a serotonin 5-HT2 receptor, such as a central nervous system (CNS) disorder, a psychological disorder, or an autonomic nervous system (ANS), comprising administering a compound of the present disclosure (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt thereof) via a transdermal patch. Here, the compound of the present disclosure is one that is capable of diffusing from the matrix of the transdermal patch (e.g., from the pressure sensitive adhesive layer) across the skin of the subject and into the bloodstream of the subject. In some embodiments, the compound can be administered for treatment of CNS disease or other disorder. In some embodiments, the compound can be administered to treat depression including, but not limited to major depression, melancholic depression, atypical depression, or dysthymia. In some embodiments, the compound can be administered to treat psychological disorders including anxiety disorder, obsessive compulsive disorder, addiction (narcotic addiction, tobacco addiction, opioid addiction), alcoholism, depression and anxiety (chronic or related to diagnosis of a life-threatening or terminal illness), compulsive behavior, or a related symptom. In some embodiments, the disease or disorder is selected from the group consisting of central nervous system (CNS) disorders, including post-traumatic stress disorder (PTSD), major depressive disorder (MDD), treatment-resistant depression (TRD), suicidal ideation, suicidal behavior, major depressive disorder with suicidal ideation or suicidal behavior, non-suicidal self-injury disorder (NSSID), bipolar and related disorders including bipolar I disorder, bipolar II disorder, cyclothymic disorder, obsessive-compulsive disorder (OCD), generalized anxiety disorder (GAD), social anxiety disorder, substance use disorders including alcohol use disorder, opioid use disorder, amphetamine use disorder, nicotine use disorder, and cocaine use disorder, anorexia nervosa, bulimia nervosa, binge eating disorder, Alzheimer’s disease, cluster headache and migraine, attention deficit hyperactivity disorder (ADHD), pain and neuropathic pain, aphantasia, childhood-onset fluency disorder, major neurocognitive disorder, mild neurocognitive disorder, sexual dysfunction, chronic fatigue syndrome, Lyme disease, and obesity. In some embodiments, the disease or disorder may include conditions of the autonomic nervous system (ANS). In some embodiments, the disease or disorder may include pulmonary disorders (e.g., asthma and chronic obstructive pulmonary disorder (COPD). In some embodiments, the disease or disorder may include cardiovascular disorders (e.g., atherosclerosis).

Automatic injection devices offer a method for delivery of the compositions disclosed herein to patients. The compositions disclosed herein may be administered to a patient using automatic injection devices through a number of known devices, a non-limiting list of which includes transdermal, subcutaneous, and intramuscular delivery.

In some transdermal, subcutaneous, or intramuscular applications, a composition disclosed herein is absorbed through the skin. Passive transdermal patch devices often include an absorbent layer or membrane that is placed on the outer layer of the skin. The membrane typically contains a dose of a substance that is allowed to be absorbed through the skin to deliver the composition to the patient. Typically, only substances that are readily absorbed through the outer layer of the skin may be delivered with such transdermal patch devices.

Other automatic injection devices disclosed herein are configured to provide for increased skin permeability to improve delivery of the disclosed compositions. Non-limiting examples of structures used to increase permeability to improve transfer of a composition into the skin, across the skin, or intramuscularly include the use of one or more microneedles, which in some embodiments may be coated with a composition disclosed herein. Alternatively, hollow microneedles may be used to provide a fluid channel for delivery of the disclosed compositions below the outer layer of the skin. Other devices disclosed herein include transdermal delivery by iontophoresis, sonophoresis, reverse iontophoresis, or combinations thereof, and other technologies known in the art to increase skin permeability to facilitate drug delivery.

The pharmaceutical compositions may also be administered topically by electroporation, iontophoresis, phonophoresis, sonophoresis and microneedle or needle-free injection, such as POWDERJECT™ (Chiron Corp., Emeryville, Calif.), and BIOJECT™ (Bioject Medical Technologies Inc., Tualatin, Oreg.).

The pharmaceutical compositions disclosed herein may be disclosed in the forms of ointments, creams, and gels. Suitable ointment excipients include oleaginous or hydrocarbon vehicles, including such as lard, benzoinated lard, olive oil, cottonseed oil, and other oils, white petrolatum; emulsifiable or absorption vehicles, such as hydrophilic petrolatum, hydroxystearin sulfate, and anhydrous lanolin; water-removable vehicles, such as hydrophilic ointment; water-soluble ointment vehicles, including polyethylene glycols of varying molecular weight; emulsion vehicles, either water-in-oil (W/O) emulsions or oil-in-water (O/W) emulsions, including cetyl alcohol, glyceryl monostearate, lanolin, and stearic acid (see, Remington: The Science and Practice of Pharmacy, supra). These vehicles are emollient but generally require addition of antioxidants and preservatives.

Suitable cream base can be oil-in-water or water-in-oil. Cream vehicles may be water-washable, and contain an oil phase, an emulsifier, and an aqueous phase. The oil phase is also called the “internal” phase, which is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol. The aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant. The emulsifier in a cream formulation may be a nonionic, anionic, cationic, or amphoteric surfactant.

Gels are semisolid, suspension-type systems. Single-phase gels contain organic macromolecules distributed substantially uniformly throughout the liquid carrier. Suitable gelling agents include crosslinked acrylic acid polymers, such as carbomers, carboxypolyalkylenes, Carbopol®; hydrophilic polymers, such as polyethylene oxides, polyoxyethylene-polyoxypropylene copolymers, and polyvinylalcohol; cellulosic polymers, such as hydroxypropyl cellulose, hydroxyethyl cellulose, hydroxypropyl methylcellulose, hydroxypropyl methylcellulose phthalate, and methylcellulose; gums, such as tragacanth and xanthan gum; sodium alginate; and gelatin. In order to prepare a uniform gel, dispersing agents such as alcohol or glycerin can be added, or the gelling agent can be dispersed by trituration, mechanical mixing, and/or stirring.

The pharmaceutical compositions disclosed herein may be administered rectally, urethrally, vaginally, or perivaginally in the forms of suppositories, pessaries, bougies, poultices or cataplasm, pastes, powders, dressings, creams, plasters, contraceptives, ointments, solutions, emulsions, suspensions, tampons, gels, foams, sprays, or enemas. These dosage forms can be manufactured using conventional processes as described in Remington: The Science and Practice of Pharmacy, supra.

Rectal, urethral, and vaginal suppositories are solid bodies for insertion into body orifices, which are solid at ordinary temperatures but melt or soften at body temperature to release the active ingredient(s) inside the orifices. Pharmaceutically acceptable carriers utilized in rectal and vaginal suppositories include bases or vehicles, such as stiffening agents, which produce a melting point in the proximity of body temperature, when formulated with the pharmaceutical compositions disclosed herein; and antioxidants as described herein, including bisulfite and sodium metabisulfite. Suitable vehicles include, but are not limited to, cocoa butter (theobroma oil), glycerin-gelatin, carbowax (polyoxyethylene glycol), spermaceti, paraffin, white and yellow wax, and appropriate mixtures of mono-, di- and triglycerides of fatty acids, hydrogels, such as polyvinyl alcohol, hydroxyethyl methacrylate, polyacrylic acid; glycerinated gelatin. Combinations of the various vehicles may be used. Rectal and vaginal suppositories may be prepared by the compressed method or molding. For example, a low melting wax, such as a mixture of fatty acid glycerides or cocoa butter, or other pharmaceutically acceptable excipient is first melted and the active ingredient is dispersed homogeneously therein, as by stirring. The molten homogeneous mixture is then poured into convenient sized molds, allowed to cool, and thereby to solidify. The typical weight of a rectal and vaginal suppository is about 2 to about 3 g.

The pharmaceutical compositions disclosed herein may be administered ophthalmically in the forms of solutions, suspensions, ointments, emulsions, gel-forming solutions, powders for solutions, gels, ocular inserts, and implants.

The pharmaceutical compositions disclosed herein may be administered intranasally. The terms “nasal,” “intranasal,” and the like refers to a route of administration, or dosage forms adapted for a route of administration, wherein the pharmaceutical dosage form is taken to, or through, the nose (e.g., nasal cavity). Similarly, a “nasal delivery device” or an “intranasal delivery device” is intended to mean an apparatus that administers an active ingredient into the nasal cavity. In some embodiments, the intranasal dosage form may be in the form of an aqueous or non-aqueous solution, suspension, liposomal dispersion, emulsion, microemulsion or sol-gel. Non-limiting examples of intranasal administration include introduction of a solution or suspension in the form of a nasal spray or drops (direct instillation) or intranasal application of a gel, emulsion or ointment. Relative to an oral dosage form such as a tablet or capsule, intranasal delivery provides for rapid absorption, faster onset of therapeutic action and avoidance of first pass metabolism. The amount of active ingredient absorbed depends on many factors. These factors include, but are not limited to, the drug concentration, the drug delivery vehicle, mucosal contact time, the venous drainage of the mucosal tissues, the degree that the drug is ionized at the pH of the absorption site, the size of the drug molecule, and its relative lipid solubility.

The pharmaceutical compositions of the present disclosure for nasal administration include a compound of the present disclosure and optionally a pharmaceutically acceptable excipient including, but not limited to, permeation agents/absorption enhancers which promote nasal absorption of the active ingredient after nasal administration and agents to improve brain penetration of the drug following nasal administration, diluents, binders, lubricants, glidants, disintegrants, desensitizing agents, emulsifying agents, bioadhesive agents, solubilizing agents, suspending and dispersing agents, thickening or viscosity building agents, isotonic agents, pH adjusting agents, buffering agents, carriers, flavoring agents, sweetening agents, and mixtures thereof. In some embodiments, the active ingredient is present in the pharmaceutical composition in particulate form. In some embodiments, the particle size of the active ingredient is less than or equal to about 60 microns, which can help to ensure uniformity of any blends of the particles with other ingredients, or to provide an adequate dispersion in a liquid vehicle.

The transport of the active ingredient across normal mucosal surfaces (such as the nasal mucosa) can be enhanced by optionally combining it with a permeation agent/absorption enhancer. Examples of these permeation agents/absorption enhancers include, but are not limited to, cationic polymers, surface active agents, chelating agents, mucolytic agents, cyclodextrin, polymeric hydrogels, combinations thereof, and any other similar absorption promoting agents known to those of skill in the art. Representative examples of permeation agents/absorption enhancers include, but are not limited to, phospholipids, such as phosphatidylglycerol or phosphatidylcholine, lysophosphatidyl derivatives, such as lysophosphatidylethanolamine, lysophosphatidylcholine, lysophosphatidylglycerol, lysophosphatidylserine, or lysophosphatidic acid, polyols, such as glycerol or propylene glycol, fatty acid esters thereof such as glycerides, amino acids, and esters thereof, cyclodextrins, or others set forth herein. Gelling excipients or viscosity-increasing excipients can also be used.

The transport of the active ingredient across normal mucosal surfaces can also be enhanced by increasing the time in which the formulations adhere to the mucosal surfaces. Bioadhesive agents, for example, those which form hydrogels, exhibit muco-adhesion and controlled drug release properties and can be included in the intranasal compositions described herein. Representative bioadhesive agents capable of binding to the nasal mucosa include, but are not limited to, polycarbophil, polylysine, methylcellulose, sodium carboxymethylcellulose, hydroxypropylmethylcellulose, hydroxyethyl cellulose, pectin, Carbopol 934P, polyethylene oxide 600K, one or more poloxomers such as Pluronic F127 and/or Pluronic F-68, polyisobutylene (PIB), polyisoprene (PIP), polyvinyl pyrrolidone (PVP), polyvinyl alcohol (PVA), xanthan gum, guar gum, and locust bean gum. Other nasal delivery compositions are chitosan-based and are suitable to increase the residence time of the active ingredient on mucosal surfaces, which results in increasing its bioavailability. Thiolated polymeric vehicles that form covalent bonds with the cysteine-rich subdomains of the mucus membrane can also provide mucoadhesion, which prolongs the contact time between the active ingredient and the membrane.

The intranasal compositions can also include one or more preservatives. Representative preservatives include quaternary ammonium salts such as lauralkonium chloride, benzalkonium chloride, benzododecinium chloride, cetyl pyridium chloride, cetrimide, domiphen bromide; alcohols such as benzyl alcohol, chlorobutanol, o-cresol, phenyl ethyl alcohol; organic acids or salts thereof such as benzoic acid, sodium benzoate, potassium sorbate, parabens; or complex forming agents such as EDTA.

Intranasal dosage forms may also include ion-exchange resins, e.g., microspheres, which carry suitable anionic groups such as carboxylic acid residues, carboxymethyl groups, sulfopropyl groups and methylsulfonate groups. Ion-exchange resins, such as cation exchangers, can also be used. For example, pharmaceutical compositions may be formulated with chitosan, which is partially deacetylated chitin, or poly-N-acetyl-D-glucosamine, or a pharmaceutically acceptable salt thereof such as hydrochloride, lactate, glutamate, maleate, acetate, formate, propionate, malate, malonate, adipate, or succinate. Examples of non-ion-exchange resins (e.g., microspheres) which may be used include, but are not limited to starch, gelatin, collagen and albumin.

The pharmaceutical composition can also include an appropriate pH adjusting agent, including, but not limited to, sodium hydroxide, hydrochloric acid, citric acid, lactic acid, glutamic acid, maleic acid, acetic acid, formic acid, propionic acid, malic acid, malonic acid, adipic acid, and succinic acid.

Other ingredients such as diluents are cellulose, microcrystalline cellulose, hydroxypropyl cellulose, starch, hydroxypropyl methyl cellulose, dicalcium phosphate, calcium sulfate, lactose, sorbitol, sucrose, inositol, kaolin, mannitol, sodium chloride, and powdered sugar and the like.

Isotonic agents to adjust the tonicity of the composition may be added, including, but not limited to, sodium chloride, glucose, dextrose, mannitol, sorbitol, lactose, and the like.

Acidic, neutral, or basic buffering agents can also be added to the intranasal composition to control the pH, including, but not limited to, phosphate buffers, acetate buffers, and citrate buffers.

In addition to using permeation agents/absorption enhancers, which increase the transport of the active ingredient through the mucosa, and bioadhesive agents, which prolong the contact time of the active agent along the mucosa, the administration of the active ingredient can be controlled by using controlled release formulations. There are numerous particulate drug delivery vehicles known to those of skill in the art which can include the active ingredients and deliver them in a controlled manner. Examples include particulate polymeric drug delivery vehicles, for example, biodegradable polymers, and particles formed of non-polymeric components. These particulate drug delivery vehicles can be in the form of powders, microparticles, nanoparticles, microcapsules, liposomes, and the like. Typically, if the active ingredient is in particulate form without added components, its release rate depends on the release of the active ingredient itself. Typically, the rate of absorption is enhanced by presenting the drug in a micronized form, wherein particles are below 20 microns in diameter. In contrast, if the active ingredient is in particulate form as a blend of the active agent and a polymer, the release of the active agent is controlled, at least in part, by the removal of the polymer, typically by dissolution, biodegradation, or diffusion from the polymer matrix. In some embodiments, the pharmaceutical composition is in the form of a viscous aqueous solution/suspension for intranasal administration to provide a slow/sustained release and absorption. Here, pharmaceutically acceptable excipients which build viscosity may be used, such as thickening or viscosity building agents including, but not limited to, polyvinyl alcohol, polyvinyl pyrrolidone, methyl cellulose, hydroxypropyl methylcellulose, hydroxyethyl cellulose, carboxymethyl cellulose (e.g., sodium carboxymethyl cellulose), hydroxypropyl cellulose, chondroitin sulfate and salts thereof, hyaluronic acid and salts thereof, including crosslinked variants of any of the forgoing, and combinations of the foregoing. In some embodiments, the pharmaceutically acceptable excipient comprises sodium carboxymethyl cellulose, hyaluronic acid and salts thereof, or a combination thereof. Such viscous aqueous solution/suspension dosage forms may be particularly well suited for intranasal dosage forms whereby the active ingredient is relatively short acting and/or where longer acting formulations are desirable, in that the active ingredient can be slowly released from the administration site and absorbed over sustained periods.

In some embodiments, the pharmaceutical composition is formulated with a pharmaceutically acceptable salt of a compound of Formula (I) through (V) with poor aqueous solubility (e.g., a water solubility at 22°C of less than 5 mg/mL, less than 4 mg/mL, less than 3 mg/mL, less than 2 mg/mL, less than 1 mg/mL, less than 0.5 mg/mL, less than 0.1 mg/mL), such as a fatty acid salt of a compound of Formula (I) through (V). Examples of fatty acid salt forms include, but are not limited to, those formed by contacting a compound of Formula (I) with adipic (hexandioic) acid, lauric (dodecanoic) acid, linoleic acid, myristic (tetradecanoic) acid, capric (decanoic) acid, stearic (octadecanoic) acid, oleic acid, caprylic (octanoic) acid, palmitic (hexadecenoic) acid, sebacic acid, undecylenic acid, or caproic acid. Such pharmaceutical compositions may be particularly well suited for intranasal dosage forms whereby the active ingredient is relatively short acting and/or where longer acting formulations are desirable, in that the active ingredient can be slowly released from the administration site and absorbed over sustained periods.

Other intranasal dosage forms and methods contemplated herein are disclosed in van Woensel M, et al. Formulations for Intranasal Delivery of Pharmacological Agents to Combat Brain Disease: A New Opportunity to Tackle GBM? Cancers (Basel). 2013 Aug 14;5(3): 1020-48, incorporated herein by reference in its entirety.

Intranasal delivery devices are known in the art. Thus, any device suitable for delivery of drug to nasal mucosa may be used. Non-limiting examples of devices useful for the administration of liquid dosage forms include vapor devices (e.g., vapor inhalers), drop devices (e.g., catheters, single-dose droppers, multi-dose droppers, and unit-dose pipettes), mechanical spray pump devices (e.g., squeeze bottles, multi-dose metered-dose spray pumps, and single/duo-dose spray pumps), bi-directional spray pumps (e.g., breath-actuated nasal delivery devices), gas-driven spray systems/atomizers (e.g., single - or multi-dose HFA or nitrogen propellant-driven metered-dose inhalers, including traditional and circumferential velocity inhalers), and electrically powered nebulizers/atomizers (e.g., pulsation membrane nebulizers, vibrating mechanical nebulizers, and hand-held mechanical nebulizers). Nonlimiting examples of devices useful for the administration of powder compositions (e.g., lyophilized or otherwise dried pooled compositions) include, but are not limited to, mechanical powder sprayers (e.g., handactuated capsule-based powder spray devices and handactuated powder spray devices, hand actuated gel delivery devices), breath-actuated inhalers (e.g., single- or multi-dose nasal inhalers and capsule-based single- or multi-dose nasal inhalers), and insufflators (e.g., breath-actuated nasal delivery devices).

Use of metered sprays for intranasal delivery can also be accomplished by including the active ingredient in a solution or dispersion in a suitable medium which can be administered as a spray. Representative devices of this type are disclosed in the following patents, patent applications, and publications: W02003026559, W02002011800, W0200051672, W02002068029, W02002068030, W02002068031, W02002068032, W02003000310, W02003020350, W02003082393,

W02003084591, W02003090812, W0200041755, and the pharmaceutical literature (See e.g., Bell, A. Intranasal Delivery Devices, in Drug Delivery Devices Fundamentals and Applications, Tyle P. (ed), Dekker, New York, 1988), Remington's Pharmaceutical Sciences, Mack Publishing Co., 1975, all of which are incorporated herein by reference.

The pharmaceutical compositions may be disclosed in the form of an aerosol or solution for delivery using a pressurized container, pump, spray, atomizer, such as an atomizer using electrohydrodynamics to produce a fine mist, or nebulizer, alone or in combination with a suitable propellant, including, but not limited to, fluorohydrocarbons, chlorofluorohydrocarbons, and volatile unsubstituted hydrocarbons, such as butane, propane, 1,1,1,2-tetrafluoroethane, and/or 1, 1,1, 2, 3,3,3- heptafluoropropane. The pharmaceutical compositions may also be disclosed as a dry powder for insufflation, alone or in combination with an inert carrier such as lactose or phospholipids; and nasal drops. For intranasal use, the powder may comprise a bioadhesive agent, including chitosan or cyclodextrin.

Solutions or suspensions for use in a pressurized container, pump, spray, atomizer, or nebulizer may be formulated to contain ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilizing, or extending release of the active ingredient disclosed herein, a propellant as solvent; and/or a surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.

The pharmaceutical compositions disclosed herein may be micronized to a size suitable for delivery, such as about 50 micrometers or less, or about 10 micrometers or less. Particles of such sizes may be prepared using a comminuting method known to those skilled in the art, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenization, or spray drying.

Capsules, blisters and cartridges for use in an inhaler or insufflator may be formulated to contain a powder mix of the pharmaceutical compositions disclosed herein; a suitable powder base, such as lactose or starch; and a performance modifier, such as 1-leucine, mannitol, or magnesium stearate. The lactose may be anhydrous or in the form of the monohydrate. Other suitable excipients or carriers include dextran, glucose, maltose, sorbitol, xylitol, fructose, sucrose, and trehalose. The pharmaceutical compositions disclosed herein for inhaled/intranasal administration may further comprise a suitable flavor, such as menthol and levomenthol, or sweeteners, such as saccharin or saccharin sodium.

The pharmaceutical compositions disclosed herein for topical administration may be formulated to be immediate release or modified release, including delayed-, sustained-, pulsed-, controlled-, targeted, and programmed release.

D. Modified Release

The pharmaceutical compositions disclosed herein may be formulated as a modified release dosage form. As used herein, the term “modified release” refers to a dosage form in which the rate or place of release of the active ingredient(s) is different from that of an immediate dosage form when administered by the same route. Such modified release formulations may provide the steady release of a therapeutically effective concentration of any of the compounds of the present disclosure from the dosage form, without sedative or psychotomimetic toxic spikes in plasma concentration of any of the compounds. The pharmaceutical compositions in modified release dosage forms can be prepared using a variety of modified release devices and methods known to those skilled in the art, including, but not limited to, matrix controlled release devices, osmotic controlled release devices, multiparticulate controlled release devices, ion-exchange resins, enteric coatings, multilayered coatings, microspheres, liposomes, and combinations thereof. The release rate of the active ingredient(s) can also be modified by varying the particle sizes and polymorphism of the active ingredient(s).

In some embodiments, the pharmaceutical composition comprises an amount of any of the compounds described herein and a matrix which provides a release rate of 0.05-2 mg/kg/h over a period of 12-24 hours, e.g., 24 hours.

In some embodiments, the pharmaceutical composition achieves a combined concentration of any of the compounds described herein (e.g., a compound of Formula (I) through (V)), in plasma in the range of about 10-500 ng/ml, or about 10-300 ng/ml, or about 10-100 ng/ml, or about 10-20 ng/ml, or about 20-500 ng/ml, or about 30-400 ng/ml, or about 40-300 ng/ml, or about 50-100 ng/ml, and maintains this concentration for duration of the release period.

In some embodiments, the tablet composition is a modified-release tablet adapted for sustained release and preferably maximum sustained release. In some embodiments, the release period of any of the compounds described herein (e.g., a compound of Formula (I) through (V)), in the formulations of the disclosure is greater than 4 hours, greater than 6 hours, greater than 8 hours, greater than 10 hours, greater than 12 hours, greater than 16 hours, greater than 20 hours, greater than 24 hours, greater than 28 hours, greater than 32 hours, greater than 36 hours, greater than 48 hours. 1. Matrix Controlled Release Devices

The pharmaceutical compositions disclosed herein in a modified release dosage form may be fabricated using a matrix controlled release device known to those skilled in the art (see, Takada et al in “Encyclopedia of Controlled Drug Delivery,” Vol. 2, Mathiowitz ed., Wiley, 1999).

In some embodiments, the pharmaceutical compositions disclosed herein in a modified release dosage form is formulated using an erodible matrix device, which is water-swellable, erodible, or soluble polymers, including synthetic polymers, and naturally occurring polymers and derivatives, such as polysaccharides and proteins.

Materials useful in forming an erodible matrix include, but are not limited to, chitin, chitosan, dextran, and pullulan; gum agar, gum arabic, gum karaya, locust bean gum, gum tragacanth, carrageenans, gum ghatti, guar gum, xanthan gum, and scleroglucan; starches, such as dextrin and maltodextrin; hydrophilic colloids, such as pectin; phosphatides, such as lecithin; alginates; propylene glycol alginate; gelatin; collagen; and cellulosics, such as ethyl cellulose (EC), methylethyl cellulose (MEC), carboxymethyl cellulose (CMC), CMEC, hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), cellulose acetate (CA), cellulose propionate (CP), cellulose butyrate (CB), cellulose acetate butyrate (CAB), CAP, CAT, hydroxypropyl methyl cellulose (HPMC), HPMCP, HPMCAS, hydroxypropyl methyl cellulose acetate trimellitate (HPMCAT), and ethylhydroxy ethylcellulose (EHEC); polyvinyl pyrrolidone; polyvinyl alcohol; polyvinyl acetate; glycerol fatty acid esters; polyacrylamide; polyacrylic acid; copolymers of ethacrylic acid or methacrylic acid (EUDRAGIT®, Rohm America, Inc., Piscataway, N.J.); poly(2-hydroxyethyl-methacrylate); polylactides; copolymers of L-glutamic acid and ethyl-L-glutamate; degradable lactic acid-glycolic acid copolymers; poly-D-(-)-3-hydroxybutyric acid; and other acrylic acid derivatives, such as homopolymers and copolymers of butylmethacrylate, methylmethacrylate, ethylmethacrylate, ethylacrylate, (2- dimethylaminoethyl)methacrylate, and (trimethylaminoethyl)methacrylate chloride.

In some embodiments, the pharmaceutical compositions are formulated with a non-erodible matrix device. The active ingredient(s) is dissolved or dispersed in an inert matrix and is released primarily by diffusion through the inert matrix once administered. Materials suitable for use as a non- erodible matrix device included, but are not limited to, insoluble plastics, such as polyethylene, polypropylene, polyisoprene, polyisobutylene, polybutadiene, polymethylmethacrylate, polybutylmethacrylate, chlorinated polyethylene, polyvinylchloride, methyl acrylate-methyl methacrylate copolymers, ethylene- vinylacetate copolymers, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, vinylchloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer, polyvinyl chloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, and; hydrophilic polymers, such as ethyl cellulose, cellulose acetate, crospovidone, and cross-linked partially hydrolyzed polyvinyl acetate, and fatty compounds, such as carnauba wax, microcrystalline wax, and triglycerides.

In a matrix controlled release system, the desired release kinetics can be controlled, for example, via the polymer type employed, the polymer viscosity, the particle sizes of the polymer and/or the active ingredient(s), the ratio of the active ingredient(s) versus the polymer, and other excipients or carriers in the compositions.

The pharmaceutical compositions disclosed herein in a modified release dosage form may be prepared by methods known to those skilled in the art, including direct compression, dry or wet granulation followed by compression, melt-granulation followed by compression.

2. Osmotic Controlled Release Devices

The pharmaceutical compositions disclosed herein in a modified release dosage form may be fabricated using an osmotic controlled release device, including one-chamber system, two-chamber system, asymmetric membrane technology (AMT) (e.g., technology directed to a single-layer tablet, caplet or granules coated with an insoluble, asymmetric microporous membrane produced by controlled phase separation), extruding core system (ECS), elementary osmotic pump (EOP), and controlled- porosity osmotic pump (CPOP). The osmotic controlled release device may be in the form of a tablet, caplet or granules, for example. In general, such devices have at least two components: (a) the core which contains the active ingredient(s); and (b) a semipermeable membrane with at least one delivery port, which encapsulates the core. Single or multi-layer release systems may be utilized. The semipermeable membrane controls the influx of water to the core from an aqueous environment of use so as to cause drug release by extrusion through the delivery port(s). Without wishing to be bound by theory, because these systems use water osmotic pressure for the controlled delivery of the active ingredient, delivery rates are expected to be independent of gastrointestinal conditions.

In addition to the active ingredient(s), the core of the osmotic device optionally includes an osmotic agent, which creates a driving force for transport of water from the environment of use into the core of the device. One class of osmotic agents water-swellable hydrophilic polymers, which are also referred to as “osmopolymers” and “hydrogels,” including, but not limited to, hydrophilic vinyl and acrylic polymers, polysaccharides such as calcium alginate, polyethylene oxide (PEO) (or polyethylene glycol (PEG)), polypropylene glycol (PPG), poly(2 -hydroxyethyl methacrylate), poly(acrylic) acid, poly(methacrylic) acid, polyvinylpyrrolidone (PVP), crosslinked PVP, polyvinyl alcohol (PVA), PVA/PVP copolymers, PVA/PVP copolymers with hydrophobic monomers such as methyl methacrylate and vinyl acetate, hydrophilic polyurethanes containing large PEO blocks, sodium croscarmellose, carrageenan, hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), carboxymethyl cellulose (CMC) and carboxyethyl, cellulose (CEC), sodium alginate, polycarbophil, gelatin, xanthan gum, and sodium starch glycolate.

The other class of osmotic agents are osmogens, which are capable of imbibing water to affect an osmotic pressure gradient across the barrier of the surrounding coating. Suitable osmogens include, but are not limited to, inorganic salts, such as magnesium sulfate, magnesium chloride, calcium chloride, sodium chloride, lithium chloride, potassium sulfate, potassium phosphates, sodium carbonate, sodium sulfite, lithium sulfate, potassium chloride, and sodium sulfate; sugars, such as dextrose, fructose, glucose, inositol, lactose, maltose, mannitol, raffinose, sorbitol, sucrose, trehalose, and xylitol, organic acids, such as ascorbic acid, benzoic acid, fumaric acid, citric acid, maleic acid, sebacic acid, sorbic acid, adipic acid, edetic acid, glutamic acid, p-toluenesulfonic acid, succinic acid, and tartaric acid; urea; and mixtures thereof.

Osmotic agents of different dissolution rates may be employed to influence how rapidly the active ingredient(s) is initially delivered from the dosage form. For example, amorphous sugars, such as Mannogeme EZ (SPI Pharma, Lewes, Del.) can be used to provide faster delivery during the first couple of hours to promptly produce the desired therapeutic effect, and gradually and continually release of the remaining amount to maintain the desired level of therapeutic or prophylactic effect over an extended period of time. In this case, the active ingredient(s) is released at such a rate to replace the amount of the active ingredient metabolized and excreted.

The core may also include a wide variety of other excipients and carriers as described herein to enhance the performance of the dosage form or to promote stability or processing.

Materials useful in forming the semipermeable membrane include various grades of acrylics, vinyls, ethers, polyamides, polyesters, and cellulosic derivatives that are water-permeable and waterinsoluble at physiologically relevant pHs, or are susceptible to being rendered water-insoluble by chemical alteration, such as crosslinking. Examples of suitable polymers useful in forming the coating, include plasticized, unplasticized, and reinforced cellulose acetate (CA), cellulose diacetate, cellulose triacetate, CA propionate, cellulose nitrate, cellulose acetate butyrate (CAB), CA ethyl carbamate, CAP, CA methyl carbamate, CA succinate, cellulose acetate trimellitate (CAT), CA dimethylaminoacetate, CA ethyl carbonate, CA chloroacetate, CA ethyl oxalate, CA methyl sulfonate, CA butyl sulfonate, CA p-toluene sulfonate, agar acetate, amylose triacetate, beta glucan acetate, beta glucan triacetate, acetaldehyde dimethyl acetate, triacetate of locust bean gum, hydroxylated ethylene-vinylacetate, EC, PEG, PPG, PEG/PPG copolymers, PVP, HEC, HPC, CMC, CMEC, HPMC, HPMCP, HPMCAS, HPMCAT, poly(acrylic) acids and esters and poly-(methacrylic) acids and esters and copolymers thereof, starch, dextran, dextrin, chitosan, collagen, gelatin, polyalkenes, polyethers, polysulfones, polyethersulfones, polystyrenes, polyvinyl halides, polyvinyl esters and ethers, natural waxes, and synthetic waxes.

Semipermeable membrane may also be a hydrophobic microporous membrane, wherein the pores are substantially filled with a gas and are not wetted by the aqueous medium but are permeable to water vapor, as disclosed in U.S. Pat. No. 5,798,119. Such hydrophobic but water-vapor permeable membrane are typically composed of hydrophobic polymers such as polyalkenes, polyethylene, polypropylene, polytetrafluoroethylene, polyacrylic acid derivatives, polyethers, polysulfones, polyethersulfones, polystyrenes, polyvinyl halides, polyvinylidene fluoride, polyvinyl esters and ethers, natural waxes, and synthetic waxes.

The delivery port(s) on the semipermeable membrane may be formed post-coating by mechanical or laser drilling. Delivery port(s) may also be formed in situ by erosion of a plug of water- soluble material or by rupture of a thinner portion of the membrane over an indentation in the core. In addition, delivery ports may be formed during coating process, as in the case of asymmetric membrane coatings of the type disclosed in U.S. Pat. Nos. 5,612,059 and 5,698,220.

The total amount of the active ingredient(s) released and the release rate can substantially by modulated via the thickness and porosity of the semipermeable membrane, the composition of the core, and the number, size, and position of the delivery ports.

The pharmaceutical compositions in an osmotic controlled-release dosage form may further comprise additional conventional excipients or carriers as described herein to promote performance or processing of the composition.

The osmotic controlled-release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy , supra; Santus and Baker, J. Controlled Release 1995, 35, 1-21; Verma et al., Drug Development and Industrial Pharmacy 2000, 26, 695-708; Verma et al., J. Controlled Release 2002, 79, 7-27).

In some embodiments, the pharmaceutical compositions disclosed herein are formulated as AMT controlled-release dosage forms, which comprises an asymmetric osmotic membrane that coats a core comprising the active ingredient(s) and other pharmaceutically acceptable excipients. The AMT controlled-release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art, including direct compression, dry granulation, wet granulation, and a dip-coating method.

In some embodiments, the pharmaceutical compositions disclosed herein are formulated as ESC controlled-release dosage form, which comprises an osmotic membrane that coats a core comprising the active ingredient(s), a hydroxylethyl cellulose, and other pharmaceutically acceptable excipients or carriers.

In some embodiments, the disclosure provides a method of formulating any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), to ensure the steady release of a therapeutically effective concentration of any of the compounds from a modified release dosage form without sedative or psychotomimetic toxic spikes in plasma concentration of any of the compounds. In some embodiments, the method comprises formulation of any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), in an osmotic controlled release tablet. In these formulations, the single core layer containing any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), is surrounded by semi- permeable membrane with or without drug delivery orifice. In some embodiments, combination with the novel and inventive pharmaceutical compositions containing any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), of the disclosure and osmotic asymmetric-membrane technology or AMT (e.g., technology directed to a single-layer tablet coated with an insoluble, asymmetric microporous membrane produced by controlled phase separation) may be used to produce formulations useful in the methods and kits described herein.

3. Multiparticulate Controlled Release Devices

The pharmaceutical compositions disclosed herein in a modified release dosage form may be fabricated a multiparticulate controlled release device, which comprises a multiplicity of particles, granules, or pellets, ranging from about 10 pm to about 3 mm, about 50 m to about 2.5 mm, or from about 100 m to about 1 mm in diameter. Such multiparticulates may be made by the processes know to those skilled in the art, including wet- and dry-granulation, extrusion/spheronization, roller-compaction, melt-congealing, and by spray-coating seed cores. See, for example, Multiparticulate Oral Drug Delivery, Marcel Dekker: 1994; and Pharmaceutical Pelletization Technology, Marcel Dekker: 1989.

Other excipients or carriers as described herein may be blended with the pharmaceutical compositions to aid in processing and forming the multiparticulates. The resulting particles may themselves constitute the multiparticulate device or may be coated by various film-forming materials, such as enteric polymers, water-swellable, and water-soluble polymers. The multiparticulates can be further processed as a capsule or a tablet. 4. Targeted Delivery

The pharmaceutical compositions disclosed herein may also be formulated to be targeted to a particular tissue, receptor, or other area of the body of the subject to be treated, including liposome-, resealed erythrocyte-, and antibody-based delivery systems.

E. Inhalation Administration

The pharmaceutical compositions disclosed herein may be formulated for inhalation administration, e.g., for pulmonary absorption. Drugs, including psychedelic drugs, that can be used for inhalation administration include the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof). Suitable preparations may include liquid form preparations such as those described above, e.g., solutions and emulsions, wherein the solvent or carrier is, for example, water, water/ water-miscible vehicles such as water/propylene glycol solutions, or organic solvents, with optional buffering agents, which can be delivered as an aerosol, preferably a mist, with a carrier gas, such as air, oxygen, a mixture of helium and oxygen, or other gases and gas mixtures. The pharmaceutical compositions may also be formulated as a dry powder for insufflation, alone or in combination with an inert carrier such as lactose or phospholipids.

The pharmaceutical compositions may be in the form of an aerosol or solution for delivery using a pressurized container, pump, spray, atomizer, such as an atomizer using electrohydrodynamics to produce a fine mist, or nebulizer, alone or in combination with a suitable propellant, such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, a hydrofluoroalkane such as 1,1,1,2-tetrafluoroethane (HFA 134A) and 1,1,1,2,3,3,3-heptafluoropropane (HFA 227), carbon dioxide, perfluorinated hydrocarbons such as perflubron, and other suitable gases.

Aqueous solutions suitable for inhalation use can be prepared by dissolving the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, in water or other water-based medium. Suitable stabilizers and thickening agents can also be added. Emulsions suitable for inhalation use can be made by solubilizing the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, in an aqueous medium and dispersing the solubilized form in a hydrophobic medium, optionally with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, and other suspending agents.

Solutions or suspensions, including those for use in a pressurized container, pump, spray, atomizer, or nebulizer, may be formulated to contain a surfactant or other appropriate co-solvent, or a suitable alternative agent for dispersing, solubilizing, or extending release of the active ingredient disclosed herein, and optionally a propellant. Such surfactants or co-solvents may include, but are not limited to, Polysorbate 20, 60, and 80; Pluronic F-68, F-84, and P-103; cyclodextrin; polyoxyl 35 castor oil; sorbitan trioleate, oleic acid, or an oligolactic acid. Surfactants and co-solvents may be optionally employed at a level between about 0.001 %, about 0.01 %, about 0.1 %, about 1 %, and about 5%, about 4%, about 3%, about 2% by weight, based on a total amount of the pharmaceutical composition, or any range therebetween. Viscosity greater than that of simple aqueous solutions may be desirable in some cases to decrease variability in dispensing the formulations, to decrease physical separation of components of an emulsion of formulation, and/or otherwise to improve the formulation. Such viscosity building agents include, for example, polyvinyl alcohol, polyvinyl pyrrolidone, methyl cellulose, hydroxy propyl methylcellulose, hydroxyethyl cellulose, carboxymethyl cellulose, hydroxy propyl cellulose, chondroitin sulfate and salts thereof, hyaluronic acid and salts thereof, and combinations of the foregoing. Such viscosity building agents, when desirable, are typically employed at a level between about 0.001 %, about 0.01 %, about 0.1 %, about 1 %, and about 5%, about 4%, about 3%, about 2% by weight, based on a total amount of the pharmaceutical composition, or any range therebetween.

The compounds of the present disclosure can also be dissolved in organic solvents or aqueous mixtures of organic solvents. Organic solvents can be, for example, acetonitrile, chlorobenzene, chloroform, cyclohexane, 1 ,2-dichloroethane (DCE), dichloromethane (DCM), 1 ,2-dimethoxyethane, N,N-dimethylacetamide, N,N-dimethylformamide, 1,4-dioxane, 2-ethoxyethanol, ethylene glycol, formamide, hexane, methanol, ethanol, 2-methoxyethanol, methybutylketone, methylcyclohexane, N- methylpyrrolidone, nitromethane, pyridine, sulfolane, tetralin, toluene, 1,1,2-trichloroethylene, or xylene, and like, including combinations thereof. Organic solvents can belong to functional group categories such as ester solvents, ketone solvents, alcohol solvents, amide solvents, ether solvents, hydrocarbon solvents, etc. each of which can be used.

The compounds of the present disclosure (e.g., a compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof) can be delivered as an aerosol, preferably a mist, via inhalation, for systemic administration to the patient’s central nervous system. Preferably, the aerosol is generated without externally added heat (this does not exclude minor temperature increases caused by the formation of the aerosol itself, such as with a vibrating mesh or other nebulizer. However, such minor temperature increases can often be offset by vaporization of the drug, which results in cooling of the composition). The compounds of the present disclosure can be delivered as an aerosol, preferably a mist, with a carrier, such as air, oxygen, or a mixture of helium and oxygen, or other gas mixtures including therapeutic gas mixtures. The carrier gas, e.g., air, oxygen, a mixture of helium and oxygen, or other gases and gas mixtures, can be heated to about 50°C to about 60°C, or to about 55°C to about 56°C. When a mixture of helium and oxygen is used as the carrier, the helium can be present in the mixture of oxygen and helium at about 50%, 60%, 70%, 80% or 90% by volume, and the oxygen can be present in the mixture at about 50%, 40%, 30%, or 10% by volume, or any range therebetween.

Inhalation delivery can further comprise administering a pretreatment inhalation therapy prior to administration of the aerosol comprising the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof. The pretreatment can comprise administering via inhalation of a mixture of helium and oxygen heated to about 90°C, to about 92°C, to about 94 °C, to about 96°C, to about 98°C, to about 100°C, to about 105°C, to about 110°C, to about 115°C, to about 120°C, or any range therebetween, to the patient. For example, an inhalation procedure may involve (i) administering via inhalation a mixture of helium and oxygen heated to about 90°C to about 120°C to the patient, followed by (ii) administering via inhalation a mixture of helium and oxygen heated to about 50°C to about 60°C and the aerosol comprising the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof to the patient and then repeating steps (i) and (ii). Steps (i) and (ii) can be repeated 1, 2, 3, 4, 5, or more times.

The compounds of the present disclosure (e.g., compound of Formula (I) through (V)) can, in some embodiments, be administered via aerosol inhalation at doses of about 1 μg to about 200 mg or more (or any range between about 1 μg to about 200 mg), e.g., about 1 μg, 2 μg, 5 μg, 6 μg, 10 μg, 13 μg, 15 μg, 20 μg, 30 μg, 40 μg, 50 μg, 60 μg, 70 μg, 80 μg, 90 μg, 100 μg, 110 μg, 120 μg, 130 μg, 140 μg, 150 μg, 160 μg, 170 μg, 180 μg, 190 μg, 200 μg, 210 μg, 220 μg, 230 μg, 240 μg, 250 μg, 260 μg, 270 μg, 280 μg, 290 μg, 300 μg, 400 μg, 500 μg, 1.0 mg, 2.0 mg, 3.0 mg, 4.0 mg, 5.0 mg, 6.0 mg, 7.0 mg, 8.0 mg, 9.0 mg, 10.0 mg, 20.0 mg, 30.0 mg, 40.0 mg, 50.0 mg, 60.0 mg, 70.0 mg, 80.0 mg, 90.0 mg, 100.0 mg, 150.0 mg, 200.0 mg, or more, per inhalation session. In some embodiments, a subject can have 1, 2, 3, 4, 5 or more inhalation sessions a day. In some embodiments, a subject can have 1, 2, 3, 4, 5 or more inhalation sessions every other day, once a week, twice a week, or three times a week. In some embodiments, a subject can have 1, 2, 3, 4, 5 or more inhalation sessions every other month, twice a month, three times a month, or four times a month. In some embodiments, a subject can have 1, 2, 3, 4, 5, 6, 7, 8, or more inhalation sessions per treatment course, such as within a 28-day time period.

Aerosols

An aerosol, preferably a mist, can be delivered using, air, oxygen, a mixture of helium and oxygen, or other gases and gas mixtures, as a carrier gas. The carrier gas can be delivered at room temperature or heated. In some embodiments, an aerosol, preferably a mist comprising a compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof is delivered via inhalation using heated helium-oxygen (HELIOX) mixtures. Due to very low viscosity of helium the helium-oxygen mixtures generate gaseous streams characterized by laminar flow that is a highly desirable feature for reaching out into the deep lung areas and reducing deposition of the drug in the respiratory tract, one of the major obstacles in dose delivery via inhalation. A patient can inhale a dissolved compound disclosed herein as a mist into an alveolar region of the patient's lungs. The compound of Formula (I) through (V) can then be delivered to a fluid lining of the alveolar region of the lungs and can be systemically absorbed into patient blood circulation. Advantageously, these formulations can be effectively delivered to the blood stream upon inhalation to the alveolar regions of the lungs.

Devices suitable for delivery of heated or unheated carrier gas (e.g., air, oxygen, or heliumoxygen mixtures) include, for example, continuous mode nebulizers Flo-Mist (Phillips) and Hope (B&B Medical Technologies) and the accessories such as regulators, e.g., Medipure™ Heliox-LCQ System (PraxAir) and control box, e.g., Precision Control Flow (PraxAir). In some embodiments, a full delivery setup can be a device as described in, for example, Russian patent RU199823U1.

The term “heliox” as used herein refers to breathing gas mixtures of helium gas (He) and oxygen gas (O2). In some embodiments, the heliox mixture can contain helium in the mixture of helium and oxygen at about 50%, 60%, 70%, 80% or 90% by volume, and contain oxygen in the mixture of helium and oxygen at about 50%, 40%, 30%, or 10% by volume, or any range therebetween. The heliox mixture can thus contain helium and oxygen in a volume ratio of 50:50, 60:40, 70:30, 80:20, 90:10, or any range therebetween. In some embodiments, heliox can generate less airway resistance through increased tendency to laminar flow and reduced resistance in turbulent flow.

The use of heat in heliox mixtures can further enhance drug delivery by increasing permeability of key physical barriers for drug absorption. Heating of mucosal surfaces can increase permeability by enhancing peripheral blood circulation and relaxing the interstitial junction, as well as other mechanisms. Helium has a thermal conductivity almost 10 times higher than oxygen and nitrogen and can facilitate heat transfer more efficiently. A dry heliox mixture can be used safely as a pretreatment step when warmed up to as high as 110°C, which can enable the dry heliox mixture to heat mucosal surfaces of the lung and respiratory tract more efficiently.

Various types of personal vaporizers are known in the art. In general, personal vaporizers are characterized by heating a solid drug or compound. Vaporizers can work by directly heating a solid drug or compound to a smoldering point. Vaporizing a solid or solid concentrate can be done by convection or conduction. Convection heating of solid concentrate involves a heating element coming into contact with water, or another liquid, which then vaporizes. The hot vapor in turn directly heats the solid or solid concentrate to a smoldering point, releasing a vapor to be inhaled by a user. Conduction heating involves direct contact between the solid or solid concentrate and the heating element, which brings the solid to a smoldering point, releasing vapor to be inhaled by a user. Though vaporizers present advantages over smoking in terms of lung damage, the drug/active ingredient that is vaporized can be substantially deteriorated by the vaporizing heat.

In some embodiments, the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, is delivered via a nebulizer, which generates an aqueous-droplet aerosol, preferably a mist, containing the compound, which is optionally combined with a heated helium-oxygen mixture. In some embodiments, the disclosed compounds are delivered via a nebulizer, which generates an aqueous-droplet aerosol, preferably a mist, containing the compound, which is combined with a driving gas comprising nitrous oxide. The driving gas comprising nitrous oxide may be nitrous oxide gas itself or a therapeutic gas mixture, such as a N2O-O2 mixture or a N2O-air mixture. The therapeutic gas mixture may further include other gases such as one or more of N2, Ar, CO2, Ne, CH4, He, Kr, H2, Xe, H2O (e.g., vapor), etc. In some embodiments, the driving gas is a therapeutic gas mixture comprising N2O, which is present at a concentration ranging from 5 vol%, from 10 vol%, from 15 vol%, from 20 vol%, from 25 vol%, from 30 vol%, from 35 vol%, from 40 vol%, from 45 vol%, and up to 75 vol%, up to 70 vol%, up to 65 vol%, up to 60 vol%, up to 55 vol%, up to 50 vol%, relative to a total volume of the therapeutic gas mixture, or any range in between. The presence of nitrous oxide (being an NMDA receptor antagonist) in (or as) the driving gas can augment the effect of the disclosed compounds and provide the ability to use lower doses thereof to obtain similar levels of effect.

For example, a preparation of compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, can be placed into a liquid medium and put into an aerosol by a device, such as a nebulizer. In some embodiments, a nebulizer can be, for example, a pneumatic compressor nebulizer, an ultrasonic nebulizer, a vibrating mesh or horn nebulizer, or a microprocessor-controlled breath-actuated nebulizer. In some embodiments, a nebulizer device can be a device as described in, for example, Russian patent RU199823U1.

A nebulizer is a device that turns a drug, such as a compound of Formula (I) through (V), in solution or suspension into a fine aerosol, such as a mist, for delivery to the lungs. A nebulizer can also be referred to as an atomizer. To atomize is to put a dissolved drug into an aerosol, such as a mist, form. To deliver a drug by nebulization, a drug can be dispersed in a liquid medium, for example, water, ethanol, or propylene glycol. Additionally, the disclosed compounds can be carried in an excipient such as, for example liposomes, polymers, emulsions, micelles, nanoparticles, or polyethylenimine (PEI). Liquid drug formations for nebulizers can be, for example, aqueous solutions or viscous solutions. After application of a dispersing forcer (e.g., jet of gas, ultrasonic waves, or vibration of mesh), the dissolved drug is contained within liquid droplets, which are then inhaled. A mist can contain liquid droplets containing the drug in air or another gaseous mixture (e.g., a mixture of helium and oxygen). Jet nebulizers (also known as pneumatic nebulizers or compressor nebulizers) use compressed gas to make a mist. In some embodiments, a jet nebulizer is a microprocessor-controlled breath-actuated nebulizer, also called a breath-actuated nebulizer. A breath-actuated nebulizer creates a mist only when a patient is inhaling, rather than creating a mist continuously. A mist can be generated by, for example, passing air flow through a Venturi in a nebulizer bowl or cup. A Venturi is a system for speeding the flow of a fluid by constricting fluid in a cone shape tube. In the restriction, the fluid must increase its velocity, thereby reducing its pressure and producing a partial vacuum. As the fluid exits the constriction point, its pressure increases back to the ambient or pipe level pressure. This can form a low-pressure zone that pulls up droplets through a feed tube from a solution of drug in a nebulizer bowl, and in turn this creates a stream of atomized droplets, which flow to a mouthpiece. Higher air flows lead to a decrease in particle size and an increase in output. Due to droplets and solvent that saturates the outgoing gas, jet nebulizers can cool a drug solution in the nebulizer and increase solute concentration in the residual volume. A baffle in a nebulizer bowl or cup can be impacted by larger particles, retaining them and returning them to the solution in the nebulizer bowl or cup to be reatomized. Entrainment of air through a nebulizer bowl as the subject inhales can increase mist output during inspiration. Generation of a mist can occur with a smaller particle size distribution, but using smaller particle sizes can result in an increased nebulization time.

The unit of measurement generally used for droplet size is mass median diameter (MMD), which is defined as the average droplet diameter by mass. This unit can also be referred to as the mass mean aerodynamic diameter, or MMAD. The MMD droplet size for jet nebulizers can be about 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 5.0, 6.0, 7.0, 8.0, 9.0, 10.0 pm or more (or any range between about 1.0 and 10.0 pm), which can be smaller than that of ultrasonic nebulizers.

Ultrasonic nebulizers generate mists by using the vibration of a piezoelectric crystal, which converts alternating current to high-frequency (about 1 to about 3 MHz) acoustic energy. The solution breaks up into droplets at the surface, and the resulting mist is drawn out of the device by the patient's inhalation or pushed out by gas flow through the device generated by a small compressor. Ultrasonic nebulizers can include large-volume ultrasonic nebulizers and small-volume ultrasonic nebulizers. Droplet sizes tend to be larger with ultrasonic nebulizers than with jet nebulizers. The MMD droplet size for ultrasonic nebulizers can be about 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 9.0, 10.0 pm or more (or any range between about 2.0 and 10.0 pm). Ultrasonic nebulizers can create a dense mist, with droplets at about 100, 150, 200, 250, 300 pm/L or more.

Mesh nebulizer devices use the vibration of a piezoelectric crystal to indirectly generate a mist. Mesh nebulizers include, for example, active mesh nebulizers and passive mesh nebulizers. Active mesh nebulizers use a piezo element that contracts and expands on application of an electric current and vibrates a precisely drilled mesh in contact with the drug solution to generate a mist. The vibration of a piezoelectric crystal can be used to vibrate a thin metal plate perforated by several thousand holes. One side of the plate is in contact with the liquid to be atomized, and the vibration forces this liquid through the holes, generating a mist of tiny droplets. Passive mesh nebulizers use a transducer horn that induces passive vibrations in the perforated plate with tapered holes to produce a mist. Examples of active mesh nebulizers include the Aeroneb® (Aerogen, Galway, Ireland) and the eFlow® (PARI, Starnberg, Germany), while the Microair NE-U22® (Omron, Bannockburn, IL) is a passive mesh nebulizer. Mesh nebulizers are precise and customizable. By altering the pore size of the mesh, the device can be tailored for use with drug solutions of different viscosities, and the output rate changed. Use of this method of atomization can offer several advantages. The size of the droplets can be extremely precise because droplet size can be determined by the size of the holes in the mesh (which may be tailor-made to suit the application). Nebulizer meshes can be manufactured using methods such as electrodeposition, electroplating, and laser cutting to produce a liquid particle in gas in the respirable range. Mesh can be made of metal alloy. The metals used in mesh manufacture can include platinum, palladium, nickel, and stainless steel. The size of the droplet is about twice the size of the mesh hole. Mesh holes, therefore, can be about 0.1, 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0 pm or more (or any value in between about 0.1 and 5.0 pm). Mist generation in mesh nebulizers can vary based on the shape of the mesh, the material that the mesh is made of, and also the way that the mesh is created. In other words, different meshes can produce different sized liquid particles suspended in gas. Generally, MMD droplet size for mesh nebulizers can be about 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5., 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6,

6.7, 6.8, 6.9, 7.0pm or more (or any value in between about 1.0 and 7.0pm).

Additionally, droplet size can be programmable. In particular, geometric changes can be made to a nebulizer to provide a specific desired droplet size. Additionally, droplet size can be controlled independently of droplet velocity. The volume of liquid atomized, and the droplet velocity can also be precisely controlled by adjusting the frequency and amplitude of the mesh vibration. Furthermore, the number of holes in the mesh and their layout on the mesh can be tailored. Mesh nebulizers can be powered either by electricity or by battery.

A mist output rate in standing cloud mL per minute (for any atomization methodology described herein) can range from, for example, 0.1, 0.2. 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9 mL/minute or more (or any range between about 0.1 and 0.9 mL/minute) and the residual volume in any type of nebulizer reservoir can range from a about 0.01, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7,

1.8, 1.9, 2.0 mL or more (or any range between about 0.01 and 2.0 mL). Precise droplet size control can be advantageous since droplet size can correlate directly to kinetic drug release (KDR). Precise control of KDR can be achievable with precise control of droplet size. The compounds herein can be delivered via a mist using any methodology with an MMD droplet size of about 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 5.0, 6.0, 7.0, 8.0, 9.0, 10.0 pm or more (or any range between about 0.5 and 10.0 pm).

In some embodiments, the compounds of the present disclosure (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof) can be delivered via a continuous positive airway pressure (CPAP) or other pressure-assisted breathing device. A pressure-assisted breathing device forces a continuous column of compressed air or other gas at a fixed designated pressure against the face and nose of the patient, who is wearing a mask or nasal cap. When the patient's glottis opens to inhale, the pressure is transmitted throughout the airway, helping to open it. When the patient exhales, pressure from the deflating lungs and chest wall pushes air out against the continuous pressure, until the two pressures are equal. The air pressure in the airway at the end of exhalation is equal to the external air pressure of the machine, and this helps “splint” the airway open, allowing better oxygenation and airway recruitment. A pressure-assisted breathing device can be coupled with a means for introducing mist particles into the gas flow in the respiratory circuit and or a means for discontinuing the introduction of mist particles into the respiratory circuit when the patient exhales. See, e.g. US Pat. No. 7,267,121.

In some embodiments, a mist can be delivered by a device such as a metered dose inhaler (MDI) (also referred to as a pressurized metered dose inhaler or pMDI), which generates an organic solventdroplet mist containing a compound of Formula (I) through (V), which is optionally combined with a heated helium-oxygen mixture. In some embodiments, the compound can be delivered via a metered dose inhaler, MDI. MDI devices can include a canister which contains the compound of Formula (I) through (V) and a propellant, a metering valve which dispenses the medicament from the canister, an actuator body that receives the canister and which forms an opening for oral inhalation, and an actuator stem which receives the drug from the canister and directs it out the opening in the actuator body. A non-limiting example of a metering valve and actuator is Bespak’s BK357 valve and actuator (orfice d=0.22 mm) by Recipharm. Moving the drug canister relative to the actuator body and actuator stem causes the metering valve to release the predetermined amount of the drug. In some embodiments, the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, can be dissolved in a liquid propellant mixture (sometimes including small amounts of a volatile organic solvent) stored in a pressurized container of the MDI. The “metered dose” is the dose that is prepackaged in a single-dose inhaler, or which in a multidose inhaler is automatically measured out of a reservoir in preparation for inhalation. MDI devices can be aided with spacers. An MDI spacer is a spacer that goes between the MDI and the mouth of a user of the MDI. An MDI spacer allows droplets in the atomized dose to settle out a bit and mix with air or other gas, thus allowing for more effective delivery of a metered dose into a user's lungs when inhaled. An MDI spacer assists in preventing a user from inhaling the metered dose directly from an MDI where the dose would be traveling so fast that the droplets of the atomized spray from the MDI hit and stick to the back of the user's throat rather than being inhaled into the user's lungs where the drug of the metered dose is designed to be delivered. MDI devices offer the advantage of regular dosing, which can be controlled in the manufacture of the drug.

Drugs can also be delivered by dry powder inhalers (DPI). In such DPI devices, the drug itself can form the powder or the powder can be formed from a pharmaceutically acceptable excipient or carrier and the drug is releasably bound to a surface of the carrier powder such that upon inhalation, the moisture in the lungs releases the drug from the surface to make the drug available for systemic absorption. In some embodiments, the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, is delivered by use of a dry powder inhaler (DPI). Depending on the compound or form used, the drug can be formed into the necessary powder itself, or can be releasably bound to a surface of a carrier powder. Such carrier powders are known in the art (see, e.g., H. Hamishehkar, et al., “The Role of Carrier in Dry Powder Inhaler”, Recent Advances in Novel Drug Carrier Systems, 2012, pp.39-66).

DPI is generally formulated as a powder mixture of coarse carrier particles and micronized drug particles with aerodynamic particle diameters of 1-5 pm (see e.g., lida, Kotaro, et al. “Preparation of dry powder inhalation by surface treatment of lactose carrier particles” Chemical and pharmaceutical bulletin 51.1 (2003): 1-5). Carrier particles are often used to improve drug particle flowability, thus improving dosing accuracy and minimizing the dose variability observed with drug formulations alone while making them easier to handle during manufacturing operations. Carrier particles should have several characteristics such as physico-chemical stability, biocompatibility and biodegradability, compatible with the drug substance and must be inert, available and economical. The choice of carrier particle (both content and size) is well within the purview of one of ordinary skill in the art. The most common carrier particles are made of lactose or other sugars, with a-lactose monohydrate being the most common lactose grade used in the inhalation field for such particulate carriers.

Delivery with Helium Oxygen Mixtures

Systemic delivery of the compounds of the present disclosure (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof) can be carried out via inhalation of an aerosol comprising the compound and a carrier gas such as air, oxygen, helium, a mixture of helium and oxygen (i.e., a heliox mixture), other gases or other gas mixtures. In particular, the compounds of the present disclosure can be delivered to a patient’s CNS. Accordingly, methods of treating various central nervous system (CNS) diseases and other conditions are described herein, in which doses can be optimized for individual patients’ metabolism and treatment needs. In some embodiments, the carrier gas can be heated. The method can further comprise using a device containing a balloon with an oxygen-helium mixture equipped with a reducer and a mask connected to each other by a gas or air connecting tube, which contains an additional heating element capable of heating the gas mixture up to 120 °C, a nebulizer with a vibrating porous plate or mesh, ensuring the passage of droplets with a size of less than 5 microns through it, and a disinfection unit.

In some embodiments, the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof is delivered to the lower respiratory tract, for instance, to a pulmonary compartment such as alveoli, alveolar ducts and/or bronchioles. From there, the drug can enter the blood stream and travel to the central nervous system. In some embodiments, inhalation of a mist can deliver the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof to the patient’s CNS without passing through the liver. Administration via inhalation can allow gaseous drugs or those dispersed in a liquid or a mist, to rapidly reach the blood stream, bypassing first-pass metabolism. First-pass metabolism, also known as “first-pass effect” or “presystemic metabolism” describes drugs that enter the liver and undergo extensive biotransformation.

In some embodiments, the present disclosure provides a treatment step, in which the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof can be administered to a patient in need thereof by administering via inhalation a mixture of helium and oxygen heated to about 50°C, 51°C, 52°C, 53°C, 54°C, 55°C, 56°C, 57°C, 58°C, 59°C, 60°C, or more (or any range between 50°C to 60°C) and the atomized compound. In some embodiments, a mist or vapor of the compound can have a particle size from about 0.1 microns to about 10 microns (e.g., about 10, 5, 4, 3, 2, 1, 0.1 or less microns). In some embodiments, atomization is performed via a nebulizer creating an inhalant that is a mist. In some embodiments, the atomized compound is driven down the patient delivery line by the patient’ s inhalation. In some embodiments, the atomized compound is driven down the patient delivery line by the patient’s inhalation using a carrier gas. The carrier gas can be air, oxygen, a mix of oxygen and helium, heated air, heated oxygen, a heated helium and oxygen mixture, among others.

In some embodiments, the treatment step can be preceded by a pretreatment step. In some embodiments, the pretreatment step can comprise first administering a pretreatment inhalation therapy prior to administration of the mist of the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof. In some embodiments, the pretreatment inhalation step can comprise (i) administering via inhalation air, oxygen, or mixture of helium and oxygen heated to about 90°C, 91 °C, 92°C, 93°C, 94°C, 95°C, 96°C, 97°C, 98°C, 99°C, 100°C, 101°C, 102°C, 103°C, 104°C, 105°C, 106°C, 107°C, 108°C, 109°C, 110°C, 111°C, 112°C, 113°C, 114°C, 115°C, 116°C, 117°C, 118°C, 119°C, 120°C, or more (or any range between about 90°C and 120°C) and no drug, and then (ii) administering a treatment step of inhalation air, oxygen, a mix of oxygen and helium, heated air, heated oxygen, or heated helium and oxygen mixture, with the atomized compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof. Heated air, heated oxygen, or heated helium and oxygen mixture, in combination with the atomized compound of the present disclosure, can be heated to about 50°C, 51°C, 52°C, 53°C, 54°C, 55°C, 56°C, 57°C, 58°C, 59°C, 60°C, or more (or any range between about 50°C and 60°C).

In some embodiments of the present disclosure, a pretreatment step (i) and a treatment step (ii) can be repeated 0, 1, 2, 3, 4, 5, or more times. In some embodiments of the present disclosure, steps (i) and (ii) can be repeated 0, 1, 2, 3, 4, 5, or more times followed by the treatment step, which can be repeated 0, 1, 2, 3, 4, 5, or more times. In some embodiments of the present disclosure, the treatment step can be repeated 0, 1, 2, 3, 4, 5, or more times with no pretreatment step.

Treatment, with optional pretreatment, can be administered once a week, twice a week, once a day, twice a day, three times a day or more, a defined number of treatments per treatment course (e.g., 1, 2, 3, or 4 treatments per treatment course) or other treatment schedules as set forth herein.

A drug delivery procedure can comprise an inhaled priming no-drug hot heliox mixture to effectively preheat the mucosal bed followed by inhaling an atomized compound of the present disclosure, again driven by the heated heliox, but at lower temperatures, that are now dictated by lower heat tolerance to the wet vs. dry inhaled gas stream. Consequently, this procedure can be conducted in multiple repeated cycles, wherein a target PK and drug exposure is controlled by the concentration of the drug, temperature, flow rate of the helium oxygen mixture, composition of the mixture, number and durations of cycles, time and combinations of the above.

In some embodiments, the present disclosure provides methods of treating a central nervous system (CNS) disorder or psychological disorder comprising administering via inhalation a heated mixture of helium and oxygen heated and an atomized compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof. The treatment can alleviate one or more symptoms of the disorder. In some embodiments, the compound of the present disclosure can be administered for treatment of CNS disease or other disorder. In some embodiments, the compound of the present disclosure can be administered to treat depression including, but not limited to major depression, melancholic depression, atypical depression, or dysthymia. In some embodiments the compound of the present disclosure can be administered to treat psychological disorders including anxiety disorder, obsessive compulsive disorder, addiction (narcotic addiction, tobacco addiction, opioid addiction), alcoholism, depression and anxiety (chronic or related to diagnosis of a life-threatening or terminal illness), compulsive behavior, or a related symptom. In some embodiments, the disease or disorder is selected from the group consisting of central nervous system (CNS) disorders, including post-traumatic stress disorder (PTSD), major depressive disorder (MDD), treatment-resistant depression (TRD), suicidal ideation, suicidal behavior, major depressive disorder with suicidal ideation or suicidal behavior, non-suicidal self-injury disorder (NSSID), bipolar and related disorders including bipolar I disorder, bipolar II disorder, cyclothymic disorder, obsessive-compulsive disorder (OCD), generalized anxiety disorder (GAD), social anxiety disorder, substance use disorders including alcohol use disorder, opioid use disorder, amphetamine use disorder, nicotine use disorder, and cocaine use disorder, anorexia nervosa, bulimia nervosa, binge eating disorder, Alzheimer’s disease, cluster headache and migraine, attention deficit hyperactivity disorder (ADHD), pain and neuropathic pain, aphantasia, childhood-onset fluency disorder, major neurocognitive disorder, mild neurocognitive disorder, sexual dysfunction, chronic fatigue syndrome, Lyme disease, and obesity. In some embodiments, the disease or disorder may include conditions of the autonomic nervous system (ANS). In some embodiments, the disease or disorder may include pulmonary disorders (e.g., asthma and chronic obstructive pulmonary disorder (COPD). In some embodiments, the disease or disorder may include cardiovascular disorders (e.g., atherosclerosis).

The methods of delivering a compound of the present disclosure to the CNS (systemic drug delivery) via nebulizer (including, for example, using a heated helium-oxygen mixture), can lead to advantageous improvements in multiple PK parameters as compared to oral delivery. In particular, the compound can cross the blood brain barrier and be delivered to the brain. As compared to oral delivery, the method of delivering a compound of the present disclosure to the CNS via nebulizer, optionally with a heated heliox mixture, can increase bioavailability by at least about 10%, 25%, 30%, 35%, 40%, 50%, 55%, 60%, 65%, 70%, 80%, 85%, 90%, 95%, 99%, 99.9%, or more as compared to oral delivery. The method of delivering a compound of the present disclosure to the CNS via nebulizer as described herein, can reduce Tmaxby at least 30%, 40%, 50%, 55%, 60%, 65%, 70%, 80%, 85%, 90%, 95%, 99%, 99.9%, or more as compared to oral delivery. In some embodiments, the method of delivering the compound of the present disclosure to the CNS via nebulizer as described herein, can increase Cmax by at least 10%, 25%, 30%, 35%, 40%, 50%, 55%, 60%, 65%, 70%, 80%, 85%, 90%, 95%, 99%, 99.9%, or more as compared to oral delivery. Furthermore, a method of delivering a compound of the present disclosure to the CNS via nebulizer as described herein, can allow clinical protocols enabling dose titration and more controlled exposure. Controlled exposure enables adjusting the patient experience and providing overall improved therapeutic outcomes.

In some embodiments, a system is provided for administering a compound of the present disclosure (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof) that includes a container comprising a solution of the compound and a nebulizer physically coupled or co-packaged with the container and adapted to produce a mist of the solution having a particle size from about 0.1 microns to about 10 microns (e.g., about 10, 5, 4, 3, 2, 1, 0.1 or less microns).

Combination therapy with an NMDA receptor antagonist

Also disclosed are combination drug therapies based on administration of both a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, (as a 5-HT2A receptor agonist) and a A-methyl-D-aspartate (NMDA) receptor antagonist. The combination drug therapy may enhance activity and improve patient experience when treating diseases or disorders associated with 5-HT2A and/or NMDA receptors (e.g., a neuropsychiatric disease or disorder, a central nervous system (CNS) disorder, a psychological disorder, etc.), for example, by providing therapeutic efficacy while reducing or eliminating psychiatric adverse effects such as acute psychedelic crisis (bad trip) and dissociative effects from hallucinogens (out of body experience).

Non-limiting examples of NMDA receptor antagonists include, but are not limited to, ketamine, nitrous oxide, memantine, amantadine, dextromethorphan (DXM), phencyclidine (PCP), methoxetamine (MXE), dizocilpine (MK-801), esmethadone, or a combination thereof. In particular, nitrous oxide (N2O), commonly known as laughing gas, is a rapid and effective analgesic gas that has a fast onset and rarely produces side effects when administered under proper medical supervision. Nitrous oxide is also a dissociative inhalant known to cause euphoria during inhalation. Prominent effects of nitrous oxide are increased feelings of euphoria, a heightened pain threshold, and involuntary laughing. Furthermore, unlike ketamine, nitrous oxide is not addictive. For these reasons, the use of nitrous oxide as the NMDA receptor antagonist is preferred.

In some embodiments, the combination drug therapy involves providing the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, and the NMDA receptor antagonist as a single dosage form for administration to a patient (e.g., each is combined to provide a single aerosol that is inhaled by the patient; or each is combined into a single transdermal patch and delivered transdermally or subcutaneously to the patient). For example, when the NMDA receptor antagonist is nitrous oxide, the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, may be present in the liquid phase of the aerosol, while the nitrous oxide may be present in the gas phase of the aerosol. The nitrous oxide (or therapeutic gas mixture comprising nitrous oxide) may be used in the generation of the aerosol or as a carrier gas used to deliver a generated aerosol to the patient. When a generated aerosol is combined with a carrier gas, the carrier gas becomes a part of the gas phase of the aerosol, i.e., the liquid phase of the aerosol becomes entrained in/diluted by the carrier gas. In some embodiments, the combination drug therapy involves providing the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, and the NMDA receptor antagonist as separate dosage forms. For example, the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, may be provided as an aerosol, preferably a mist, while the NMDA receptor antagonist is provided separately as a therapeutic gas mixture. Alternatively, the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, may be provided as an injectable (e.g., intravenous, intradermal, etc.), bolus, infusion, perfusion, etc., while the NMDA receptor antagonist is provided as a therapeutic gas mixture for inhalation delivery.

The co-action of the compound of Formula (I) through (V) (as a 5-HT2A receptor agonist) and a NMDA receptor antagonist (e.g., nitrous oxide, ketamine, etc.) may provide multiple benefits. For example, the NMDA receptor antagonist may control and/or reduce the activating effects of the 5- HT2RS, thereby reducing the risk of overstimulation and occurrences of psychiatric adverse effects such as acute psychedelic crisis. Additionally, administration of the NMDA receptor antagonist may enable the use of a reduced therapeutic dose of the compound of Formula (I) through (V), thereby decreasing the likelihood of a negative patient experience or dose-dependent side effects. Similarly, administration of the compound of Formula (I) through (V) may reduce the amount of NMDA receptor antagonist necessary for a therapeutic effect, which in the case of NMDA receptor antagonists such as nitrous oxide may alleviate certain side effects such as induced involuntary laughter and the general feelings of anxiety associated therewith. Thus, it is believed that co-administration would reduce the likelihood of a negative experience from the psychedelic administration, either because less psychedelic would be administered or the NMDA receptor antagonist (e.g., nitrous oxide, ketamine, etc.) would enable more efficient functioning of the psychedelic. Similarly, such co-administration would reduce the time or amount of NMDA receptor antagonist (e.g., nitrous oxide, ketamine, etc.) necessary for a therapeutic effect.

NMDA receptor antagonists (e.g., nitrous oxide) and 5-HT2A receptor agonists function via different pharmacological pathways. However, both pathways appear to ultimately converge in a cascade at mTOR (mammalian target of rapamycin, or mechanistic target of rapamycin). Thus, a shared mechanism of action appears to exist between NMDA receptor antagonists and 5-HT2A receptor agonists. Specifically, mTOR’s signaling pathway may be modulated by 5-HT2A receptor activation and NMDA antagonism. Without being bound by theory, such modulation of the mTOR pathway may underpin the immediate and long-lasting therapeutic and synergistic benefits of combined administration of both agents. As such, in some embodiments, administration of both agents at psychedelic or sub-psychedelic doses enables therapeutic efficacy without or minimizing psychiatric adverse effects. In addition, it has been found that atrophy of neurons in the prefrontal cortex (PFC) plays a key role in the pathophysiology of depression and related disorders. The ability to promote both structural and functional plasticity in the PFC has been hypothesized to underlie the fast-acting antidepressant properties of the dissociative anesthetic ketamine but also the long-lasting effect after a single administration. Without being bound by theory, it is believed that the combination drug therapy disclosed herein may function by synergistically increasing neuritogenesis and spinogenesis, including increased density of dendritic spines, thereby providing or contributing to long-lasting therapeutic benefits.

A ratio of the compound of Formula (I) through (V) and the NMDA receptor antagonist administered in the combination drug therapy may vary depending on the patient (i.e., subject), the identity of the active ingredient(s) selections of the combination, the dosage form(s), and the specific disease or condition being treated. It should be understood that a specific ratio of the combination for any particular patient will depend upon a variety of factors, such as the activity of the specific compounds employed, the age, sex, general health of the patient, time of administration, rate of excretion, and the severity of the particular disease or condition being treated. In some embodiments, a weight ratio of the compound of Formula (I) through (V) and the NMDA receptor antagonist administered to the patient may range from about 1:100 to about 100:1, or any range therebetween, e.g., from about 1:75, from about 1:50, from about 1:40, from about 1:30, from about 1:20, from about 1:10, from about 1:8, from about 1:6, from about 1:5, from about 1:4, from about 1:3, from about 1:2, from about 2:3, from about 1:1, and up to about 100:1, up to about 75:1, up to about 50:1, up to about 40:1, up to about 30:1, up to about 20:1, up to about 10:1, up to about 8:1, up to about 6:1, up to about 5:1, up to about 4:1, up to about 3:1, up to about 2:1. Ratios outside of this range may also be employed, in certain circumstances.

The combination drug therapy is intended to embrace administration of the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof and the NMDA receptor antagonist (e.g., nitrous oxide) in a sequential manner, that is, wherein each active ingredient is administered at a different time, as well as administration of these active ingredients, or at least two of the active ingredients, in a concurrent manner. Concurrent administration can be accomplished, for example, by administering to the subject a single dosage form having a fixed ratio of each active ingredient or in multiple, single dosage forms for each of the active ingredients. Administration of the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof and a NMDA receptor antagonist (e.g., nitrous oxide), whether in a single dosage form or separate dosage forms, can be carried out by any administration route set forth herein. In some embodiments, both the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof and the NMDA receptor antagonist are administered via inhalation, preferably in aerosol (e.g., mist) form. In some embodiments, the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof is administered intravenously (IV), and the NMDA receptor antagonist is administered via inhalation. In some embodiments, the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof is administered orally, and the NMDA receptor antagonist is administered via inhalation. In some embodiments, both the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof and the NMDA receptor antagonist are administered transdermally or subcutaneously. The compositions for inhalation such as pharmaceutically acceptable excipients, etc. for the single or separate dosage forms are set forth herein.

In some embodiments, the NMDA receptor antagonist used in the combination drug therapy is nitrous oxide. Nitrous oxide may be administered alone, or as a therapeutic gas mixture, e.g., N2O and O2; N2O and air; N2O and medical air (medical air being 78% nitrogen, 21% oxygen, 1% other gases); N2O and a N2/O2 mix; N2O and O2 enriched medical air; N2O and a He/Ch mix etc. Thus, in addition to nitrous oxide and oxygen, the therapeutic gas mixture may further include other gases such as one or more of N2, Ar, CO2, Ne, CH4, He, Kr, H2, Xe, H2O (e.g., vapor), etc. For example, nitrous oxide may be administered using a blending system that combines N2O, O2 and optionally other gases from separate compressed gas cylinders into a therapeutic gas mixture which is delivered to a patient via inhalation. Alternatively, the therapeutic gas mixture containing nitrous oxide may be packaged, for example, in a pressurized tank or in small, pressurized canisters which are easy to use and/or portable. The blending system and/or pressurized tanks/canisters may be adapted to fluidly connect to an inhalation device such as a device capable of generating an aerosol of the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof. Nitrous oxide itself, or the therapeutic gas mixture comprising nitrous oxide may be used for the generation of the aerosol (i.e., as the gas phase component of the aerosol) or as a carrier gas to facilitate the transfer of a generated aerosol to a patient’s lungs. In some embodiments, N2O is present in the therapeutic gas mixture at a concentration ranging from 5 vol%, from 10 vol%, from 15 vol%, from 20 vol%, from 25 vol%, from 30 vol%, from 35 vol%, from 40 vol%, from 45 vol%, and up to 75 vol%, up to 70 vol%, up to 65 vol%, up to 60 vol%, up to 55 vol%, up to 50 vol%, relative to a total volume of the therapeutic gas mixture. The therapeutic gas mixture containing nitrous oxide can be administered over any desired duration, e.g., 5 minutes, 10 minutes 15 minutes, 20 minutes, 30 minutes, 40 minutes, 45 minutes, 50 minutes, 60 minutes, 90 minutes, 120 minutes, 150 minutes, 180 minutes, or any range therebetween. In some embodiments, the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof and the NMDA receptor antagonist (e.g., nitrous oxide) are each delivered by aerosol inhalation, as a single dosage form or as separate dosage forms. The aerosol, preferably a mist, may be generated by any capable device (e.g., a pressurized container, pump, spray, atomizer, or nebulizer), such as those devices disclosed herein, with or without the use of a propellant. When nitrous oxide is administered concurrently with the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, the nitrous oxide may dually act as a carrier gas or propellant for the aerosol generation and as a therapeutic agent (an NMDA receptor antagonist).

In some embodiments, the delivery device is an inhalation delivery device for delivery of the combination of the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof and nitrous oxide by inhalation to a patient in need thereof, comprising an inhalation outlet portal for administration of the combination to the patient; a container configured to deliver nitrous oxide, e.g., in a therapeutic gas mixture, to the inhalation outlet portal; and a device configured to generate and deliver an aerosol comprising the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof to the inhalation outlet portal. In some embodiments, the inhalation outlet portal is selected from a mouthpiece or a mask covering the patient’s nose and mouth. In some embodiments, the device configured to generate and deliver the aerosol to the inhalation outlet portal is a nebulizer. In some embodiments, the nebulizer is a jet nebulizer and the nitrous oxide gas, alone, or in combination with other gases (therapeutic gas mixture containing nitrous oxide), acts as a driving gas for the jet nebulizer. Accordingly, nitrous oxide delivered using a nebulizer (e.g., jet nebulizer) may dually act as a therapeutic agent and as a driving gas to entrain the nebulized form of the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof.

In some embodiments, the device is a dual delivery device configured to administer the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, preferably in the form of an aerosol, and to simultaneously administer a controlled amount of nitrous oxide, either alone or as a therapeutic gas mixture. Any of the above aerosol delivery devices can be used for such a device, with the addition of a source of nitrous oxide (or a source of a therapeutic gas mixture containing nitrous oxide) configured to provide a metered, controlled dose/flow rate of nitrous oxide through the same administration outlet as the aerosol delivery device. In some embodiments, the driving gas for the nebulization of the compound of Formula (I) through (V) or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof is the nitrous oxide or therapeutic gas mixture containing nitrous oxide. Any of the delivery devices above, e.g., controlled release device, implant, patch, pump, depot, inhaler, inhalation delivery device, etc., can be optionally manufactured with smart technology, e.g., electronics, configured to provide remote activation and operational control of the drug delivery. The remote activation can be performed via computer or mobile app. To ensure security, the remote activation device can be password encoded. This technology enables a healthcare provider to perform telehealth sessions with a patient, during which the healthcare provider can remotely activate and administer the compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof, via the desired delivery device while supervising the patient on the televisit.

Kits

In some embodiments, a kit is provided for the treatment of a subject comprising 1) a pharmaceutical composition disclosed herein comprising any of the compounds described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof), such as an oral dosage form (e.g., a pill or single-layered orally administered tablet), a parenteral dosage form (e.g., an injectable), a topical/transdermal dosage form, a modified release dosage form, or a dosage form to be delivered via inhalation, and (2) instructions for use in the treatment, prevention or management of a disease, disorder or condition described herein.

In some embodiments, the disease, disorder or condition is pain, examples of which include, but are not limited to, cancer pain, e.g., refractory cancer pain, post-surgical pain, orthopedic pain, back pain, neuropathic pain, dental pain, chronic pain, and chronic pain in opioid-tolerant patients. In some embodiments, the disease, disorder or condition is associated with a serotonin 5-HT2 receptor. In some embodiments, the disease, disorder or condition is selected from the group consisting of central nervous system (CNS) disorders, including post-traumatic stress disorder (PTSD), major depressive disorder (MDD), treatment-resistant depression (TRD), suicidal ideation, suicidal behavior, major depressive disorder with suicidal ideation or suicidal behavior, non-suicidal self-injury disorder (NSSID), bipolar and related disorders including bipolar I disorder, bipolar II disorder, cyclothymic disorder, obsessive- compulsive disorder (OCD), generalized anxiety disorder (GAD), social anxiety disorder, substance use disorders including alcohol use disorder, opioid use disorder, amphetamine use disorder, nicotine use disorder, and cocaine use disorder, anorexia nervosa, bulimia nervosa, binge eating disorder, Alzheimer’s disease, cluster headache and migraine, attention deficit hyperactivity disorder (ADHD), pain and neuropathic pain, aphantasia, childhood-onset fluency disorder, major neurocognitive disorder, mild neurocognitive disorder, sexual dysfunction, chronic fatigue syndrome, Lyme disease, and obesity. In some embodiments, the disease, disorder or condition includes conditions of the autonomic nervous system (ANS). In some embodiments, the disease, disorder or condition includes pulmonary disorders including asthma and chronic obstructive pulmonary disorder (COPD). In some embodiments, the disease, disorder or condition includes cardiovascular disorders including atherosclerosis. In some embodiments, the disease, disorder or condition is brain injury. In some embodiments, the disease, disorder or condition is depression. In some embodiments, the disease, disorder or condition is migraine, e.g., with aura. In some embodiments, the disease, disorder or condition is refractory asthma. In some embodiments, the disease, disorder or condition is stroke. In some embodiments, the disease, disorder or condition is alcohol dependence.

In some embodiments, the instructions for use form an integrated component of the packaging for the pharmaceutical composition, e.g., single-layered orally administered tablet composition.

In some embodiments, the disclosure features an oral, modified-release pharmaceutical composition for oral administration to a subject for treating the subject diagnosed with, suffering from or susceptible to a disease, disorder or condition, such as those for which phenethylamine treatment may be indicated, considered or recommended, wherein the subject is in need of treatment with said oral, modified-release pharmaceutical composition, said oral, modified-release pharmaceutical composition comprising:

(a) a compound described herein (e.g., a compound of Formula (I) through (V), or a pharmaceutically acceptable salt, solvate, stereoisomer, or prodrug thereof) in an effective amount for treating, preventing and/or managing the disease, disorder, or condition in the subject; and

(b) a pharmaceutically acceptable excipient; whereby, upon oral administration of the modified-release pharmaceutical composition to the subject, a steady release of said compound from the modified-release pharmaceutical composition is maintained so that no neurologically toxic spike in the subject's plasma occurs during the release period of said drug from said pharmaceutical composition.

Compliance with Monographs

In some embodiments, the pharmaceutical compositions of the disclosure conform to certain industry accepted monographs to afford compliance with the Federal Food Drug and Cosmetic Act. In particular, the pharmaceutical compositions of the disclosure conform and are considered acceptable under visual inspection, uniformity of mass analysis, uniformity of content analysis, and/or dissolution/disintegration analysis all of which are established by a relevant monograph.

In some embodiments, throughout manufacturing certain procedures are validated and monitored by carrying out appropriate in-process controls. These are designed to guarantee the effectiveness of each stage of production. In-process controls during tablet production may include the moisture content of the final lubricated blend, the size of granules, the flow of the final mixture and, where relevant, the uniformity of mass of tablet cores before coating. In-process controls during tablet production may also include the dimensions (thickness, diameter), uniformity of mass, hardness and/or crushing force, friability, disintegration or dissolution rate (for example, for modified-release tablets) of the finished dosage form. Suitable test methods that may be used to demonstrate certain of these attributes arc known in the art.

In some embodiments, packaging maybe or is required to be adequate to protect the pharmaceutical compositions, including tablets, from light, moisture and damage during transportation.

In some embodiments, the commercially available formulation (e.g., kit) complies with the labeling requirements established under Good Manufacturing Practices (GMP). Such label includes:

(1) the name of the pharmaceutical product;

(2) the name(s) of the active ingredient(s); International Nonproprietary Names (INN) should be used wherever possible;

(3) the amount of the active ingredient(s) in each tablet and the number of tablets in the container;

(4) the batch (lot) number assigned by the manufacturer;

(5) the expiry date and, when required, the date of manufacture;

(6) any special storage conditions or handling precautions that may be necessary;

(7) directions for use, warnings, and precautions that may be necessary;

(8) the name and address of the manufacturer or the person responsible for placing the product on the market;

(9) for scored tablets where the directions for use include subdivision to provide doses of less than one tablet, the label should also include: the storage conditions for and the period of use of those subdivided part(s) not immediately taken or administered.

In some embodiments, the pharmaceutical compositions, e.g., tablets, can withstand handling, including packaging and transportation, without losing their integrity.

EXAMPLES

I. Synthetic routes

Compounds of the present disclosure and Reference Compounds may generally be prepared according to, or analogous to, the following synthetic procedures as depicted in Figs. 1-22.

Example 1

2-(benzo [d] [ 1 ,3 ]dioxol-5 -yl-2,2-<72)ethan- 1 -amine (II- 11)

Synthesis of 2-(benzo[d][l,3]dioxol-5-yl-2,2-r/2)ethan-l-amine (11-11) was carried out according to Fig. 1. To a solution of 3,4-dihydroxybenzaldehyde (II-lla) (2.5 g, 18.1 mmol) in THF (10 mL, 4 V) was added D2O (10 mL, 4V) and the reaction mass was stirred overnight at room temperature under nitrogen atmosphere. The solvent was removed and dried under vacuum to give II- 11b (2.5 g, 98.56%); J H NMR (400 MHz, DMSO-rfo) 5 9.703 (s, 1H), 7.28-7.23 (m, 2H), 6.92 (d, J=8Hz, 1H).

To a suspension of K2CO3 (0.394 g, 2.85 mmol) in N-methylpyrrolidinone (3.6 mL, 18 V) at 110°C under nitrogen atmosphere, a solution of II-llb (0.2 g, 1.43 mmol) in CD2CI2 (0.9 mL, 1.1 mmol) and NMP (0.4 mL, 2V) was added dropwise over 45 minutes. The reaction mixture was stirred at 110 °C for an additional 90 min. The mixture was allowed to cool and filtered. The filtrate was poured into water (30 mL) and extracted with dichloromethane (3 x 40 mL). The combined organic layer was washed with water (2 x 30 mL), dried over sodium sulfate, and concentrated under vacuum. The residue was purified by silica-gel chromatography (4:1 Hexane / EtOAc) to provide II-llc (0.2 g, 92.11 %) as a pale brown oil that solidified on standing; LCMS: [M+H]+ =153.15.

A suspension of II-llc (0.15 g, 0.98 mmol), nitromethane (0.18, 2.9 mmol) and ammonium acetate (0.189 g, 2.46 mmol) in acetic acid (1.5 mL, 10 V) was refluxed for 4 h. After 4 h, reaction mass was cooled to room temperature and poured into ice-water (50mL) and extracted with dichloromethane (DCM) (3 x 30 ml). The combined organic layer was evaporated under vacuum to give II-lld (0.1 g, 52%); ' H NMR (400 MHz, DMSO-Je): 8.16-8.05 (m, 2H), 7.55 (s, 1H), 7.41-7.39 (d, J=8Hz, 1H), 7.05- 7.03 (d, J=8Hz, 1H).

A solution of II-lld (0.1 g, 0.512 mmol) in THF (2 ml, 20 V) was added dropwise to stirred suspension of LiAlH4 (IM in THF, 2 mL, 2.04 mmol) in Et20 (1 ml, 10 V) and the reaction mass was refluxed for 2 h. After 2 h, the reaction mass was cooled on ice bath and quenched with dropwise addition of water (5 mL), followed by the addition of a 15% aqueous NaOH solution (5 mL). The reaction mass was allowed to stir for 30 min, filtered and organic was removed under vacuum. The aqueous residue was extracted with DCM (3 x 20 mL). The combined organic layer was dried over sodium sulfate and concentrated in vacuum to give free base of 2-(benzo[d][l,3]dioxol-5-yl-2,2- d2)ethan-l -amine (11-11). To this, DCM (1 mL) was added and cooled to 0° C, and 2M HC1 in Et20 (0.09 ml, 0.179 mmol) was added and the reaction mass was stirred at same temperature for 30 minutes. After 30 min, the reaction mass was concentrated and dried under vacuum to provide 11-11, as HC1 salt (20 mg, 20%); LCMS: [M+H]+ =168; ' H NMR (400 MHz, DMSO-r/s): 7.98 (bs, 2H), 6.88-6.86 (m, 2H), 6.72-6.71 (d, H=8Hz, 1H), 2.88 (s, 2H), 2.80 (m, 2H).

Example 2 2-(benzo[d][l,3]oxathiol-6-yl)ethan-l-amine (11-14) Synthesis of 2-(benzo[d][1,3]oxathiol-6-yl)ethan-1-amine (II-14) was carried out according to Fig.2. To a solution of potassium hydroxide (2.20 g, 39.28 mmol) in water (10 mL, 10 V), vanillin (II- 14a) (3 g, 19.73 mmol) was added and the reaction mass was cooled on ice-salt bath. To this, N,N- dimethylthiocarbamoylchloride (2.52 g, 20.39 mmol) in tetrahydrofuran (12.5 mL, 5V) was added dropwise and the reaction mass was stirred at room temperature for 3 h. After 3h, the white precipitate formed was filtered and dried under vacuum to give II-14b (3.3g, 70%); LCMS: [M+H]+ = 239.29. A suspension of II-14b (1 g, 4.2 mmol) in diphenyl ether (10 ml, 10 V) was heated under reflux under nitrogen atmosphere for 1 h. After 1 h, reaction mass was cooled and the reaction mass was directly loaded in glass column and product was eluted in 70% ethyl acetate in hexane to give II-14c as off white solid (0.8 g, 80%); 1 H NMR (400 MHz, DMSO-d 6 $ k +*(*,- #\& +=$& 1(01'1(0/ #N& ?52=c& 1H), 7.55-7.52 (m, 2H), 3.83 (s, 3H), 3.07 (s, 3H), 2.92 (s, 3H). To a cooled solution of II-14c (1.5 g, 6.27 mmol) was dissolved in dichloromethane (15 ml, 10 V) at - 40 o C, BBr3 (1M in DCM, 15 mL, 15.7 mmol) was added dropwise under nitrogen atmosphere and the reaction mass was stirred at room temperature for 1 h. After completion of reaction, reaction was quenched by sat. solution of NaHCO3 (30 mL) and extracted with dichloromethane (3 x 50 mL). The combined organic layer was dried over anhydrous sodium sulfate and concentrated under vacuum to obtain II-14d (0.8g, 56%); LCMS: [M+H]+ = 225.26. To a solution of II-14d (0.8 g, 3.55 mmol) in methanol (16 mL, 20 V), a solution of potassium hydroxide (0.9 g, 17 mmol) in water (8 mL, 10 V) was added dropwise and the reaction mass was refluxed for 1.5 h. After completion of reaction, reaction mass was acidified with acetic acid (20 mL) and the yellow precipitate thus formed was filtered and discarded. The acidic layer was evaporated in vacuum to get crude II-14e (0.45 g, 82%), which was used as such in next step; LCMS: [M+H]+ = 153.0. To a suspension of K 2 CO 3 (0.425 g, 3 mmol) in N-methylpyrrolidinone (4 mL, 10 V) at 110°C under nitrogen atmosphere, a solution of II-14e (0.35 g, 1.5 mmol) in CH2Cl2 (1.2 mL, 4 V) and NMP (0.6 mL, 2V) were added dropwise over 45 minutes. The reaction mixture was stirred at 110 °C for an additional 90 min. The mixture was allowed to cool and filtered. The filtrate was poured into water (30 mL) and extracted with dichloromethane (3 x 40 mL). The combined organic layer was washed with water (2 x 30 mL), dried over sodium sulfate and concentrated under vacuum. The residue was purified by silica-gel chromatography (4:1 Hexane / EtOAc) to provide II-14f (0.35 g, 92%) as a pale brown oil that solidified on standing; 1 H NMR (400 MHz, DMSO-d6): 8.85 (s, 1H), 7.55-7.49 (m, 2H), 7.27 (s, 1H), 5.88 (s, 2H). A suspension of II-14f (0.15 g, 0.9 mmol), nitromethane (0.165, 2.7 mmol) and ammonium acetate (0.173 g, 2.2 mmol) in acetic acid (1.5 mL, 10 V) was refluxed for 4 h. After 4 h, reaction mass was cooled to room temperature and poured into ice-water (50mL) and the precipitated solid was filtered and dried to obtain II-14g (0.1 g, 53%); 1 H NMR (400 MHz, DMSO-d6): 8.22-8.19 (d, J=13.4 Hz, 1H), 8.07-8.04 (d, J=13.4 Hz, 1H), 7.54 (s, 1H), 7.42-7.38(m, 2H), 5.85 (s, 2H). A solution of II-14g (0.1 g, 0.477 mmol) in THF (2 ml, 20 V ) was added dropwise to stirred suspension of LiAlH4 (1M in THF, 1.9 mL, 1.9 mmol) in Et2O (1 ml, 10 V) and the reaction mass was refluxed for 2 h. After 2 h, the reaction mass was cooled on ice bath and quenched with dropwise addition of water (5 mL), followed by the addition of a 15% aqueous NaOH solution (5 mL). The reaction mass was allowed to stir for 30 min, filtered and organic was removed under vacuum. The aqueous residue was extracted with DCM (3 x 20 mL). The combined organic layer was dried over sodium sulfate and concentrated in vacuum to give free base of II-14. To this, DCM (1 mL) was added and cooled to 0 o C, and 2M HCl in Et 2 O (0.08 ml, 0.165 mmol) was added and the reaction mass was stirred at same temperature for 30 minutes. After 30 min, the reaction mass was concentrated and dried under vacuum to give II-14, as HCl salt (20 mg, 20%); LCMS: [M+H]+ = 182; 1 H NMR (400 MHz, DMSO-d6): 7.88 (bs, 2H), 7.26 (m, 1H), 6.84-6.80 (m, 2H), 5.76 (s, 2H), 3.00 (s, 2H), 2.79 (m, 2H). Example 3 2-(benzo[d][1,3]oxathiol-6-yl-2,2-d2)ethan-1-amine (II-15) Synthesis of 2-(benzo[d][1,3]oxathiol-6-yl-2,2-d2)ethan-1-amine (II-15) was carried out according to Fig. 3. To a suspension of K2CO3 (2.6 g, 19 mmol) in N-methylpyrrolidinone (26 mL, 10 V) at 110°C under nitrogen atmosphere, a solution of II-14e (1.5 g, 9.72 mmol) in CD 2 Cl 2 (6 mL, 4V) and NMP (3 mL, 2V) were added dropwise over 45 minutes. The reaction mixture was stirred at 110 °C for an additional 90 min. The mixture was allowed to cool and filtered. The filtrate was poured into water (100 mL) and extracted with dichloromethane (3 x 100 mL). The combined organic layer was washed with water (2 x 50 mL), dried over sodium sulfate and concentrated under vacuum. The residue was purified by silica-gel chromatography (30% EtOAc in hexane) to provide II-15a (0.6 g, 36%); LCMS: [M+H]+ =168.98. A suspension of II-15a (0.35 g, 2.08 mmol), nitromethane (0.38, 6.2 mmol) and ammonium acetate (0.4 g, 5.1 mmol) in acetic acid (3.5 mL, 10 V) was refluxed for 4 h. After 4 h, the reaction mass was cooled to room temperature and poured into ice-water (50mL) and the solid precipitate was filtered and dried to obtain II-15b as yellow solid (0.15g, 34%); 1 H NMR (400 MHz, DMSO-d6): 8.23 (m, 1H), 8.09 (m, 1H), 7.45-7.43 (m, 3H). A solution of II-15b as yellow solid (0.15 g, 0.71 mmol) in THF (3 ml, 20 V ) was added dropwise to a stirred suspension of LiAlH4 (1M in THF, 3 mL, 3.00 mmol) in Et2O (1.5 ml, 10 V) and the reaction mass was refluxed for 2 h. After 2 h, the reaction mass was cooled in an ice bath and quenched with dropwise addition of water (5 mL), followed by the addition of a 15% aqueous NaOH solution (5 mL). The reaction mass was allowed to stir for 30 min, filtered, and the organic was removed under vacuum. The aqueous residue was extracted with DCM (3 x 20 mL). The combined organic layer was dried over sodium sulfate and concentrated in vacuum to give free base of II-15. To this, DCM (1 mL) was and cooled to 0 o C, and 2M HCl in Et2O (0.1 mL, 0.22 mmol) was added and the reaction mass was stirred at same temperature for 30 minutes. After 30 min, the reaction mass was concentrated and dried under vacuum to give II-15, as HCl salt (30 mg, 23%); LCMS: [M+H]+ =184.1; 1 H NMR (400 MHz, DMSO-d 6 ): 7.89 (bs, 2H), 7.25-7.23 (d, J=8Hz 1H), 6.83-6.79 (m, 3H), 2.99 (s, 2H), 2.80 (m, 2H). Example 4 1-(benzo[d][1,3]oxathiol-6-yl)propan-2-amine (II-16) Synthesis of 1-(benzo[d][1,3]oxathiol-6-yl)propan-2-amine (II-16) was carried out according to Fig.4. A suspension of II-14f (0.15 g, 0.9 mmol), nitroethane (0.2 g, 2.7 mmol) and ammonium acetate (0.2 g, 2.2 mmol) in acetic acid (1.5 mL, 10 V) was refluxed for 4 h. After 4 h, the reaction mass was cooled to room temperature and poured into ice-water (30 mL) and the resulting precipitate was filtered and dried to obtain II-16a (0.11 g, 54.59%); LCMS: [M-NO2] = 178. A solution of II-16a (0.11 g, 0.49 mmol) in THF (2.2 ml, 20 V) was added dropwise to a stirred suspension of LiAlH4 (1M in THF, 2 mL, 1.96 mmol) in Et2O (1.1 ml, 10 V) and the reaction mass was refluxed for 2 h. After 2 h, the reaction mass was cooled on ice bath and quenched with dropwise addition of water (5 mL), followed by the addition of a 15% aqueous NaOH solution (5 mL). The reaction mass was allowed to stir for 30 min, filtered, and the organic was removed under vacuum. The aqueous residue was extracted with DCM (3 x 20 mL). The combined organic layer was dried over sodium sulfate and concentrated in vacuum to give free base of II-16. To this, DCM (1 mL) was added and cooled to 0 o C, and 2M HCl in Et2O (0.8 mL, 0.153 mmol) was added and the reaction mass was stirred at same temperature for 30 minutes. After 30 min, the reaction mass was concentrated and dried under vacuum to give II-16, as HCl salt (21 mg, 18.39%); LCMS: [M+H]+ =196.09; 1 H NMR (400 MHz, MeOD-d4): 7.19-7.17 (d, J=8Hz, 1H), 6.81-6.77 (m, 2H), 5.72 (s, 2H), 3.51 (m, 1H), 2.91 (m, 1H), 2.78 (m, 1H), 1.28-1.26 (d, J=6.8Hz, 3H). Example 5 1-(benzo[d][1,3]oxathiol-6-yl-2,2-d2)propan-2-amine (II-17) The synthesis of 1-(benzo[d][1,3]oxathiol-6-yl-2,2-d2)propan-2-amine (II-17) was carried out according to Fig. 5. A suspension of II-15a (0.35 g, 2.08 mmol), nitroethane (0.47g, 6.2 mmol) and ammonium acetate (0.4 g, 5.1 mmol) in acetic acid (3.5 mL, 10 V) was refluxed for 4 h. After 4 h, the reaction mass was cooled to room temperature and poured into ice-water (50 mL) and the solid precipitate was filtered and dried to obtain II-17a as yellow solid (0.14 g, 30%); 1 H NMR (400 MHz, DMSO-d6): 8.03 (s, 1H), 7.46-7.44 (d, J=8Hz, 1H), 7.19 (m, 2H), 2.41 (s, 3H). A solution of II-17a (0.14 g, 0.62 mmol) in THF (3 ml, 20 V) was added dropwise to a stirred suspension of LiAlH4 (1M in THF, 2.5 mL, 2.4 mmol) in Et2O (1.4 mL, 10 V) and the reaction mass was reflux for 2 h. After 2 h, the reaction mass was cooled in an ice bath and quenched with dropwise addition of water (5 mL), followed by the addition of a 15% aqueous NaOH solution (5 mL). The reaction mass was allowed to stir for 30 min, filtered, and the organics removed under vacuum. The aqueous residue was extracted with DCM (3 x 20 mL). The combined organic layer was dried over sodium sulfate and concentrated in vacuum to give free base of II-17. To this, DCM (1 mL) was added and cooled to 0 o C, and 2M HCl in Et2O (0.1 mL, 0.2 mmol) was added and the reaction mass was stirred at same temperature for 30 minutes. After 30 min, the reaction mass was concentrated and dried under vacuum to give II-17, as HCl salt (25 mg, 17 %); LCMS: [M+H] + =198.1; 1 H NMR (400 MHz, MeOD- d4): 7.14-7.12 (d, J=8Hz, 1H), 6.75-6.71 (m, 2H), 3.44 (m, 1H), 2.85 (m, 1H), 2.73 (m, 1H), 1.23-1.21 (d, J=6.8Hz, 3H). Example 6 1-(5-methoxybenzo[d][1,3]oxathiol-6-yl)propan-2-amine (II-20) The synthesis of 1-(5-methoxybenzo[d][1,3]oxathiol-6-yl)propan-2-amine (II-20) was carried out according to Fig.6. To a solution of commercially available 4-methoxyphenol (II-20a) (5.0 g, 40.3 mmol, 1 eq) and tributylamine (10.6 mL, 44.3 mmol, 1.1 eq) in anhydrous DCM (160 mL) at 0 °C under N 2 was added chlorocarbonylsulfenyl chloride (3.7 mL, 44.3 mmol, 1.1 eq). After warming to room temperature and stirring 2 h, the reaction mixture was cooled 0 °C before adding AlCl 3 (12.9 g, 96.7 mmol, 2.4 eq) portion wise over 30 min. After stirring overnight at room temperature, the reaction mixture was carefully poured into ice water (200 mL) and extracted with DCM (3 x 200 mL). The combined organic phase was washed with brine (50 mL), dried over Na2SO4, and concentrated. The crude material was purified on silica (Hex to 30% EtOAc/Hex) to afford II-20b (3.83 g, 52% yield) as a white solid; the structure was confirmed by 1 H NMR in CDCl3. To a pressure vessel was added II-20b (1.17 g, 6.41 mmol, 1 eq), tetrabutylammonium bromide (207 mg, 0.641 mmol, 0.1 eq), aqueous NaOH (5M, 6.4 mL, 32.06 mmol, 5 eq), and dibromomethane (4.5 mL, 64.12 mmol, 10 eq). The vessel was sealed and heated at 100 °C overnight. The reaction mixture was poured into water (50 mL) and extracted with DCM (3 x 50 mL). The combined organic phase was washed with brine (20 mL), dried over Na 2 SO 4 , and concentrated. The crude material was purified on silica (Hex to 10% EtOAc/Hex) to afford II-20c (0.78 g, 72% yield) as a colorless oil; the structure was confirmed by 1 H NMR in CDCl3, with minor impurities present. To POCl3 (2.60 mL, 27.86 mmol, 4.3 eq) was slowly added N-methylformanilide (2.96 mL, 23.98 mmol, 3.7 eq) at room temperature under N2. After 30 min, II-20c (1.09 g, 6.48 mmol, 1 eq) was slowly added and reaction heated to 70 °C. After 2 h, the reaction mixture was carefully poured into ice water (50 mL) and extracted with DCM (3 x 50 mL). The combined organic phase was washed with brine (20 mL), dried over Na 2 SO 4 , and concentrated. The crude material was purified on silica (Hex to 20% EtOAc/Hex). The impurity in the combined fractions was removed by trituration with EtOAc/Hex to afford II-20d (0.99 g, 78% yield) as a yellow solid; the structure was confirmed by 1 H NMR in CDCl 3 . To II-20d (0.99 g, 5.02 mmol, 1 eq) was added anhydrous ammonium acetate (387 mg, 5.02 mmol, 1 eq), nitroethane (1.8 mL, 25.12 mmol, 5 eq), acetic acid (glacial, 4.0 mL, 70.28 mmol, 14 eq) and activated 3Å molecular sieves (2.5 g wet weight) under N2 at room temperature. The reaction mixture was heated at 95 °C for 1 h. The molecular sieves were removed, reaction mixture poured into brine (40 mL) and extracted with EtOAc (3 x 40 mL). The combined organic phase was washed with water (20 mL), then brine (20 mL), dried over Na2SO4, and concentrated to afford II-20e (1.13 g, 89% yield) as an orange solid; the structure was confirmed by 1 H NMR in CDCl3. The crude material was used in the next step without further purification. To a solution of II-20e (1.17 g, 4.62 mmol, 1 eq) in MeOH (17 mL), water (7 mL), and concentrated HCl (12.1 M, 7 mL) was added iron (powder, reduced) (1.29 g, 23.10 mmol, 5 eq) portionwise over 10 min at room temperature under N 2 . The reaction was heated at 70 °C for 2 h before adding additional concentrated HCl (12.1 M, 7 mL) and iron (powder, reduced) (1.29 g, 23.10 mmol, 5 eq). After heating for 2 h, the reaction mixture was poured into water (50 mL) and extracted with DCM (3 x 50 mL). The combined organic phase was washed with brine (30 mL), dried over Na 2 SO 4 , and concentrated. The crude material was purified on silica (Hex to 30% EtOAc/Hex) to afford II-20f (414 mg, 40% yield) as a white solid; the structure was confirmed by 1 H NMR in CDCl3. To a solution of II-20f (345 mg, 1.54 mmol, 1 eq) and titanium(IV) ethoxide (0.65 mL, 3.08 mmol, 2 eq) in anhydrous THF (8 mL) was added (R/S)-2-methyl-2-propanesulfinamide (373 mg, 3.08 mmol, 2 eq) in anhydrous THF (3 mL) at room temperature under N2. The reaction was heated at 70 °C for 4 h before cooling to -45 °C and slowly transferring into a solution of NaBH4 (233 mg, 6.15 mmol, 4 eq) in anhydrous THF (8 mL) at -45 °C under N2. After 45 min, the reaction was carefully quenched with MeOH (5 mL) and allowed to warm to room temperature. Saturated aqueous NH4Cl (10 mL) was added and mixture filtered through Celite with EtOAc. The filtrate was poured into water (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic phase was washed with brine (10 mL), dried over Na 2 SO 4 , and concentrated. The crude material was purified on silica (Hex to EtOAc) to afford II- 20g (279 mg, 55% yield) as a colorless oil; the structure was confirmed by 1 H NMR in CDCl3. To a solution of II-20g (390 mg, 1.18 mmol, 1 eq) in a mixture of anhydrous MeOH (2.1 mL) and anhydrous Et2O (21 mL) was added anhydrous HCl (2M in Et2O, 2.4 mL, 4.73 mmol, 4 eq) dropwise at room temperature under N2. After stirring overnight, the reaction mixture was concentrated, solid washed with Et2O, and dried to afford II-20 (314 mg, 100% yield) as a white solid; the structure was confirmed by 1 H NMR in CD3CN. Example 7 1-(5-methoxybenzo[d][1,3]oxathiol-6-yl-2,2-d2)propan-2-amine (II-21) The synthesis of 1-(5-methoxybenzo[d][1,3]oxathiol-6-yl-2,2-d2)propan-2-amine (II-21) was carried out according to Fig. 7. To a pressure vessel was added II-20b (1.17 g, 6.41 mmol, 1 eq), tetrabutylammonium bromide (207 mg, 0.641 mmol, 0.1 eq), aqueous NaOH (5M, 6.4 mL, 32.06 mmol, 5 eq), and dibromomethane-d2 (2.25 mL, 32.06 mmol, 5 eq). The vessel was sealed and heated at 100 °C for 3 days. The reaction mixture was poured into water (50 mL) and extracted with DCM (3 x 50 mL). The combined organic phase was washed with brine (20 mL), dried over Na2SO4, and concentrated. The crude material was purified on silica (Hex to 10% EtOAc/Hex) to afford II-21a (0.61 g, 59% yield) as a colorless oil; the structure was confirmed by 1 H NMR in CDCl3, with some H/D exchange of methylene protons, and some minor impurities present. To POCl3 (2.60 mL, 27.86 mmol, 4.3 eq) was slowly added N-methylformanilide (2.96 mL, 23.98 mmol, 3.7 eq) at room temperature under N 2 . After 30 min, II-21a (1.09 g, 6.48 mmol, 1 eq) was slowly added and reaction heated to 70 °C. After 2 h, the reaction mixture was carefully poured into ice water (50 mL) and extracted with DCM (3 x 50 mL). The combined organic phase was washed with brine (20 mL), dried over Na 2 SO 4 , and concentrated. The crude material was purified on silica (Hex to 20% EtOAc/Hex). The impurity in the combined fractions was removed by trituration with EtOAc/Hex to afford II-21b (0.99 g, 79% yield) as a yellow solid; the structure was confirmed by 1 H NMR in CDCl3. To II-21b (0.99 g, 5.02 mmol, 1 eq) was added anhydrous ammonium acetate (387 mg, 5.02 mmol, 1 eq), nitroethane (1.8 mL, 25.12 mmol, 5 eq), acetic acid (glacial, 4.0 mL, 70.28 mmol, 14 eq) and activated 3Å molecular sieves (2.5 g wet weight) under N2 at room temperature. The reaction mixture was heated at 95 °C for 1 h. The molecular sieves were removed, reaction mixture poured into brine (40 mL) and extracted with EtOAc (3 x 40 mL). The combined organic phase was washed with water (20 mL), then brine (20 mL), dried over Na2SO4, and concentrated to afford II-21c (97% yield) as an orange solid; the structure was confirmed by 1 H NMR in CDCl 3 . The crude material was used in the next step without further purification. To a solution of II-21c (1.17 g, 4.62 mmol, 1 eq) in MeOH (17 mL), water (7 mL), and concentrated HCl (12.1 M, 7 mL) was added iron (powder, reduced) (1.29 g, 23.10 mmol, 5 eq) portionwise over 10 min at room temperature under N2. The reaction was heated at 70 °C for 2 h before adding additional concentrated HCl (12.1 M, 7 mL) and iron (powder, reduced) (1.29 g, 23.10 mmol, 5 eq). After heating for 2 h, the reaction mixture was poured into water (50 mL) and extracted with DCM (3 x 50 mL). The combined organic phase was washed with brine (30 mL), dried over Na2SO4, and concentrated. The crude material was purified on silica (Hex to 30% EtOAc/Hex) to afford II-21d (50% yield) as a white solid; the structure was confirmed by 1 H NMR in CDCl 3 . To a solution of II-21d (345 mg, 1.54 mmol, 1 eq) and titanium(IV) ethoxide (0.65 mL, 3.08 mmol, 2 eq) in anhydrous THF (8 mL) was added (R/S)-2-methyl-2-propanesulfinamide (373 mg, 3.08 mmol, 2 eq) in anhydrous THF (3 mL) at room temperature under N 2 . The reaction was heated at 70 °C for 4 h before cooling to -45 °C and slowly transferring into a solution of NaBH 4 (233 mg, 6.15 mmol, 4 eq) in anhydrous THF (8 mL) at -45 °C under N2. After 45 min, the reaction was carefully quenched with MeOH (5 mL) and allowed to warm to room temperature. Saturated aqueous NH4Cl (10 mL) was added and mixture filtered through Celite with EtOAc. The filtrate was poured into water (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic phase was washed with brine (10 mL), dried over Na2SO4, and concentrated. The crude material was purified on silica (Hex to EtOAc) to afford II- 21e (65% yield) as a colorless oil; the structure was confirmed by 1 H NMR in CDCl3. To a solution of II-21e (390 mg, 1.18 mmol, 1 eq) in a mixture of anhydrous MeOH (2.1 mL) and anhydrous Et2O (21 mL) was added anhydrous HCl (2M in Et2O, 2.4 mL, 4.73 mmol, 4 eq) dropwise at room temperature under N 2 . After stirring overnight, the reaction mixture was concentrated, solid washed with Et 2 O, and dried to afford II-21 (213 mg, 91% yield) as a white solid; the structure was confirmed by 1 H NMR in CD 3 CN, with some H/D exchange of methylene protons occurring in the conversion of II-20b to II-21a as noted above. Example 8 2-(2,2-difluorobenzo[d][1,3]oxathiol-6-yl)ethan-1-amine (II-58) Synthesis of 2-(2,2-difluorobenzo[d][1,3]oxathiol-6-yl)ethan-1-amine (II-58) was carried out according to Fig.8. To a dried pressure vessel at room temperature was added commercially available 4-bromo-3-hydroxybenzaldehyde (II-58a) (797 mg, 3.96 mmol, 1 eq), trifluoromethylthiolato(2,2- bipyridine)copper(I) (1.53 g, 4.76 mmol, 1.2 eq), and potassium fluoride (230 mg, 3.96 mmol, 1 eq) in anhydrous MeCN (sparged with N2, 33 mL). After sealing vessel, reaction heated at 100 °C for 1 day. The reaction mixture was poured into hexane (200 mL) and washed with brine (3 x 50 mL). The combined organic phase was dried over Na2SO4 and concentrated. The crude material was purified on silica (Hex to 10% EtOAc/Hex) to afford II-58b (235 mg, 29% yield) as a white solid; the structure was confirmed by 1 H NMR in CDCl3. To II-58b (235 mg, 1.16 mmol, 1 eq) was added anhydrous ammonium acetate (134 mg, 1.74 mmol, 1.5 eq), nitromethane (0.31 mL, 5.80 mmol, 5 eq), acetic acid (glacial, 0.93 mL, 16.24 mmol, 14 eq) and activated 3Å molecular sieves (0.65 g wet weight) under N2 at room temperature. The reaction mixture was heated at 95 °C for 5 h. The molecular sieves were removed, reaction mixture poured into brine (20 mL) and extracted with EtOAc (3 x 30 mL). The combined organic phase was washed with water (10 mL), then brine (10 mL), dried over Na 2 SO 4 , and concentrated to afford II-58c (192 mg, 68% yield) as an orange solid. The crude material was used in the next step without further purification; the structure was confirmed by 1 H NMR in CDCl 3 . To anhydrous MeOH (13 mL) at 0 °C under N 2 was added II-58c (192 mg, 0.78 mmol, 1 eq), zinc dust (614 mg, 9.40 mmol, 12 eq), and concentrated HCl (12.1 M, 1.55 mL, 18.79 mmol, 24 eq) portionwise over 10 min. Reaction mixture was allowed to warm to room temperature and stirred for 2 h before adding additional zinc dust (614 mg, 9.40 mmol, 12 eq) and concentrated HCl (12.1 M, 1.55 mL, 18.79 mmol, 24 eq). After 3 h, the excess zinc was removed, reaction mixture was cooled to 0 °C, basified with 20% NH4OH/MeOH, and partially concentrated. The mixture was poured into water (25 mL) and extracted with DCM (3 x 25 mL). The combined organic phase was washed with brine (20 mL), dried over Na2SO4, and concentrated. The crude material was purified on silica (DCM to 90:10:1 DCM/MeOH/NH4OH) to afford II-58 (11 mg, 6% yield) as a yellow oil; the structure was confirmed by 1 H NMR in CDCl3. To a solution of II-58 (19 mg, 0.087 mmol, 1 eq) in a mixture of anhydrous MeOH (1 mL) and anhydrous Et 2 O (1 mL) was added anhydrous HCl (2M in Et 2 O, 48 µL, 0.096 mmol, 1.1 eq) dropwise at room temperature under N 2 . After 10 min, the reaction mixture was concentrated, solid washed with Et 2 O, and dried to afford II-58 as an HCl salt (18 mg, 81% yield) as a beige solid; the structure was confirmed by 1 H NMR and 19 F NMR in DMSO-d 6 . Examples 9-20 Examples 9-20 are prepared according to general Fig. 9, using a modified general procedure as reported by Shulgin (Shulgin, A., and Shulgin, Ann. (1991) Pihkal: a chemical love story, Transform Press, Berkeley, CA) and later modified by Maresh (Maresh, J. J., Ralko, A. A., Speltz, T. E., Burke, J. L., Murphy, C. M., Gaskell, Z., Girel, J. K., Terranova, E., Richtscheidt, C., and Krzeszowiec, M. (2014) Chemoselective Zinc/HCl Reduction of Halogenated beta-Nitrostyrenes: Synthesis of Halogenated Dopamine Analogues, Synlett 25, 2891-2894). Suitable starting material A is treated with cesium carbonate and either bromochloromethane or deuterated bromochloromethane in DMF to form cyclized intermediate B according to a procedure outlined by Zelle (Zelle, R. E., and Mcclellan, W. J., 1991, A Simple, High-Yielding Method for the Methylenation of Catechols, Tetrahedron Letters 32, 2461- 2464). Next, a nitroaldol condensation with either nitromethane or nitroethane and buffered acetic acid affords intermediate C. GX \bW]RO\ScO KWKUXQ\ `S]R j'NO^]O[K]SXW& SW]O[VONSK]O C is reduced with sodium borohydride and silicone dioxide to selectively reduce the alkene to intermediate D (Sinhababu, A. K., and Borchardt, R. T., 1983, Silica Gel-Assisted Reduction of Nitrostyrenes to 2-Aryl-1-Nitroalkanes with Sodium- Borohydride, Tetrahedron Letters 24& ,,1',-*$( 9O^]O[S^V OaMRKWQO K] ]RO j'YX\S]SXW S\ MK[[SON X^] using the basic resin WA30 and deuterated water developed by Yamada (Yamada, T., Kuwata, M., Takakura, R., Monguchi, Y., Sajiki, H., and Sawama, Y. (2018) Organocatalytic Nitroaldol Reaction Associated with Deuterium-Labeling, Adv Synth Catal 360, 637-641) to form intermediate E. Reduction of the nitro group with zinc dust in methanol containing hydrochloric acid yields the final product F, as an HCl salt. GX \bW]RO\ScO j'RbN[XQOW KWKUXQ\& SW]O[VONSK]O C is subjected bis-reduction of the nitro group and alkene with zinc dust in methanol containing hydrochloric acid, to yield the final product G, as an HCl salt. Example 9: 2-(benzo[d][1,3]dioxol-5-yl)ethan-1,1-d2-1-amine (II-1). The structure of the product will be confirmed by 1 H NMR. Example 10: 1-(benzo[d][1,3]dioxol-5-yl-2,2-d2)propan-2-amine (II-4). The structure of the product will be confirmed by 1 H NMR. Example 11: 2-(benzo[d][1,3]dioxol-5-yl-2,2-d2)ethan-1,1-d2-1-amine (II-5). The structure of the product will be confirmed by 1 H NMR. Example 12: 2-(7-methoxybenzo[d][1,3]dioxol-5-yl)ethan-1,1-d2-1-amine (II-9). The structure of the product will be confirmed by 1 H NMR. Example 13: 2-(7-methoxybenzo[d][1,3]dioxol-5-yl-2,2-d2)ethan-1,1-d2-1-a mine (II-10). The structure of the product will be confirmed by 1 H NMR. Example 14: 2-(6-methoxybenzo[d][1,3]dioxol-5-yl)ethan-1,1-d2-1-amine (II-12). The structure of the product will be confirmed by 1 H NMR. Example 15: 2-(6-methoxybenzo[d][1,3]dioxol-5-yl-2,2-d2)ethan-1-amine (II-13). The structure of the product will be confirmed by 1 H NMR. Example 16: 2-(5-methoxybenzo[d][1,3]oxathiol-6-yl)ethan-1-amine (II-18). The structure of the product will be confirmed by 1 H NMR. Example 17: 2-(5-methoxybenzo[d][1,3]oxathiol-6-yl-2,2-d2)ethan-1-amine (II-19). The structure of the product will be confirmed by 1 H NMR. Example 18: 1-(benzo[d][1,3]dioxol-5-yl)propan-2-d-2-amine (II-27). The structure of the product will be confirmed by 1 H NMR. Example 19: 1-(benzo[d][1,3]dioxol-5-yl-2,2-d2)propan-2-d-2-amine (II-29). The structure of the product will be confirmed by 1 H NMR. Example 20: 1-(7-methoxybenzo[d][1,3]dioxol-5-yl)propan-2-d-2-amine (II-44). The structure of the product will be confirmed by 1 H NMR. Examples 21-33 Examples 21-33 are prepared according to general Fig. 10. Compounds F or G (see Fig. 9) are subjected to reductive amination with either (i) sodium cyanoborodeuteride and deuterated formaldehyde (CD 2 O) or (ii) sodium cyanoborohydride and formaldehyde (CH 2 O), to yield product H or I. Example 21: 2-(benzo[d][1,3]dioxol-5-yl)-N-methylethan-1,1-d2-1-amine (II-2). The structure of the product will be confirmed by 1 H NMR. Example 22: 2-(benzo[d][1,3]dioxol-5-yl)-N-(methyl-d3)ethan-1,1-d2-1-ami ne (II-3). The structure of the product will be confirmed by 1 H NMR. Example 23: 2-(benzo[d][1,3]dioxol-5-yl-2,2-d2)-N-methylethan-1,1-d2-1-a mine (II-6). The structure of the product will be confirmed by 1 H NMR. Example 24: 2-(benzo[d][1,3]dioxol-5-yl-2,2-d2)-N-(methyl-d3)ethan-1,1-d 2-1-amine (II-7). The structure of the product will be confirmed by 1 H NMR. Example 25: 1-(benzo[d][1,3]dioxol-5-yl-2,2-d2)-N-methylpropan-2-amine (II-8). The structure of the product will be confirmed by 1 H NMR. Example 26: 1-(benzo[d][1,3]dioxol-5-yl)-N-methylpropan-2-d-2-amine (II-31). The structure of the product will be confirmed by 1 H NMR. Example 27: 1-(benzo[d][1,3]dioxol-5-yl-2,2-d2)-N-(methyl-d3)propan-2-d- 2-amine (II-33). The structure of the product will be confirmed by 1 H NMR. Example 28: 1-(benzo[d][1,3]dioxol-5-yl)-N-(methyl-d3)propan-2-d-2-amine (II-35). The structure of the product will be confirmed by 1 H NMR. Example 29: 1-(benzo[d][1,3]dioxol-5-yl-2,2-d2)-N-(methyl-d3)propan-2-am ine (II-40). The structure of the product will be confirmed by 1 H NMR. Example 30: 1-(benzo[d][1,3]dioxol-5-yl)-N-(methyl-d3)propan-2-amine (II-42). The structure of the product will be confirmed by 1 H NMR. Example 31: 1-(7-methoxybenzo[d][1,3]dioxol-5-yl)-N-methylpropan-2-d-2-a mine (II-45). The structure of the product will be confirmed by 1 H NMR. Example 32: 1-(7-methoxybenzo[d][1,3]dioxol-5-yl)-N-(methyl-d3)propan-2- d-2-amine (II-47). The structure of the product will be confirmed by 1 H NMR. Example 33: 2-(7-methoxybenzo[d][1,3]dioxol-5-yl)-N-(methyl-d3)ethan-1,1 -d2-1-amine (II-48). The structure of the product will be confirmed by 1 H NMR. Example 34 2-(3,4,5-trimethoxyphenyl)ethan-1,1-d2-1-amine (III-1) Synthesis of 2-(3,4,5-trimethoxyphenyl)ethan-1,1-d2-1-amine (III-1) is carried out according to Fig. 11, using a modified general procedure as reported by Shulgin (Shulgin, A., and Shulgin, Ann. (1991) Pihkal: a chemical love story, Transform Press, Berkeley, CA) and later modified by Maresh (Maresh, J. J., Ralko, A. A., Speltz, T. E., Burke, J. L., Murphy, C. M., Gaskell, Z., Girel, J. K., Terranova, E., Richtscheidt, C., and Krzeszowiec, M. (2014) Chemoselective Zinc/HCl Reduction of Halogenated beta-Nitrostyrenes: Synthesis of Halogenated Dopamine Analogues, Synlett 25, 2891- 2894). The starting material 3,4,5-trimethoxybenzaldehyde (III-1a) undergoes a nitroaldol condensation with nitromethane and buffered acetic acid to form intermediate III-1b, followed by a selective alkene reduction with sodium borohydride and silicone dioxide to selectively reduce the alkene to intermediate III-1c (Sinhababu, A. K., and Borchardt, R. T., 1983, Silica Gel-Assisted Reduction of Nitrostyrenes to 2-Aryl-1-Nitroalkanes with Sodium-Borohydride, Tetrahedron Letters 24, 227-230). Deuterium OaMRKWQO K] ]RO j'YX\S]SXW S\ MK[[SON X^] ^\SWQ ]RO LK\SM [O\SW I6-* KWN NO^]O[K]ON `K]O[ NO_OUXYON Lb Yamada (Yamada, T., Kuwata, M., Takakura, R., Monguchi, Y., Sajiki, H., and Sawama, Y. (2018) Organocatalytic Nitroaldol Reaction Associated with Deuterium-Labeling, Adv Synth Catal 360, 637- 641) to form intermediate III-1d. Reduction of the nitro group with zinc dust in methanol containing hydrochloric acid yields the final product (III-1), as an HCl salt. The structure of the product will be confirmed by 1 H NMR. Alternatively, the compound (III-1) is produced according to Fig. 11, whereby reduction of starting material III-1a with sodium borohydride yields benzyl alcohol III-1e, which is then converted with PBr 3 to benzyl bromide III-1f. Displacement with potassium cyanide then yields the benzyl cyanide III-1g, followed by reduction with lithium aluminum deuteride in the presence of aluminum chloride to yield the title compound (III-1). Example 35 2-(3,4,5-trimethoxyphenyl)ethan-1,1,2,2-d4-1-amine (III-2) Synthesis of 2-(3,4,5-trimethoxyphenyl)ethan-1,1,2,2-d4-1-amine (III-2) is carried out according to Fig. 12, using a modified general procedure as reported by Shulgin (Shulgin, A., and Shulgin, Ann. (1991) Pihkal: a chemical love story, Transform Press, Berkeley, CA) and later modified by Maresh (Maresh, J. J., Ralko, A. A., Speltz, T. E., Burke, J. L., Murphy, C. M., Gaskell, Z., Girel, J. K., Terranova, E., Richtscheidt, C., and Krzeszowiec, M. (2014) Chemoselective Zinc/HCl Reduction of Halogenated beta-Nitrostyrenes: Synthesis of Halogenated Dopamine Analogues, Synlett 25, 2891- 2894). 3,4,5-trimethoxybenzonitrile (III-2a) is deuterated by reduction using Lithium tris(dihexylamino)aluminum deuteride (Li(hex 2 N) 3 AlD) to the deuterated benzaldehyde III-2b using a method developed by Cha (Cha, J. S., Lee, S. E., and Lee, H. S., 1992, Selective Conversion of Aromatic Nitriles to Aldehydes by Lithium Tris(Dihexylamino)Aluminum Hydride, Org Prep Proced Int 24, 331- --.$( n'WS][X\]b[OWO III-2c is formed using a nitroaldol condensation with nitromethane in buffered acidic conditions. Subsequent treatment with sodium borodeuteride and silicone dioxide selectively reduces the alkene to intermediate III-2d (Sinhababu, A. K., and Borchardt, R. T. (1983) Silica Gel- Assisted Reduction of Nitrostyrenes to 2-Aryl-1-Nitroalkanes with Sodium-Borohydride, Tetrahedron Letters 24& ,,1',-*$& PXUUX`ON Lb NO^]O[S^V OaMRKWQO K] ]RO j'YX\S]SXW ^\SWQ ]RO LK\SM [O\SW I6-* KWN deuterated water developed by Yamada (Yamada, T., Kuwata, M., Takakura, R., Monguchi, Y., Sajiki, H., and Sawama, Y. (2018) Organocatalytic Nitroaldol Reaction Associated with Deuterium-Labeling, Adv Synth Catal 360, 637-641) to form intermediate III-2e. Reduction of the nitro group with zinc dust in methanol containing hydrochloric acid yields the final product (III-2), as an HCl salt. The structure of the product will be confirmed by 1 H NMR. Alternatively, the compound III-2 is produced according to Fig.12, whereby benzylic deuterium exchange of intermediate III-1g (see Fig.11, Example 34) with deuterated water under basic conditions affords the benzyl cyanide III-2f, followed by reduction with lithium aluminum deuteride in the presence of aluminum chloride to yield the title compound (III-2). Example 36 2-(3,4,5-tris(methoxy-d3)phenyl)ethan-1,1-d2-1-amine (III-3) Synthesis of 2-(3,4,5-tris(methoxy-d3)phenyl)ethan-1,1-d2-1-amine (III-3) is carried out according to Fig. 13, using a modified general procedure as reported by Shulgin (Shulgin, A., and Shulgin, Ann. (1991) Pihkal: a chemical love story, Transform Press, Berkeley, CA) and later modified by Maresh (Maresh, J. J., Ralko, A. A., Speltz, T. E., Burke, J. L., Murphy, C. M., Gaskell, Z., Girel, J. K., Terranova, E., Richtscheidt, C., and Krzeszowiec, M. (2014) Chemoselective Zinc/HCl Reduction of Halogenated beta-Nitrostyrenes: Synthesis of Halogenated Dopamine Analogues, Synlett 25, 2891- 2894). Methylation of 3,4,5-hydroxybenzaldehyde (III-3a) is carried out with neat deuterated dimethylcarbonate according to Ouk (Ouk, S., Thiebaud, S., Borredon, E., and Le Gars, P., 2003, High performance method for O-methylation of phenol with dimethyl carbonate, Appl Catal A-Gen 241, 227- 233). Benzaldehyde III-3b then undergoes a nitroaldol condensation with nitromethane and buffered acetic acid followed by a selective alkene reduction of intermediate III-3c with sodium borohydride and silicone dioxide to selectively reduce the alkene to intermediate III-3d (Sinhababu, A. K., and Borchardt, R. T., 1983, Silica Gel-Assisted Reduction of Nitrostyrenes to 2-Aryl-1-Nitroalkanes with Sodium-Borohydride, Tetrahedron Letters 24& ,,1',-*$( 9O^]O[S^V OaMRKWQO K] ]RO j'YX\S]SXW S\ carried out using the basic resin WA30 and deuterated water developed by Yamada (Yamada, T., Kuwata, M., Takakura, R., Monguchi, Y., Sajiki, H., and Sawama, Y. (2018) Organocatalytic Nitroaldol Reaction Associated with Deuterium-Labeling, Adv Synth Catal 360, 637-641) to form intermediate III- 3e. Reduction of the nitro group with zinc dust in methanol containing hydrochloric acid yields the final product (III-3), as an HCl salt. The structure of the product will be confirmed by 1 H NMR. Alternatively, the compound III-3 is produced according to Fig. 13, whereby starting material III-3a is per-alkylated with CD3I forming intermediate III-3f, which is then reduced with sodium borohydride to yield benzyl alcohol III-3g. Conversion to benzyl bromide III-3h is accomplished with PBr3. Displacement with potassium cyanide then yields the benzyl cyanide III-3i, followed by reduction with lithium aluminum deuteride in the presence of aluminum chloride to yield the title compound (III- 3). Example 37 2-(3,5-dimethoxy-4-(trifluoromethoxy)phenyl)ethan-1,1-d2-1-a mine (III-4) Synthesis of 2-(3,5-dimethoxy-4-(trifluoromethoxy)phenyl)ethan-1,1-d2-1-a mine (III-4) is carried out according to Fig. 14, using a modified general nitrostyrene route reported by Shulgin (Shulgin, A., and Shulgin, Ann., 1991, Pihkal: a chemical love story, Transform Press, Berkeley, CA.) and later modified by Sinhababu for the selective alkene reduction in the presence of nitro groups (Sinhababu, A. K., and Borchardt, R. T., 1983, Silica Gel-Assisted Reduction of Nitrostyrenes to 2- Aryl-1-Nitroalkanes with Sodium-Borohydride, Tetrahedron Letters 24, 227-230). 3,5-Dimethoxy-4- hydroxybenzaldehyde (III-4a) is treated sequentially with sodium hydride, carbon disulfide, and methyl iodide to form xanthate ester III-4b. Reaction with HF/pyridine and dibromohydantoin (DBH) forms intermediate III-4c, which then undergoes a nitroaldol condensation with nitromethane and buffered acetic acid followed by a selective alkene reduction with sodium borohydride and silicone dioxide to selectively reduce the alkene to intermediate III-4d. Alpha protons relative to the nitro group are exchanged for deuterium using a basic resin WA30 developed by Yamada (Yamada, T., Kuwata, M., Takakura, R., Monguchi, Y., Sajiki, H., and Sawama, Y., 2018, Organocatalytic Nitroaldol Reaction Associated with Deuterium-Labeling, Adv Synth Catal 360, 637-641). Reduction of the nitro group in intermediate III-4e is completed using zinc dust in methanol containing hydrochloric acid according to Maresh (Maresh, J. J., Ralko, A. A., Speltz, T. E., Burke, J. L., Murphy, C. M., Gaskell, Z., Girel, J. K., Terranova, E., Richtscheidt, C., and Krzeszowiec, M., 2014, Chemoselective Zinc/HCl Reduction of Halogenated beta-Nitrostyrenes: Synthesis of Halogenated Dopamine Analogues, Synlett 25, 2891- 2894), to afford the final product (III-4), as an HCl salt. The structure of the product will be confirmed by 1 H NMR. Alternatively, the compound III-4 is produced according to Fig. 14, whereby reduction of intermediate III-4c with sodium borohydride yields benzyl alcohol III-4f, which is then converted with PBr 3 to benzyl bromide III-4g. Displacement with potassium cyanide then yields the benzyl cyanide III-4h, followed by reduction with lithium aluminum deuteride in the presence of aluminum chloride to yield the title compound (III-4). Example 38 2-(3,5-dimethoxy-4-((trifluoromethyl)thio)phenyl)ethan-1-ami ne (III-5) Synthesis of 2-(3,5-dimethoxy-4-((trifluoromethyl)thio)phenyl)ethan-1-ami ne (III-5) was carried out according to Fig. 15. To a solution of commercially available 3,5-dihydroxybenzoic acid (III-5a) (3.00 g, 19.46 mmol, 1 eq) in anhydrous MeOH (13 mL) at 0 °C under N2 was added N- iodosuccinimide (4.60 g, 20.44 mmol, 1.05 eq) in anhydrous MeOH (13 mL). After warming to room temperature and stirring overnight, the reaction mixture was poured into ice water (100 mL) and extracted with EtOAc (3 x 100 mL). The combined organic phase was washed with aqueous Na 2 S 2 O 5 (1M, 50 mL) then brine (20 mL), dried over Na2SO4, and concentrated to afford III-5b (5.57 g, 100% yield) as a beige solid. The crude material was used in the next step without further purification; the structure was confirmed by 1 H NMR in DMSO-d6. To a solution of III-5b (5.57 g, 19.9 mmol, 1 eq) in anhydrous DMF (20 mL) was added potassium carbonate (13.8 g, 99.5 mmol, 5 eq) and iodomethane (6.2 mL, 99.5 mmol, 5 eq) under N2 at room temperature. After 3 days, the reaction mixture was diluted with EtOAc (300 mL) and washed with water (3 x 100 mL) then brine (100 mL). The combined organic phase was dried over Na 2 SO 4 and concentrated to afford III-5c (5.80 g, 90% yield) as a beige solid. The crude material was used in the next step without further purification; the structure was confirmed by 1 H NMR in CDCl 3 . To a solution of III-5c (5.75 g, 17.9 mmol, 1 eq) in anhydrous PhMe (89 mL) under N 2 at 0 °C was slowly added DIBAL-H (1M in THF, 54 mL, 53.6 mmol, 3 eq) over 10 min. After 1 h, reaction warmed to room temperature. After 1 h, reaction mixture cooled to 0 °C and carefully quenched with MeOH (25 mL), then poured into aqueous HCl (0.5 M, 250 mL) and extracted with DCM (3 x 250 mL). The combined organic phase was washed with brine (100 mL), dried over Na2SO4, and concentrated to afford III-5d (4.84 g, 92% yield) as a white solid. The crude material was used in the next step without further purification; the structure was confirmed by 1 H NMR in CDCl3. To a solution of III-5d (4.84 g, 16.5 mmol, 1 eq) in anhydrous DCM (82 mL) was added activated manganese(IV) oxide (14.3 g, 165 mmol, 10 eq) under N2 at room temperature. After 1 day, the reaction mixture was filtered through Celite with DCM and concentrated. The crude material was purified on silica (Hex to 50% EtOAc/Hex) to afford III-5e (3.37 g, 70% yield) as a white solid; the structure was confirmed by 1 H NMR in CDCl 3 . To a dried pressure vessel was added III-5e (1.25 g, 4.28 mmol, 1 eq), XPhos (347 mg, 0.73 mmol, 0.17 eq) and [(cod)Pd(CH 2 TMS) 2 ] (250 mg, 0.64 mmol, 0.15 eq) and the vessel was purged with N 2 before adding anhydrous PhMe (sparged with N 2 , 21 mL). The mixture was then heated at 100 °C for 1 min before adding AgSCF3 (1.16 g, 5.57 mmol, 1.3 eq) and Ph(Et)3NI (1.70 g, 5.57 mmol, 1.3 eq), purging with N2, and sealing the pressure vessel. After heating at 100 °C for 3 days, the reaction mixture was filtered through silica with DCM/EtOAc and concentrated. The crude material was purified on silica (Hex to 50% DCM/Hex) to afford III-5f (0.82 g, 72% yield) as a tan solid; the structure was confirmed by 1 H NMR and 19 F NMR in CDCl3. To III-5f (1.10 g, 4.13 mmol, 1 eq) was added anhydrous ammonium acetate (320 mg, 4.13 mmol, 1 eq), nitromethane (1.12 mL, 20.65 mmol, 5 eq), acetic acid (glacial, 3.32 mL, 57.82 mmol, 14 eq) and activated 3Å molecular sieves (2.3 g wet weight) under N 2 at room temperature. The reaction mixture was heated at 95 °C for 1 h. The molecular sieves were removed, reaction mixture poured into brine (30 mL) and extracted with EtOAc (3 x 30 mL). The combined organic phase was washed with water (20 mL), then brine (20 mL), dried over Na2SO4, and concentrated. The crude material was purified on silica (Hex to 50% EtOAc/Hex) to afford III-5g (1.01 g, 80% yield) as a tan solid; the structure was confirmed by 1 H NMR in in CDCl3. To anhydrous MeOH (20 mL) at 0 °C under N2 was added III-5g (0.81 g, 2.62 mmol, 1 eq), zinc dust (2.06 g, 31.43 mmol, 12 eq), and concentrated HCl (12.1 M, 5.2 mL, 62.9 mmol, 24 eq) portionwise over 20 min. The reaction mixture allowed to stir at 0 °C for 4 h. The excess zinc was removed and the reaction mixture was cooled to 0 °C before basifying with saturated methanolic NaOH. The mixture was diluted with DCM, dried over Na 2 SO 4 , and partially concentrated. The crude mixture was filtered through Celite with DCM to remove insoluble and concentrated. The crude material was purified on silica (DCM to 90:10:1 DCM/MeOH/NH 4 OH) to afford III-5 (177 mg, 24% yield) as a beige solid; the structure was confirmed by 1 H NMR in in CDCl 3 . To a solution of III-5 (270 mg, 0.960 mmol, 1 eq) in a mixture of anhydrous MeOH (10 mL) and anhydrous Et2O (10 mL) was added anhydrous HCl (2M in Et2O, 0.53 mL, 1.056 mmol, 1.1 eq) dropwise at room temperature under N2. After 10 min, the reaction mixture was concentrated, solid washed with Et2O, and dried to afford III-5, as an HCl salt (290 mg, 95% yield) as a beige solid; the structure was confirmed by 1 H NMR and 19 F NMR in DMSO-d6. Example 39 2-(3,5-dimethoxy-4-((trifluoromethyl)thio)phenyl)ethan-1,1-d 2-1-amine (III-7) Synthesis of 2-(3,5-dimethoxy-4-((trifluoromethyl)thio)phenyl)ethan-1,1-d 2-1-amine (III-7) is carried out according to Fig. 16, using a modified general nitrostyrene route reported by Shulgin (Shulgin, A., and Shulgin, Ann., 1991, Pihkal: a chemical love story, Transform Press, Berkeley, CA.) and later modified by Sinhababu for the selective alkene reduction in the presence of nitro groups (Sinhababu, A. K., and Borchardt, R. T., 1983, Silica Gel-Assisted Reduction of Nitrostyrenes to 2- Aryl-1-Nitroalkanes with Sodium-Borohydride, Tetrahedron Letters 24, 227-230). 4-mercapto-3,5- dimethoxybenzaldehyde (III-7a) is fluoroalkylated with potassium t-butoxide and trifluoromethyl bromide to give intermediate III-7b, which then undergoes a nitroaldol condensation with nitromethane and buffered acetic acid followed by a selective alkene reduction with sodium borohydride and silicone dioxide to selectively reduce the alkene to intermediate III-7c. Alpha protons relative to the nitro group are exchanged for deuterium using a basic resin WA30 developed by Yamada (Yamada, T., Kuwata, M., Takakura, R., Monguchi, Y., Sajiki, H., and Sawama, Y., 2018, Organocatalytic Nitroaldol Reaction Associated with Deuterium-Labeling, Adv Synth Catal 360, 637-641). Reduction of the nitro group in intermediate III-7d is completed using zinc dust in methanol containing hydrochloric acid according to Maresh (Maresh, J. J., Ralko, A. A., Speltz, T. E., Burke, J. L., Murphy, C. M., Gaskell, Z., Girel, J. K., Terranova, E., Richtscheidt, C., and Krzeszowiec, M., 2014, Chemoselective Zinc/HCl Reduction of Halogenated beta-Nitrostyrenes: Synthesis of Halogenated Dopamine Analogues, Synlett 25, 2891- 2894), to afford the final product III-7, as an HCl salt. The structure of the product will be confirmed by 1 H NMR. Alternatively, the compound III-7 is produced according to Fig. 16, whereby a Pd/XPhos- catalyzed cross coupling between iodobenzaldehyde starting material III-7e and AgSCF3 using (1,5- cyclooctadiene)bis(trimethylsilylmethyl)palladium(II) catalyst in the presence of phenyltriethylammonium iodide provides intermediate III-7b. Reduction with sodium borohydride yields benzyl alcohol III-7f, which is then converted with PBr 3 to benzyl bromide III-7g. Displacement with potassium cyanide then yields the benzyl cyanide III-7h, followed by reduction with lithium aluminum deuteride in the presence of aluminum chloride to yield the title compound (III-7). Example 40 1-(2,5-dimethoxy-4-((trifluoromethyl)thio)phenyl)propan-2-am ine (IV-1) Synthesis of 1-(2,5-dimethoxy-4-((trifluoromethyl)thio)phenyl)propan-2-am ine (IV-1) was carried out according to Fig. 17. To a solution of commercially available 2,5-dimethoxybenzaldehyde IV-1a (5.00 g, 30.1 mmol, 1 eq) in anhydrous MeOH (200 mL) was added AgNO3 (5.11 g, 30.1 mmol, 1 eq) and iodine (8.40 g, 33.1 mmol, 1.1 eq) at room temperature under N2. After 2 days, the precipitate was removed by filtration and the filtrate was poured into aqueous Na2S2O5 (1M, 250 mL). The resulting precipitate was collected by filtration, washed with water then hexane, and dried to afford IV-1b (7.80 g, 89% yield) as a beige solid. The crude material was used in the next step without further purification; the structure was confirmed by 1 H NMR in CDCl 3 . To a dried pressure vessel was added IV-1b (1.25 g, 4.28 mmol, 1 eq), XPhos (347 mg, 0.73 mmol, 0.17 eq) and [(cod)Pd(CH 2 TMS) 2 ] (250 mg, 0.64 mmol, 0.15 eq) and vessel purged with N 2 before adding anhydrous PhMe (sparged with N2, 21 mL). The mixture was then heated at 100 °C for 1 min before adding AgSCF3 (1.16 g, 5.57 mmol, 1.3 eq) and Ph(Et)3NI (1.70 g, 5.57 mmol, 1.3 eq), purging with N2, and sealing the pressure vessel. After heating at 100 °C for 3 days, the reaction mixture was filtered through silica with DCM/EtOAc and concentrated. The crude material was purified on silica (Hex to 5% EtOAc/Hex) to afford IV-1c (0.84 g, 74% yield) as a beige solid; the structure was confirmed by 1 H NMR in CDCl3. To IV-1c (0.83 g, 3.12 mmol, 1 eq) was added anhydrous ammonium acetate (240 mg, 3.12 mmol, 1 eq), nitroethane (1.12 mL, 15.6 mmol, 5 eq), acetic acid (glacial, 2.5 mL, 43.7 mmol, 14 eq) and activated 3Å molecular sieves (1.7 g wet weight) under N2 at room temperature. The reaction mixture was heated at 95 °C for 1 h. The molecular sieves were removed, reaction mixture poured into brine (30 mL) and extracted with EtOAc (3 x 30 mL). The combined organic phase was washed with water (20 mL), then brine (20 mL), dried over Na2SO4, and concentrated. The crude material was purified on silica (Hex to 50% EtOAc/Hex) to afford IV-1d (0.63 g, 63% yield) as a yellow solid; the structure was confirmed by 1 H NMR in CDCl3. To a solution of IV-1d (0.43 g, 1.33 mmol, 1 eq) in MeOH (5 mL), water (2 mL), and concentrated HCl (12.1 M, 2 mL) was added iron (powder, reduced) (0.30 g, 5.32 mmol, 4 eq) portionwise over 10 min at room temperature under N 2 . The reaction was heated at 70 °C for 2 h before adding additional concentrated HCl (12.1 M, 2 mL) and iron (powder, reduced) (0.30 g, 5.32 mmol, 4 eq). After heating for 2 h, the reaction mixture was poured into water (30 mL) and extracted with DCM (3 x 30 mL). The combined organic phase was washed with brine (20 mL), dried over Na 2 SO 4 , and concentrated. The crude material was purified on silica (Hex to 30% EtOAc/Hex) to afford IV-1e (245 mg, 63% yield) as a white solid; the structure was confirmed by 1 H NMR in CDCl3. To a solution of IV-1e (245 mg, 0.833 mmol, 1 eq) and titanium(IV) ethoxide (0.35 mL, 1.67 mmol, 2 eq) in anhydrous THF (4.2 mL) was added (R/S)-2-methyl-2-propanesulfinamide (202 mg, 1.67 mmol, 2 eq) in anhydrous THF (1.7 mL) at room temperature under N2. The reaction was heated at 70 °C for 4 h before cooling to -45 °C and slowly transferred into a solution of NaBH4 (126 mg, 3.33 mmol, 4 eq) in anhydrous THF (4.2 mL) at -45 °C under N2. After 45 min, the reaction was carefully quenched with MeOH (5 mL) and allowed to warm to room temperature. Saturated aqueous NH4Cl (10 mL) was added and mixture filtered through Celite with EtOAc. The filtrate was poured into water (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic phase was washed with brine (10 mL), dried over Na 2 SO 4 , and concentrated. The crude material was purified on silica (Hex to 50% EtOAc/Hex) to afford IV-1f (196 mg, 59% yield) as a white solid; the structure was confirmed by 1 H NMR in CDCl 3 . To a solution of IV-1f (190 mg, 0.476 mmol, 1 eq) in a mixture of anhydrous MeOH (0.9 mL) and anhydrous Et 2 O (9 mL) was added anhydrous HCl (2M in Et 2 O, 0.48 mL, 1.90 mmol, 4 eq) dropwise at room temperature under N2. After stirring overnight, the reaction mixture was concentrated, solid washed with Et2O, and dried to afford IV-1, as an HCl salt (102 mg, 64% yield) as a white solid; the structure was confirmed by 1 H NMR and 19 F NMR in DMSO-d6. Example 41 1-(2,5-bis(methoxy-d3)-4-(methylthio)phenyl)propan-2-amine (IV-9) Synthesis of (R)-1-(2,5-bis(methoxy-d3)-4-(methylthio)phenyl)propan-2-ami ne (R)-(IV-9) was carried out according to Fig. 18. 2,5-dihydroxybenzaldehyde (IV-9a) was bis-alkylated with CD 3 I and K2CO3 in DMF forming intermediate I-9b. To a solution of IV-9b in anhydrous MeOH was added AgNO 3 and iodine at room temperature under N 2 to provide iodobenzaldehyde intermediate IV-9c. To a dried pressure vessel was added IV-9c (2.59 g, 8.67 mmol, 1 eq), Xantphos (552 mg, 0.954 mmol, 0.11 eq) and Pd2(dba)3 (794 mg, 0.867 mmol, 0.10 eq) and vessel purged with N2 before adding anhydrous PhMe (sparged with N2, 60 mL). After stirring 20 min at room temperature, NaSMe (1.22 g, 17.34 mmol, 2 eq) was added, vessel purged with N2 and sealed. After heating at 110 °C overnight, the reaction mixture was filtered through Celite with DCM and concentrated. The crude material was purified on silica (Hex to 10% EtOAc/Hex) to afford IV-9d (0.93 g, 49% yield) as a yellow solid; the structure was confirmed by 1 H NMR in CDCl 3 . To IV-9d (0.677 g, 3.10 mmol, 1 eq) was added anhydrous ammonium acetate (239 mg, 3.10 mmol, 1 eq), nitroethane (1.11 mL, 15.51 mmol, 5 eq), acetic acid (glacial, 2.5 mL, 43.40 mmol, 14 eq) and activated 3Å molecular sieves (1.5 g wet weight) under N 2 at room temperature. The reaction mixture was heated at 95 °C for 1 h. The molecular sieves were removed, reaction mixture poured into brine (30 mL) and extracted with EtOAc (3 x 30 mL). The combined organic phase was washed with water (20 mL), then brine (20 mL), dried over Na2SO4, and concentrated. The crude material was purified on silica (Hex to 20% EtOAc/Hex) to afford IV-9e (0.726 g, 85% yield) as an orange solid; the structure was confirmed by 1 H NMR in CDCl3. To a solution of IV-9e (0.99 g, 3.60 mmol, 1 eq) in MeOH (13.3 mL), water (5.3 mL), and concentrated HCl (12.1 M, 5.3 mL) was added iron (powder, reduced) (0.80 g, 14.4 mmol, 4 eq) portionwise over 10 min at room temperature under N2. The reaction was heated at 70 °C for 2 h before adding additional concentrated HCl (12.1 M, 5.3 mL) and iron (powder, reduced) (0.80 g, 14.4 mmol, 4 eq). After heating for 4 h, the reaction mixture was poured into water (30 mL) and extracted with DCM (3 x 30 mL). The combined organic phase was washed with brine (20 mL), dried over Na 2 SO 4 , and concentrated. The crude material was purified on silica (Hex to 30% EtOAc/Hex) to afford IV-9f (456 mg, 51% yield) as a white solid; the structure was confirmed by 1 H NMR in CDCl 3 . To a solution of IV-9f (279 mg, 1.13 mmol, 1 eq) and titanium(IV) ethoxide (0.48 mL, 2.27 mmol, 2 eq) in anhydrous THF (5.7 mL) was added (R)-2-methyl-2-propanesulfinamide (275 mg, 2.27 mmol, 2 eq) in anhydrous THF (2.3 mL) at room temperature under N2. The reaction was heated at 70 °C for 4 h before cooling to -45 °C and was slowly transferred into a solution of NaBH4 (171 mg, 4.53 mmol, 4 eq) in anhydrous THF (5.7 mL) at -45 °C under N2. After 45 min, the reaction was carefully quenched with MeOH (5 mL) and allowed to warm to room temperature. Saturated aqueous NH4Cl (10 mL) was added and the mixture was filtered through Celite with EtOAc. The filtrate was poured into water (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic phase was washed with brine (10 mL), dried over Na 2 SO 4 , and concentrated. The crude material was purified on silica (Hex to 50% EtOAc/Hex) to afford IV-9g (221 mg, 56% yield) as a white solid; the structure was confirmed by 1 H NMR in CDCl 3 . To a solution of IV-9g (211 mg, 0.60 mmol, 1 eq) in a mixture of anhydrous MeOH (1.1 mL) and anhydrous Et2O (11 mL) was added anhydrous HCl (2M in Et2O, 1.20 mL, 2.40 mmol, 4 eq) dropwise at room temperature under N2. After stirring overnight, the reaction mixture was concentrated, the solid was washed with Et2O, and dried to afford (R)-IV-9 as an HCl salt (162 mg, 95% yield) as a white solid; the structure was confirmed by 1 H NMR in DMSO-d6. Synthesis of (S)-1-(2,5-bis(methoxy-d3)-4-(methylthio)phenyl)propan-2-ami ne (S)-(IV-9) was carried out according to Fig. 18. To a solution of IV-9f (279 mg, 1.13 mmol, 1 eq) and titanium(IV) ethoxide (0.48 mL, 2.27 mmol, 2 eq) in anhydrous THF (5.7 mL) was added (S)-2-methyl-2- propanesulfinamide (275 mg, 2.27 mmol, 2 eq) in anhydrous THF (2.3 mL) at room temperature under N 2 . The reaction was heated at 70 °C for 4 h before cooling to -45 °C and was slowly transferred into a solution of NaBH 4 (171 mg, 4.53 mmol, 4 eq) in anhydrous THF (5.7 mL) at -45 °C under N 2 . After 45 min, the reaction was carefully quenched with MeOH (5 mL) and allowed to warm to room temperature. Saturated aqueous NH4Cl (10 mL) was added and the mixture was filtered through Celite with EtOAc. The filtrate was poured into water (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic phase was washed with brine (10 mL), dried over Na2SO4, and concentrated. The crude material was purified on silica (Hex to 50% EtOAc/Hex) to afford IV-9h (226 mg, 57% yield) as a white solid; the structure was confirmed by 1 H NMR in CDCl3. To a solution of IV-9h (216 mg, 0.61 mmol, 1 eq) in a mixture of anhydrous MeOH (1.1 mL) and anhydrous Et2O (11 mL) was added anhydrous HCl (2M in Et2O, 1.23 mL, 2.46 mmol, 4 eq) dropwise at room temperature under N 2 . After stirring overnight, the reaction mixture was concentrated, the solid was washed with Et 2 O, and dried to afford (S)-IV-9 as an HCl salt (169 mg, 97% yield) as a white solid; the structure was confirmed by 1 H NMR in DMSO-d 6 . Example 42 1-(2,4,5-trimethoxyphenyl)propan-2-d-2-amine (IV-36) Synthesis of 1-(2,4,5-trimethoxyphenyl)propan-2-d-2-amine (IV-36) is carried out according to Fig. 19, using a modified general procedure as reported by Shulgin (Shulgin, A., and Shulgin, Ann. (1991) Pihkal: a chemical love story, Transform Press, Berkeley, CA) and later modified by Maresh (Maresh, J. J., Ralko, A. A., Speltz, T. E., Burke, J. L., Murphy, C. M., Gaskell, Z., Girel, J. K., Terranova, E., Richtscheidt, C., and Krzeszowiec, M. (2014) Chemoselective Zinc/HCl Reduction of Halogenated beta-Nitrostyrenes: Synthesis of Halogenated Dopamine Analogues, Synlett 25, 2891- 2894). The starting material 2,4,5-trimethoxybenzaldehyde (IV-36a) undergoes a nitroaldol condensation with nitroethane and buffered acetic acid followed by a selective alkene reduction of intermediate IV-36b with sodium borohydride and silicone dioxide to selectively reduce the alkene to intermediate IV-36c (Sinhababu, A. K., and Borchardt, R. T., 1983, Silica Gel-Assisted Reduction of Nitrostyrenes to 2-Aryl-1-Nitroalkanes with Sodium-Borohydride, Tetrahedron Letters 24, 227-230). 9O^]O[S^V OaMRKWQO K] ]RO j'YX\S]SXW S\ MK[[SON X^] ^\SWQ ]RO LK\SM [O\SW I6-* KWN NO^]O[K]ON `K]O[ developed by Yamada (Yamada, T., Kuwata, M., Takakura, R., Monguchi, Y., Sajiki, H., and Sawama, Y. (2018) Organocatalytic Nitroaldol Reaction Associated with Deuterium-Labeling, Adv Synth Catal 360, 637-641) to form intermediate IV-36d. Reduction of the nitro group with zinc dust in methanol containing hydrochloric acid yields the final product (IV-36), as an HCl salt. The structure of the product will be confirmed by 1 H NMR. Example 43 General procedures for resolution of phenylpropan-2-amine (e.g., amphetamine) enantiomers. If desired, phenylpropan-2-amine enantiomers may be resolved with chiral chromatography, by crystallization as the tartrate salt, or via the fractional crystallization procedure as outlined by Aldous (Aldous, F. A., Barrass, B. C., Brewster, K., Buxton, D. A., Green, D. M., Pinder, R. M., Rich, P., Skeels, M., and Tutt, K. J., 1974, Structure-activity relationships in psychotomimetic phenylalkylamines, J Med Chem 17, 1100-1111), see Fig. 20. In this scheme, phenylisopropylamines undergo a reaction with N-benzyloxycarbonyl-L-(or D-) phenylalanine p-nitrophenylester and the resulting diastereomeric amides are resolved by precipitation of the insoluble fraction. Recovery of the desired phenylisopropylamine is carried out by catalytic hydrogenation followed by the Edman degradation. Reference Compound 1 3,4,5-trimethoxyphenethylamine (Reference Compound 1)(mescaline). Synthesis of mescaline (Reference Compound 1) was carried out according to Fig.21, using a modified general procedure as reported by Shulgin (Shulgin, A., and Shulgin, Ann. (1991) Pihkal: a chemical love story, Transform Press, Berkeley, CA) and later modified by Maresh (Maresh, J. J., Ralko, A. A., Speltz, T. E., Burke, J. L., Murphy, C. M., Gaskell, Z., Girel, J. K., Terranova, E., Richtscheidt, C., and Krzeszowiec, M. (2014) Chemoselective Zinc/HCl Reduction of Halogenated beta- Nitrostyrenes: Synthesis of Halogenated Dopamine Analogues, Synlett 25, 2891-2894). The starting material 3,4,5-trimethoxybenzaldehyde A underwent a nitroaldol condensation with nitromethane and buffered acetic acid, followed by bis-reduction of the nitro group and alkene in intermediate B with zinc dust in methanol containing hydrochloric acid, to yield the final product (Reference Compound 1), as an HCl salt. The structure of the product was confirmed by 1 H NMR; total yield, 90%. Reference Compound 2

Synthesis of l-(2,5-dimethoxy-4-(methylthio)phenyl)propan-2-amine (Reference Compound 2) (“DOT”).

Enantioselective synthesis of l-(2,5-dimethoxy-4-(methylthio)phenyl)propan-2-amine (Reference Compound 2) was carried out according to Fig. 22. A Pd2(dba)3/xanphos-catalyzed cross coupling between iodobenzaldehyde starting material C and sodium methanethiolate provided intermediate D, which then underwent a nitroaldol condensation with nitroethane in buffered acidic conditions to form nitro intermediate E. Subsequent reduction with iron and hydrochloric acid formed an oxime (not shown), which was hydrolyzed during work-up to yield methyl benzyl ketone intermediate F using procedures reported by Pearl (Pearl, I.A., Beyer, D.L. J. Org. Chem. 1951, 16, 2, 221-224). Methyl benzyl ketone intermediate F was next condensed with either enantiomer of Ellman’s sulfinamide (t-butanesulfinamide) to form intermediate G, which was then selectively reduced with sodium borohydride to either the (R,R) or (S,S) diastereomer of H, and hydrolyzed with HC1 in methanol using the procedures described in Cinelli (Cinelli, M.A. et al. J. Med. Chem. 2017, 60, 9, 3958-3979) to yield the final product (Reference Compound 2), as an HC1 salt, in either enantiomeric form, i.e., (R)-

1-(2,5-dimethoxy-4-(methylthio)phenyl)propan-2-amine or (S)-l-(2,5-dimethoxy-4-

(methylthio)phenyl)propan-2-amine. The structure of the product(s) was confirmed by NMR.

Reference Compound 3

2-(2,5-dimethoxy-4-(methylthio)phenyl)ethan-l-amine (Reference Compound 3) (“2C-T”), HC1 salt, is commercially available and was purchased from Cayman Chemical Co.

Reference Compound 4 2-(2,5-dimethoxy-4-(trifluoromethyl)phenyl)ethan-l-amine (Reference Compound 4) (“2C-TFM”), HC1 salt, is commercially available and was purchased from Cayman Chemical Co.

Reference Compound 5 3,4-Methylenedioxyphenethylamine (Reference Compound 5) (“MDPEA”), HC1 salt, is commercially available and was purchased from Oakwood Products Inc.

II. Formulations

Preparation of Ion-Exchange Resin Complex

Freebase of the compounds are complexed with a strong cation-exchange resin (sodium form, Amberlite IRP69, Rohm & Haas, Sodium Polystyrene Sulfonate, pharmaceutical grade USP, particle size 75-150 microns). The maximum load of the above resin is known to be about 5 meqv/g. In a typical procedure, the compound free base (10 mmol) is dissolved in 20 ml of ethanol. To this solution, 2 g of the IRP69 resin (washed with 3x50 ml of ethanol) is added at room temperature using a magnetic stirrer and kept stirring for 2 h. The resin is then filtered and washed with ethanol (2x20 ml). Compound release from the resin complex is studied using a Type I (basket) dissolution apparatus at pH 1 (0.1 M HC1) and 7.4 (0.1 M phosphate buffer). In an acidic environment, the release is a fast process, with >90% of the drug leaching out within 30 min. At neutral pH, the release is substantially slower, with about 50% of the drug released in 1 h and 80% at 2 h. The drug concentration is determined by HPLC using an Agilux 1100 setup and UV detection.

Preparation of Beads of Ion Exchange Resin Complex Coated by Enteric Coating

Seal Coating

Compound-ion-exchange resin Complex beads are seal-coated at a 2% weight gain using Opadry 03K19229 coating (Colorcon, NJ, USA, reconstituted at 6% solids) in a hydro-alcoholic solvent system (88:12, isopropanol: water) on a Niro-Aeromatic STREA 1 fluidized bed machine equipped with a Wurster coating module (bottom feed).

Enteric Coating

The resulting beads are then coated using Opadry Enteric 940 white coating (Colorcon, NJ, USA). Coating dispersions are reconstituted at 10% solids in a hydro-alcoholic solvent system (88:12, isopropanol: water) and applied to either a 5 or 12% weight gain. Enteric coating of placebo tablets is carried out without the preceding seal coating step. Samples are drawn at 5, 6, 7, 8, 10 and 12% weight gains.

Drug Release Testing

Drug release at low pH is determined using a Type I apparatus 1 (basket), at 100 rpm. In the first stage, the dissolution medium is 1000 ml of 0.1 N HC1 at 37°C (±0.5°C) and the bead load 2 g. After 1 h of operation in this medium, an aliquot is withdrawn and the drug content is determined by HPLC to be less than 1% total, confirming the integrity of the applied enteric coating. Drug release at neutral pH is determined using a Type I apparatus 1 (basket), at 100 rpm. In the second stage, the dissolution medium is 1000 mL of 0.1 M phosphate buffer at pH 7.4 at 37°C (±0.5°C) and a 2 g bead load. The medium aliquots are withdrawn at 15, 30, 60, 90, and 120 min, and the drug content is determined by HPLC using an Agilux 1100 setup and UV detection. The release of the drug is found to be ca. 50 % at 1 h and 80 % at 2 h, with a release profile over time similar to the uncoated resin beads.

Manufacturing of Orally Disintegrating Tablets

Orally disintegrated tablets are designed by incorporating micro-beads of the drug-ion exchange resin complex with extended release characteristics into a matrix of the fast orally disintegrating components that help dispersing active material in the oral cavity and facilitate subsequent swallowing without use of water for more convenient administration of the drug.

Compounds are formulated into an orally disintegrating release tablet form, composition PI- ODT-1 by dry granulation using sugar based, fast-disintegrating matrix Pharmaburst 500 (SPI, PA, USA). 100 g of Pharmaburst 500 is mixed up with 20 g of the enterically coated compound-ion- exchange resin complex beads and sieved via 40 mesh sieves, to break agglomerates, and then the mixture is blended in a 400 ml tube blender for 15 minutes at 200 rev/min. After blending, magnesium stearate (200 mg) is added and blended for additional 3 minutes. The 250 mg convex-shaped tablets containing about 20 mg of active compound are compressed using a TDP tablet press and 9 mm dye. By applying a compression force of 8 kN, tablets of the hardness in the range 10-15 kP are generated. The tablet disintegration time is determined to be 60-75 seconds. The tablet dissolution is carried out in a Type II dissolution apparatus (paddle) (Distek Premiere 5100 Dissolution System, Distek Inc., North Brunswick, USA) at 100 rpm, 37°C, using lx PBS buffer, pH=6.8 as an immersion media. At predetermined time intervals, 1 ml samples are withdrawn (not replaced), filtered and assayed. The amount of compound released is measured by HPLC using an Agilent 1100 setup (Nagy, J., and Veress, T., 2016, HPLC Analysis of Hallucinogenic Mushroom Alkaloids (Psilocin and Psilocybin) Applying Hydrophilic Interaction Chromatography (HILIC), J Forensic Res 7, 356). Solutions of known concentrations of compound are used to calculate the amount of drug released.

Fatty Acid Salts of Compounds

Compound free bases (10 mmol) are dissolved in 30 ml of acetone and 10 mmol of decanoic acid is added and mixed for 5 min. A white crystalline precipitate of the 1:1 salt is formed upon cooling the mixture in a refrigerator overnight. The salt composition will be confirmed by elemental analysis.

III. Testing

Radioligand Competition Binding

5-HT2 Receptor Binding Assays. Serotonin receptor (5-HTR) affinities were determined by radioligand competition binding as previously described (see Canal, C. E., Cordova-Sintjago, T., Liu, Y., Kim, M. S., Morgan, D., and Booth, R. G., 2013, Molecular pharmacology and ligand docking studies reveal a single amino acid difference between mouse and human serotonin 5-HT2A receptors that impacts behavioral translation of novel 4-phenyl-2-dimethylaminotetralin ligands, J Pharmacol Exp Ther 347, 705-716; Armstrong, J. L., Casey, A. B., Saraf, T. S., Mukherjee, M., Booth, R. G., and Canal, C. E., 2020, (S)-5-(2'-Fluorophenyl)-N,N-dimethyl-l,2,3,4-tetrahydronapht halen-2-amine, a Serotonin Receptor Modulator, Possesses Anticonvulsant, Prosocial, and Anxiolytic-like Properties in an Fmrl Knockout Mouse Model of Fragile X Syndrome and Autism Spectrum Disorder, ACS Pharmacol Transl Sci 3, 509-523; Saraf, T. S., Felsing, D. E., Armstrong, J. L., Booth, R. G., and Canal, C. E., 2021, Evaluation of lorcaserin as an anticonvulsant in juvenile Fmrl knockout mice, Epilepsy Res 175, 106677), with minor modifications.

Briefly, cDNA plasmids encoding human 5-HTRs were procured from the cDNA Resource Center. Human embryonic kidney cells (HEK293, ATCC CRL-1573) were grown in a cell incubator in 100 mm dishes with antibiotic-free Dulbecco’s modified Eagle’s medium containing 10% fetal bovine serum. Cells were transfected at -85% confluency with 5-15 μg cDNA using one of various transfection reagents (e.g., TransIT-2020 reagent, Mirus Bio, Madison, Wisconsin). After approximately 48 hours, cell membrane was collected via homogenization in 50 mM Tris buffer and centrifugation steps. For all experiments, serotonin (5-HT) hydrochloride was used as a positive control, and an appropriate high affinity, cold ligand (e.g., mianserin hydrochloride, 10 pM, for 5-HT2RS) was used to define nonspecific binding. Each cell membrane homogenate expressing a 5-HTR was incubated at room temperature in 96-well plates with [ 3 H]Lysergic acid diethylamide ([ 3 H]LSD) for labeling 5-HT2RS, in the absence or presence of test articles (e.g., at 10 concentrations from 0.3 nM to 10 pM in half-log units) in a buffer for 90 min. Following equilibration, each sample was filtered rapidly under vacuum through fiberglass filters presoaked in a buffer and washed by vacuum several times with an ice-cold buffer. Filters were soaked with scintillation fluid and counts per minute were detected using photodetectors (Microbeta Scintillation Counter). The Kd for [ 3 H]LSD was set to 0.78 and the IC50 values were computed using nonlinear, least squares regression analyses and then converted to binding affinity (Ki) values using the Cheng-Prusoff equation (GraphPad Prism 9.0, San Diego, California). Data shown are the results from all experiments combined. As is known, G protein-coupled receptors (GPCR), including the 5-HT2AR, can exist in multiple conformations, and ligands, typically agonist ligands, have unique affinities for these conformations of the receptor (Kenakin, T., 2017, Theoretical Aspects of GPCR-Ligand Complex Pharmacology, Chem Rev 117, 1, 4-20). The competition binding data from one of the tested compounds fit best to a two-site fit K model. The data point for -10 samples is total specific binding (no compound present), and -4 was interpolated to complete the binding curves to 0 specific binding. In another series of 5-HT2A, 2B, and 2c radioligand binding assays, cell membrane homogenates derived from HEK-293 or CHO-K1 cells expressing the human wild-type 5-HT2RS receptors were incubated with [ 125 I](±)DOI at either 0.1 or 0.2 nM for 60 min at room temperature or 37 °C in the presence or absence of 10 pM of the test article (see Table 4; Mulheron, J.G., Casanas, S.J., Arthur, J.M., Garnovskaya, M.N., Gettys, T.W. and Raymond, J.R., 1994, Human 5-HT1A receptor expressed in insect cells activates endogenous Go-like G protein, J Biol Chem 269, 12954; Bryant, H.U., Nelson, D.L., Button, D., Cole, H.W., Baez, M.B., Lucaites, V.L., Wainscott, D.B., Whitesitt, C., Reel, J., Simon, R. and Koppel, G.A., 1996, A novel class of 5-HT2 Areceptor antagonist: aryl aminoguanidines, Life Sci 15, 1259; Choi, D.S., Birraux, G., Launay, J.M. and Maroteaux, L., 1994, The human serotonin 5-HT2B receptor: pharmacological link between 5-HT2 and 5-HT1D receptors, FEBS Lett 352, 393.) Non-specific binding was determined in the presence of 10 p M (±)DOI. Samples were then analyzed as described above.

Monoamine Transporter Binding Assays. Human norepinephrine transporter, human dopamine transporter, and human 5-HT transporter binding assays were performed as summarized in Table 4 and described in Pacholczyk, T. et al. (1991), Nature, 350: 350-354; Pristupa, Z.B. et al. (1994), Mol. Pharmacol., 45: 125-135; 566. Tatsumi, M. et al. (1999), Eur. J. Pharmacol., 368: 277-283. The reference compound results for binding assays with 5-HTRs and monoamine transporters are presented in Table 5.

Table 4. Methods In Vitro Pharmacology: Binding Assays

Table 5. 5-HT2 and Monoamine Transporter Binding Assays: Reference Compound Results

Serotonin (5-HT2) Receptor Functional Assays

5-HT2A ,2B,2C receptor-mediated Gq activation and phosphoinositide hydrolysis leading to the production of inositol phosphate 1 (IPi, IPOne, Cisbio assay), canonical signaling pathways, was determined as previously described by measuring IPI production using Fluorescence Resonance Energy Transfer (FRET) technology (Porter, R.H.P., Benwell, K.R., Lamb, H., Malcolm, C.S., Allen, N.F., Revell, D.F., Adams, D.R. and Sheardown, M.J., 1999, Functional characterization of agonists at recombinant human 5-HT2a, 5HT2b and 5-HT2c receptors in CHO- KI cells, Brit J Pharmacol 128, 13; Olsen, R.H.J., DiBerto, J.F., English, J.G. et al.). Briefly, for 5-HT2A.2B, 2c Rs, CHO or HEK-293 cells were incubated with test articles and 5-HT (positive control) in stimulation buffer. After incubation, cells were lysed and fluorescence acceptor and donor were added. FRET was measured after 30 min at room temperature using a microplate reader (Envision, Perkin Elmer). The results are expressed as a percent of control response to 10 p M 5- HT and data were fit to non-linear curves to calculate potencies (e.g., EC50) and efficacies (e.g., Emax), relative to positive controls (e.g., 5-HT). Details of the methods for 5-HT2 functional assays and reference compound results are presented in Table 6 and 7, respectively. Table 6. Methods In Vitro Pharmacology: 5-HT2 Functional Assays

Table 7. 5-HT2 Functional Assays: Reference Compound Results Monoamine transporter uptake assays

Human norepinephrine transporter (NET), human dopamine transporter (DAT) and rat serotonin transporter (SERT) uptake assays were performed as outlined in Table 8 and described in Perovic S., Muller W.E., “Pharmacological profile of hypericum extract. Effect on serotonin uptake by postsynaptic receptors” Arzneimittelforschung, 1995, 45 (11), 1145-8 and Verrico C. et al. (2005), “MDMA (Ecstasy) and human dopamine, norepinephrine, and serotonin transporters: implications for MDMA-induced neurotoxicity and treatment” Psychopharmacology (Berl), 1-15, in press. Reference compound results are presented in Table 9.

Table 8. Methods In Vitro Pharmacology: Monoamine Transporter Uptake Assays

Table 9. Monoamine Transporter Update Inhibition Assays: Reference Compound Results In Vitro Metabolism and Kinetic Deuterium Isotope Effects

Rat Liver Microsomes (RLM). Male Sprague-Dawley rat liver microsomes were purchased from XenoTech. The reaction mixture, minus NADPH, was prepared as described in Table 10. The test compound was added into the reaction mixture at a final concentration of 1 pM. The control compound, testosterone, was run simultaneously with the test compound in a separate reaction. The reaction mixture (without cofactor) was equilibrated in a shaking water bath at 37°C for 5 minutes. The reaction was initiated by the addition of the cofactor, and the mixture was incubated in a shaking water bath at 37°C. Aliquots (150 μL) were withdrawn at 0, 5, 10, 20, 30, 60, and for some assays at 120 minutes. Test compound and testosterone samples were immediately combined with 150 μL of ice-cold acetonitrile (ACN) containing 0.1% formic acid and internal standard to terminate the reaction. The samples were then mixed and centrifuged to precipitate proteins. All samples were assayed by LC-MS/MS using electrospray ionization. The peak area response ratio (PARR) of analyte to internal standard at each time point was compared to the PARR at time 0 to determine the percent remaining at each time point. Half-lives were calculated using GraphPad software, fitting to a single -phase exponential decay equation.

Table 10. Reaction mixture composition

In a different series of reactions using RLM, a 10 mM stock solution of the test compound was prepared in DMSO. From this stock solution, an intermediate stock solution of 1 mM was prepared by diluting the compound in DMSO and from this intermediate stock solution, a working solution of 0.30 mM was prepared by diluting the compound in ACN:Water (50:50) as a cocktail of two compounds (i.e., 30 μL the non-deuterated and the deuterated compound in 40 μL diluent in 100 μL total volume). Following this, a master mix of rat liver microsomes (final concentration 0.5 mg/ml, Xenotech Cat # R1000; Lot# 1610290) and 0.1 M potassium phosphate buffer was prepared and pre-incubated for 5 min at 37 °C. The test compound mixture was then added to achieve a final test article concentration of 1 pM (for each analog if co-dosed; 0.2% DMSO); verapamil was used as a positive control. Subsequently, 10 mM NADPH prepared in 0.1 M potassium phosphate buffer was added (final concentration 1 mM) to initiate the reaction and samples were incubated at 37°C for the desired time points (0, 5, 15 , 30, 45, 60 min). At each time point, 40 μL of the samples were withdrawn and reactions were stopped using 200 μL chilled ACN containing 0.1 % formic acid and internal standard carbamazepine (10 ng/mL). The samples were centrifuged, and the supernatant was analyzed by LC-MS/MS. The percent compound remaining at each time point was calculated with respect to that of the 0 min sample. The data were then analyzed to calculate half-life. Blank samples were prepared using DMSO (without the test compound). Recombinant human MAO-A. hrMAO-A and MAO control were purchased from XenoTech. The reaction mixture was prepared as described in Table 11. The test compound was added into the reaction mixture at a final concentration of 1 pM. The positive control kynuramine (25 pM) was run simultaneously with the test compound in a separate reaction. The reaction mixture (without test compounds or kynuramine) was equilibrated in a shaking water bath at 37 °C for 5 minutes. The reaction was initiated by the addition of the test compound or kynuramine, and the mixture was incubated in a shaking water bath at 37°C. Aliquots (150 μL) of the test compound reaction mixture were withdrawn at 0, 5, 10, 20, 30,60, and for some assays at 120 minutes. Aliquots (150 μL) of the positive control reaction mixture were withdrawn at 0 and 30 minutes. Test compound and kynuramine samples were immediately combined with 150μL of ice-cold 100% acetonitrile containing 0.1% formic acid and internal standard (0.2 pM metoprolol) to terminate the reaction. The samples were then mixed and centrifuged to precipitate proteins. All samples were assayed by LC-MS/MS. The peak area response ratio (PARR) of analyte to internal standard at each time point was compared to the PARR at time 0 to determine the percent remaining at each time point. Half-lives were calculated using GraphPad software, fitting to a single -phase exponential decay equation.

Table 11. Reaction mixture composition

In a different series of reactions using human MAO-A and MAO-B, a 10 mM stock solution of the test compound was prepared in DMSO. From this stock solution, an intermediate stock solution of 1 mM was prepared by diluting the compound in DMSO and from this intermediate stock solution, a working solution of 0.30 mM was prepared by diluting the compound in ACN:Water (50:50) as cocktail of 2 compounds (30 μL each compound in 40 μL diluent for lOOμL). Following this, a master mix of 0.1 M potassium phosphate buffer and test compound mixture i.e., the non-deuterated and the deuterated analog, were prepared to achieve final drug concentration of 1 pM (0.2% DMSO) for the required number of reactions. The aforementioned master mix was pre-incubated at 37°C for 5 min. To this master mix, MAO-A (final protein cone. 0.1 mg/mL; Corning Cat # 456283; Lot# 1173003) or MAO-B (final protein cone. 0.1 mg/mL; Corning Cat # 456284; Lot# 0318001) was added to initiate the reaction. The samples were then incubated at 37°C for the desired time points. At each time point (0, 5, 15 , 30, 45, 60 ), 40 μL of the samples were withdrawn and reactions were stopped using 200 p L chilled ACN containing 0.1 % formic acid and internal standard Carbamazepine (10 ng/mL). The samples were centrifuged, and the supernatant was analyzed by LC-MS/MS. The percent compound remaining at each time point was calculated with respect to that of the 0 min sample. The data were then analyzed to calculate half-life and intrinsic clearance (CLint). Blank samples were prepared using DMSO (without the test compound).

Metabolite identification in rat and human hepatocytes

To evaluate metabolites in rat and human hepatocytes, a 10 mM stock solution of the test compound was prepared in DSMO. From this stock solution, 0.2 mM of working stock was prepared in incubation buffer. Incubation buffer was prepared as per manufacturer's recommendations using Williams E (Cat#A1217601), dexamethasone (Cat#A13449) and cell maintenance supplement b kit (Cat# A 13448). Cryopreserved rat or human hepatocytes vials were taken out from the liquid nitrogen vapor phase and thawed using hepatocyte thaw media. Working stock of each test compound was added to freshly thawed rat or human hepatocyte ( lx 10 6 cells/mL suspension in incubation media) and incubated at 37 °C at400 rpm in a thermo plate shaker. The final concentration of compound(s) was 3 pM and hepatocyte density 0.5 x 10 6 cells/mL. At each time point (0, 15, 30, 45 and 60 min), samples were withdrawn, and reactions were stopped using chilled 100% ACN containing 0.1% formic acid, samples were centrifuged at 10,000 RPM for 10 mins and the supernatants were analyzed by LC-MS/MS.

Data section.

In vitro metabolism of MDMA/MDA analogs

Compounds 11-11, 11-14, 11-15, 11-16, and 11-17 are MDMA/MDA analogs and were subjected to in vitro metabolism assays using rat liver microsomes (RLM), hrMAO-A and hrMAO-B, and the results are presented in Table 12. In RLM, compounds 11-16 and 11-17 were rapidly metabolized with a ti/2 of ~12 minutes, whereas the disappearance of Reference Compound 5 and compounds 11-11, 11-14, and 11-16 appeared to be NADPH-independent. Compounds 11-11, 11-14, 11-15, and Reference Compound 5 were substrates for MAO-A metabolism with half-lives of about 8 minutes or less, whereas the alpha-methyl analogs 11-16 and 11-17 appeared to be stable in the presence of MAO-A. Last, the same pattern and trends were observed for these compounds in MAO-B assays. Table 12. In vitro metabolism

Metabolite identification with compounds 11-16 and 11-17

Compound 11-16 and its deuterated analog 11-17 were subjected to metabolite identification in rat and human hepatocytes with samples taken after 0, 15, 30, 45, and 60 min of incubation, and the results are presented in Tables 13-16. Two metabolites were identified for the non-deuterated analog 11-16, Ml and M2, neither of which have been conclusively confirmed. For Ml, the metabolite could either be the loss of H2O (dehydration) or the loss of NH3 whereas for M2, it could be the addition of CH3 (methylation) or the loss of two hydrogens with the concomitant gain of an oxygen (oxidation). In contrast, for 11-17, the deuterated analog of 11-16, only one metabolite was identified in both human and rat metabolites, namely Ml, which again could be predicted to be MI-H2O or MI-NH3.

Table 13. Fragmentation Pattern for Major Metabolites of Compound 11-16 in Rat Hepatocytes

Table 14. Fragmentation Pattern for Major Metabolites of Compound 11-17 in Rat Hepatocytes Table 15. Fragmentation Pattern for Major Metabolites of Compound 11-16 in Human Hepatocytes

Table 16. Fragmentation Pattern for Major Metabolites of Compound 11-17 in Human Hepatocytes Radioligand competition binding and in vitro metabolism of compound III-5

Compound III-5 is a 3,4,5-substituted phenethylamine, and thus belongs to the same class as Reference Compound 1 (“mescaline”). Radioligand competition binding at 5-HT2A receptors was performed as previously described (Saraf, T. S., Felsing, D. E., Armstrong, J. L., Booth, R. G., and Canal, C. E., 2021, Evaluation of lorcaserin as an anticonvulsant in juvenile Fmrl knockout mice, Epilepsy Res 175, 106677), with minor modifications. The competition binding data from compound III-5 fit best to a two-site fit K model. The affinity of this compound is thus reported herein for both the “high- affinity active state” and “low-affinity inactive state” of the receptor. As can be seen in Table 17, compound III-5 shows superior affinity at 5-HT2A receptors compared to Reference Compound 1. Additionally, the technological modifications performed rendered compound III-5 with affinity equal to or better than classic 2,4,5-substituted 5-HT2A agonists, such as Reference Compounds 2-4. Furthermore, compound III-5 appears to be stable in the presence of rat liver microsome with a half-life of longer than 120 min in contrast to the reference compounds with ti/2 of less than 17 min. The competition binding curve for compound III-5 is represented graphically in Fig. 23.

Table 17. a agonist radioligand: [ 3 H]LSD b 5HT2A Ki, nM, at the high-affinity active state c 5HT2A K, nM, at the low-affinity inactive state d Literature value reported in Rickli A, et al. Receptor interaction profiles of novel psychoactive tryptamines compared with classic hallucinogens. Eur Neuropsychopharmacol. 2016 Aug;26(8): 1327-37

Radioligand competition binding and in vitro metabolism of DOx compounds

In the DOx (4-substituted-2,5-dimethoxyamphetamines) series of analogs, compounds IV-1 and both enantiomers of compound IV -9 were subjected to radioligand competition binding assays and in vitro metabolism. As can be seen in Table 18, compound IV-1 (racemate) shows affinity at 5-HT2A receptors equal or superior to both enantiomers of Reference Compound 2. The competition binding curves for compound IV-1, both enantiomers of compound IV-9, and both enantiomers of Reference Compound 2 are represented graphically in Figs. 24-26, respectively. Additionally, the technological modifications performed rendered compound IV-1 with greater stability in RLMs than Reference Compound 2.

The deuterated analogs of the two enantiomers of Reference Compound 2, the R- and S- enantiomer of compound IV-9, showed a slight decrease in affinity at the human 5-HT2A receptor. Of note, the fold differences in affinities between enantiomers stayed consistent for the nondeuterated and the deuterated analogs with a 2.2-2.4-fold higher potency of the R enantiomer than the S enantiomer. Regarding in vitro metabolism, the half-lives for the two deuterated enantiomers did not change in RLMs or MAO-A assays compared to the non-deuterated enantiomers of Reference Compound 2.

Table 18.

Activity of compounds 11-14 and 11-16 at human 5-HT2A,2B,2C receptors

Compounds 11-14 and 11-16 are analogs of known entactogens (e.g., MDA). As can be seen in Table 19 and detailed below, compounds 11-14 and 11-16 have superior affinity and functional potency at 5-HT2 receptors compared to MDA and maintain activity at the serotonin transporter (SERT). The pharmacology data — affinity and functional pharmacology at human monoamine transporters and serotonin 5-HT2A, 2B, and 2c receptors — show that compound 11-14 and its a-methyl analog 11-16 have similar pharmacodynamic properties as the entactogens MDMA and MDA, with a few notable exceptions, one being that compounds 11-14 and 11-16 are efficacious 5-HT2 receptor agonists, suggesting they may possess classic psychedelic effects in addition to entactogenic effects. Compounds 11-14 and 11-16 were tested at four concentrations 1, 3, 10, and 100 pM for their efficacy to stimulate each of the serotonin 5-HT2 receptor subtypes, 5- HT 2 A, 5-HT2B, and 5-HT2C- EMAX values are relative to 5-HT. Each compound was a nearly full or full agonist at each of the 5-HT2 receptor subtypes.

Activity of compounds 11-14 and 11-16 at human monoamine transporters

Affinity Screen at 10 /iM: Compounds 11-14 and 11-16 were tested at a single concentration of 10 p M for their affinities at each of the monoamine transporters, i.e., the dopamine (DA) transporter (DAT), the norepinephrine (NE) transporter (NET), and the serotonin transporter (SERT). Both compounds showed substantial selectivity for binding SERT over DAT and NET (see Table 19).

Function Screen at 1, 3 (SERT only), 10, 100 /iM: Compounds 11-14 and 11-16 were tested at three concentrations 1, 10, and 100 pM for their efficacy at inhibiting the uptake of NE through NET, and DA through DAT. They were also tested at four concentrations 1, 3, 10, and 100 pM for their efficacy at inhibiting the uptake of 5-HT through SERT. As shown in Table 19, compounds 11-14 and 11-16 showed full efficacy for blocking monoamine uptake through each transporter. However, both showed selectivity (higher potency) for blocking 5-HT reuptake relative to NE and DA at their respective transporters.

Compounds 11-14 and 11-16 have similar, albeit some unique pharmacological properties, relative to the classic entactogens MDMA and MDA; they are full agonists at the 5 -HT2 receptors and have selectivity (~5-fold) and appreciable potency for inhibiting SERT, relative to DAT and NET, distinguishing them from MDMA and MDA (see Oeri, H. E., Beyond ecstasy: Alternative entactogens to 3,4-methylenedioxymethamphetamine with potential applications in psychotherapy. J Psychopharmacol 2021, 35 (5), 512-536).

Compound 11-16, the a-methyl amphetamine analog of 11-14, was generally more potent than 11-14 at all targets tested. There was a disconnect in the potencies of compounds 11-14 and 11-16 to bind the monoamine transporters compared to their potencies to inhibit monoamine uptake through them. Specifically, 11-14 and II- 16 were more potent at inhibiting the transporters than binding to them; this suggests they bind uniquely to the transporters relative to the selective antagonist (inhibitor) radioligands with which they were “competing” for access to the transporters. Collectively, compounds 11-14 and 11-16 have pharmacological properties indicative of entactogen and classic psychedelic effects, distinguishing them from the classic entactogens, MDMA and MDA. Compound binding was calculated as a % inhibition of the binding of a radioactively labeled ligand specific for each target. Cellular agonist effect was calculated as a % of control response to a known reference agonist for each target and cellular antagonist effect was calculated as a % inhibition of control reference agonist response for each target. Results showing an inhibition or stimulation higher than 50% were considered to represent significant effects of the test compounds. Such effects were observed here and are listed in Table 19.

Table 19. Activity of compounds 11-14 and 11-16 at human 5-HT2 receptors and monoamine transporters. MDMA and MPA data are from the literature 3, b c Table 19 (Continued). a Oeri, H. E., Beyond ecstasy: Alternative entactogens to 3,4-methylenedioxymethamphetamine with potential applications in psychotherapy. J Psychopharmacol 2021, 35 (5), 512-536. b Dunlap, L. E.; Andrews, A. M.; Olson, D. E., Dark Classics in Chemical Neuroscience: 3,4- Methylenedioxymethamphetamine. ACS Chemical Neuroscience 2018, 9 (10), 2408-2427. c Nash, J. F.; Roth, B. L.; Brodkin, J. D.; Nichols, D. E.; Gudelsky, G. A., Effect of the R(-) and S(+) isomers of MDA and MDMA on phosphatidyl inositol turnover in cultured cells expressing 5-HT2A or 5-HT2C receptors. Neurosci Lett 1994, 177 (1-2), 111-5.

ND = no data

* Numbers represent racemic MDMA unless otherwise noted

** Numbers represent racemic MDA unless otherwise noted

All patents, patent applications, and other scientific or technical writings referred to anywhere herein are incorporated by reference herein in their entirety. The embodiments illustratively described herein suitably can be practiced in the absence of any element or elements, limitation or limitations that are specifically or not specifically disclosed herein. Thus, for example, in each instance herein any of the terms "comprising," "consisting essentially of," and "consisting of" can be replaced with either of the other two terms, while retaining their ordinary meanings. The terms and expressions which have been employed are used as terms of description and not of limitation, and there is no intention that in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the claims. Thus, it should be understood that although the present methods and compositions have been specifically disclosed by embodiments and optional features, modifications and variations of the concepts herein disclosed can be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of the compositions and methods as defined by the description and the appended claims.

Any single term, single element, single phrase, group of terms, group of phrases, or group of elements described herein can each be specifically excluded from the claims.

Whenever a range is given in the specification, for example, a temperature range, a time range, a composition, or concentration range, all intermediate ranges and subranges, as well as all individual values included in the ranges given are intended to be included in the disclosure. It will be understood that any subranges or individual values in a range or subrange that are included in the description herein can be excluded from the aspects herein. It will be understood that any elements or steps that are included in the description herein can be excluded from the claimed compositions or methods.

In addition, where features or aspects of the compositions and methods are described in terms of Markush groups or other grouping of alternatives, those skilled in the art will recognize that the compositions and methods are also thereby described in terms of any individual member or subgroup of members of the Markush group or other group.

Accordingly, the preceding merely illustrates the principles of the methods and compositions. It will be appreciated that those skilled in the art will be able to devise various arrangements which, although not explicitly described or shown herein, embody the principles of the disclosure and are included within its spirit and scope. Furthermore, all examples and conditional language recited herein are principally intended to aid the reader in understanding the principles of the disclosure and the concepts contributed by the inventors to furthering the art, and are to be construed as being without limitation to such specifically recited examples and conditions. Moreover, all statements herein reciting principles, aspects, and embodiments of the disclosure as well as specific examples thereof, are intended to encompass both structural and functional equivalents thereof. Additionally, it is intended that such equivalents include both currently known equivalents and equivalents developed in the future, i.e., any elements developed that perform the same function, regardless of structure. The scope of the present disclosure, therefore, is not intended to be limited to the exemplary embodiments shown and described herein. Rather, the scope and spirit of present disclosure is embodied by the following.